1
|
Gerges SH, El-Kadi AOS. Changes in cardiovascular arachidonic acid metabolism in experimental models of menopause and implications on postmenopausal cardiac hypertrophy. Prostaglandins Other Lipid Mediat 2024; 173:106851. [PMID: 38740361 DOI: 10.1016/j.prostaglandins.2024.106851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Menopause is a normal stage in the human female aging process characterized by the cessation of menstruation and the ovarian production of estrogen and progesterone hormones. Menopause is associated with an increased risk of several different diseases. Cardiovascular diseases are generally less common in females than in age-matched males. However, this female advantage is lost after menopause. Cardiac hypertrophy is a disease characterized by increased cardiac size that develops as a response to chronic overload or stress. Similar to other cardiovascular diseases, the risk of cardiac hypertrophy significantly increases after menopause. However, the exact underlying mechanisms are not yet fully elucidated. Several studies have shown that surgical or chemical induction of menopause in experimental animals is associated with cardiac hypertrophy, or aggravates cardiac hypertrophy induced by other stressors. Arachidonic acid (AA) released from the myocardial phospholipids is metabolized by cardiac cytochrome P450 (CYP), cyclooxygenase (COX), and lipoxygenase (LOX) enzymes to produce several eicosanoids. AA-metabolizing enzymes and their respective metabolites play an important role in the pathogenesis of cardiac hypertrophy. Menopause is associated with changes in the cardiovascular levels of CYP, COX, and LOX enzymes and the levels of their metabolites. It is possible that these changes might play a role in the increased risk of cardiac hypertrophy after menopause.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
2
|
Ricciotti E, Haines PG, Chai W, FitzGerald GA. Prostanoids in Cardiac and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:558-583. [PMID: 38269585 PMCID: PMC10922399 DOI: 10.1161/atvbaha.123.320045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Prostanoids are biologically active lipids generated from arachidonic acid by the action of the COX (cyclooxygenase) isozymes. NSAIDs, which reduce the biosynthesis of prostanoids by inhibiting COX activity, are effective anti-inflammatory, antipyretic, and analgesic drugs. However, their use is limited by cardiovascular adverse effects, including myocardial infarction, stroke, hypertension, and heart failure. While it is well established that NSAIDs increase the risk of atherothrombotic events and hypertension by suppressing vasoprotective prostanoids, less is known about the link between NSAIDs and heart failure risk. Current evidence indicates that NSAIDs may increase the risk for heart failure by promoting adverse myocardial and vascular remodeling. Indeed, prostanoids play an important role in modulating structural and functional changes occurring in the myocardium and in the vasculature in response to physiological and pathological stimuli. This review will summarize current knowledge of the role of the different prostanoids in myocardial and vascular remodeling and explore how maladaptive remodeling can be counteracted by targeting specific prostanoids.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Philip G Haines
- Rhode Island Hospital, Department of Medicine, Warren Alpert Medical School of Brown University, Providence (P.G.H.)
| | - William Chai
- Health and Human Biology, Division of Biology and Medicine, Brown University, Providence, RI (W.C.)
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Medicine (G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
3
|
Maxwell DL, Bryson TD, Taube D, Xu J, Peterson E, Harding P. Deleterious effects of cardiomyocyte-specific prostaglandin E2 EP3 receptor overexpression on cardiac function after myocardial infarction. Life Sci 2023; 313:121277. [PMID: 36521546 PMCID: PMC9805516 DOI: 10.1016/j.lfs.2022.121277] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
AIMS Prostaglandin E2 (PGE2) is a lipid hormone that signals through 4 different G-protein coupled receptor subtypes which act to regulate key physiological processes. Our laboratory has previously reported that PGE2 through its EP3 receptor reduces cardiac contractility at the level of isolated cardiomyocytes and in the isolated working heart preparation. We therefore hypothesized that cardiomyocyte specific overexpression of the PGE2 EP3 receptor further decreases cardiac function in a mouse model of heart failure produced by myocardial infarction. MAIN METHODS Our study tested this hypothesis using EP3 transgenic mice (EP3 TG), which overexpress the porcine analogue of human EP3 in the cardiomyocytes, and their wildtype (WT) littermates. Mice were analyzed 2 wks after myocardial infarction (MI) or sham operation by echocardiography, RT-PCR, immunohistochemistry, and histology. KEY FINDINGS We found that the EP3 TG sham controls had a reduced ejection fraction, reduced fractional shortening, and an increased left ventricular dimension at systole and diastole compared to the WT sham controls. Moreover, there was a further reduction in the EP3 TG mice after myocardial infarction. Additionally, single-cell analysis of cardiomyocytes isolated from EP3 TG mice showed reduced contractility under basal conditions. Overexpression of EP3 significantly increased cardiac hypertrophy, interstitial collagen fraction, macrophage, and T-cell infiltration in the sham operated group. Interestingly, after MI, there were no changes in hypertrophy but there were changes in collagen fraction, and inflammatory cell infiltration. SIGNIFICANCE Overexpression of EP3 reduces cardiac function under basal conditions and this is exacerbated after myocardial infarction.
Collapse
Affiliation(s)
- DruAnne L Maxwell
- Department of Physiology, Wayne State University School of Medicine, USA; Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA
| | - Timothy D Bryson
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA
| | - David Taube
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA
| | - Jiang Xu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA
| | - Edward Peterson
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI, USA
| | - Pamela Harding
- Department of Physiology, Wayne State University School of Medicine, USA; Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA.
| |
Collapse
|
4
|
COX-2/sEH Dual Inhibitor PTUPB Attenuates Epithelial-Mesenchymal Transformation of Alveolar Epithelial Cells via Nrf2-Mediated Inhibition of TGF- β1/Smad Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5759626. [PMID: 35509835 PMCID: PMC9060975 DOI: 10.1155/2022/5759626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/02/2022] [Accepted: 04/09/2022] [Indexed: 12/15/2022]
Abstract
Background Arachidonic acid (ARA) metabolites are involved in the pathogenesis of epithelial-mesenchymal transformation (EMT). However, the role of ARA metabolism in the progression of EMT during pulmonary fibrosis (PF) has not been fully elucidated. The purpose of this study was to investigate the role of cytochrome P450 oxidase (CYP)/soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) metabolic disorders of ARA in EMT during PF. Methods A signal intratracheal injection of bleomycin (BLM) was given to induce PF in C57BL/6 J mice. A COX-2/sEH dual inhibitor PTUPB was used to establish the function of CYPs/COX-2 dysregulation to EMT in PF mice. In vitro experiments, murine alveolar epithelial cells (MLE12) and human alveolar epithelial cells (A549) were used to explore the roles and mechanisms of PTUPB on transforming growth factor (TGF)-β1-induced EMT. Results PTUPB treatment reversed the increase of mesenchymal marker molecule α-smooth muscle actin (α-SMA) and the loss of epithelial marker molecule E-cadherin in lung tissue of PF mice. In vitro, COX-2 and sEH protein levels were increased in TGF-β1-treated alveolar epithelial cells (AECs). PTUPB decreased the expression of α-SMA and restored the expression of E-cadherin in TGF-β1-treated AECs, accompanied by reduced migration and collagen synthesis. Moreover, PTUPB attenuated TGF-β1-Smad2/3 pathway activation in AECs via Nrf2 antioxidant cascade. Conclusion PTUPB inhibits EMT in AECs via Nrf2-mediated inhibition of the TGF-β1-Smad2/3 pathway, which holds great promise for the clinical treatment of PF.
Collapse
|
5
|
Dedifferentiation of Human Cardiac Myofibroblasts Is Independent of Activation of COX-2/PGE 2 Pathway. Int J Mol Sci 2022; 23:ijms23063023. [PMID: 35328443 PMCID: PMC8952377 DOI: 10.3390/ijms23063023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/24/2021] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
The differentiation of cardiac fibroblasts to myofibroblasts is considered to be a critical step in activation and progression of cardiac fibrosis in heart disease. TGF-β is one of the key cytokines that promotes transition of fibroblasts to myofibroblasts. Dedifferentiation of formed myofibroblasts or reversal of formed myofibroblasts to fibroblasts remains incompletely understood. Prostaglandin E2 (PGE2) has been shown to dedifferentiate human lung myofibroblasts. The role of activation of the COX-2/PGE2 pathway in dedifferentiation of cardiac myofibroblasts remains unknown. Here, we show that phorbol 12-myristate 13-acetate (PMA) but not PGE2 induces dedifferentiation of de novo adult human cardiac myofibroblasts stimulated by TGF-β1 from human cardiac fibroblasts as evidenced by reduced expression of α-smooth muscle actin (α-SMA). PMA remarkably increased endogenous levels of PGE2 in human cardiac myofibroblasts. Pretreatment of myofibroblasts with NS-398, a selective COX-2 inhibitor, and PF-04418948, a selective PGE2 receptor type 2 (EP2) antagonist, had no effect on expression of α-SMA nor abolished the dedifferentiation induced by PMA. Our results indicated that endogenous and exogenous PGE2 has no effects on dedifferentiation of cardiac myofibroblasts. PMA-induced dedifferentiation of cardiac myofibroblast is independent of activation of COX-2 and PGE2 pathway. The mechanism in PMA-induced reversal of cardiac myofibroblasts needs to be explored further.
Collapse
|
6
|
Khan H, Sharma K, Kumar A, Kaur A, Singh TG. Therapeutic implications of cyclooxygenase (COX) inhibitors in ischemic injury. Inflamm Res 2022; 71:277-292. [PMID: 35175358 DOI: 10.1007/s00011-022-01546-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Ischemia-reperfusion injury (IRI) is the inexplicable aggravation of cellular dysfunction that results in blood flow restoration to previously ischemic tissues. COX mediates the oxidative conversion of AA to various prostaglandins and thromboxanes, which are involved in various physiological and pathological processes. In the pathophysiology of I/R injuries, COX has been found to play an important role. I/R injuries affect most vital organs and are characterized by inflammation, oxidative stress, cell death, and apoptosis, leading to morbidity and mortality. MATERIALS AND METHODS A systematic literature review of Bentham, Scopus, PubMed, Medline, and EMBASE (Elsevier) databases was carried out to understand the Nature and mechanistic interventions of the Cyclooxygenase modulations in ischemic injury. Here, we have discussed the COX Physiology and downstream signalling pathways modulated by COX, e.g., Camp Pathway, Peroxisome Proliferator-Activated Receptor Activity, NF-kB Signalling, PI3K/Akt Signalling in ischemic injury. CONCLUSION This review will discuss the various COX types, specifically COX-1 and COX-2, which are involved in developing I/R injury in organs such as the brain, spinal cord, heart, kidney, liver, and intestine.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kunal Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amit Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
7
|
Sasaki T, Izumaru K, Hata J, Sakata S, Oishi E, Nagata T, Tsuboi N, Oda Y, Kitazono T, Yokoo T, Ninomiya T. Serum NT-proBNP levels and histopathological myocardial fibrosis in autopsied cases from a Japanese community: The Hisayama Study. J Cardiol 2021; 78:237-243. [PMID: 33863625 DOI: 10.1016/j.jjcc.2021.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND natriuretic peptide is associated with myocardial fibrosis in animal models and among patients with heart disease. However, it remains unclear whether serum N-terminal pro-B-type peptide (NT-proBNP) levels are associated with histopathologically proven myocardial fibrosis among individuals without apparent heart disease. This study aimed to evaluate the association between serum NT-proBNP levels and the histopathologically estimated myocardial fibrotic area in autopsied samples from a community. METHODS we selected 63 cases without apparent heart disease with available data of serum NT-proBNP concentrations within six years before death (average age: 82 years; male: 52%) from autopsied cases in a community, and evaluated the percentage areas of myocardial fibrosis in four cardiac segments from each case (i.e. 252 cardiac segments in total). The association between serum NT-proBNP levels and the percentage area of myocardial fibrosis was estimated using a linear mixed model for repeated measures. RESULTS serum NT-proBNP levels were positively correlated with myocardial fibrotic area [Pearson's correlation coefficient: r = 0.49 (95% confidence interval: 0.28-0.66), p <0.001]. Serum NT-proBNP levels were significantly associated with the percentage areas of myocardial fibrosis after adjusting for potential confounding factors. There was no evidence of heterogeneity in the association between the subgroups with and without reduced estimated glomerular filtration rate (<60 mL/min/1.73 m2). CONCLUSIONS the present study demonstrated that elevated serum NT-proBNP levels were associated with the histopathologically measured myocardial fibrotic area in autopsied cases from a community. These findings may help clarify the association between elevated serum NT-proBNP levels and myocardial fibrosis and their influence on prognosis.
Collapse
Affiliation(s)
- Takaya Sasaki
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoko Sakata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Emi Oishi
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Nagata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuo Tsuboi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
8
|
Celecoxib alleviates pathological cardiac hypertrophy and fibrosis via M1-like macrophage infiltration in neonatal mice. iScience 2021; 24:102233. [PMID: 33748715 PMCID: PMC7967012 DOI: 10.1016/j.isci.2021.102233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
Cardiac hypertrophy is an adaptive response to all forms of heart disease, including hypertension, myocardial infarction, and cardiomyopathy. Cyclooxygenase-2 (COX-2) overexpression results in inflammatory response, cardiac cell apoptosis, and hypertrophy in adult heart after injury. However, immune response-mediated cardiac hypertrophy and fibrosis have not been well documented in injured neonatal heart. This study showed that cardiac hypertrophy and fibrosis are significantly attenuated in celecoxib (a selective COX-2 inhibitor)-treated P8 ICR mice after cryoinjury. Molecular and cellular profiling of immune response shows that celecoxib inhibits the production of cytokines and the expression of adhesion molecular genes, increases the recruitment of M1-like macrophage at wound site, and alleviates cardiac hypertrophy and fibrosis. Furthermore, celecoxib administration improves cardiac function at 4 weeks after injury. These results demonstrate that COX-2 inhibition promotes the recruitment of M1-like macrophages during early wound healing, which may contribute to the suppression of cardiac hypertrophy and fibrosis after injury. Cryoinjury successfully induces cardiac hypertrophy and fibrosis in P8 ICR mice COX-2 inhibition alleviates cardiac hypertrophy and fibrosis after cryoinjury MCP-1 significantly increases in COX-2 inhibition COX-2 inhibition improves cardiac repair in P8 ICR mice by recruiting M1-like macrophages
Collapse
|
9
|
Bryson TD, Pandrangi TS, Khan SZ, Xu J, Pavlov TS, Ortiz PA, Peterson E, Harding P. The deleterious role of the prostaglandin E 2 EP 3 receptor in angiotensin II hypertension. Am J Physiol Heart Circ Physiol 2020; 318:H867-H882. [PMID: 32142358 DOI: 10.1152/ajpheart.00538.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Timothy D Bryson
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Teja S Pandrangi
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Safa Z Khan
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Jiang Xu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Tengis S Pavlov
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Edward Peterson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan
| | - Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
10
|
Hidden Cardiotoxicity of Rofecoxib Can be Revealed in Experimental Models of Ischemia/Reperfusion. Cells 2020; 9:cells9030551. [PMID: 32111102 PMCID: PMC7140447 DOI: 10.3390/cells9030551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiac adverse effects are among the leading causes of the discontinuation of clinical trials and the withdrawal of drugs from the market. The novel concept of ‘hidden cardiotoxicity’ is defined as cardiotoxicity of a drug that manifests in the diseased (e.g., ischemic/reperfused), but not in the healthy heart or as a drug-induced deterioration of cardiac stress adaptation (e.g., ischemic conditioning). Here, we aimed to test if the cardiotoxicity of a selective COX-2 inhibitor rofecoxib that was revealed during its clinical use, i.e., increased occurrence of proarrhythmic and thrombotic events, could have been revealed in early phases of drug development by using preclinical models of ischemia/reperfusion (I/R) injury. Rats that were treated with rofecoxib or vehicle for four weeks were subjected to 30 min. coronary artery occlusion and 120 min. reperfusion with or without cardioprotection that is induced by ischemic preconditioning (IPC). Rofecoxib increased overall the arrhythmias including ventricular fibrillation (VF) during I/R. The proarrhythmic effect of rofecoxib during I/R was not observed in the IPC group. Rofecoxib prolonged the action potential duration (APD) in isolated papillary muscles, which was not seen in the simulated IPC group. Interestingly, while showing hidden cardiotoxicity manifested as a proarrhythmic effect during I/R, rofecoxib decreased the infarct size and increased the survival of adult rat cardiac myocytes that were subjected to simulated I/R injury. This is the first demonstration that rofecoxib increased acute mortality due to its proarrhythmic effect via increased APD during I/R. Rofecoxib did not interfere with the cardiprotective effect of IPC; moreover, IPC was able to protect against rofecoxib-induced hidden cardiotoxicity. These results show that cardiac safety testing with simple preclinical models of I/R injury uncovers hidden cardiotoxicity of rofecoxib and might reveal the hidden cardiotoxicity of other drugs.
Collapse
|
11
|
Zhang L, Deng M, Lu A, Chen Y, Chen Y, Wu C, Tan Z, Boini KM, Yang T, Zhu Q, Wang L. Sodium butyrate attenuates angiotensin II-induced cardiac hypertrophy by inhibiting COX2/PGE2 pathway via a HDAC5/HDAC6-dependent mechanism. J Cell Mol Med 2019; 23:8139-8150. [PMID: 31565858 PMCID: PMC6850921 DOI: 10.1111/jcmm.14684] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/13/2019] [Accepted: 04/22/2019] [Indexed: 12/24/2022] Open
Abstract
Sodium butyrate (NaBu) is reported to play important roles in a number of chronic diseases. The present work is aimed to investigate the effect of NaBu on angiotensin II (Ang II)-induced cardiac hypertrophy and the underlying mechanism in in vivo and in vitro models. Sprague Dawley rats were infused with vehicle or Ang II (200 ng/kg/min) and orally administrated with or without NaBu (1 g/kg/d) for two weeks. Cardiac hypertrophy parameters and COX2/PGE2 pathway were analysed by real-time PCR, ELISA, immunostaining and Western blot. The cardiomyocytes H9C2 cells were used as in vitro model to investigate the role of NaBu (2 mmol/L) in inhibition of Ang II-induced cardiac hypertrophy. NaBu significantly attenuated Ang II-induced increase in the mean arterial pressure. Ang II treatment remarkably increased cardiac hypertrophy as indicated by increased ratio of heart weight/body weight and enlarged cardiomyocyte size, extensive fibrosis and inflammation, as well as enhanced expression of hypertrophic markers, whereas hearts from NaBu-treated rats exhibited a significant reduction in these hypertrophic responses. Mechanistically, NaBu inhibited the expression of COX2/PGE2 along with production of ANP and phosphorylated ERK (pERK) stimulated by Ang II in in vivo and in vitro, which was accompanied by the suppression of HDAC5 and HDAC6 activities. Additionally, knocking down the expression of HDAC5 and HDAC6 via gene-editing strategy dramatically blocked Ang II-induced hypertrophic responses through COX2/PGE2 pathway. These results provide solid evidence that NaBu attenuates Ang II-induced cardiac hypertrophy by inhibiting the activation of COX2/PGE2 pathway in a HDAC5/HDAC6-dependent manner.
Collapse
Affiliation(s)
- Linlin Zhang
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Mokan Deng
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Aihua Lu
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Yanting Chen
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Yang Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunying Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhi Tan
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Krishna M Boini
- School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tianxin Yang
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Qing Zhu
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.,School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lei Wang
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.,School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
12
|
Gonzalez NC, Kuwahira I. Systemic Oxygen Transport with Rest, Exercise, and Hypoxia: A Comparison of Humans, Rats, and Mice. Compr Physiol 2018; 8:1537-1573. [PMID: 30215861 DOI: 10.1002/cphy.c170051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The objective of this article is to compare and contrast the known characteristics of the systemic O2 transport of humans, rats, and mice at rest and during exercise in normoxia and hypoxia. This analysis should help understand when rodent O2 transport findings can-and cannot-be applied to human responses to similar conditions. The O2 -transport system was analyzed as composed of four linked conductances: ventilation, alveolo-capillary diffusion, circulatory convection, and tissue capillary-cell diffusion. While the mechanisms of O2 transport are similar in the three species, the quantitative differences are naturally large. There are abundant data on total O2 consumption and on ventilatory and pulmonary diffusive conductances under resting conditions in the three species; however, there is much less available information on pulmonary gas exchange, circulatory O2 convection, and tissue O2 diffusion in mice. The scarcity of data largely derives from the difficulty of obtaining blood samples in these small animals and highlights the need for additional research in this area. In spite of the large quantitative differences in absolute and mass-specific O2 flux, available evidence indicates that resting alveolar and arterial and venous blood PO2 values under normoxia are similar in the three species. Additionally, at least in rats, alveolar and arterial blood PO2 under hypoxia and exercise remain closer to the resting values than those observed in humans. This is achieved by a greater ventilatory response, coupled with a closer value of arterial to alveolar PO2 , suggesting a greater efficacy of gas exchange in the rats. © 2018 American Physiological Society. Compr Physiol 8:1537-1573, 2018.
Collapse
Affiliation(s)
- Norberto C Gonzalez
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ichiro Kuwahira
- Department of Pulmonary Medicine, Tokai University School of Medicine, Tokai University Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
13
|
Huang S, Frangogiannis NG. Anti-inflammatory therapies in myocardial infarction: failures, hopes and challenges. Br J Pharmacol 2018; 175:1377-1400. [PMID: 29394499 PMCID: PMC5901181 DOI: 10.1111/bph.14155] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 12/14/2022] Open
Abstract
In the infarcted heart, the damage-associated molecular pattern proteins released by necrotic cells trigger both myocardial and systemic inflammatory responses. Induction of chemokines and cytokines and up-regulation of endothelial adhesion molecules mediate leukocyte recruitment in the infarcted myocardium. Inflammatory cells clear the infarct of dead cells and matrix debris and activate repair by myofibroblasts and vascular cells, but may also contribute to adverse fibrotic remodelling of viable segments, accentuate cardiomyocyte apoptosis and exert arrhythmogenic actions. Excessive, prolonged and dysregulated inflammation has been implicated in the pathogenesis of complications and may be involved in the development of heart failure following infarction. Studies in animal models of myocardial infarction (MI) have suggested the effectiveness of pharmacological interventions targeting the inflammatory response. This article provides a brief overview of the cell biology of the post-infarction inflammatory response and discusses the use of pharmacological interventions targeting inflammation following infarction. Therapy with broad anti-inflammatory and immunomodulatory agents may also inhibit important repair pathways, thus exerting detrimental actions in patients with MI. Extensive experimental evidence suggests that targeting specific inflammatory signals, such as the complement cascade, chemokines, cytokines, proteases, selectins and leukocyte integrins, may hold promise. However, clinical translation has proved challenging. Targeting IL-1 may benefit patients with exaggerated post-MI inflammatory responses following infarction, not only by attenuating adverse remodelling but also by stabilizing the atherosclerotic plaque and by inhibiting arrhythmia generation. Identification of the therapeutic window for specific interventions and pathophysiological stratification of MI patients using inflammatory biomarkers and imaging strategies are critical for optimal therapeutic design.
Collapse
Affiliation(s)
- Shuaibo Huang
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology)Albert Einstein College of MedicineBronxNY10461USA
- Department of Cardiology, Changzheng HospitalSecond Military Medical UniversityShanghai200003China
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology)Albert Einstein College of MedicineBronxNY10461USA
| |
Collapse
|
14
|
Abstract
BACKGROUND Prostaglandin E2 (PGE2) signals through 4 separate G-protein coupled receptor sub-types to elicit a variety of physiologic and pathophysiological effects. We recently reported that PGE2 via its EP3 receptor could reduce cardiac contractility of isolated myocytes and the working heart preparation. We thus hypothesized that there is an imbalance in the EP3/EP4 ratio towards EP3 in the failing heart and that overexpression of EP4 in a mouse model of heart failure would improve cardiac function. METHODS AND RESULTS Our hypothesis was tested in a mouse model of myocardial infarction (MI) with the use of AAV9-EP4 driven by the myosin heavy chain promoter to overexpress EP4 in the cardiac myocytes. Echocardiography was performed to assess cardiac function. We found that overexpression of EP4 improved shortening fraction (p = 0.0025), ejection fraction (p = 0.0003), and reduced left ventricular dimension at systole (p = 0.0013). Overexpression of EP4 also significantly reduced indices of cardiac hypertrophy and interstitial collagen fraction. Animals treated with AAV9-EP4 also had a significant decrease in TNFα mRNA expression and in the number of macrophages and T cells migrated post MI coupled with a reduction in the expression of iNOS. CONCLUSION Overexpression of EP4 improves cardiac function post MI. This may be mediated through reductions in adverse cardiac remodeling or via inhibition of cytokine/chemokine production.
Collapse
|
15
|
Ibrahim KS, El-Yazbi AF, El-Gowelli HM, El-Mas MM. Opposite Modulatory Effects of Selective and Non-Selective Cyclooxygenase Inhibition on Cardiovascular and Autonomic Consequences of Cyclosporine in Female Rats. Basic Clin Pharmacol Toxicol 2017; 120:571-581. [DOI: 10.1111/bcpt.12754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/22/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Karim S. Ibrahim
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Alexandria University; Alexandria Egypt
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Alexandria University; Alexandria Egypt
| | - Hanan M. El-Gowelli
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Alexandria University; Alexandria Egypt
| | - Mahmoud M. El-Mas
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Alexandria University; Alexandria Egypt
| |
Collapse
|
16
|
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 2016; 173:3176-3195. [PMID: 27535007 DOI: 10.1111/bph.13579] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Oxidative stress plays a key role in the vascular and metabolic abnormalities associated with obesity. Herein, we assessed whether obesity can increase coronary vasoconstriction induced by hydrogen peroxide (H2 O2 ) and the signalling pathways involving COX-2 and superoxide (O2.- ) generation. EXPERIMENTAL APPROACH Contractile responses to H2 O2 and O2.- generation were measured in coronary arteries from genetically obese Zucker rats (OZR) and compared to lean Zucker rats (LZR). KEY RESULTS Both basal and H2 O2 -stimulated O2.- production were enhanced in coronary arteries from OZR, but H2 O2 -induced vasoconstriction was unchanged. The selective COX-2 inhibitor NS398 significantly reduced H2 O2 -induced contractions in endothelium-denuded arteries from LZR and OZR, but only in endothelium-intact arteries from LZR. PGI2 (IP) receptor antagonism modestly reduced the vasoconstrictor action of H2 O2 while antagonism of the PGE2 receptor 4 (EP4 ) enhanced H2 O2 contractions in arteries from OZR but not LZR. Basal release of COX-2-derived PGE2 was higher in coronary arteries from OZR where the selective agonist of EP4 receptors TCS 2519 evoked potent relaxations. COX-2 was up-regulated after acute exposure to H2 O2 in coronary endothelium and vascular smooth muscle (VSM) and inhibition of COX-2 markedly reduced H2 O2 -elicited O2.- generation in coronary arteries and myocardium. Expression of Nox subunits in VSM and NADPH-stimulated O2.- generation was enhanced and contributed to H2 O2 vasoconstriction in arteries from obese rats. CONCLUSION AND IMPLICATIONS COX-2 contributes to cardiac oxidative stress and to the endothelium-independent O2.- -mediated coronary vasoconstriction induced by H2 O2 in obesity, which is offset by the release of COX-2-derived endothelial PGE2 acting on EP4 vasodilator receptors.
Collapse
Affiliation(s)
- Elvira Santiago
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Maria Pilar Martínez
- Departamento de Anatomía and Anatomía Patológica Comparadas, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Belén Climent
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Mercedes Muñoz
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana María Briones
- Departamento de Farmacología, Facultad de Medicina, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Albino García-Sacristán
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Rivera
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
17
|
Cao Y, Wang J, Su G, Wu Y, Bai R, Zhang Q, Gao X, Li C, Chen S, Tu P, Chai X. Anti-myocardial ischemia effect of Syringa pinnatifolia Hemsl. by inhibiting expression of cyclooxygenase-1 and -2 in myocardial tissues of mice. JOURNAL OF ETHNOPHARMACOLOGY 2016; 187:259-268. [PMID: 27130642 DOI: 10.1016/j.jep.2016.04.039] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/23/2016] [Accepted: 04/23/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The peeled stem of Syringa pinnatifolia Hemsl. (SP) is a traditional medicine in Inner Mongolia, China. The powder form of SP has been widely used for hundreds of years to relieve "He-Yi" related myocardial ischemia independently or in a traditional Chinese medicine preparation. MATERIALS AND METHODS SP was extracted with 95% and 80% ethanol. Chemical profiling was performed using HPLC-DAD and IT-TOF-ESI-MS analyses. Myocardial ischemia was produced by ligation of the left anterior descending (LAD) coronary artery to evaluate the anti-myocardial ischemia effect of SP. Male C57BL/6 mice were randomly divided into six groups (n=10 per group): a sham group, a model group, groups pretreated with SP at three dosages (20mg/kg, 40mg/kg, and 80mg/kg, intragastrically), and a positive control group (acetylsalicylic acid, ASA, 53mg/kg, intragastrically). Echocardiography was performed to determine heart function by measuring ejection fraction and fractional shortening. The levels of creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH) in serum, and 6-keto-PGF1α and TXB2 both in plasma and in protein homogenate of myocardial tissue were also measured. The levels of cyclooxygenase (COX)-1 and -2 in the heart tissue and their expressions in mouse myocardial tissue were determined using Western blot and an immunofluorescence assay, respectively. Inflammatory cell infiltration and collagen deposition changes in the myocardial ischemic tissue were observed by pathological examination. RESULTS Intragastric pretreatment with SP produced a dose-dependent increase in cardiac function. SP at 80mg/kg significantly improved the EF (p<0.001) and FS (p<0.01) compared with the model group, as well as the levels of serum CK-MB and LDH decreased obviously (p<0.001), approaching those in the sham group. Besides, an obvious reduction in inflammatory cells infiltration and collagen deposition in the infarcted myocardial tissue was shown in each SP treatment group. In addition, SP increased 6-keto-PGF1α and decreased TXB2 levels in the plasma, whereas the opposite pattern was observed in the protein homogenate from the myocardial tissues at the infarction edge, but keeping balance the ratio of 6-keto-PGF1α and TXB2, which is better than ASA in plasma. The mechanisms is associated with the downregulated expressions of COX-1 (p<0.05) and COX-2 (p<0.001). CONCLUSIONS Ethanol extract of SP has a protective effect against myocardial ischemia via down regulation of COX-1 and COX-2 expression and by adjusting the ischemia-induced imbalance between 6-keto-PGF1α and TXB2. This study shows substantial evidence to support the clinical application of SP and indicates that such medicine has great potential for treating ischemia-induced heart disease.
Collapse
Affiliation(s)
- Yuan Cao
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, PR China
| | - Jing Wang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, PR China
| | - Guozhu Su
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, PR China
| | - Yan Wu
- Center of Scientific Experiment, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Ruifeng Bai
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, PR China
| | - Qian Zhang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Xiaoli Gao
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China.
| | - Suyile Chen
- Alashan Mongolian Hospital, Alashan East Banner of Alashan, Inner Mongolia 750306, PR China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Xingyun Chai
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China.
| |
Collapse
|
18
|
Chang H, Wang Q, Shi T, Huo K, Li C, Zhang Q, Wang G, Wang Y, Tang B, Wang W, Wang Y. Effect of DanQi Pill on PPARα, lipid disorders and arachidonic acid pathway in rat model of coronary heart disease. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:103. [PMID: 27000070 PMCID: PMC4802898 DOI: 10.1186/s12906-016-1083-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/16/2016] [Indexed: 12/13/2022]
Abstract
Background Danqi pill (DQP) is one of the most widely prescribed formulas and has been shown to have remarkable protective effect on coronary heart disease (CHD). However, its regulatory effects on lipid metabolism disorders haven’t been comprehensively studied so far. We aimed to explore the effects of DQP on Peroxisome Proliferator activated receptors α (PPARα), lipid uptake-transportation-metabolism pathway and arachidonic acid (AA)-mediated inflammation pathway in rats with CHD. Methods 80 Sprague-Dawley (SD) Rats were randomly divided into sham group, model group, positive control group and DQP group. Rat model of CHD was induced by ligation of left ventricle anterior descending artery and fed with high fat diet in all but the sham group. Rats in sham group only underwent thoracotomy. After surgery, rats in the positive control and DQP group received daily treatments of pravastatin and DQP respectively. At 28 days after surgery, rats were sacrificed and plasma lipids were evaluated by plasma biochemical detection. Western blot and PCR were applied to evaluate the expressions of PPARα, proteins involved in lipid metabolism and AA pathways. Results Twenty eight days after surgery, dyslipidemia developed in CHD model rats, as illustrated by elevated plasma lipid levels. Expressions of apolipoprotein A-I (ApoA-I), cluster of differentiation 36 (CD36) and fatty acid binding protein (FABP) in the heart tissues of model group were down-regulated compared with those in sham group. Expressions of carnitine palmitoyl transferase I (CPT-1A) and lipoproteinlipase (LPL) were also reduced significantly. In addition, levels of phospholipase A2 (PLA2) and cyclooxygenase 2 (COX-2) were up-regulated. Expressions of Nuclear factor-κB (NF- κB) and signal transducer and activator of transcription 3 (STAT3) also increased. Furthermore, Expression of PPARα decreased in the model group. DQP significantly up-regulated expressions of ApoA-I and FABP, as well as the expressions of CPT-1A and CD36. In addition, DQP down-regulated expressions of PLA2, COX-2 and NF-κB in inflammation pathway. Levels of STAT3 and LPL were not affected by DQP treatment. In particular, DQP up-regulated PPARα level significantly. Conclusions DQP could effectively regulate lipid uptake-transportation-metabolism process in CHD model rats, and the effect is achieved mainly by activating ApoA-I-CD36-CPT-1A molecules. Interestingly, DQP can up-regulate expression of PPARα significantly. The anti-inflammatory effect of DQP is partly exerted by inhibiting expressions of PLA2-COX2 -NF-κB pathway.
Collapse
|
19
|
Avendaño MS, Martínez-Revelles S, Aguado A, Simões MR, González-Amor M, Palacios R, Guillem-Llobat P, Vassallo DV, Vila L, García-Puig J, Beltrán LM, Alonso MJ, Cachofeiro MV, Salaices M, Briones AM. Role of COX-2-derived PGE2 on vascular stiffness and function in hypertension. Br J Pharmacol 2016; 173:1541-55. [PMID: 26856544 DOI: 10.1111/bph.13457] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 01/08/2016] [Accepted: 01/29/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Prostanoids derived from COX-2 and EP receptors are involved in vascular remodelling in different cardiovascular pathologies. This study evaluates the contribution of COX-2 and EP1 receptors to vascular remodelling and function in hypertension. EXPERIMENTAL APPROACH Spontaneously hypertensive rats (SHR) and angiotensin II (AngII)-infused (1.44 mg · kg(-1) · day(-1), 2 weeks) mice were treated with the COX-2 inhibitor celecoxib (25 mg · kg(-1) · day(-1) i.p) or with the EP1 receptor antagonist SC19220 (10 mg · kg(-1) · day(-1) i.p.). COX-2(-/-) mice with or without AngII infusion were also used. KEY RESULTS Celecoxib and SC19220 treatment did not modify the altered lumen diameter and wall : lumen ratio in mesenteric resistance arteries from SHR-infused and/or AngII-infused animals. However, both treatments and COX-2 deficiency decreased the augmented vascular stiffness in vessels from hypertensive animals. This was accompanied by diminished vascular collagen deposition, normalization of altered elastin structure and decreased connective tissue growth factor and plasminogen activator inhibitor-1 gene expression. COX-2 deficiency and SC19220 treatment diminished the increased vasoconstrictor responses and endothelial dysfunction induced by AngII infusion. Hypertensive animals showed increased mPGES-1 expression and PGE2 production in vascular tissue, normalized by celecoxib. Celecoxib treatment also decreased AngII-induced macrophage infiltration and TNF-α expression. Macrophage conditioned media (MCM) increased COX-2 and collagen type I expression in vascular smooth muscle cells; the latter was reduced by celecoxib treatment. CONCLUSIONS AND IMPLICATIONS COX-2 and EP1 receptors participate in the increased extracellular matrix deposition and vascular stiffness, the impaired vascular function and inflammation in hypertension. Targeting PGE2 receptors might have benefits in hypertension-associated vascular damage.
Collapse
Affiliation(s)
- M S Avendaño
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - S Martínez-Revelles
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - A Aguado
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - M R Simões
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain.,Dept. Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - M González-Amor
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - R Palacios
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - P Guillem-Llobat
- Centro de Biología Molecular "Severo Ochoa", UAM-CSIC, Madrid, Spain
| | - D V Vassallo
- Dept. Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - L Vila
- Laboratorio de Angiología, Biología Vascular e Inflamación, Instituto de Investigación Biomédica (IIB Sant Pau), Barcelona, Spain
| | - J García-Puig
- Servicio de Medicina Interna, Hospital Universitario La Paz, UAM, IdiPaz, Madrid, Spain
| | - L M Beltrán
- Servicio de Medicina Interna, Hospital Universitario La Paz, UAM, IdiPaz, Madrid, Spain
| | - M J Alonso
- Dept Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - M V Cachofeiro
- Dept. Fisiología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - M Salaices
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - A M Briones
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
20
|
Pang L, Cai Y, Tang EHC, Irwin MG, Ma H, Xia Z. Prostaglandin E Receptor Subtype 4 Signaling in the Heart: Role in Ischemia/Reperfusion Injury and Cardiac Hypertrophy. J Diabetes Res 2016; 2016:1324347. [PMID: 27190998 PMCID: PMC4846751 DOI: 10.1155/2016/1324347] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/23/2016] [Indexed: 01/08/2023] Open
Abstract
Prostaglandin E2 (PGE2) is an endogenous lipid mediator, produced from the metabolism of arachidonic acids, upon the sequential actions of phospholipase A2, cyclooxygenases, and prostaglandin E synthases. The various biological functions governed by PGE2 are mediated through its four distinct prostaglandin E receptors (EPs), designated as EP1, EP2, EP3, and EP4, among which the EP4 receptor is the one most widely distributed in the heart. The availability of global or cardiac-specific EP4 knockout mice and the development of selective EP4 agonists/antagonists have provided substantial evidence to support the role of EP4 receptor in the heart. However, like any good drama, activation of PGE2-EP4 signaling exerts both protective and detrimental effects in the ischemic heart disease. Thus, the primary object of this review is to provide a comprehensive overview of the current progress of the PGE2-EP4 signaling in ischemic heart diseases, including cardiac hypertrophy and myocardial ischemia/reperfusion injury. A better understanding of PGE2-EP4 signaling should promote the development of more effective therapeutic approaches to treat the ischemic heart diseases without triggering unwanted side effects.
Collapse
Affiliation(s)
- Lei Pang
- Department of Anesthesiology, The First Hospital, Jilin University, Jilin 130021, China
| | - Yin Cai
- Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Eva Hoi Ching Tang
- Department of Pharmacology and Pharmacy and State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Michael G. Irwin
- Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Haichun Ma
- Department of Anesthesiology, The First Hospital, Jilin University, Jilin 130021, China
- *Haichun Ma:
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
21
|
Chien PTY, Lin CC, Hsiao LD, Yang CM. c-Src/Pyk2/EGFR/PI3K/Akt/CREB-activated pathway contributes to human cardiomyocyte hypertrophy: Role of COX-2 induction. Mol Cell Endocrinol 2015; 409:59-72. [PMID: 25869400 DOI: 10.1016/j.mce.2015.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/23/2015] [Accepted: 04/07/2015] [Indexed: 12/27/2022]
Abstract
Thrombin and COX-2 regulating cardiac hypertrophy are via various signaling cascades. Several transcriptional factors including CREB involve in COX-2 expression. However, the interplay among thrombin, CREB, and COX-2 in primary human neonatal ventricular cardiomyocytes remains unclear. In this study, thrombin-induced COX-2 promoter activity, mRNA and protein expression, and PGE2 synthesis were attenuated by pretreatment with the inhibitors of c-Src (PP1), Pyk2 (PF431396), EGFR (AG1478), PI3K/Akt (LY294002/SH-5), and p300 (GR343), or transfection with siRNAs of c-Src, Pyk2, EGFR, p110, Akt, CREB, and p300. Moreover, thrombin-stimulated phosphorylation of c-Src, Pyk2, EGFR, Akt, CREB and p300 was attenuated by their respective inhibitors. These results indicate that thrombin-induced COX-2 expression is mediated through PAR-1/c-Src/Pyk2/EGFR/PI3K/Akt linking to CREB and p300 cascades. Functionally, thrombin-induced hypertrophy and ANF/BNP release were, at least in part, mediated through a PAR-1/COX-2-dependent pathway. We uncover the importance of COX-2 regarding human cardiomyocyte hypertrophy that will provide a therapeutic intervention in cardiovascular diseases.
Collapse
Affiliation(s)
- Peter Tzu-Yu Chien
- Graduate Institute of Biomedical Sciences, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Physiology and Pharmacology, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical Sciences, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Physiology and Pharmacology, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.
| |
Collapse
|
22
|
Touchberry CD, Silswal N, Tchikrizov V, Elmore CJ, Srinivas S, Akthar AS, Swan HK, Wetmore LA, Wacker MJ. Cardiac thromboxane A2 receptor activation does not directly induce cardiomyocyte hypertrophy but does cause cell death that is prevented with gentamicin and 2-APB. BMC Pharmacol Toxicol 2014; 15:73. [PMID: 25519194 PMCID: PMC4293009 DOI: 10.1186/2050-6511-15-73] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/12/2014] [Indexed: 12/31/2022] Open
Abstract
Background We have previously shown that the thromboxane (TXA2) receptor agonist, U46619, can directly induce ventricular arrhythmias that were associated with increases in intracellular calcium in cardiomyocytes. Since TXA2 is an inflammatory mediator and induces direct calcium changes in cardiomyocytes, we hypothesized that TXA2 released during ischemia or inflammation could also cause cardiac remodeling. Methods U46619 (0.1-10 μM) was applied to isolated adult mouse ventricular primary cardiomyocytes, mouse ventricular cardiac muscle strips, and cultured HL-1 cardiomyocytes and markers of hypertrophy and cell death were measured. Results We found that TXA2 receptors were expressed in ventricular cardiomyocytes and were functional via calcium imaging. U46619 treatment for 24 h did not increase expression of pathological hypertrophy genes (atrial natriuretic peptide, β-myosin heavy chain, skeletal muscle α-actin) and it did not increase protein synthesis. There was also no increase in cardiomyocyte size after 48 h treatment with U46619 as measured by flow cytometry. However, U46619 (0.1-10 μM) caused a concentration-dependent increase in cardiomyocyte death (trypan blue, MTT assays, visual cell counts and TUNEL stain) after 24 h. Treatment of cells with the TXA2 receptor antagonist SQ29548 and inhibitors of the IP3 pathway, gentamicin and 2-APB, eliminated the increase in cell death induced by U46619. Conclusions Our data suggests that TXA2 does not induce cardiac hypertrophy, but does induce cell death that is mediated in part by IP3 signaling pathways. These findings may provide important therapeutic targets for inflammatory-induced cardiac apoptosis that can lead to heart failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Michael J Wacker
- Muscle Biology Group, School of Medicine, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA.
| |
Collapse
|
23
|
Dehlin HM, Levick SP. Substance P in heart failure: the good and the bad. Int J Cardiol 2013; 170:270-7. [PMID: 24286592 DOI: 10.1016/j.ijcard.2013.11.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 06/12/2013] [Accepted: 11/02/2013] [Indexed: 12/28/2022]
Abstract
The tachykinin, substance P, is found primarily in sensory nerves. In the heart, substance P-containing nerve fibers are often found surrounding coronary vessels, making them ideally situated to sense changes in the myocardial environment. Recent studies in rodents have identified substance P as having dual roles in the heart, depending on disease etiology and/or timing. Thus far, these studies indicate that substance P may be protective acutely following ischemia-reperfusion, but damaging long-term in non-ischemic induced remodeling and heart failure. Sensory nerves may be at the apex of the cascade of events leading to heart failure, therefore, they make a promising potential therapeutic target that warrants increased investigation.
Collapse
Affiliation(s)
- Heather M Dehlin
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Scott P Levick
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States.
| |
Collapse
|
24
|
Barudzic N, Turjacanin-Pantelic D, Zivkovic V, Selakovic D, Srejovic I, Joksimovic J, Jakovljevic J, Djuric DM, Jakovljevic VL. The effects of cyclooxygenase and nitric oxide synthase inhibition on oxidative stress in isolated rat heart. Mol Cell Biochem 2013; 381:301-11. [PMID: 23749198 DOI: 10.1007/s11010-013-1712-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 05/26/2013] [Indexed: 12/17/2022]
Abstract
Despite the widespread clinical use of cyclooxygenase (COX) inhibitors, dilemmas still exist about potential impact of these drugs on cardiovascular system. The present study was aimed to estimate the effects of different COX inhibitors (meloxicam, acetylsalicylic acid [ASA], and SC-560) on oxidative stress in isolated rat heart, with special focus on L-arginine/NO system. The hearts of male Wistar albino rats (total number n = 96, each group 12 rats, 8 weeks old, body mass 180-200 g) were retrogradely perfused according to the Langendorff technique at gradually increased perfusion pressure (40-120 cmH2O). After control experiments the hearts were perfused with the following drugs: 100 μmol/l ASA (Aspirin), alone or in combination with 30 μmol/l L-NAME, 0.3 μmol/l meloxicam (movalis) with or without 30 μmol/l L-NAME, 3 μmol/l meloxicam (alone or in combination with 30 μmol/l L-NAME), 30 μmol/l L-NAME, and administration of 0.25 μmol/l SC-560. In samples of coronary venous effluent the following oxidative stress markers were measured spectrophotometrically: index of lipid peroxidation (measured as thiobarbituric acid reactive substances [TBARS]), superoxide anion radical release (O2(-)), and hydrogen peroxide (H2O2). While ASA was found to have an adverse influence on redox balance in coronary circulation, and coronary perfusion, meloxicam and SC-560 do not negatively affect the intact model of the heart. Furthermore, all effects were modulated by NOS inhibition. It seems that interaction between COX and L-arginine/NO system truly exists in coronary circulation, and can be one of the possible causes for achieved effects. That means: those effects induced by different inhibitors of COX are modulated by subsequent inhibition of NOS.
Collapse
Affiliation(s)
- Nevena Barudzic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
A Model of Left Ventricular Dysfunction Complicated by CAWS Arteritis in DBA/2 Mice. Int J Vasc Med 2012; 2012:570297. [PMID: 22830029 PMCID: PMC3399367 DOI: 10.1155/2012/570297] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/24/2012] [Accepted: 05/14/2012] [Indexed: 11/18/2022] Open
Abstract
It was reported previously that a Candida albicans water-soluble fraction (CAWS), including a mannoprotein and β-glucan complex, has strong potency in inducing fatal necrotizing arteritis in DBA/2 mice. In this study, histopathological changes and cardiac function were investigated in this system. One mg/day of CAWS was given to DBA/2 mice via peritoneal injection for five days. The CAWS-treated DBA/2 mice were induced aortitis and died at an incidence of 100% within several weeks. Histological findings included stenosis in the left ventricular outflow tract (LVOT) and severe inflammatory changes of the aortic valve with fibrinoid necrosis. Cardiomegaly was observed and heart weight increased 1.62 fold (P < 0.01). Echocardiography revealed a severe reduction in contractility and dilatation of the cavity in the left ventricle (LV): LV fractional shortening (LVFS) decreased from 71% to 38% (P < 0.01), and the LV end-diastolic diameter (LVDd) increased from 2.21 mm to 3.26 mm (P < 0.01). The titer of BNP mRNA increased in the CAWS-treated group. Severe inflammatory changes resulting from CAWS brought about lethal LV dysfunction by aortic valve deformation with LVOT stenosis. This system is proposed as an easy and useful experimental model of heart failure because CAWS arteritis can be induced by CAWS injection alone.
Collapse
|
26
|
Degousee N, Simpson J, Fazel S, Scholich K, Angoulvant D, Angioni C, Schmidt H, Korotkova M, Stefanski E, Wang XH, Lindsay TF, Ofek E, Pierre S, Butany J, Jakobsson PJ, Keating A, Li RK, Nahrendorf M, Geisslinger G, Backx PH, Rubin BB. Lack of Microsomal Prostaglandin E
2
Synthase-1 in Bone Marrow–Derived Myeloid Cells Impairs Left Ventricular Function and Increases Mortality After Acute Myocardial Infarction. Circulation 2012; 125:2904-13. [DOI: 10.1161/circulationaha.112.099754] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background—
Microsomal prostaglandin E
2
synthase-1 (mPGES-1), encoded by the
Ptges
gene, catalyzes prostaglandin E
2
biosynthesis and is expressed by leukocytes, cardiac myocytes, and cardiac fibroblasts.
Ptges
−/−
mice develop more left ventricle (LV) dilation, worse LV contractile function, and higher LV end-diastolic pressure than
Ptges
+/+
mice after myocardial infarction. In this study, we define the role of mPGES-1 in bone marrow–derived leukocytes in the recovery of LV function after coronary ligation.
Methods and Results—
Cardiac structure and function in
Ptges
+/+
mice with
Ptges
+/+
bone marrow (
BM
+/+
) and
Ptges
+/+
mice with
Ptges
−/−
BM (
BM
−/−
) were assessed by morphometric analysis, echocardiography, and invasive hemodynamics before and 7 and 28 days after myocardial infarction. Prostaglandin levels and prostaglandin biosynthetic enzyme gene expression were measured by liquid chromatography–tandem mass spectrometry and real-time polymerase chain reaction, immunoblotting, immunohistochemistry, and immunofluorescence microscopy, respectively. After myocardial infarction,
BM
−/−
mice had more LV dilation, worse LV systolic and diastolic function, higher LV end-diastolic pressure, more cardiomyocyte hypertrophy, and higher mortality but similar infarct size and pulmonary edema compared with
BM
+/+
mice.
BM
−/−
mice also had higher levels of COX-1 protein and more leukocytes in the infarct, but not the viable LV, than
BM
+/+
mice. Levels of prostaglandin E
2
were higher in the infarct and viable myocardium of
BM
−/−
mice than in
BM
+/+
mice.
Conclusions—
Lack of mPGES-1 in bone marrow–derived leukocytes negatively regulates COX-1 expression, prostaglandin E
2
biosynthesis, and inflammation in the infarct and leads to impaired LV function, adverse LV remodeling, and decreased survival after acute myocardial infarction.
Collapse
Affiliation(s)
- Norbert Degousee
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Jeremy Simpson
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Shafie Fazel
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Klaus Scholich
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Denis Angoulvant
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Carlo Angioni
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Helmut Schmidt
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Marina Korotkova
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Eva Stefanski
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Xing-Hua Wang
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Thomas F. Lindsay
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Efrat Ofek
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Sandra Pierre
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Jagdish Butany
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Per-Johan Jakobsson
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Armand Keating
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Ren-Ke Li
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Matthias Nahrendorf
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Gerd Geisslinger
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Peter H. Backx
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Barry B. Rubin
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| |
Collapse
|
27
|
Catalán M, Smolic C, Contreras A, Ayala P, Olmedo I, Copaja M, Boza P, Vivar R, Avalos Y, Lavandero S, Velarde V, Díaz-Araya G. Differential regulation of collagen secretion by kinin receptors in cardiac fibroblast and myofibroblast. Toxicol Appl Pharmacol 2012; 261:300-8. [DOI: 10.1016/j.taap.2012.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 03/28/2012] [Accepted: 04/09/2012] [Indexed: 01/08/2023]
|
28
|
McCollum LT, Gallagher PE, Tallant EA. Angiotensin-(1-7) abrogates mitogen-stimulated proliferation of cardiac fibroblasts. Peptides 2012; 34:380-8. [PMID: 22326709 PMCID: PMC3326596 DOI: 10.1016/j.peptides.2012.01.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 12/16/2022]
Abstract
Previous studies showed that angiotensin-(1-7) [Ang-(1-7)] attenuates cardiac remodeling by reducing both interstitial and perivascular fibrosis. Although a high affinity binding site for Ang-(1-7) was identified on cardiac fibroblasts, the molecular mechanisms activated by the heptapeptide hormone were not identified. We isolated cardiac fibroblasts from neonatal rat hearts to investigate signaling pathways activated by Ang-(1-7) that participate in fibroblast proliferation. Ang-(1-7) reduced (3)H-thymidine, -leucine and -proline incorporation into cardiac fibroblasts stimulated with serum or the mitogen endothelin-1 (ET-1), demonstrating that the heptapeptide hormone decreases DNA, protein and collagen synthesis. The reduction in DNA synthesis by Ang-(1-7) was blocked by the AT((1-7)) receptor antagonist [d-Ala(7)]-Ang-(1-7), showing specificity of the response. Treatment of cardiac fibroblasts with Ang-(1-7) reduced the Ang II- or ET-1-stimulated increase in phospho-ERK1 and -ERK2. In contrast, Ang-(1-7) increased dual-specificity phosphatase DUSP1 immunoreactivity and mRNA, suggesting that the heptapeptide hormone increases DUSP1 to reduce MAP kinase phosphorylation and activity. Incubation of cardiac fibroblasts with ET-1 increased cyclooxygenase 2 (COX-2) and prostaglandin synthase (PGES) mRNAs, while Ang-(1-7) blocked the increase in both enzymes, suggesting that the heptapeptide hormone alters the concentration and the balance between the proliferative and anti-proliferative prostaglandins. Collectively, these results indicate that Ang-(1-7) participates in maintaining cardiac homeostasis by reducing proliferation and collagen production by cardiac fibroblasts in association with up-regulation of DUSP1 to reduce MAP kinase activities and attenuation of the synthesis of mitogenic prostaglandins. Increased Ang-(1-7) or agents that enhance production of the heptapeptide hormone may prevent abnormal fibrosis that occurs during cardiac pathologies.
Collapse
Affiliation(s)
- LaTronya T McCollum
- The Hypertension and Vascular Research Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1032, USA
| | | | | |
Collapse
|
29
|
Ibrahim MA, Morsy MA, Hafez HM, Gomaa WM, Abdelrahman AM. Effect of selective and non-selective cyclooxygenase inhibitors on doxorubicin-induced cardiotoxicity and nephrotoxicity in rats. Toxicol Mech Methods 2012; 22:424-31. [PMID: 22394338 DOI: 10.3109/15376516.2012.666658] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Doxorubicin (DX) is a highly effective chemotherapeutic agent used widely in the treatment of solid tumors; however, its optimal use was associated with cardiotoxicity and nephrotoxicity. The exact mechanism of DX-induced cardiotoxicity and nephrotoxicity is not fully explored. Induction of cyclooxygenase-2 (COX-2) activity in either cardiac or renal tissue by DX has been previously reported, indicating a possible role of COX-2 in DX-induced tissue injury. However, the nature of this role in either tissue injury is an issue of controversy. OBJECTIVE This study was the first that simultaneously evaluated the effects of a selective COX-2 inhibitor, nimesulide, and a non-selective COX-inhibitor, indomethacin, on DX-induced cardiotoxicity and nephrotoxicity in male Wistar rats. MATERIALS AND METHODS Rats were allocated into four groups. Control group, DX group (received 15 mg/kg, ip), DX + nimesulide (10 mg/kg/day, po) group, and DX + indomethacin (2 mg/kg/day, po) group. Nimesulide and indomethacin were started at the same day of DX injection and continued for 5 days. RESULTS The results of the present study showed that inhibition of COX-2 either by selective or non-selective COX-2 inhibitor ameliorated DX-induced cardiotoxicity but aggravated DX-induced nephrotoxicity in rats, as evidenced biochemically and histopathologically. DISCUSSION AND CONCLUSION Our study indicates that production of COX-2 is organ specific; consequently, the differential effect of COX-inhibitors should be considered in DX-treated patients. However, a wide scale experiment is needed for further confirmation and testing other members of COX-inhibitors (e.g. celecoxib and diclofenac).
Collapse
Affiliation(s)
- Mohamed A Ibrahim
- Department of Pharmacology, Faculty of Medicine, El-Minia University, El-Minia, Egypt
| | | | | | | | | |
Collapse
|
30
|
Zhao M, He X, Zhao M, Bi XY, Zhang HL, Yu XJ, Liu JJ, Li DL, Ma X, Zang WJ. Low-dose celecoxib improves coronary function after acute myocardial ischaemia in rabbits. Clin Exp Pharmacol Physiol 2012; 39:233-40. [DOI: 10.1111/j.1440-1681.2011.05664.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Ming Zhao
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Xi He
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Mei Zhao
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Xue-Yuan Bi
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Hong-Li Zhang
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Xiao-Jiang Yu
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Jin-Jun Liu
- Department Physiology and Pathophysiology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Dong-Ling Li
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Xin Ma
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| | - Wei-Jin Zang
- Department of Pharmacology; Xi'an Jiaotong University; College of Medicine; Xi'an; China
| |
Collapse
|
31
|
Nagalla KT, Gole M, Claudino MA, Gardner JD, Murray DB. Alteration in myocardial prostaglandin D synthase expression in pressure overload-induced left ventricular remodeling in rats. Exp Biol Med (Maywood) 2012; 237:24-30. [PMID: 22228706 DOI: 10.1258/ebm.2011.011180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We hypothesized that acute pharmacological blockade of cyclooxygenase-2 (COX-2) using nimesulide (Nime) would prevent maladaptive changes in left ventricular (LV) structure and function secondary to abdominal aortic coarctation-induced pressure overload (PO). In vivo LV chamber dimension and function were assessed by pressure/volume admittance catheter at 14 days' postsurgery in three groups (n ≥ 6/group): sham-operated (Sham); untreated PO; and selective COX-2 inhibitor nimesulide-treated PO (PO + Nime; 25 mg/kg/d). Treatment was initiated 24 h prior to surgical induction of PO. Relative to Sham, there was a marked increase in LV mass index in the PO groups (2.2 ± 0.01 mg/g versus 2.9 ± 0.10 mg/g Sham versus PO, PO+Nime: 2.5 ± 0.03 mg/g). End diastolic volume, an indicator of chamber size, was significantly decreased in the PO animals compared with Sham (202 ± 17μL versus 143 ± 16 μL Sham versus PO, PO + Nime: 226 ± 9 μL). Collagen levels in PO rats assessed by hydroxyproline analysis were significantly elevated relative to Sham values. Nimesulide treatment attenuated: (1) the increase in LV mass index; (2) the reduction in end diastolic volume; and (3) the PO-induced increase in myocardial collagen. In summary, acute COX-2 inhibition with nimesulide attenuated the maladaptive changes in the LV after PO. Acknowledging the clinical failure of chronic COX-2 inhibitor use, we propose that acute treatment with COX-2 inhibition during the initial stages of cardiac remodeling can be beneficial in maintaining the normal cardiac structure and function during PO.
Collapse
Affiliation(s)
- Krishna T Nagalla
- Department of Pharmacology, University of Mississippi, University, MS 38677, USA
| | | | | | | | | |
Collapse
|
32
|
Harding P, Murray DB. The contribution of prostaglandins versus prostacyclin in ventricular remodeling during heart failure. Life Sci 2011; 89:671-6. [PMID: 21855554 DOI: 10.1016/j.lfs.2011.07.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 07/26/2011] [Accepted: 07/28/2011] [Indexed: 12/20/2022]
Abstract
Although the role of Cox-2 in the heart's response to physiologic stress remains controversial (i.e. expression in myocytes versus other resident myocardial cells) the ever expanding role of prostanoids in multiple models of heart failure cannot be denied. Due to the fact that prostanoids are metabolized rather quickly (half life of seconds to minutes) it is believed these signaling mediators act in a paracrine fashion at the site of production. Evidence to date is quite convincing that these bioactive lipid derivatives are involved in physiologic homeostatic regulation as well as beneficial and maladaptive ventricular remodeling in heart failure. Thus, this review will assess the direct contribution of each PG on remodeling in the left ventricle (e.g. hypertrophy, functional effects, and fibrosis).
Collapse
Affiliation(s)
- Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | | |
Collapse
|
33
|
Murray DB, McLarty-Williams J, Nagalla KT, Janicki JS. Tryptase activates isolated adult cardiac fibroblasts via protease activated receptor-2 (PAR-2). J Cell Commun Signal 2011; 6:45-51. [PMID: 21786087 DOI: 10.1007/s12079-011-0146-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 07/12/2011] [Indexed: 10/18/2022] Open
Abstract
Protease activated receptor-2 (PAR-2) derived cycloxygenase-2 (COX-2) was recently implicated in a cardiac mast cell and fibroblast cross-talk signaling cascade mediating myocardial remodeling secondary to mechanical stress. We designed this study to investigate in vitro assays of isolated adult cardiac fibroblasts to determine whether binding of tryptase to the PAR-2 receptor on cardiac fibroblasts will lead to increased expression of COX-2 and subsequent formation of the arachodonic acid metabolite 15-d-Prostaglandin J(2) (15-d-PGJ(2)). The effects of tryptase (100 mU) and co-incubation with PAR-2 inhibitor peptide sequence FSLLRY-NH(2) (10(-6)M) on proliferation, hydroxyproline concentration, 15-d-PGJ(2) formation and PAR-2/COX-2 expression were investigated in fibroblasts isolated from 9 week old SD rats. Tryptase induced a significant increase in fibroproliferation, hydroxyproline, 15-d-PGJ(2) formation and PAR-2 expression which were markedly attenuated by FSLLRY. Tryptase-induced changes in cardiac fibroblast function utilize a PAR-2 dependent mechanism.
Collapse
Affiliation(s)
- David B Murray
- Department of Pharmacology, University of Mississippi School of Pharmacy, 311 Faser Hall, University, MS, 38677, USA,
| | | | | | | |
Collapse
|
34
|
Baba HA, Wohlschlaeger J. Morphological and molecular changes of the myocardium after left ventricular mechanical support. Curr Cardiol Rev 2011; 4:157-69. [PMID: 19936192 PMCID: PMC2780817 DOI: 10.2174/157340308785160606] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2008] [Revised: 04/17/2008] [Accepted: 04/17/2008] [Indexed: 12/31/2022] Open
Abstract
Left ventricular assist devices (LVAD) are currently used to either “bridge” patients with terminal congestive heart failure (CHF) until cardiac transplantation is possible or optionally for patients with contraindications for transplantation (“destination therapy”). Mechanical support is associated with a marked decrease of cardiac dilation and hypertrophy as well as numerous cellular and molecular changes (“reverse cardiac remodeling”), which can be accompanied by improved cardiac function (“bridge to recovery”) in a relatively small subset of patients with heart transplantation no longer necessary even after removal of the device (“weaning”). In the recent past, novel pharmacological strategies have been developed and are combined with mechanical support, which has increased the percentage of patients with improved clinical status and cardiac performance. Gene expression profiles have demonstrated that individuals who recover after LVAD show different gene expression compared to individuals who do not respond to unloading. This methodology holds promise for the future to develop read out frames to identify individuals who can recover after support. Aside from describing the morphological changes associated with “reverse cardiac remodeling”, this review will focus on signal transduction, transcriptional regulation, apoptosis, cell stress proteins, matrix remodeling, inflammatory mediators and aspects of neurohormonal activation in the failing human heart before and after ventricular unloading.
Collapse
Affiliation(s)
- Hideo A Baba
- Institute of Pathology and Neuropathology, University Hospital of Essen, University of Duisburg-Essen, Germany
| | | |
Collapse
|
35
|
Harding P, LaPointe MC. Prostaglandin E2 increases cardiac fibroblast proliferation and increases cyclin D expression via EP1 receptor. Prostaglandins Leukot Essent Fatty Acids 2011; 84:147-52. [PMID: 21342756 PMCID: PMC3071899 DOI: 10.1016/j.plefa.2011.01.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 01/12/2011] [Accepted: 01/12/2011] [Indexed: 10/18/2022]
Abstract
PGE(2) affects growth of many cell types. Thus, we hypothesized that PGE(2) would stimulate growth of cardiac fibroblasts. To test our hypothesis we used neonatal rat ventricular fibroblasts (NVF). RT-PCR demonstrated the presence of all 4 PGE(2) receptor (EPs) mRNAs in NVF. Using flow cytometry, we found that PGE(2) decreased the percentage of cells in G0/G1 and increased the number of cells in S phase. PGE(2) also increased expression of cyclin D3, a known regulator of the cell cycle and this effect was mimicked by the EP1/EP3 agonist sulprostone. Next, we found that treatment of NVF with PGE(2) increased phosphorylation of p42/44 MAPK and Akt and that PGE(2)-stimulation of cyclin D3 was antagonized with both a MEK inhibitor and a PI3 kinase inhibitor. In conclusion, PGE(2) stimulates cardiac fibroblast proliferation via EP1 and/or EP3, p42/44 MAPK and Akt-regulation of cyclin D3. These results may be relevant to cardiac fibrosis.
Collapse
Affiliation(s)
- Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA.
| | | |
Collapse
|
36
|
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 2010; 300:H1053-61. [PMID: 21193590 DOI: 10.1152/ajpheart.00772.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Our laboratory previously reported that inducible PGE(2) synthase, mPGES-1, contributes to micromolar production of PGE(2) in neonatal ventricular myocytes in vitro, which stimulates their growth. We therefore hypothesized that mPGES-1 contributes to cardiac hypertrophy following angiotensin II (ANG II) infusion. To test this hypothesis, we used 10- to 12-wk-old mPGES-1 knockout mice (mPGES-1 KO) and C57Bl/6 control mice infused for 8 wk with either 1.4 mg · kg(-1) · day(-1) ANG II or vehicle subcutaneously. Blood pressure [systolic blood pressure (SBP)] was measured throughout the study, and cardiac function was assessed by M-mode echocardiography at baseline and at 8 wk of infusion. At the conclusion of the study, immunohistochemistry was used to evaluate collagen fraction, myocyte cross-sectional area (MCSA), and apoptosis. At baseline, there was no difference in SBP between mPGES-1 KO mice and C57BL/6 controls. ANG II infusion increased SBP to similar levels in both strains. In control mice, infusion of ANG II increased MCSA and posterior wall thickness at diastole (PWTd) but had little effect on cardiac function, consistent with compensatory hypertrophy. In contrast, cardiac function was worse in mPGES-1 KO mice after ANG II treatment. Ejection fraction declined from 76.2 ± 2.7 to 63.3 ± 3.4% after ANG II, and left ventricular dimension at systole and diastole increased from 1.29 ± 0.02 to 1.78 ± 0.15 mm and from 2.57 ± 0.03 to 2.90 ± 0.13 mm, respectively. Infusion of ANG II increased both the LV-to-body weight and the mass-to-body weight ratios to a similar extent in both strains. However, PWTd increased by a lesser extent in KO mice, suggesting an impaired hypertrophic response. ANG II infusion increased collagen staining similarly in both strains, but TdT-dUTP nick end labeling staining was greater in mPGES-1 KO mice. Overall, these results are consistent with a beneficial effect for mPGES-1 in the maintenance of cardiac function in ANG II-dependent hypertension.
Collapse
Affiliation(s)
- Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, USA.
| | | | | | | |
Collapse
|
37
|
Shibata K, Yatera Y, Furuno Y, Sabanai K, Morisada N, Nakata S, Morishita T, Yamazaki F, Tanimoto A, Sasaguri Y, Tasaki H, Nakashima Y, Shimokawa H, Yanagihara N, Otsuji Y, Tsutsui M. Spontaneous development of left ventricular hypertrophy and diastolic dysfunction in mice lacking all nitric oxide synthases. Circ J 2010; 74:2681-92. [PMID: 20966596 DOI: 10.1253/circj.cj-10-0277] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The role of the nitric oxide synthase (NOS) system in cardiac architecture and function remains unknown. This point was addressed in mice that lack all 3 NOS genes. METHODS AND RESULTS Morphological, echocardiographic, and hemodynamic analyses were performed in wild-type (WT), singly nNOS(-/-), iNOS(-/-), eNOS(-/-), and triply n/i/eNOS(-/-) mice. At 5 months of age, but not at 2 months of age, significant left ventricular (LV) hypertrophy was noted in n/i/eNOS(-/-) mice and to a lesser extent in eNOS(-/-) mice, but not in nNOS(-/-) or iNOS(-/-) mice, compared with WT mice. Importantly, significant LV diastolic dysfunction (as evaluated by echocardiographic E/A wave ratio and hemodynamic -dP/dt and Tau), with preserved LV systolic function (as assessed by echocardiographic fractional shortening and hemodynamic +dP/dt), was noted only in n/i/eNOS(-/-) mice, and this was associated with enhanced LV end-diastolic pressure and increased lung wet weight, all of which are characteristics consistent with diastolic heart failure in humans. Finally, long-term oral treatment with an angiotensin II type 1 (AT(1)) receptor blocker, olmesartan, significantly prevented all these abnormalities of n/i/eNOS(-/-) mice. CONCLUSIONS These results provide the first direct evidence that the complete disruption of all NOSs results in LV hypertrophy and diastolic dysfunction in mice in vivo through the AT(1) receptor pathway, demonstrating a pivotal role of the endogenous NOS system in maintaining cardiac homeostasis.
Collapse
Affiliation(s)
- Kiyoko Shibata
- Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyusyu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Branski RC, Zhou H, Sandulache VC, Chen J, Felsen D, Kraus DH. Cyclooxygenase-2 signaling in vocal fold fibroblasts. Laryngoscope 2010; 120:1826-31. [PMID: 20717945 DOI: 10.1002/lary.21017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVES/HYPOTHESIS Inflammation and its role in a coordinated fibroplastic response, which disrupts the structure of the vocal folds following injury, is critical. Cyclooxygenase-2 (COX-2) is an important enzyme involved in both inflammation and fibrosis; in addition, it is a prime target for therapeutic intervention. We sought to study this pathway in vocal fold fibroblasts to provide a foundation for future interventional studies. STUDY DESIGN In vitro. METHODS Human vocal fold fibroblasts were incubated with IL-1 beta to determine the effects on COX-2 signaling, along with upstream regulatory mechanisms and downstream mediators of wound healing. In vitro methods to assess mRNA expression, as well as intracellular and secreted protein (sodium dodecyl sulfate polyacrylamide gel electrophoresis and enzyme-linked immunosorbent assay) were employed. RESULTS IL-1 beta regulation of COX-2 mRNA and protein levels was dose and time dependent and IL-1 beta altered PGE(2) metabolism, via regulation of both synthetic and degradative enzymes. IL-1 beta increased nuclear factor (NF)-kappaB activation and nuclear translocation. Inhibition of the p50 and p65 subunits of NF-kappaB decreased IL-1 beta-induced COX-2 transcription. IL-1 beta also altered mRNA expression of four cell-surface prostaglandin receptors. CONCLUSIONS Inflammation and fibrosis are important in the vocal fold pathophysiologic response to injury. Our data suggest that COX-2 and PGE(2) are inducible in human vocal fold fibroblasts, and this response appears to be NF-kappaB-dependent. We purport this fundamental investigation will lead to increased insight regarding injury and repair in the vocal folds, with the ultimate goal of developing novel clinical care paradigms.
Collapse
Affiliation(s)
- Ryan C Branski
- Department of Head and Neck Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Wang BH, Bertucci MC, Ma JY, Adrahtas A, Cheung RY, Krum H. Celecoxib, but not rofecoxib or naproxen, attenuates cardiac hypertrophy and fibrosis induced in vitro by angiotensin and aldosterone. Clin Exp Pharmacol Physiol 2010; 37:912-8. [DOI: 10.1111/j.1440-1681.2010.05405.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
40
|
Qu C, Leung SWS, Vanhoutte PM, Man RYK. Chronic inhibition of nitric-oxide synthase potentiates endothelium-dependent contractions in the rat aorta by augmenting the expression of cyclooxygenase-2. J Pharmacol Exp Ther 2010; 334:373-80. [PMID: 20444882 DOI: 10.1124/jpet.110.167098] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acute inhibition of nitric-oxide synthase (NOS) unmasks the release of endothelium-derived contracting factors (EDCFs). The present study investigated whether chronic inhibition of NOS modulates endothelium-dependent contractions. Eighteen-week-old male Sprague-Dawley rats were treated by daily gavage for 6 weeks with the NOS inhibitor N(omega)-nitro-L-arginine methyl ester (L-NAME) (60 mg/kg) or vehicle (distilled water; 1 ml/kg). Chronic treatment with L-NAME increased arterial blood pressure. Isometric tension was measured in aortic rings with or without endothelium. Endothelium-dependent relaxations to acetylcholine and the calcium ionophore 5-(methylamino)-2-[(2R,3R,6S,8S,9R,11R)-3,9,11-trimethyl-8-[(1S)-1-methyl-2-oxo-2-(1H-pyrrol-2-yl)-ethyl]-1,7-dioxaspiro[5.5]undec-2-yl]methyl]-4-benzoxazolecarboxylic acid (A23187) were reduced in preparations from L-NAME-treated rats. The reduction in relaxation to A23187 was partially reversed by L-arginine (1 mM). In quiescent aortic rings, A23187 caused contractions in the presence of L-NAME and intact endothelium. The A23187-induced contractions were greater in rings from the L-NAME-treated rats than in those from the control group. These contractions were abolished by the cyclooxygenase (COX)-2 inhibitor N-[2-cyclohexyloxy-4-nitrophenyl]methanesulfonamide (NS-398) and the thromboxane-prostanoid (TP) receptor antagonist 3-((6R)-6-{[(4-chlorophenyl)sulfonyl]amido}-2-methyl-5,6,7,8-tetrahydronaphthalen-1-yl)propanoate (S18886), but not by the COX-1 inhibitor 5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazole (SC-560). Chronic L-NAME treatment reduced the level of nitric oxide in the plasma but increased COX activity in the aortic rings. Western blotting and immunohistochemical staining showed that endothelial NOS expression was reduced in the aortae of the chronic L-NAME-treated group. COX-1 expression was augmented slightly, whereas COX-2 expression was up-regulated markedly. The TP receptor expression was comparable with control. These experiments demonstrate that chronic NOS inhibition increases endothelium-dependent contractions of the rat aorta by inducing COX-2 expression and augmenting the production of EDCF.
Collapse
Affiliation(s)
- Chen Qu
- Department of Pharmacology and Pharmacy, University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.
| | | | | | | |
Collapse
|
41
|
Papanicolaou KN, Streicher JM, Ishikawa TO, Herschman H, Wang Y, Walsh K. Preserved heart function and maintained response to cardiac stresses in a genetic model of cardiomyocyte-targeted deficiency of cyclooxygenase-2. J Mol Cell Cardiol 2010; 49:196-209. [PMID: 20399788 DOI: 10.1016/j.yjmcc.2010.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/31/2010] [Accepted: 04/06/2010] [Indexed: 12/12/2022]
Abstract
Cyclooxygenase-1 and -2 are rate-limiting enzymes in the formation of a wide array of bioactive lipid mediators collectively known as prostanoids (prostaglandins, prostacyclins, and thromboxanes). Evidence from clinical trials shows that selective inhibition of the second isoenzyme (cyclooxygenase-2, or Cox-2) is associated with increased risk for serious cardiovascular events and findings from animal-based studies have suggested protective roles of Cox-2 for the heart. To further characterize the function of Cox-2 in the heart, mice with loxP sites flanking exons 4 and 5 of Cox-2 were rendered knockout specifically in cardiac myocytes (Cox-2 CKO mice) via cre-mediated recombination. Baseline cardiac performance of CKO mice remained unchanged and closely resembled that of control mice. Furthermore, myocardial infarct size induced after in vivo ischemia/reperfusion (I/R) injury was comparable between CKO and control mice. In addition, cardiac hypertrophy and function four weeks after transverse aortic constriction (TAC) was found to be similar between the two groups. Assessment of Cox-2 expression in purified adult cardiac cells isolated after I/R and TAC suggests that the dominant source of Cox-2 is found in the non-myocyte fraction. In conclusion, our animal-based analyses together with the cell-based observations portray a limited role of cardiomyocyte-produced Cox-2 at baseline and in the context of ischemic or hemodynamic challenge.
Collapse
Affiliation(s)
- Kyriakos N Papanicolaou
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | |
Collapse
|
42
|
Sellers RS, Radi ZA, Khan NK. Pathophysiology of cyclooxygenases in cardiovascular homeostasis. Vet Pathol 2010; 47:601-13. [PMID: 20418470 DOI: 10.1177/0300985810364389] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cyclooxygenase (COX) catalyzes the conversion of arachidonic acid into prostaglandin H(2) (PGH(2)), which is subsequently converted to the prostanoids PGE(2), PGI(2), PGF(2alpha), and thromboxane A(2). COX has 2 distinct membrane-anchored isoenzymes: COX-1 and COX-2. COX-1 is constitutively expressed in most normal tissues; COX-2 is highly induced by proinflammatory mediators in the setting of inflammation, injury, and pain. Inhibitors of COX activity include conventional nonselective nonsteroidal anti-inflammatory drugs and selective nonsteroidal anti-inflammatory drugs, such as COX-2 inhibitors. The adverse effects of COX inhibitors on the cardiovascular system have been addressed in the last few years. In general, COX inhibitors have many effects, but those most important to the cardiovascular system can be direct (through the effects of prostanoids) and indirect (through alterations in fluid dynamics). Despite reports of detrimental human cardiovascular events associated with COX inhibitors, short, long, and lifetime preclinical toxicology studies in rodents and nonrodents have failed to identify these risks. This article focuses on the expression and function of COX enzymes in normal and pathologic conditions of the cardiovascular system and discusses the cardiovascular pathophysiologic complications associated with COX inhibition.
Collapse
Affiliation(s)
- R S Sellers
- Albert Einstein College of Medicine Cancer Center, Histology and Comparative Pathology Facility, 158 Price Center, 1301 Morris Park Ave, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
43
|
Streicher JM, Ren S, Herschman H, Wang Y. MAPK-activated protein kinase-2 in cardiac hypertrophy and cyclooxygenase-2 regulation in heart. Circ Res 2010; 106:1434-43. [PMID: 20339119 DOI: 10.1161/circresaha.109.213199] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Activation of p38 mitogen-activated protein kinase (MAPK) has a significant impact on cardiac gene expression, contractility, extracellular matrix remodeling, and inflammatory response in heart. The p38 kinase pathway also has a controversial role in cardiac hypertrophy. MAPK-activated protein kinase-2 (MK2) is a well-established p38 downstream kinase, yet its contribution to p38-mediated pathological response in heart has not been investigated. OBJECTIVE We examined the specific contribution of MK2 to the pathological remodeling induced by p38. METHODS AND RESULTS We used a cardiomyocyte specific and inducible transgenic approach to determine the functional and molecular impact of acute activation of the p38 pathway in heart in either a MK2 wild-type or a MK2-null background. p38 activation in wild-type mice led to a rapid onset of lethal cardiomyopathy associated with cardiomyocyte hypertrophy, interstitial fibrosis, and contractile dysfunction. Inactivation of MK2 partially but significantly reduced cardiomyocyte hypertrophy, improved contractile performance, and prevented early lethality. MK2 inactivation had no effect on the mRNA levels of hypertrophic marker genes or the proinflammatory gene cyclooxygenase (COX)-2. However, MK2 had a major role in COX-2 protein synthesis without affecting the mRNA level or protein stability. CONCLUSIONS p38 activity in adult myocytes can contribute to pathological hypertrophy and remodeling in adult heart and that MK2 is an important downstream molecule responsible for specific features of p38-induced cardiac pathology.
Collapse
Affiliation(s)
- John M Streicher
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, CA 91301, USA
| | | | | | | |
Collapse
|
44
|
Cyclooxygenase-2 inhibition provides lasting protection against neonatal hypoxic-ischemic brain injury. Crit Care Med 2010; 38:572-8. [PMID: 20029340 DOI: 10.1097/ccm.0b013e3181cb1158] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate whether inhibition of cyclooxygenase-2, a critical component of the inflammatory pathway, is neuroprotective in a neonatal rat model of cerebral hypoxia-ischemia. The development of brain inflammation largely contributes to neonatal brain injury that may lead to a lifetime of neurologic deficits. DESIGN Laboratory investigation. SETTING University research laboratory. SUBJECTS Postnatal day ten Sprague-Dawley rats. INTERVENTIONS Neonatal hypoxia-ischemia was induced by ligation of the right common carotid artery followed by 2 hrs of hypoxia (8% oxygen). The pups in treatment groups were administered 10 mg/kg (low dose) or 30 mg/kg (high dose) of a known selective cyclooxygenase-2 inhibitor (NS398). Animals were euthanized at three time points: 72 hrs, 2 wks, or 6 wks. Inflammation outcomes were assessed at 72 hrs; brain damage was assessed at 2 wks and 6 wks along with other organs (heart, spleen). Detailed neurobehavioral examination was performed at 6 wks. MEASUREMENTS AND MAIN RESULTS Pharmacologic inhibition of cyclooxygenase-2 markedly increased survivability within the first 72 hrs compared with untreated rats (100% vs. 72%). Low- and high-dose NS398 significantly attenuated the loss of brain and body weights observed after hypoxia-ischemia. Neurobehavioral outcomes were significantly improved in some parameters with low-dose treatment, whereas high-dose treatment consistently improved all neurologic deficits. Immunohistochemical results showed a marked decrease in macrophage, microglial, and neutrophil abundance in ipsilateral hemisphere of the NS398-treated group along with a reduction in interleukin-6 expression. CONCLUSIONS Selective cyclooxygenase-2 inhibition protected neonatal rats against death, progression of brain injury, growth retardation, and neurobehavioral deficits after a hypoxic-ischemic insult.
Collapse
|
45
|
Kwak HJ, Park KM, Choi HE, Park HY. Protective mechanisms of NO preconditioning against NO-induced apoptosis in H9c2 cells: role of PKC and COX-2. Free Radic Res 2010; 43:744-52. [DOI: 10.1080/10715760903040602] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
46
|
Kwak HJ, Park KM, Choi HE, Lim HJ, Park JH, Park HY. The cardioprotective effects of zileuton, a 5-lipoxygenase inhibitor, are mediated by COX-2 via activation of PKCδ. Cell Signal 2010; 22:80-7. [DOI: 10.1016/j.cellsig.2009.09.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 09/12/2009] [Accepted: 09/14/2009] [Indexed: 10/20/2022]
|
47
|
He Q, Harding P, LaPointe MC. PKA, Rap1, ERK1/2, and p90RSK mediate PGE2 and EP4 signaling in neonatal ventricular myocytes. Am J Physiol Heart Circ Physiol 2009; 298:H136-43. [PMID: 19880670 DOI: 10.1152/ajpheart.00251.2009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously reported that 1) inhibition of cyclooxygenase-2 and PGE(2) production reduces hypertrophy after myocardial infarction in mice and 2) PGE(2) acting through its EP4 receptor causes hypertrophy of neonatal ventricular myocytes (NVMs) via ERK1/2. It is known that EP4 couples to adenylate cyclase, cAMP, and PKA. The present study was designed to determine interactions between the cAMP-PKA pathway and ERK1/2 and to further characterize events downstream of ERK1/2. We hypothesized that PKA and the small GTPase Rap are upstream of ERK1/2 and that 90-kDa ribosomal S6 kinase (p90RSK) is activated downstream. Treatment of NVMs with PGE(2) activated Rap, and this activation was inhibited in part by an EP4 antagonist and PKA inhibition. Transfection of a dominant negative mutant of Rap reduced PGE(2) activation of ERK1/2. PGE(2) activation of p90RSK was also dependent on EP4, PKA, and Rap. We also tested the involvement of Rap, ERK1/2, and p90RSK in PGE(2) regulation of gene expression. PGE(2) stimulation of brain natriuretic peptide promoter activity was blocked by either ERK1/2 inhibition or a dominant negative mutation of p90RSK. PGE(2) stimulation of c-Fos was dependent on EP4, PKA, ERK1/2, and p90RSK, whereas only the latter two kinases were involved in PGE(2) regulation of early growth response-1. Finally, we tested the involvement of EP4-dependent signaling in the NVM growth response and found that the overexpression of EP4 increased NVM cell size. We conclude that EP4-dependent signaling in NVMs in part involves PKA, Rap, ERK1/2, and p90RSK and results in the increased expression of brain natriuretic peptide and c-Fos.
Collapse
Affiliation(s)
- Quan He
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202-2689, USA
| | | | | |
Collapse
|
48
|
Parecoxib inhibits apoptosis in acute myocardial infarction due to permanent coronary ligation but not due to ischemia-reperfusion. J Cardiovasc Pharmacol 2009; 53:495-8. [PMID: 19455055 DOI: 10.1097/fjc.0b013e3181a7b5b6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE Myocardial ischemia induces cyclooxygenase 2 (COX-2) expression. We evaluated the effects of parecoxib, a COX-2 inhibitor, in 2 different mouse models of myocardial ischemia: permanent left coronary artery ligation (PI) and transient ligation (30 minutes ischemia) followed by reperfusion (I/R). METHODS Forty adult male Institute of Cancer Research mice underwent PI (n = 24) or I/R (n = 16), followed by randomization to parecoxib (0.75 mg/kg intraperitoneal daily) or normal saline for 7 days. RESULTS Parecoxib significantly reduced apoptosis [0.8% vs. 3.4% (saline), P < 0.001] and 7-day mortality [0% vs. 57% (saline), P = 0.040] in the PI group but showed no benefit in the I/R group. Parecoxib-treated mice also exhibited greater fractional shortening in the PI group [22% vs. 14% (saline), P = 0.045) but not in the I/R group. Parecoxib did not affect infarct size in either group. CONCLUSIONS COX-2 may play a pivotal role in mediating apoptosis in the ischemic peri-infarct myocardium that is not reperfused after infarct.
Collapse
|
49
|
Wu D, Mennerich D, Arndt K, Sugiyama K, Ozaki N, Schwarz K, Wei J, Wu H, Bishopric NH, Doods H. Comparison of microsomal prostaglandin E synthase-1 deletion and COX-2 inhibition in acute cardiac ischemia in mice. Prostaglandins Other Lipid Mediat 2009; 90:21-5. [PMID: 19559811 DOI: 10.1016/j.prostaglandins.2009.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 06/16/2009] [Accepted: 06/18/2009] [Indexed: 01/26/2023]
Abstract
The aim of the present study was to compare the effects of genetic mPGES-1 loss and COX-2 inhibition on myocardial damage after coronary occlusion. mPGES-1(-/-) mice and their wild-type littermates were injected with vehicle or COX-2 inhibitor (celecoxib), and 30min later the left coronary artery was surgically occluded. At 24h, myocardial infarct (MI) volume was measured histologically. Post-MI survival was reduced in WT mice receiving celecoxib (12/20) compared with vehicle-treated controls (12/12) or the loss of mPGES-1 (13/13) together with increased phosphokinase (CPK) and cardiac troponin-I release. Endogenous mPGES-1 expression was unchanged by ischemia in WT mice and absent in mPGES-1(-/-) hearts. COX-2 expression was markedly increased at 24h after MI in WT hearts; this upregulation was largely attenuated in mPGES-1(-/-) mice. We conclude that loss of mPGES-1 prevents the upregulation of COX-2 after myocardial infarct, and in contrast to inhibition of COX-2, does not increase ischemic myocardial damage.
Collapse
Affiliation(s)
- Dongmei Wu
- Department of Research, Division of Neonatology, Mount Sinai Medical Center, 4300 Alton Road, Miami Beach, FL 33140, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Carnieto A, Dourado PMM, Luz PLD, Chagas ACP. Selective cyclooxygenase-2 inhibition protects against myocardial damage in experimental acute ischemia. Clinics (Sao Paulo) 2009; 64:245-52. [PMID: 19330252 PMCID: PMC2666458 DOI: 10.1590/s1807-59322009000300016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Accepted: 12/05/2008] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Acute myocardial infarction is associated with tissue inflammation. Early coronary reperfusion clearly improves the outcome but may help propagate the inflammatory response and enhance tissue damage. Cyclooxygenase-2 is an enzyme that catalyzes the initial step in the formation of inflammatory prostaglandins from arachidonic acid. Cyclooxygenase-2 levels are increased when ischemic cardiac events occur. The overall function of COX-2 in the inflammatory process generated by myocardial ischemic damage has not yet been elucidated. GOAL The objective of this study was to determine whether a selective cyclooxygenase-2 inhibitor (rofecoxib) could alter the evolution of acute myocardial infarction after reperfusion. METHODS AND RESULTS This study was performed with 48 mongrel dogs divided into two groups: controls and those treated with the drug. All animals were prepared for left anterior descending coronary artery occlusion. The dogs then underwent 180 minutes of coronary occlusion, followed by 30 minutes of reperfusion. Blood samples were collected from the venous sinus immediately before coronary occlusion and after 30 minutes of reperfusion for measurements of CPK-MB, CPK-MBm and troponin I. During the experiment we observed the mean blood pressure, heart rate and coronary flow. The coronary flow and heart rate did not change, but in the control group, there was blood pressure instability, in addition to maximal levels of CPK-MB post-infarction. The same results were observed for CPK-MBm and troponin I. CONCLUSION In a canine model of myocardial ischemia-reperfusion, selective inhibition of Cyclooxygenase-2 with rofecoxib was not associated with early detrimental effects on the hemodynamic profile or the gross extent of infarction; in fact, it may be beneficial by limiting cell necrosis.
Collapse
Affiliation(s)
- Alberto Carnieto
- Atherosclerosis Unit, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo - São Paulo/SP, Brasil.
| | | | | | | |
Collapse
|