1
|
Sidorkiewicz M. The Cardioprotective Effects of Polyunsaturated Fatty Acids Depends on the Balance Between Their Anti- and Pro-Oxidative Properties. Nutrients 2024; 16:3937. [PMID: 39599723 PMCID: PMC11597422 DOI: 10.3390/nu16223937] [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: 10/16/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are not only structural components of membrane phospholipids and energy storage molecules in cells. PUFAs are important factors that regulate various biological functions, including inflammation, oxidation, and immunity. Both n-3 and n-6 PUFAs from cell membranes can be metabolized into pro-inflammatory and anti-inflammatory metabolites that, in turn, influence cardiovascular health in humans. The role that PUFAs play in organisms depends primarily on their structure, quantity, and the availability of enzymes responsible for their metabolism. n-3 PUFAs, such as eicosapentaenoic (EPA) and docosahexaenoic (DHA), are generally known for anti-inflammatory and atheroprotective properties. On the other hand, n-6 FAs, such as arachidonic acid (AA), are precursors of lipid mediators that display mostly pro-inflammatory properties and may attenuate the efficacy of n-3 by competition for the same enzymes. However, a completely different light on the role of PUFAs was shed due to studies on the influence of PUFAs on new-onset atrial fibrillation. This review analyzes the role of PUFAs and PUFA derivatives in health-related effects, considering both confirmed benefits and newly arising controversies.
Collapse
Affiliation(s)
- Malgorzata Sidorkiewicz
- Department of Medical Biochemistry, Faculty of Health Sciences, Medical University of Lodz, 90-419 Lodz, Poland
| |
Collapse
|
2
|
Xu L, Yang Q, Zhou J. Mechanisms of Abnormal Lipid Metabolism in the Pathogenesis of Disease. Int J Mol Sci 2024; 25:8465. [PMID: 39126035 PMCID: PMC11312913 DOI: 10.3390/ijms25158465] [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: 06/25/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Lipid metabolism is a critical component in preserving homeostasis and health, and lipids are significant chemicals involved in energy metabolism in living things. With the growing interest in lipid metabolism in recent years, an increasing number of studies have demonstrated the close relationship between abnormalities in lipid metabolism and the development of numerous human diseases, including cancer, cardiovascular, neurological, and endocrine system diseases. Thus, understanding how aberrant lipid metabolism contributes to the development of related diseases and how it works offers a theoretical foundation for treating and preventing related human diseases as well as new avenues for the targeted treatment of related diseases. Therefore, we discuss the processes of aberrant lipid metabolism in various human diseases in this review, including diseases of the cardiovascular system, neurodegenerative diseases, endocrine system diseases (such as obesity and type 2 diabetes mellitus), and other diseases including cancer.
Collapse
Affiliation(s)
| | | | - Jinghua Zhou
- School of Basic Medicine Sciences, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
3
|
Chang M, Wu G, Bao P, Yao S, Du M, Chu C, Wang D, Jia H, Sun Y, Yan Y, Zhang X, Hu G, Man Z, Guo T, Luo W, Li H, Wang Y, Mu J. Associations of E-proteinoid 3 receptor genetic polymorphisms with salt sensitivity, longitudinal blood pressure changes, and hypertension incidence in Chinese adults. J Clin Hypertens (Greenwich) 2024; 26:955-963. [PMID: 38952049 PMCID: PMC11301432 DOI: 10.1111/jch.14859] [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: 12/20/2023] [Revised: 04/13/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024]
Abstract
The E-proteinoid 3 receptor (PTGER3), a member of the prostaglandin E2 (PGE2) subtype receptor, belongs to the G-protein-coupled superfamily of receptors. Animal studies have demonstrated its involvement in salt sensitivity by regulating sodium reabsorption. This study aimed to investigate the association between genetic variants of PTGER3 and salt sensitivity, longitudinal blood pressure (BP) changes, and the incidence of hypertension in Chinese adults. A chronic salt intake intervention was conducted involving 514 adults from 124 families in the 2004 Baoji Salt-Sensitivity Study Cohort in northern China. These participants followed a 3-day regular baseline diet, followed by a 7-day low-salt diet (3.0 g/d) and a 7-day high-salt diet (18 g/d), and were subsequently followed for 14 years. The findings revealed a significant relationship between the single nucleotide polymorphism (SNP) rs17482751 of PTGER3 and diastolic blood pressure (DBP) response to high salt intervention. Additionally, SNPs rs11209733, rs3765894, and rs2268062 were significantly associated with longitudinal changes in systolic blood pressure (SBP), DBP, and mean arterial pressure (MAP) during the 14-year follow-up period. SNP rs6424414 was significantly associated with longitudinal changes in DBP over 14 years. Finally, SNP rs17482751 showed a significant correlation with the incidence of hypertension over 14 years. These results emphasize the significant role of PTGER3 gene polymorphism in salt sensitivity, longitudinal BP changes, and the development of hypertension in the Chinese population.
Collapse
Affiliation(s)
- Ming‐Ke Chang
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Guan‐Ji Wu
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Department of CardiologyXi'an Central Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Peng Bao
- Department of General PracticeXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shi Yao
- Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral DiseasesAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongChina
| | - Ming‐Fei Du
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Chao Chu
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Dan Wang
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Hao Jia
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Yue Sun
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Yu Yan
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Xi Zhang
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Gui‐Lin Hu
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Zi‐Yue Man
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Tong‐Shuai Guo
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Wen‐Jing Luo
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Hao Li
- Department of Critical Care MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yang Wang
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Jian‐Jun Mu
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| |
Collapse
|
4
|
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
|
5
|
Ye L, Wang B, Xu H, Zhang X. The Emerging Therapeutic Role of Prostaglandin E2 Signaling in Pulmonary Hypertension. Metabolites 2023; 13:1152. [PMID: 37999248 PMCID: PMC10672796 DOI: 10.3390/metabo13111152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/28/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Mild-to-moderate pulmonary hypertension (PH) is a common complication of chronic obstructive pulmonary disease (COPD). It is characterized by narrowing and thickening of the pulmonary arteries, resulting in increased pulmonary vascular resistance (PVR) and ultimately leading to right ventricular dysfunction. Pulmonary vascular remodeling in COPD is the main reason for the increase of pulmonary artery pressure (PAP). The pathogenesis of PH in COPD is complex and multifactorial, involving chronic inflammation, hypoxia, and oxidative stress. To date, prostacyclin and its analogues are widely used to prevent PH progression in clinical. These drugs have potent anti-proliferative, anti-inflammatory, and stimulating endothelial regeneration properties, bringing therapeutic benefits to the slowing, stabilization, and even some reversal of vascular remodeling. As another well-known and extensively researched prostaglandins, prostaglandin E2 (PGE2) and its downstream signaling have been found to play an important role in various biological processes. Emerging evidence has revealed that PGE2 and its receptors (i.e., EP1-4) are involved in the regulation of pulmonary vascular homeostasis and remodeling. This review focuses on the research progress of the PGE2 signaling pathway in PH and discusses the possibility of treating PH based on the PGE2 signaling pathway.
Collapse
Affiliation(s)
- Lan Ye
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116041, China;
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central Hospital of Dalian University of Technology, Dalian 116000, China;
| | - Hu Xu
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Xiaoyan Zhang
- Health Science Center, East China Normal University, Shanghai 200241, China
| |
Collapse
|
6
|
Beccacece L, Abondio P, Bini C, Pelotti S, Luiselli D. The Link between Prostanoids and Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24044193. [PMID: 36835616 PMCID: PMC9962914 DOI: 10.3390/ijms24044193] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Cardiovascular diseases are the leading cause of global deaths, and many risk factors contribute to their pathogenesis. In this context, prostanoids, which derive from arachidonic acid, have attracted attention for their involvement in cardiovascular homeostasis and inflammatory processes. Prostanoids are the target of several drugs, but it has been shown that some of them increase the risk of thrombosis. Overall, many studies have shown that prostanoids are tightly associated with cardiovascular diseases and that several polymorphisms in genes involved in their synthesis and function increase the risk of developing these pathologies. In this review, we focus on molecular mechanisms linking prostanoids to cardiovascular diseases and we provide an overview of genetic polymorphisms that increase the risk for cardiovascular disease.
Collapse
Affiliation(s)
- Livia Beccacece
- Computational Genomics Lab, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
- Correspondence: (L.B.); (P.A.)
| | - Paolo Abondio
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
- Correspondence: (L.B.); (P.A.)
| | - Carla Bini
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Susi Pelotti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Donata Luiselli
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
| |
Collapse
|
7
|
Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis. Int J Mol Sci 2023; 24:ijms24021637. [PMID: 36675152 PMCID: PMC9863938 DOI: 10.3390/ijms24021637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
It is commonly believed that the inactivation of inflammation is mainly due to the decay or cessation of inducers. In reality, in connection with the development of atherosclerosis, spontaneous decay of inducers is not observed. It is now known that lipid mediators originating from polyunsaturated fatty acids (PUFAs), which are important constituents of all cell membranes, can act in the inflamed tissue and bring it to resolution. In fact, PUFAs, such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are precursors to both pro-inflammatory and anti-inflammatory compounds. In this review, we describe the lipid mediators of vascular inflammation and resolution, and their biochemical activity. In addition, we highlight data from the literature that often show a worsening of atherosclerotic disease in subjects deficient in lipid mediators of inflammation resolution, and we also report on the anti-proteasic and anti-thrombotic properties of these same lipid mediators. It should be noted that despite promising data observed in both animal and in vitro studies, contradictory clinical results have been observed for omega-3 PUFAs. Many further studies will be required in order to clarify the observed conflicts, although lifestyle habits such as smoking or other biochemical factors may often influence the normal synthesis of lipid mediators of inflammation resolution.
Collapse
|
8
|
Bothrops moojeni snake venom induces an inflammatory response in preadipocytes: Insights into a new aspect of envenomation. PLoS Negl Trop Dis 2022; 16:e0010658. [PMID: 35939519 PMCID: PMC9359566 DOI: 10.1371/journal.pntd.0010658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 07/14/2022] [Indexed: 11/19/2022] Open
Abstract
Bothrops envenomation is a public health problem in Brazil. Despite the advances in the knowledge of the pathogenesis of systemic and local effects induced by Bothrops venom, the target tissues to this venom are not completely characterised. As preadipocytes are important cells of the adipose tissue and synthesize inflammatory mediators, we investigated the ability of B. moojeni snake venom (Bmv) to stimulate an inflammatory response in 3T3-L1 preadipocytes in vitro, focusing on (1) the release of PGE2, IL-6, TNF-α, MCP-1, KC, leptin and adiponectin; (2) the mechanisms involved in PGE2 release and (3) differentiation of these cells. Cytotoxicity of Bmv was determined by MTT assay. The concentrations of PGE2, cytokines and adipokines were quantified by EIA. Participation of the COX-1 and COX-2 enzymes, NF-κB and PGE2 receptors (EP1-4) was assessed using a pharmacological approach, and protein expression of the COX enzymes and P-NF-κB was analysed by western blotting. Preadipocyte differentiation was quantified by Oil Red O staining. Bmv (1 μg/mL) induced release of PGE2, IL-6 and KC and increased expression of COX-2 in preadipocytes. Basal levels of TNF-α, MCP-1, leptin and adiponectin were not modified. Treatment of cells with SC560 (COX-1 inhibitor) and NS398 (COX-2 inhibitor) inhibited Bmv-induced PGE2 release. Bmv induced phosphorylation of NF-κB, and treatment of the cells with TPCK and SN50, which inhibit distinct NF-κB domains, significantly reduced Bmv-induced PGE2 release, as did the treatment with an antagonist of PGE2 receptor EP1, unlike treatment with antagonists of EP2, EP3 or EP4. Bmv also induced lipid accumulation in differentiating cells. These results demonstrate that Bmv can activate an inflammatory response in preadipocytes by inducing the release of inflammatory mediators; that PGE2 production is mediated by the COX-1, COX-2 and NF-κB pathways; and that engagement of EP1 potentiates PGE2 synthesis via a positive feedback mechanism. Our findings highlight the role of the adipose tissue as another target for Bmv and suggest that it contributes to Bothrops envenomation by producing inflammatory mediators.
Collapse
|
9
|
Wang L, Wu Y, Jia Z, Yu J, Huang S. Roles of EP Receptors in the Regulation of Fluid Balance and Blood Pressure. Front Endocrinol (Lausanne) 2022; 13:875425. [PMID: 35813612 PMCID: PMC9262144 DOI: 10.3389/fendo.2022.875425] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Abstract
Prostaglandin E2 (PGE2) is an important prostanoid expressing throughout the kidney and cardiovascular system. Despite the diverse effects on fluid metabolism and blood pressure, PGE2 is implicated in sustaining volume and hemodynamics homeostasis. PGE2 works through four distinct E-prostanoid (EP) receptors which are G protein-coupled receptors. To date, pharmacological specific antagonists and agonists of all four subtypes of EP receptors and genetic targeting knockout mice for each subtype have helped in uncoupling the diverse functions of PGE2 and discriminating the respective characteristics of each receptor. In this review, we summarized the functions of individual EP receptor subtypes in the renal and blood vessels and the molecular mechanism of PGE2-induced fluid metabolism and blood pressure homeostasis.
Collapse
Affiliation(s)
- Lu Wang
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yiqian Wu
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Yu
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Songming Huang, ; Jing Yu,
| | - Songming Huang
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Songming Huang, ; Jing Yu,
| |
Collapse
|
10
|
Liu N, Tang J, Xue Y, Mok V, Zhang M, Ren X, Wang Y, Fu J. EP3 Receptor Deficiency Improves Vascular Remodeling and Cognitive Impairment in Cerebral Small Vessel Disease. Aging Dis 2022; 13:313-328. [PMID: 35111376 PMCID: PMC8782563 DOI: 10.14336/ad.2021.0627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/27/2021] [Indexed: 11/21/2022] Open
Abstract
Aging and hypertension are major risk factors for cerebral small vessel disease (CSVD). Anti-hypertensive therapy has achieved effective; however, incomplete results in treating CSVD, suggesting the need for additional treatments. Targeting abnormal inflammatory responses has become a topic of research interest. Small artery remodeling is the main pathological feature of CSVD. Inhibition of the E-prostanoid 3 (EP3) receptor has been shown to attenuate vascular remodeling in peripheral organs; however, little is known about its role in CSVD. Therefore, we investigated whether the deletion of EP3 attenuates the development of CSVD in an animal model-- stroke-prone renovascular hypertensive rat (RHRsp). We found that the cerebral small arteries of RHRsp exhibited increased EP3 expression. Despite no alleviation of hypertension, the deletion of EP3 still attenuated the cerebral small artery remodeling of RHRsp, as evidenced by reduced overexpression of extracellular matrix (ECM) in the vessel. In vitro experiments indicated that EP3 deletion regulated the expression of ECM by downregulating TGF-β1/Smad signaling. Furthermore, the Morris water maze test and magnetic resonance test demonstrated that EP3 knockout attenuated cognitive impairment of the RHRsp, possibly through increased cerebral blood flow. Together, our results indicate that the deletion of EP3 attenuates vascular remodeling and vascular cognitive impairment induced by hypertension, and blockade of the EP3 receptor may be a promising strategy for the treatment of CSVD.
Collapse
Affiliation(s)
- Na Liu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jie Tang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Yang Xue
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Vincent Mok
- Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Miaoyi Zhang
- Department of Neurology, North Huashan hospital, Fudan University, No.108 Lu Xiang Road, Shanghai, China.
| | - Xue Ren
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Correspondence should be addressed to: Dr. Jianhui Fu, Huashan Hospital, Fudan University, Shanghai, China. ; Dr. Yilong Wang, Beijing Tiantan Hospital, Capital Medical University, Beijing, China. .
| | - Jianhui Fu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
- Correspondence should be addressed to: Dr. Jianhui Fu, Huashan Hospital, Fudan University, Shanghai, China. ; Dr. Yilong Wang, Beijing Tiantan Hospital, Capital Medical University, Beijing, China. .
| |
Collapse
|
11
|
Bryson TD, Harding P. Prostaglandin E2 EP receptors in cardiovascular disease: An update. Biochem Pharmacol 2021; 195:114858. [PMID: 34822808 DOI: 10.1016/j.bcp.2021.114858] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022]
Abstract
This review article provides an update for the role of prostaglandin E2 receptors (EP1, EP2, EP3 and EP4) in cardiovascular disease. Where possible we have reported citations from the last decade although this was not possible for all of the topics covered due to the paucity of publications. The authors have attempted to cover the subjects of ischemia-reperfusion injury, arrhythmias, hypertension, novel protein binding partners of the EP receptors and their pathophysiological significance, and cardiac regeneration. These latter two topics bring studies of the EP receptors into new and exciting areas of research that are just beginning to be explored. Where there is peer-reviewed literature, the authors have placed particular emphasis on clinical studies although these are limited in number.
Collapse
Affiliation(s)
- Timothy D Bryson
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI, United States; Frankel Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pamela Harding
- Hypertension & Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States.
| |
Collapse
|
12
|
Song CY, Singh P, Motiwala M, Shin JS, Lew J, Dutta SR, Gonzalez FJ, Bonventre JV, Malik KU. 2-Methoxyestradiol Ameliorates Angiotensin II-Induced Hypertension by Inhibiting Cytosolic Phospholipase A 2α Activity in Female Mice. Hypertension 2021; 78:1368-1381. [PMID: 34628937 PMCID: PMC8516072 DOI: 10.1161/hypertensionaha.121.18181] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. We tested the hypothesis that CYP1B1 (cytochrome P450 1B1)-17β-estradiol metabolite 2-methoxyestradiol protects against Ang II (angiotensin II)–induced hypertension by inhibiting group IV cPLA2α (cytosolic phospholipase A2α) activity and production of prohypertensive eicosanoids in female mice. Ang II (700 ng/kg per minute, SC) increased mean arterial blood pressure (BP), systolic and diastolic BP measured by radiotelemetry, renal fibrosis, and reactive oxygen species production in wild-type mice (cPLA2α+/+/Cyp1b1+/+) that were enhanced by ovariectomy and abolished in intact and ovariectomized-cPLA2α−/−/Cyp1b1+/+ mice. Ang II–induced increase in SBP measured by tail-cuff, renal fibrosis, reactive oxygen species production, and cPLA2α activity measured by its phosphorylation in the kidney, and urinary excretion of prostaglandin E2 and thromboxane A2 metabolites were enhanced in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. 2-Methoxyestradiol and arachidonic acid metabolism inhibitor 5,8,11,14-eicosatetraynoic acid attenuated the Ang II–induced increase in SBP, renal fibrosis, reactive oxygen species production, and urinary excretion of prostaglandin E2, and thromboxane A2 metabolites in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. Antagonists of prostaglandin E2 and thromboxane A2 receptors EP1 and EP3 and TP, respectively, inhibited Ang II–induced increases in SBP and reactive oxygen species production and renal fibrosis in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. These data suggest that CYP1B1-generated metabolite 2-methoxyestradiol mitigates Ang II–induced hypertension and renal fibrosis by inhibiting cPLA2α activity, reducing prostaglandin E2, and thromboxane A2 production and stimulating EP1 and EP3 and TP receptors, respectively. Thus, 2-methoxyestradiol and the drugs that selectively block EP1 and EP3 and TP receptors could be useful in treating hypertension and its pathogenesis in females.
Collapse
Affiliation(s)
- Chi Young Song
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Purnima Singh
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Mustafa Motiwala
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Ji Soo Shin
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Jessica Lew
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Shubha R. Dutta
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Joseph V. Bonventre
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.V.B.)
| | - Kafait U. Malik
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| |
Collapse
|
13
|
Liu B, Zhou Y. Endothelium-dependent contraction: The non-classical action of endothelial prostacyclin, its underlying mechanisms, and implications. FASEB J 2021; 35:e21877. [PMID: 34449098 DOI: 10.1096/fj.202101077r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 02/05/2023]
Abstract
Although commonly thought to produce prostacyclin (prostaglandin I2 ; PGI2 ) that evokes vasodilatation and protects vessels from the development of diseases, the endothelial cyclooxygenase (COX)-mediated metabolism has also been found to release substance(s) called endothelium-derived contracting factor(s) (EDCF) that causes endothelium-dependent contraction and implicates in endothelial dysfunction of disease conditions. Various mechanisms have been proposed for the process; however, the major endothelial COX metabolite PGI2 , which has been classically considered to activate the I prostanoid receptor (IP) that mediates vasodilatation and opposes the effects of thromboxane (Tx) A2 produced by COX in platelets, emerges as a major EDCF in health and disease conditions. Our recent studies from genetically altered mice further suggest that vasomotor reactions to PGI2 are collectively modulated by IP, the vasoconstrictor Tx-prostanoid receptor (TP; the prototype receptor of TxA2 ) and E prostanoid receptor-3 (EP3; a vasoconstrictor receptor of PGE2 ) although with differences in potency and efficacy; a contraction to PGI2 reflects activities of TP and/or EP3 outweighing that of the concurrently activated IP. Here, we discuss the history of endothelium-dependent contraction, evidences that support the above hypothesis, proposed mechanisms for the varied reactions to endothelial PGI2 synthesis as well as the relation of its dilator activity to the effect of another NO-independent vasodilator mechanism, the endothelium-derived hyperpolarizing factor. Also, we address the possible pathological and therapeutic implications as well as questions remaining to be resolved or limitations of our above findings obtained from genetically altered mouse models.
Collapse
Affiliation(s)
- Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| |
Collapse
|
14
|
Singh P, Song CY, Dutta SR, Pingili A, Shin JS, Gonzalez FJ, Bonventre JV, Malik KU. 6β-Hydroxytestosterone Promotes Angiotensin II-Induced Hypertension via Enhanced Cytosolic Phospholipase A 2α Activity. Hypertension 2021; 78:1053-1066. [PMID: 34420370 PMCID: PMC8415516 DOI: 10.1161/hypertensionaha.121.17927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Supplemental Digital Content is available in the text. This study was conducted to test the hypothesis that the CYP1B1 (cytochrome P450 1B1)-testosterone metabolite 6β-hydroxytestosterone contributes to angiotensin II-induced hypertension by promoting activation of group IV cPLA2α (cytosolic phospholipase A2α) and generation of prohypertensive eicosanoids in male mice. Eight-week-old male intact or orchidectomized cPLA2α+/+/Cyp1b1+/+ and cPLA2α–/–/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1–/– mice were infused with angiotensin II (700 ng/kg/min, subcutaneous) for 2 weeks and injected with 6β-hydroxytestosterone (15 μg/g/every third day, intraperitoneal). Systolic blood pressure was measured by tail-cuff and confirmed by radiotelemetry. Angiotensin II-induced increase in systolic blood pressure, cardiac and renal collagen deposition, and reactive oxygen species production were reduced by disruption of the cPLA2α or Cyp1b1 genes or by administration of the arachidonic acid metabolism inhibitor 5,8,11,14-eicosatetraynoic acid to cPLA2α+/+/Cyp1b1+/+ mice. 6β-hydroxytestosterone treatment restored these effects of angiotensin II in cPLA2α+/+/Cyp1b1–/– mice but not in orchidectomized cPLA2α–/–/Cyp1b1+/+ mice, which were lowered by 5,8,11,14-eicosatetraynoic acid in cPLA2α+/+/Cyp1b1–/– mice. Antagonists of prostaglandin E2-EP1/EP3 receptors and thromboxane A2-TP receptors decreased the effect of 6β-hydroxytestosterone in restoring the angiotensin II-induced increase in systolic blood pressure, cardiac and renal collagen deposition, and reactive oxygen species production in cPLA2α+/+/Cyp1b1–/– mice. These data suggest that 6β-hydroxytestosterone promotes angiotensin II-induced increase in systolic blood pressure and associated pathogenesis via cPLA2α activation and generation of eicosanoids, most likely prostaglandin E2 and thromboxane A2 that exerts prohypertensive effects by stimulating EP1/EP3 and TP receptors, respectively. Therefore, agents that selectively block these receptors could be useful in treating testosterone exacerbated angiotensin II-induced hypertension and its pathogenesis.
Collapse
Affiliation(s)
- Purnima Singh
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Chi Young Song
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Shubha R Dutta
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Ajeeth Pingili
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Ji Soo Shin
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Kafait U Malik
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| |
Collapse
|
15
|
Shen Y, Wang X, Yuan R, Pan X, Yang X, Cai J, Li Y, Yin A, Xiao Q, Ji Q, Li Y, He B, Shen L. Prostaglandin E1 attenuates AngII-induced cardiac hypertrophy via EP3 receptor activation and Netrin-1upregulation. J Mol Cell Cardiol 2021; 159:91-104. [PMID: 34147480 DOI: 10.1016/j.yjmcc.2021.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/27/2021] [Accepted: 06/13/2021] [Indexed: 01/09/2023]
Abstract
AIMS Pathological cardiac hypertrophy induced by activation of the renin-angiotensin-aldosterone system (RAAS) is one of the leading causes of heart failure. However, in current clinical practice, the strategy for targeting the RAAS is not sufficient to reverse hypertrophy. Here, we investigated the effect of prostaglandin E1 (PGE1) on angiotensin II (AngII)-induced cardiac hypertrophy and potential molecular mechanisms underlying the effect. METHODS AND RESULTS Adult male C57 mice were continuously infused with AngII or saline and treated daily with PGE1 or vehicle for two weeks. Neonatal rat cardiomyocytes were cultured to detect AngII-induced hypertrophic responses. We found that PGE1 ameliorated AngII-induced cardiac hypertrophy both in vivo and in vitro. The RNA sequencing (RNA-seq) and expression pattern analysis results suggest that Netrin-1 (Ntn1) is the specific target gene of PGE1. The protective effect of PGE1 was eliminated after knockdown of Ntn1. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the PGE1-mediated signaling pathway changes are associated with the mitogen-activated protein kinase (MAPK) pathway. PGE1 suppressed AngII-induced activation of the MAPK signaling pathway, and such an effect was attenuated by Ntn1 knockdown. Blockade of MAPK signaling rescued the phenotype of cardiomyocytes caused by Ntn1 knockdown, indicating that MAPK signaling may act as the downstream effector of Ntn1. Furthermore, inhibition of the E-prostanoid (EP) 3 receptor, as opposed to the EP1, EP2, or EP4 receptor, in cardiomyocytes reversed the effect of PGE1, and activation of EP3 by sulprostone, a specific agonist, mimicked the effect of PGE1. CONCLUSION In conclusion, PGE1 ameliorates AngII-induced cardiac hypertrophy through activation of the EP3 receptor and upregulation of Ntn1, which inhibits the downstream MAPK signaling pathway. Thus, targeting EP3, as well as the Ntn1-MAPK axis, may represent a novel approach for treating pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Yejiao Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ruosen Yuan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Pan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxiao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiali Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqing Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqi Ji
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanjie Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
16
|
Wang FF, Ba J, Yu XJ, Shi XL, Liu JJ, Liu KL, Fu LY, Su Q, Li HB, Kang KB, Yi QY, Wang SQ, Gao HL, Qi J, Li Y, Zhu GQ, Kang YM. Central Blockade of E-Prostanoid 3 Receptor Ameliorated Hypertension Partially by Attenuating Oxidative Stress and Inflammation in the Hypothalamic Paraventricular Nucleus of Spontaneously Hypertensive Rats. Cardiovasc Toxicol 2021; 21:286-300. [PMID: 33165770 DOI: 10.1007/s12012-020-09619-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/24/2020] [Indexed: 12/27/2022]
Abstract
Hypertension, as one of the major risk factors for cardiovascular disease, significantly affects human health. Prostaglandin E2 (PGE2) and the E3-class prostanoid (EP3) receptor have previously been demonstrated to modulate blood pressure and hemodynamics in various animal models of hypertension. The PGE2-evoked pressor and biochemical responses can be blocked with the EP3 receptor antagonist, L-798106 (N-[(5-bromo-2methoxyphenyl)sulfonyl]-3-[2-(2-naphthalenylmethyl) phenyl]-2-propenamide). In the hypothalamic paraventricular nucleus (PVN), sympathetic excitation can be introduced by PGE2, which can activate EP3 receptors located in the PVN. In such a case, the central knockdown of EP3 receptor can be considered as a potential therapeutic modality for hypertension management. The present study examined the efficacy of the PVN infusion of L-798106, by performing experiments on spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto rats (WKYs). The rats were administered with chronic bilateral PVN infusion of L-798106 (10 μg/day) or the vehicle for 28 days. The results indicated that the SHRs had a higher mean arterial pressure (MAP), an increased Fra-like (Fra-LI) activity in the PVN, as well as a higher expression of gp91phox, mitogen-activated protein kinase (MAPK), and proinflammatory cytokines in the PVN compared with the WKYs. Additionally, the expression of Cu/Zn-SOD in the PVN of the SHRs was reduced compared with the WKYs. The bilateral PVN infusion of L-798106 significantly reduced MAP, as well as plasma norepinephrine (NE) levels in the SHRs. It also inhibited Fra-LI activity and reduced the expression of gp91phox, proinflammatory cytokines, and MAPK, whereas it increased the expression of Cu/Zn-SOD in the PVN of SHRs. In addition, L-798106 restored the balance of the neurotransmitters in the PVN. On the whole, the findings of the present study demonstrate that the PVN blockade of EP3 receptor can ameliorate hypertension and cardiac hypertrophy partially by attenuating ROS and proinflammatory cytokines, and modulating neurotransmitters in the PVN.
Collapse
Affiliation(s)
- Fang-Fang Wang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Functional Medicine, School of Basic Medical Sciences, Jiamusi University, Jiamusi, 154007, China
| | - Juan Ba
- Department of Anesthesiology, Center for Brian Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiao-Lian Shi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jin-Jun Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Kai-Li Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li-Yan Fu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qing Su
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Kai B Kang
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Qiu-Yue Yi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shu-Qiu Wang
- Department of Functional Medicine, School of Basic Medical Sciences, Jiamusi University, Jiamusi, 154007, China
| | - Hong-Li Gao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jie Qi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ying Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine; Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
17
|
Zhang Y, Luo W, Li H, Yu G, Luo H, Leng J, Ge J, Zeng R, Guo T, Yin Y, Zhou Y, Liu B. Larger endothelium-dependent contractions in iliac arteries of adult SHRs are attributed to differential downregulation of TP and EP3 receptors in the vessels of WKYs and SHRs during the transition from adolescence to adulthood. Eur J Pharmacol 2021; 893:173828. [PMID: 33347824 DOI: 10.1016/j.ejphar.2020.173828] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 02/05/2023]
Abstract
This study was to determine how endothelium-dependent contractions (EDCs) change in iliac arteries of Wistar-Kyoto (WKYs) and spontaneously hypertensive rats (SHRs) during the transition from adolescence to adulthood and the underlying mechanism(s). We also aimed to elucidate effects of L-798106, an EP3 receptor antagonist, on EDCs and the blood pressure increase in adolescent SHRs. Blood vessels were isolated for functional and biochemical analyses. EDCs were comparable in adolescent iliac arteries of both strains, and contractions to ACh, prostacyclin (PGI2), the EP3 receptor agonist sulprostone and the TP receptor agonist U46619 in adult vessels were less prominent compared with those in the adolescents, while the attenuation of vasoconstrictions to ACh, PGI2 or U46619 with age was to a lesser extent in SHRs. PGI2 production was decreased to a similar level in adult arteries. TP and EP3 expressions were downregulated in adult vessels, whereas the extent of TP downregulation was less in SHRs. L-798106 partially suppressed the vasoconstrictions to U46619 and attenuated EDCs to a greater extent than SQ29548, and administration of L-798106 blunted the blood pressure increase with age in prehypertensive SHRs. These results demonstrate the comparable EDCs in iliac arteries of the adolescents are decreased in the adults, but relatively larger EDCs in adult SHRs can be a reflection of differential downregulation of TP and EP3 receptors during the transition from adolescence to adulthood. Also, our data suggest that blockade of both TP and EP3 receptors starting from the prehypertensive stage suppresses EDCs and the development of hypertension in SHRs.
Collapse
MESH Headings
- Age Factors
- Animals
- Antihypertensive Agents/pharmacology
- Blood Pressure/drug effects
- Disease Models, Animal
- Down-Regulation
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Hypertension/prevention & control
- Iliac Artery/metabolism
- Iliac Artery/physiopathology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Rats, Inbred WKY
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Thromboxane/antagonists & inhibitors
- Receptors, Thromboxane/genetics
- Receptors, Thromboxane/metabolism
- Signal Transduction
- Vasoconstriction/drug effects
- Rats
Collapse
Affiliation(s)
- Yingzhan Zhang
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Wenhong Luo
- Bio-analytical Laboratory, Shantou University Medical College, Shantou, China
| | - Hui Li
- Bio-analytical Laboratory, Shantou University Medical College, Shantou, China
| | - Gang Yu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Hongjun Luo
- Bio-analytical Laboratory, Shantou University Medical College, Shantou, China
| | - Jing Leng
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Jiahui Ge
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Ruhui Zeng
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Tingting Guo
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yehu Yin
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China.
| | - Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China.
| |
Collapse
|
18
|
Schaid MD, Green CL, Peter DC, Gallagher SJ, Guthery E, Carbajal KA, Harrington JM, Kelly GM, Reuter A, Wehner ML, Brill AL, Neuman JC, Lamming DW, Kimple ME. Agonist-independent Gα z activity negatively regulates beta-cell compensation in a diet-induced obesity model of type 2 diabetes. J Biol Chem 2020; 296:100056. [PMID: 33172888 PMCID: PMC7948463 DOI: 10.1074/jbc.ra120.015585] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
The inhibitory G protein alpha-subunit (Gαz) is an important modulator of beta-cell function. Full-body Gαz-null mice are protected from hyperglycemia and glucose intolerance after long-term high-fat diet (HFD) feeding. In this study, at a time point in the feeding regimen where WT mice are only mildly glucose intolerant, transcriptomics analyses reveal islets from HFD-fed Gαz KO mice have a dramatically altered gene expression pattern as compared with WT HFD-fed mice, with entire gene pathways not only being more strongly upregulated or downregulated versus control-diet fed groups but actually reversed in direction. Genes involved in the “pancreatic secretion” pathway are the most strongly differentially regulated: a finding that correlates with enhanced islet insulin secretion and decreased glucagon secretion at the study end. The protection of Gαz-null mice from HFD-induced diabetes is beta-cell autonomous, as beta cell–specific Gαz-null mice phenocopy the full-body KOs. The glucose-stimulated and incretin-potentiated insulin secretion response of islets from HFD-fed beta cell–specific Gαz-null mice is significantly improved as compared with islets from HFD-fed WT controls, which, along with no impact of Gαz loss or HFD feeding on beta-cell proliferation or surrogates of beta-cell mass, supports a secretion-specific mechanism. Gαz is coupled to the prostaglandin EP3 receptor in pancreatic beta cells. We confirm the EP3γ splice variant has both constitutive and agonist-sensitive activity to inhibit cAMP production and downstream beta-cell function, with both activities being dependent on the presence of beta-cell Gαz.
Collapse
Affiliation(s)
- Michael D Schaid
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Cara L Green
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Darby C Peter
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Shannon J Gallagher
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Erin Guthery
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kathryn A Carbajal
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jeffrey M Harrington
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Grant M Kelly
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Austin Reuter
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Molly L Wehner
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Allison L Brill
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Joshua C Neuman
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Dudley W Lamming
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Michelle E Kimple
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Cell and Regenerative Biology, University of Wisconsin- Madison School of Medicine and Public Health, Madison, Wisconsin, USA.
| |
Collapse
|
19
|
A Representative GIIA Phospholipase A 2 Activates Preadipocytes to Produce Inflammatory Mediators Implicated in Obesity Development. Biomolecules 2020; 10:biom10121593. [PMID: 33255269 PMCID: PMC7760919 DOI: 10.3390/biom10121593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 12/27/2022] Open
Abstract
Adipose tissue secretes proinflammatory mediators which promote systemic and adipose tissue inflammation seen in obesity. Group IIA (GIIA)-secreted phospholipase A2 (sPLA2) enzymes are found to be elevated in plasma and adipose tissue from obese patients and are active during inflammation, generating proinflammatory mediators, including prostaglandin E2 (PGE2). PGE2 exerts anti-lipolytic actions and increases triacylglycerol levels in adipose tissue. However, the inflammatory actions of GIIA sPLA2s in adipose tissue cells and mechanisms leading to increased PGE2 levels in these cells are unclear. This study investigates the ability of a representative GIIA sPLA2, MT-III, to activate proinflammatory responses in preadipocytes, focusing on the biosynthesis of prostaglandins, adipocytokines and mechanisms involved in these effects. Our results showed that MT-III induced biosynthesis of PGE2, PGI2, MCP-1, IL-6 and gene expression of leptin and adiponectin in preadipocytes. The MT-III-induced PGE2 biosynthesis was dependent on cytosolic PLA2 (cPLA2)-α, cyclooxygenases (COX)-1 and COX-2 pathways and regulated by a positive loop via the EP4 receptor. Moreover, MT-III upregulated COX-2 and microsomal prostaglandin synthase (mPGES)-1 protein expression. MCP-1 biosynthesis induced by MT-III was dependent on the EP4 receptor, while IL-6 biosynthesis was dependent on EP3 receptor engagement by PGE2. These data highlight preadipocytes as targets for GIIA sPLA2s and provide insight into the roles played by this group of sPLA2s in obesity.
Collapse
|
20
|
Xu H, Fang B, Du S, Wang S, Li Q, Jia X, Bao C, Ye L, Sui X, Qian L, Luan Z, Yang G, Zheng F, Wang N, Chen L, Zhang X, Guan Y. Endothelial cell prostaglandin E2 receptor EP4 is essential for blood pressure homeostasis. JCI Insight 2020; 5:138505. [PMID: 32641583 DOI: 10.1172/jci.insight.138505] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/03/2020] [Indexed: 01/07/2023] Open
Abstract
Prostaglandin E2 and its cognate EP1-4 receptors play important roles in blood pressure (BP) regulation. Herein, we show that endothelial cell-specific (EC-specific) EP4 gene-knockout mice (EC-EP4-/-) exhibited elevated, while EC-specific EP4-overexpression mice (EC-hEP4OE) displayed reduced, BP levels compared with the control mice under both basal and high-salt diet-fed conditions. The altered BP was completely abolished by treatment with l-NG-nitro-l-arginine methyl ester (l-NAME), a competitive inhibitor of endothelial nitric oxide synthase (eNOS). The mesenteric arteries of the EC-EP4-/- mice showed increased vasoconstrictive response to angiotensin II and reduced vasorelaxant response to acetylcholine, both of which were eliminated by l-NAME. Furthermore, EP4 activation significantly reduced BP levels in hypertensive rats. Mechanistically, EP4 deletion markedly decreased NO contents in blood vessels via reducing eNOS phosphorylation at Ser1177. EP4 enhanced NO production mainly through the AMPK pathway in cultured ECs. Collectively, our findings demonstrate that endothelial EP4 is essential for BP homeostasis.
Collapse
Affiliation(s)
- Hu Xu
- Advanced Institute for Medical Sciences and.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Liaoning Engineering and Technology Research Center of Nuclear Receptors and Major Metabolic Diseases, Dalian, China
| | | | - Shengnan Du
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | | | - Qingwei Li
- Advanced Institute for Medical Sciences and
| | - Xiao Jia
- Advanced Institute for Medical Sciences and
| | | | - Lan Ye
- Advanced Institute for Medical Sciences and
| | - Xue Sui
- Advanced Institute for Medical Sciences and
| | - Lei Qian
- Advanced Institute for Medical Sciences and
| | | | - Guangrui Yang
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Feng Zheng
- Advanced Institute for Medical Sciences and.,Liaoning Engineering and Technology Research Center of Nuclear Receptors and Major Metabolic Diseases, Dalian, China
| | - Nanping Wang
- Advanced Institute for Medical Sciences and.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Liaoning Engineering and Technology Research Center of Nuclear Receptors and Major Metabolic Diseases, Dalian, China
| | - Lihong Chen
- Advanced Institute for Medical Sciences and.,Liaoning Engineering and Technology Research Center of Nuclear Receptors and Major Metabolic Diseases, Dalian, China
| | - Xiaoyan Zhang
- Advanced Institute for Medical Sciences and.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Liaoning Engineering and Technology Research Center of Nuclear Receptors and Major Metabolic Diseases, Dalian, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences and.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Liaoning Engineering and Technology Research Center of Nuclear Receptors and Major Metabolic Diseases, Dalian, China
| |
Collapse
|
21
|
Zhu L, Zhang Y, Guo Z, Wang M. Cardiovascular Biology of Prostanoids and Drug Discovery. Arterioscler Thromb Vasc Biol 2020; 40:1454-1463. [PMID: 32295420 DOI: 10.1161/atvbaha.119.313234] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Prostanoids are a group of bioactive lipids that are synthesized de novo from membrane phospholipid-released arachidonic acid and have diverse functions in normal physiology and disease. NSAIDs (non-steroidal anti-inflammatory drugs), which are among the most commonly used medications, ameliorate pain, fever, and inflammation by inhibiting COX (cyclooxygenase), which is the rate-limiting enzyme in the biosynthetic cascade of prostanoids. The use of NSAIDs selective for COX-2 inhibition increases the risk of a thrombotic event (eg, myocardial infarction and stroke). All NSAIDs are associated with an increased risk of heart failure. Substantial variation in clinical responses to aspirin exists and is associated with cardiovascular risk. Limited clinical studies suggest the involvement of prostanoids in vascular restenosis in patients who received angioplasty intervention. mPGES (microsomal PG [prostaglandin] E synthase)-1, an alternative target downstream of COX, has the potential to be therapeutically targeted for inflammatory disease, with diminished thrombotic risk relative to selective COX-2 inhibitors. mPGES-1-derived PGE2 critically regulates microcirculation via its receptor EP (receptor for prostanoid E) 4. This review summarizes the actions and associated mechanisms for modulating the biosynthesis of prostanoids in thrombosis, vascular remodeling, and ischemic heart disease as well as their therapeutic relevance.
Collapse
Affiliation(s)
- Liyuan Zhu
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yuze Zhang
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Ziyi Guo
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Miao Wang
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing.,Clinical Pharmacology Center (M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| |
Collapse
|
22
|
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: 2.6] [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
|
23
|
Liu B, Wu X, Zeng R, Yin Y, Guo T, Xu Y, Zhang Y, Leng J, Ge J, Yu G, Guo J, Zhou Y. Prostaglandin E 2 sequentially activates E-prostanoid receptor-3 and thromboxane prostanoid receptor to evoke contraction and increase in resistance of the mouse renal vasculature. FASEB J 2020; 34:2568-2578. [PMID: 31908041 DOI: 10.1096/fj.201901611r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/21/2019] [Accepted: 12/04/2019] [Indexed: 02/05/2023]
Abstract
Although recognized to have an in vivo vasodepressor effect blunted by the vasoconstrictor effect of E-prostanoid receptor-3 (EP3), prostaglandin E2 (PGE2 ) evokes contractions of many vascular beds that are sensitive to antagonizing the thromboxane prostanoid receptor (TP). This study aimed to determine the direct effect of PGE2 on renal arteries and/or the whole renal vasculature and how each of these two receptors is involved in the responses. Experiments were performed on isolated vessels and perfused kidneys of wild-type mice and/or mice with deficiency in TP (TP-/- ), EP3 (EP3-/- ), or both TP and EP3 (TP-/- /EP3-/- ). Here we show that PGE2 (0.001-30 μM) evoked not only contraction of main renal arteries, but also a decrease of flow in perfused kidneys. EP3-/- diminished the response to 0.001-0.3 μM PGE2 , while TP-/- reduced that to the prostanoid of higher concentrations. In TP-/- /EP3-/- vessels and perfused kidneys, PGE2 did not evoke contraction but instead resulted in vasodilator responses. These results demonstrate that PGE2 functions as an overall direct vasoconstrictor of the mouse renal vasculature with an effect reflecting the vasoconstrictor activities outweighing that of dilation. Also, our results suggest that EP3 dominates the vasoconstrictor effect of PGE2 of low concentrations (≤0.001-0.3 μM), but its effect is further added by that of TP, which has a higher efficacy, although activated by higher concentrations (from 0.01 μM) of the same prostanoid PGE2 .
Collapse
Affiliation(s)
- Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Xiangzhong Wu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Ruhui Zeng
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
- Department of Gynaecology and Obstetrics, First Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yehu Yin
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Tingting Guo
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yineng Xu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingzhan Zhang
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Jing Leng
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Jiahui Ge
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Gang Yu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Jinwei Guo
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| |
Collapse
|
24
|
Xiao L, Itani HA, do Carmo LS, Carver LS, Breyer RM, Harrison DG. Central EP3 (E Prostanoid 3) Receptors Mediate Salt-Sensitive Hypertension and Immune Activation. Hypertension 2019; 74:1507-1515. [PMID: 31679420 PMCID: PMC7040566 DOI: 10.1161/hypertensionaha.119.13850] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We recently identified a pathway underlying immune activation in hypertension. Proteins oxidatively modified by reactive isoLG (isolevuglandin) accumulate in dendritic cells (DCs). PGE2 (Prostaglandin E2) has been implicated in the inflammation associated with hypertension. We hypothesized that PGE2 via its EP (E prostanoid) 3 receptor contributes to DC activation in hypertension. EP3-/- mice and wild-type littermates were exposed to sequential hypertensive stimuli involving an initial 2-week exposure to the nitric oxide synthase inhibitor Nω-nitro-L-arginine methyl ester hydrochloride in drinking water, followed by a 2-week washout period, and a subsequent 4% high-salt diet for 3 weeks. In wild-type mice, this protocol increased systolic pressure from 123±2 to 148±8 mm Hg (P<0.05). This was associated with marked renal inflammation and a striking accumulation of isoLG adducts in splenic DCs. However, the increases in blood pressure, renal T-cell infiltration, and DC isoLG formation were completely prevented in EP3-/- mice. Similar protective effects were also observed in wild-type mice that received intracerebroventricular injection of a lentiviral vector encoding shRNA targeting the EP3 receptor. Further, in vitro experiments indicated that PGE2 also acts directly on DCs via its EP1 receptors to stimulate intracellular isoLG formation. Together, these findings provide new insight into how EP receptors in both the central nervous system and peripherally on DCs promote inflammation in salt-induced hypertension.
Collapse
Affiliation(s)
- Liang Xiao
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Hana A Itani
- Department of Pharmacology and Toxicology, American University of Beirut, Lebanon (H.A.I.)
| | - Luciana Simao do Carmo
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Lucas S Carver
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Richard M Breyer
- Division of Nephrology and Hypertension (R.M.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - David G Harrison
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
25
|
Ceddia RP, Downey JD, Morrison RD, Kraemer MP, Davis SE, Wu J, Lindsley CW, Yin H, Daniels JS, Breyer RM. The effect of the EP3 antagonist DG-041 on male mice with diet-induced obesity. Prostaglandins Other Lipid Mediat 2019; 144:106353. [PMID: 31276827 DOI: 10.1016/j.prostaglandins.2019.106353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIMS The prostaglandin E2 (PGE2) EP3 receptor has a multifaceted role in metabolism. Drugs targeting EP3 have been proposed as therapeutics for diabetes; however, studies utilizing global EP3 knockout mice suggest that EP3 blockade increases obesity and insulin resistance. The present studies attempt to determine the effect of acute EP3 antagonist treatment on the diabetic phenotype. METHODS DG-041 was confirmed to be a high affinity antagonist at the mouse EP3 receptor by competition radioligand binding and by blockade of EP3-mediated responses. DG-041 pharmacokinetic studies were performed to determine the most efficacious route of administration. Male C57BL/6 × BALB/c (CB6F1) mice were fed diets containing 10%, 45%, or 60% calories from fat to induce obesity. Changes to the metabolic phenotype in these mice were evaluated after one week treatment with DG-041. RESULTS Subcutaneous injections of DG-041 at 20 mg/kg blocked the sulprostone-evoked rise in mean arterial pressure confirming the efficacy of this administration regime. Seven day treatment with DG-041 had minimal effect on body composition or glycemic control. DG-041 administration caused a reduction in skeletal muscle triglyceride content while showing a trend toward increased hepatic triglycerides. CONCLUSION Short term EP3 administration of DG-041 produced effective blockade of the EP3 receptor and decreased skeletal muscle triglyceride content but had no significant effects on the diabetic phenotype.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jason D Downey
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ryan D Morrison
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Maria P Kraemer
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sarah E Davis
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jing Wu
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Huiyong Yin
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - J Scott Daniels
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Richard M Breyer
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, TN 37232, USA; Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
26
|
VSMC-specific EP4 deletion exacerbates angiotensin II-induced aortic dissection by increasing vascular inflammation and blood pressure. Proc Natl Acad Sci U S A 2019; 116:8457-8462. [PMID: 30948641 DOI: 10.1073/pnas.1902119116] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Prostaglandin E2 (PGE2) plays an important role in vascular homeostasis. Its receptor, E-prostanoid receptor 4 (EP4) is essential for physiological remodeling of the ductus arteriosus (DA). However, the role of EP4 in pathological vascular remodeling remains largely unknown. We found that chronic angiotensin II (AngII) infusion of mice with vascular smooth muscle cell (VSMC)-specific EP4 gene knockout (VSMC-EP4-/-) frequently developed aortic dissection (AD) with severe elastic fiber degradation and VSMC dedifferentiation. AngII-infused VSMC-EP4-/- mice also displayed more profound vascular inflammation with increased monocyte chemoattractant protein-1 (MCP-1) expression, macrophage infiltration, matrix metalloproteinase-2 and -9 (MMP2/9) levels, NADPH oxidase 1 (NOX1) activity, and reactive oxygen species production. In addition, VSMC-EP4-/- mice exhibited higher blood pressure under basal and AngII-infused conditions. Ex vivo and in vitro studies further revealed that VSMC-specific EP4 gene deficiency significantly increased AngII-elicited vasoconstriction of the mesenteric artery, likely by stimulating intracellular calcium release in VSMCs. Furthermore, EP4 gene ablation and EP4 blockade in cultured VSMCs were associated with a significant increase in MCP-1 and NOX1 expression and a marked reduction in α-SM actin (α-SMA), SM22α, and SM differentiation marker genes myosin heavy chain (SMMHC) levels and serum response factor (SRF) transcriptional activity. To summarize, the present study demonstrates that VSMC EP4 is critical for vascular homeostasis, and its dysfunction exacerbates AngII-induced pathological vascular remodeling. EP4 may therefore represent a potential therapeutic target for the treatment of AD.
Collapse
|
27
|
Li L, Lv Y, Yan D. Inhibition of Ep3 attenuates migration and promotes apoptosis of non-small cell lung cancer cells via suppression of TGF-β/Smad signaling. Oncol Lett 2018; 16:5645-5654. [PMID: 30344720 PMCID: PMC6176252 DOI: 10.3892/ol.2018.9391] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/19/2018] [Indexed: 01/05/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common cause of cancer-associated mortality worldwide. Prostaglandin E2 (PGE2) regulates various biological processes, including invasion, proliferation and apoptosis. E-prostanoid 3 (Ep3) is a PGE2 receptor, and the functional role of Ep3 in the progression of NSCLC remains unresolved. The present study investigated the effects of Ep3 in A549 cells and explored the underlying molecular mechanisms. The results revealed that the mRNA and protein expression levels of Ep3 were significantly upregulated in NSCLC tissues and A549 cells. Pharmacological inhibition of Ep3 or RNA interference against Ep3 attenuated the cell viability, migration and invasion, and promoted apoptosis in A549 cells. Ep3 deficiency also decreased the expression of transforming growth factor (TGF)-β, phosphorylated (p)-Smad2 and p-Smad3. The transfection of TGF-β overexpression plasmids reversed the effects of Ep3 deficiency on the cell viability and apoptosis in A549 cells. Finally, an in vivo experiment revealed that Ep3-siRNA transfection strongly reduced the tumor growth and tumor volume. The Ep3-siRNA transfection also inhibited tumor metastasis via suppression of the expression of metastasis-associated proteins. Taken together, these findings indicate that inhibition of Ep3 attenuates the viability and migration, and promotes the apoptosis of NSCLC through suppression of the TGF-β/Smad signaling pathway. Targeting of the Ep3/TGF-β/Smad signaling pathway may be a novel therapeutic strategy for the prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Lei Li
- Department of Respiration, Zhoukou Central Hospital, Zhoukou, Henan 466000, P.R. China
| | - Yanping Lv
- Department of Respiration, Zhoukou Central Hospital, Zhoukou, Henan 466000, P.R. China
| | - Dengfeng Yan
- Department of Respiration, Zhoukou Central Hospital, Zhoukou, Henan 466000, P.R. China
| |
Collapse
|
28
|
Avendaño MS, García-Redondo AB, Zalba G, González-Amor M, Aguado A, Martínez-Revelles S, Beltrán LM, Camacho M, Cachofeiro V, Alonso MJ, Salaices M, Briones AM. mPGES-1 (Microsomal Prostaglandin E Synthase-1) Mediates Vascular Dysfunction in Hypertension Through Oxidative Stress. Hypertension 2018; 72:492-502. [PMID: 29891646 DOI: 10.1161/hypertensionaha.118.10833] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/28/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022]
Abstract
mPGES-1 (microsomal prostaglandin E synthase-1), the downstream enzyme responsible for PGE2 (prostaglandin E2) synthesis in inflammatory conditions and oxidative stress are increased in vessels from hypertensive animals. We evaluated the role of mPGES-1-derived PGE2 in the vascular dysfunction and remodeling in hypertension and the possible contribution of oxidative stress. We used human peripheral blood mononuclear cells from asymptomatic patients, arteries from untreated and Ang II (angiotensin II)-infused mPGES-1-/- and mPGES-1+/+ mice, and vascular smooth muscle cells exposed to PGE2 In human cells, we found a positive correlation between mPGES-1 mRNA and carotid intima-media thickness (r=0.637; P<0.001) and with NADPH oxidase-dependent superoxide production (r=0.417; P<0.001). In Ang II-infused mice, mPGES-1 deletion prevented all of the following: (1) the augmented wall:lumen ratio, vascular stiffness, and altered elastin structure; (2) the increased gene expression of profibrotic and proinflammatory markers; (3) the increased vasoconstrictor responses and endothelial dysfunction; (4) the increased NADPH oxidase activity and the diminished mitochondrial membrane potential; and (5) the increased reactive oxygen species generation and reduced NO bioavailability. In vascular smooth muscle cells or aortic segments, PGE2 increased NADPH oxidase expression and activity and reduced mitochondrial membrane potential, effects that were abolished by antagonists of the PGE2 receptors (EP), EP1 and EP3, and by JNK (c-Jun N-terminal kinase) and ERK1/2 (extracellular-signal-regulated kinases 1/2) inhibition. Deletion of mPGES-1 augmented vascular production of PGI2 suggesting rediversion of the accumulated PGH2 substrate. In conclusion, mPGES-1-derived PGE2 is involved in vascular remodeling, stiffness, and endothelial dysfunction in hypertension likely through an increase of oxidative stress produced by NADPH oxidase and mitochondria.
Collapse
Affiliation(s)
- María S Avendaño
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.)
| | - Ana B García-Redondo
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.).,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| | - Guillermo Zalba
- Departamento de Bioquímica y Genética, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain (G.Z.)
| | - María González-Amor
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.)
| | - Andrea Aguado
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.)
| | - Sonia Martínez-Revelles
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.).,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| | - Luis M Beltrán
- Unidad Clínico-Experimental de Riesgo Vascular-Medicina Interna, Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, Spain (L.M.B.)
| | - Mercedes Camacho
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.).,Laboratorio de Angiología, Biología Vascular e Inflamación, Instituto de Investigación Biomédica Sant Pau, Barcelona, Spain (M.C.)
| | - Victoria Cachofeiro
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.).,Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Gregorio Marañón, Universidad Complutense de Madrid, Spain (V.C.)
| | - María J Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.).,Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain (M.J.A.)
| | - Mercedes Salaices
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.).,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| | - Ana M Briones
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.) .,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| |
Collapse
|
29
|
Song CY, Khan NS, Liao FF, Wang B, Shin JS, Bonventre JV, Malik KU. Brain Cytosolic Phospholipase A2α Mediates Angiotensin II-Induced Hypertension and Reactive Oxygen Species Production in Male Mice. Am J Hypertens 2018; 31:622-629. [PMID: 29342227 PMCID: PMC5905655 DOI: 10.1093/ajh/hpy009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/09/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Recently, we reported that angiotensin II (Ang II)-induced hypertension is mediated by group IV cytosolic phospholipase A2α (cPLA2α) via production of prohypertensive eicosanoids. Since Ang II increases blood pressure (BP) via its action in the subfornical organ (SFO), it led us to investigate the expression and possible contribution of cPLA2α to oxidative stress and development of hypertension in this brain area. METHODS Adenovirus (Ad)-green fluorescence protein (GFP) cPLA2α short hairpin (sh) RNA (Ad-cPLA2α shRNA) and its control Ad-scrambled shRNA (Ad-Scr shRNA) or Ad-enhanced cyan fluorescence protein cPLA2α DNA (Ad-cPLA2α DNA) and its control Ad-GFP DNA were transduced into SFO of cPLA2α+/+ and cPLA2α−/− male mice, respectively. Ang II (700 ng/kg/min) was infused for 14 days in these mice, and BP was measured by tail-cuff and radio telemetry. cPLA2 activity, reactive oxygen species production, and endoplasmic reticulum stress were measured in the SFO. RESULTS Transduction of SFO with Ad-cPLA2α shRNA, but not Ad-Scr shRNA in cPLA2α+/+ mice, minimized expression of cPLA2α, Ang II-induced cPLA2α activity and oxidative stress in the SFO, BP, and cardiac and renal fibrosis. In contrast, Ad-cPLA2α DNA, but not its control Ad-GFP DNA in cPLA2α−/− mice, restored the expression of cPLA2α, and Ang II-induced increase in cPLA2 activity and oxidative stress in the SFO, BP, cardiac, and renal fibrosis. CONCLUSIONS These data suggest that cPLA2α in the SFO is crucial in mediating Ang II-induced hypertension and associated pathogenesis. Therefore, development of selective cPLA2α inhibitors could be useful in treating hypertension and its pathogenesis.
Collapse
Affiliation(s)
- Chi Young Song
- Department of Pharmacology, College of Medicine, University of Tennessee HSC, Memphis, Tennessee, USA
| | - Nayaab S Khan
- Department of Pharmacology, College of Medicine, University of Tennessee HSC, Memphis, Tennessee, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, College of Medicine, University of Tennessee HSC, Memphis, Tennessee, USA
| | - Bin Wang
- Department of Pharmacology, College of Medicine, University of Tennessee HSC, Memphis, Tennessee, USA
| | - Ji Soo Shin
- Department of Pharmacology, College of Medicine, University of Tennessee HSC, Memphis, Tennessee, USA
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee HSC, Memphis, Tennessee, USA
| |
Collapse
|
30
|
Physiological and pathophysiological implications of PGE2 and the PGE2 synthases in the kidney. Prostaglandins Other Lipid Mediat 2018; 134:1-6. [DOI: 10.1016/j.prostaglandins.2017.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 10/09/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022]
|
31
|
Liu B, Zhan M, Zhang Y, Li H, Wu X, Zhuang F, Luo W, Zhou Y. Increased role of E prostanoid receptor-3 in prostacyclin-evoked contractile activity of spontaneously hypertensive rat mesenteric resistance arteries. Sci Rep 2017; 7:8927. [PMID: 28827689 PMCID: PMC5566542 DOI: 10.1038/s41598-017-09288-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/14/2017] [Indexed: 02/05/2023] Open
Abstract
This study aimed to determine whether E prostanoid receptor-3 (EP3) is involved in prostacyclin (PGI2)-evoked vasoconstrictor activity of resistance arteries and if so, how it changes under hypertensive conditions. Mesenteric resistance arteries from Wistar-Kyoto rats (WKYs) and spontaneously hypertensive rats (SHRs) were isolated for functional and biochemical studies. Here we show that in vessels from WKYs, PGI2 or the endothelial muscarinic agonist ACh (which stimulates in vitro PGI2 synthesis) evoked vasoconstrictor activity, which increased in SHRs. The thromboxane-prostanoid receptor (TP) antagonist SQ29548 partially removed the vasoconstrictor activity, and an increased contractile activity of PGI2 resistant to SQ29548 was observed in SHRs. Interestingly, L798106, an antagonist of EP3 (whose expression was higher in SHRs than in WKYs), not only added to the effect of SQ29548 but also caused relaxation to PGI2 more than that obtained with SQ29548. In accordance, EP3 deletion, which reduced PGI2-evoked contraction, together with SQ29548 resulted in relaxation evoked by the agonist in mouse aortas. These results thus demonstrate an explicit involvement of EP3 in PGI2-evoked vasoconstrictor activity in rat mesenteric resistance arteries and suggest that up-regulation of the receptor contributes significantly to the increased contractile activity evoked by PGI2 under hypertensive conditions.
Collapse
Affiliation(s)
- Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Mengyi Zhan
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingzhan Zhang
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Hui Li
- The Central Lab, Shantou University Medical College, Shantou, China
| | - Xiangzhong Wu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | | | - Wenhong Luo
- The Central Lab, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China.
| |
Collapse
|
32
|
Li Z, Zhang Y, Liu B, Luo W, Li H, Zhou Y. Role of E-type prostaglandin receptor EP3 in the vasoconstrictor activity evoked by prostacyclin in thromboxane-prostanoid receptor deficient mice. Sci Rep 2017; 7:42167. [PMID: 28165064 PMCID: PMC5292700 DOI: 10.1038/srep42167] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 01/06/2017] [Indexed: 02/05/2023] Open
Abstract
Prostacyclin, also termed as prostaglandin I2 (PGI2), evokes contraction in vessels with limited expression of the prostacyclin receptor. Although the thromboxane-prostanoid receptor (TP) is proposed to mediate such a response of PGI2, other unknown receptor(s) might also be involved. TP knockout (TP-/-) mice were thus designed and used to test the hypothesis. Vessels, which normally show contraction to PGI2, were isolated for functional and biochemical analyses. Here, we showed that the contractile response evoked by PGI2 was indeed only partially abolished in the abdominal aorta of TP-/- mice. Interestingly, further antagonizing the E-type prostaglandin receptor EP3 removed the remaining contractile activity, resulting in relaxation evoked by PGI2 in such vessels of TP-/- mice. These results suggest that EP3 along with TP contributes to vasoconstrictor responses evoked by PGI2, and hence imply a novel mechanism for endothelial cyclooxygenase metabolites (which consist mainly of PGI2) in regulating vascular functions.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Base Sequence
- Blood Pressure/drug effects
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Epoprostenol/metabolism
- Epoprostenol/pharmacology
- Female
- Gene Expression Regulation
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Thromboxane/deficiency
- Receptors, Thromboxane/genetics
- Renal Artery/drug effects
- Renal Artery/metabolism
- Signal Transduction
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/metabolism
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Zhenhua Li
- Dept of Pathology, The 2nd Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yingzhan Zhang
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Wenhong Luo
- The Central Lab, Shantou University Medical College, Shantou, China
| | - Hui Li
- The Central Lab, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| |
Collapse
|
33
|
Rezq S, Abdel-Rahman AA. Rostral Ventrolateral Medulla EP3 Receptor Mediates the Sympathoexcitatory and Pressor Effects of Prostaglandin E2 in Conscious Rats. J Pharmacol Exp Ther 2016; 359:290-299. [PMID: 27572469 PMCID: PMC5074484 DOI: 10.1124/jpet.116.233502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/25/2016] [Indexed: 12/13/2022] Open
Abstract
Whereas few studies have dealt with the central sympathoexcitatory action of the inflammatory prostanoid prostaglandin E2 (PGE2), there is no information on the expression and cardiovascular function of different PGE2 (EP) receptors in one of the major cardiovascular-regulating nuclei, the rostral ventrolateral medulla (RVLM). The current study aimed at filling this knowledge gap as well as elucidating the implicated molecular mechanisms. To achieve these goals, we showed the expression of EP2, EP3, and EP4 receptors in the RVLM and investigated their cardiovascular roles in conscious rats, ex vivo as well as in cultured PC12 cells. Intra-RVLM PGE2 significantly increased blood pressure and sympathetic dominance (spectral analysis). Studies with selective EP receptor subtype agonists and antagonists showed that these PGE2-evoked responses were only replicated by intra-RVLM activation of the EP3 receptor with its agonist sulprostone. The RVLM of PGE2-treated rats exhibited increases in c-Fos expression and extracellular signal-regulated kinase 1/2 and neuronal nitric oxide synthase phosphorylation along with oxidative stress, and PGE2 increased l-glutamate release in PC12 cells (surrogates of RVLM neurons). Abrogation of the PGE2-evoked pressor and biochemical responses only occurred following EP3 receptor blockade (N-[(5-Bromo-2-methoxyphenyl)sulfonyl]-3-[2-(2-naphthalenylmethyl)phenyl]-2-propenamide, L-798106). These findings suggest the dependence of RVLM PGE2-mediated sympathoexcitation/pressor response on local EP3 receptor signaling in conscious rats, and highlight central EP3 receptor blockade as a potential therapeutic modality for hypertension management.
Collapse
Affiliation(s)
- Samar Rezq
- Department of Pharmacology, School of Medicine, East Carolina University, Greenville, North Carolina
| | - Abdel A Abdel-Rahman
- Department of Pharmacology, School of Medicine, East Carolina University, Greenville, North Carolina
| |
Collapse
|
34
|
An Update of Microsomal Prostaglandin E Synthase-1 and PGE2 Receptors in Cardiovascular Health and Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5249086. [PMID: 27594972 PMCID: PMC4993943 DOI: 10.1155/2016/5249086] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/19/2016] [Accepted: 06/26/2016] [Indexed: 12/16/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs), especially cyclooxygenase-2 (COX-2) selective inhibitors, are among the most widely used drugs to treat pain and inflammation. However, clinical trials have revealed that these inhibitors predisposed patients to a significantly increased cardiovascular risk, consisting of thrombosis, hypertension, myocardial infarction, heart failure, and sudden cardiac death. Thus, microsomal prostaglandin E (PGE) synthase-1 (mPGES-1), the key terminal enzyme involved in the synthesis of inflammatory prostaglandin E2 (PGE2), and the four PGE2 receptors (EP1-4) have gained much attention as alternative targets for the development of novel analgesics. The cardiovascular consequences of targeting mPGES-1 and the PGE2 receptors are substantially studied. Inhibition of mPGES-1 has displayed a relatively innocuous or preferable cardiovascular profile. The modulation of the four EP receptors in cardiovascular system is diversely reported as well. In this review, we highlight the most recent advances from our and other studies on the regulation of PGE2, particularly mPGES-1 and the four PGE2 receptors, in cardiovascular function, with a particular emphasis on blood pressure regulation, atherosclerosis, thrombosis, and myocardial infarction. This might lead to new avenues to improve cardiovascular disease management strategies and to seek optimized anti-inflammatory therapeutic options.
Collapse
|
35
|
Liu S, Ji Y, Yao J, Zhao X, Xu H, Guan Y, Breyer RM, Sheng H, Zhu J. Knockout of the Prostaglandin E2 Receptor Subtype 3 Promotes Eccentric Cardiac Hypertrophy and Fibrosis in Mice. J Cardiovasc Pharmacol Ther 2016; 22:71-82. [PMID: 27093953 DOI: 10.1177/1074248416642520] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background: Prostaglandin E2 receptor subtype 3 (EP3), a Gi protein-coupled receptor activated by prostaglandin E2, plays a particular role in cardioprotection. This study aimed to investigate the impact of EP3 deletion on cardiac remodeling and further elucidate the related involvement of possible signaling pathways. Methods and Results: The animals used were EP3 receptor knockout (EP3KO) mice and wild-type (WT) litter mate controls at 16-18 weeks old. The high-resolution echocardiography and weight index indicated that eccentric cardiac hypertrophy might occur in EP3KO mice, which were having worse cardiac function than WT litter mates. Isolated adult myocytes from EP3KO hearts showed spontaneous lengthening. Cardiac fibrosis was observed in EP3KO mice through Masson trichrome staining. The elevated messenger RNA (mRNA) level in matrix genes and the reduced mRNA, protein, and activity levels of matrix metalloproteinase 2 (MMP-2) indicated an increased synthesis and suppressed degradation of matrix collagen in EP3KO mice. The phosphorylation level of extracellular signal-regulated kinase (ERK) 1/2 protein was reduced in the cardiac tissue of EP3KO mice, accompanied by no significant change in the protein level of total ERK1/2, total p38, phospho-p38, glycogen synthase kinase-3β (GSK3β), phospho-GSK3β, and calcineurin (CaN) as well as CaN activity. Conclusion: EP3 knockout in cardiac tissues could induce eccentric cardiac hypertrophy and cardiac fibrosis at 16-18 weeks old. These effects of EP3 knockout might be regulated through inactivating MAPK/ERK pathway and affecting the MMP-2 expression. Overall, PGE2-EP3 is necessary to maintain the normal growth and development of the heart.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yawei Ji
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jian Yao
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Xiaodan Zhao
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hu Xu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Youfei Guan
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Richard M. Breyer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hongzhuan Sheng
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jianhua Zhu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
36
|
Kraemer MP, Choi H, Reese J, Lamb FS, Breyer RM. Regulation of arterial reactivity by concurrent signaling through the E-prostanoid receptor 3 and angiotensin receptor 1. Vascul Pharmacol 2016; 84:47-54. [PMID: 27260940 DOI: 10.1016/j.vph.2016.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/20/2016] [Accepted: 05/30/2016] [Indexed: 12/28/2022]
Abstract
Prostaglandin E2 (PGE2), a cyclooxygenase metabolite that generally acts as a systemic vasodepressor, has been shown to have vasopressor effects under certain physiologic conditions. Previous studies have demonstrated that PGE2 receptor signaling modulates angiotensin II (Ang II)-induced hypertension, but the interaction of these two systems in the regulation of vascular reactivity is incompletely characterized. We hypothesized that Ang II, a principal effector of the renin-angiotensin-aldosterone system, potentiates PGE2-mediated vasoconstriction. Here we demonstrate that pre-treatment of arterial rings with 1nM Ang II potentiated PGE2-evoked constriction in a concentration dependent manner (AUC-Ang II 2.778±2.091, AUC+Ang II 22.830±8.560, ***P<0.001). Using genetic deletion models and pharmacological antagonists, we demonstrate that this potentiation effect is mediated via concurrent signaling between the angiotensin II receptor 1 (AT1) and the PGE2 E-prostanoid receptor 3 (EP3) in the mouse femoral artery. EP3 receptor-mediated vasoconstriction is shown to be dependent on extracellular calcium in combination with proline-rich tyrosine kinase 2 (Pyk2) and Rho-kinase. Thus, our findings reveal a novel mechanism through which Ang II and PGE2 regulate peripheral vascular reactivity.
Collapse
Affiliation(s)
- Maria P Kraemer
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, United States.
| | - Hyehun Choi
- Department of Pediatrics, Division of Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeff Reese
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Fred S Lamb
- Department of Pediatrics, Division of Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Richard M Breyer
- Department of Medicine, Veterans Affairs Hospital, Nashville, TN, United States; Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
37
|
Zhang H, Liu J, Qu D, Wang L, Luo JY, Lau CW, Liu P, Gao Z, Tipoe GL, Lee HK, Ng CF, Ma RCW, Yao X, Huang Y. Inhibition of miR-200c Restores Endothelial Function in Diabetic Mice Through Suppression of COX-2. Diabetes 2016; 65:1196-207. [PMID: 26822089 DOI: 10.2337/db15-1067] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/06/2016] [Indexed: 11/13/2022]
Abstract
Endothelial dysfunction plays a crucial role in the development of diabetic vasculopathy. Our initial quantitative PCR results showed an increased miR-200c expression in arteries from diabetic mice and patients with diabetes. However, whether miR-200c is involved in diabetic endothelial dysfunction is unknown. Overexpression of miR-200c impaired endothelium-dependent relaxations (EDRs) in nondiabetic mouse aortas, whereas suppression of miR-200c by anti-miR-200c enhanced EDRs in diabetic db/db mice. miR-200c suppressed ZEB1 expression, and ZEB1 overexpression ameliorated endothelial dysfunction induced by miR-200c or associated with diabetes. More importantly, overexpression of anti-miR-200c or ZEB1 in vivo attenuated miR-200c expression and improved EDRs in db/db mice. Mechanistic study with the use of COX-2(-/-) mice revealed that COX-2 mediated miR-200c-induced endothelial dysfunction and that miR-200c upregulated COX-2 expression in endothelial cells through suppression of ZEB1 and increased production of prostaglandin E2, which also reduced EDR. This study demonstrates for the first time to our knowledge that miR-200c is a new mediator of diabetic endothelial dysfunction and inhibition of miR-200c rescues EDRs in diabetic mice. These new findings suggest the potential usefulness of miR-200c as the target for drug intervention against diabetic vascular complications.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Cell Line
- Cells, Cultured
- Cyclooxygenase 2/chemistry
- Cyclooxygenase 2/metabolism
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Diabetes Mellitus/physiopathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Gene Expression Regulation
- Humans
- In Vitro Techniques
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/metabolism
- Middle Aged
- RNA/metabolism
- RNA Interference
- Renal Artery/metabolism
- Renal Artery/pathology
- Renal Artery/physiopathology
- Vasodilation
- Zinc Finger E-box-Binding Homeobox 1/antagonists & inhibitors
- Zinc Finger E-box-Binding Homeobox 1/genetics
- Zinc Finger E-box-Binding Homeobox 1/metabolism
Collapse
Affiliation(s)
- Huina Zhang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jian Liu
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dan Qu
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiang-Yun Luo
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Wai Lau
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhen Gao
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - George L Tipoe
- Department of Anatomy, The University of Hong Kong, Hong Kong, China
| | - Hung Kay Lee
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Fai Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald Ching Wan Ma
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoqiang Yao
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
38
|
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: 53] [Impact Index Per Article: 5.9] [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
|
39
|
Khan NS, Song CY, Thirunavukkarasu S, Fang XR, Bonventre JV, Malik KU. Cytosolic Phospholipase A2α Is Essential for Renal Dysfunction and End-Organ Damage Associated With Angiotensin II-Induced Hypertension. Am J Hypertens 2016; 29:258-65. [PMID: 26045535 DOI: 10.1093/ajh/hpv083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/13/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The kidney plays an important role in regulating blood pressure (BP). cPLA2α in the kidney is activated by various agents including angiotensin II (Ang II) and selectively releases arachidonic acid (AA) from tissue lipids, generating pro- and antihypertensive eicosanoids. Since activation of cPLA2α is the rate-limiting step in AA release, this study was conducted to determine its contribution to renal dysfunction and end-organ damage associated with Ang II-induced hypertension. METHODS cPLA2α(+/+) and cPLA2α(-/-) mice were infused with Ang II (700 ng/ kg/min) or its vehicle for 13 days. Mice were placed in metabolic cages to monitor their food and water intake, and urine was collected and its volume was measured. Doppler imaging was performed to assess renal hemodynamics. On the 13th day of Ang II infusion, mice were sacrificed and their tissues and blood collected for further analysis. RESULTS Ang II increased renal vascular resistance, water intake, and urine output and Na(+) excretion, decreased urine osmolality, and produced proteinuria in cPLA2α(+/+) mice. Ang II also caused accumulation of F4/80(+) macrophages and CD3(+) T cells and renal fibrosis, and increased oxidative stress in the kidneys of cPLA2α(+/+) mice. All these effects of Ang II were minimized in cPLA2α(-/-) mice. CONCLUSION cPLA2α contributes to renal dysfunction, inflammation, and end-organ damage, most likely via the action of pro-hypertensive eicosanoids and increased oxidative stress associated with Ang II-induced hypertension. Thus, cPLA2α could serve as a potential therapeutic target for treating renal dysfunction and end-organ damage in hypertension.
Collapse
Affiliation(s)
- Nayaab S Khan
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chi Young Song
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Shyamala Thirunavukkarasu
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Xiao R Fang
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital Boston, Harvard Medical School, Harvard Institute of Medicine, Boston, Massachusetts, USA
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA;
| |
Collapse
|
40
|
Nasrallah R, Hassouneh R, Hébert RL. PGE2, Kidney Disease, and Cardiovascular Risk: Beyond Hypertension and Diabetes. J Am Soc Nephrol 2015; 27:666-76. [PMID: 26319242 DOI: 10.1681/asn.2015050528] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
An important measure of cardiovascular health is obtained by evaluating the global cardiovascular risk, which comprises a number of factors, including hypertension and type 2 diabetes, the leading causes of illness and death in the world, as well as the metabolic syndrome. Altered immunity, inflammation, and oxidative stress underlie many of the changes associated with cardiovascular disease, diabetes, and the metabolic syndrome, and recent efforts have begun to elucidate the contribution of PGE2 in these events. This review summarizes the role of PGE2 in kidney disease outcomes that accelerate cardiovascular disease, highlights the role of cyclooxygenase-2/microsomal PGE synthase 1/PGE2 signaling in hypertension and diabetes, and outlines the contribution of PGE2 to other aspects of the metabolic syndrome, particularly abdominal adiposity, dyslipidemia, and atherogenesis. A clearer understanding of the role of PGE2 could lead to new avenues to improve therapeutic options and disease management strategies.
Collapse
Affiliation(s)
- Rania Nasrallah
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramzi Hassouneh
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
41
|
Rare SNP rs12731181 in the miR-590-3p Target Site of the Prostaglandin F
2α
Receptor Gene Confers Risk for Essential Hypertension in the Han Chinese Population. Arterioscler Thromb Vasc Biol 2015; 35:1687-95. [DOI: 10.1161/atvbaha.115.305445] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 05/01/2015] [Indexed: 11/16/2022]
|
42
|
Disruption of prostaglandin E2 receptor EP4 impairs urinary concentration via decreasing aquaporin 2 in renal collecting ducts. Proc Natl Acad Sci U S A 2015; 112:8397-402. [PMID: 26100911 DOI: 10.1073/pnas.1509565112] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The antidiuretic hormone arginine vasopressin is a systemic effector in urinary concentration. However, increasing evidence suggests that other locally produced factors may also play an important role in the regulation of water reabsorption in renal collecting ducts. Recently, prostaglandin E2 (PGE2) receptor EP4 has emerged as a potential therapeutic target for the treatment of nephrogenic diabetes insipidus, but the underlying mechanism is unknown. To evaluate the role of EP4 in regulating water homeostasis, mice with renal tubule-specific knockout of EP4 (Ksp-EP4(-/-)) and collecting duct-specific knockout of EP4 (AQP2-EP4(-/-)) were generated using the Cre-loxP recombination system. Urine concentrating defect was observed in both Ksp-EP4(-/-) and AQP2-EP4(-/-) mice. Decreased aquaporin 2 (AQP2) abundance and apical membrane targeting in renal collecting ducts were evident in Ksp-EP4(-/-) mice. In vitro studies demonstrated that AQP2 mRNA and protein levels were significantly up-regulated in mouse primary inner medullary collecting duct (IMCD) cells after pharmacological activation or adenovirus-mediated overexpression of EP4 in a cAMP/cAMP-response element binding protein-dependent manner. In addition, EP4 activation or overexpression also increased AQP2 membrane accumulation in a mouse IMCD cell line (IMCD3) stably transfected with the AQP2 gene, mainly through the cAMP/protein kinase A and extracellular signal-regulated kinase pathways. In summary, the EP4 receptor in renal collecting ducts plays an important role in regulating urinary concentration under physiological conditions. The ability of EP4 to promote AQP2 membrane targeting and increase AQP2 abundance makes it a potential therapeutic target for the treatment of clinical disorders including acquired and congenital diabetes insipidus.
Collapse
|
43
|
Khan NS, Song CY, Jennings BL, Estes AM, Fang XR, Bonventre JV, Malik KU. Cytosolic phospholipase A2α is critical for angiotensin II-induced hypertension and associated cardiovascular pathophysiology. Hypertension 2015; 65:784-92. [PMID: 25667212 DOI: 10.1161/hypertensionaha.114.04803] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiotensin II activates cytosolic phospholipase A(2)α (cPLA2α) and releases arachidonic acid from tissue phospholipids, which mediate or modulate ≥1 cardiovascular effects of angiotensin II and has been implicated in hypertension. Because arachidonic acid release is the rate limiting step in eicosanoid production, cPLA2α might play a central role in the development of angiotensin II-induced hypertension. To test this hypothesis, we investigated the effect of angiotensin II infusion for 13 days by micro-osmotic pumps on systolic blood pressure and associated pathogenesis in wild type (cPLA2α(+/+)) and cPLA2α(-/-) mice. Angiotensin II-induced increase in systolic blood pressure in cPLA2α(+/+) mice was abolished in cPLA2α(-/-) mice; increased systolic blood pressure was also abolished by the arachidonic acid metabolism inhibitor, 5,8,11,14-eicosatetraynoic acid in cPLA2α(+/+) mice. Angiotensin II in cPLA2α(+/+) mice increased cardiac cPLA2 activity and urinary eicosanoid excretion, decreased cardiac output, caused cardiovascular remodeling with endothelial dysfunction, and increased vascular reactivity in cPLA2α(+/+) mice; these changes were diminished in cPLA2α(-/-) mice. Angiotensin II also increased cardiac infiltration of F4/80(+) macrophages and CD3(+) T lymphocytes, cardiovascular oxidative stress, expression of endoplasmic reticulum stress markers p58(IPK), and CHOP in cPLA2α(+/+) but not cPLA2α(-/-) mice. Angiotensin II increased cardiac activity of ERK1/2 and cSrc in cPLA2α(+/+) but not cPLA2α(-/-) mice. These data suggest that angiotensin II-induced hypertension and associated cardiovascular pathophysiological changes are mediated by cPLA2α activation, most likely through the release of arachidonic acid and generation of eicosanoids with predominant prohypertensive effects and activation of ≥1 signaling molecules, including ERK1/2 and cSrc.
Collapse
Affiliation(s)
- Nayaab S Khan
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Chi Young Song
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Brett L Jennings
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Anne M Estes
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Xiao R Fang
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Joseph V Bonventre
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Kafait U Malik
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.).
| |
Collapse
|
44
|
Lai H, Jia X, Yu Q, Zhang C, Qiao J, Guan Y, Kang J. High-fat diet induces significant metabolic disorders in a mouse model of polycystic ovary syndrome. Biol Reprod 2014; 91:127. [PMID: 25100714 DOI: 10.1095/biolreprod.114.120063] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common female endocrinopathy associated with both reproductive and metabolic disorders. Dehydroepiandrosterone (DHEA) is currently used to induce a PCOS mouse model. High-fat diet (HFD) has been shown to cause obesity and infertility in female mice. The possible effect of an HFD on the phenotype of DHEA-induced PCOS mice is unknown. The aim of the present study was to investigate both reproductive and metabolic features of DHEA-induced PCOS mice fed a normal chow or a 60% HFD. Prepubertal C57BL/6 mice (age 25 days) on the normal chow or an HFD were injected (s.c.) daily with the vehicle sesame oil or DHEA for 20 consecutive days. At the end of the experiment, both reproductive and metabolic characteristics were assessed. Our data show that an HFD did not affect the reproductive phenotype of DHEA-treated mice. The treatment of HFD, however, caused significant metabolic alterations in DHEA-treated mice, including obesity, glucose intolerance, dyslipidemia, and pronounced liver steatosis. These findings suggest that HFD induces distinct metabolic features in DHEA-induced PCOS mice. The combined DHEA and HFD treatment may thus serve as a means of studying the mechanisms involved in metabolic derangements of this syndrome, particularly in the high prevalence of hepatic steatosis in women with PCOS.
Collapse
Affiliation(s)
- Hao Lai
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Xiao Jia
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Qiuxiao Yu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Chenglu Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Youfei Guan
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Jihong Kang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
45
|
Kimple ME, Neuman JC, Linnemann AK, Casey PJ. Inhibitory G proteins and their receptors: emerging therapeutic targets for obesity and diabetes. Exp Mol Med 2014; 46:e102. [PMID: 24946790 PMCID: PMC4081554 DOI: 10.1038/emm.2014.40] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/10/2014] [Accepted: 02/17/2014] [Indexed: 12/21/2022] Open
Abstract
The worldwide prevalence of obesity is steadily increasing, nearly doubling between 1980 and 2008. Obesity is often associated with insulin resistance, a major risk factor for type 2 diabetes mellitus (T2DM): a costly chronic disease and serious public health problem. The underlying cause of T2DM is a failure of the beta cells of the pancreas to continue to produce enough insulin to counteract insulin resistance. Most current T2DM therapeutics do not prevent continued loss of insulin secretion capacity, and those that do have the potential to preserve beta cell mass and function are not effective in all patients. Therefore, developing new methods for preventing and treating obesity and T2DM is very timely and of great significance. There is now considerable literature demonstrating a link between inhibitory guanine nucleotide-binding protein (G protein) and G protein-coupled receptor (GPCR) signaling in insulin-responsive tissues and the pathogenesis of obesity and T2DM. These studies are suggesting new and emerging therapeutic targets for these conditions. In this review, we will discuss inhibitory G proteins and GPCRs that have primary actions in the beta cell and other peripheral sites as therapeutic targets for obesity and T2DM, improving satiety, insulin resistance and/or beta cell biology.
Collapse
Affiliation(s)
- Michelle E Kimple
- Department of Medicine-Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua C Neuman
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Amelia K Linnemann
- Department of Medicine-Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI, USA
| | - Patrick J Casey
- Duke University Medical Center Department of Pharmacology and Cancer Biology, Durham, NC, USA
| |
Collapse
|
46
|
Pushpakumar SB, Kundu S, Metreveli N, Sen U. Folic acid mitigates angiotensin-II-induced blood pressure and renal remodeling. PLoS One 2013; 8:e83813. [PMID: 24386282 PMCID: PMC3873386 DOI: 10.1371/journal.pone.0083813] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 11/08/2013] [Indexed: 12/11/2022] Open
Abstract
Clinical data suggests an association between systolic hypertension, renal function and hyperhomocysteinemia (HHcy). HHcy is a state of elevated plasma homocysteine (Hcy) levels and is known to cause vascular complications. In this study, we tested the hypothesis whether Ang II-induced hypertension increases plasma Hcy levels and contributes to renovascular remodeling. We also tested whether folic acid (FA) treatment reduces plasma Hcy levels by enhancing Hcy remethylation and thus mitigating renal remodeling. Hypertension was induced in WT mice by infusing Ang II using Alzet mini osmotic pumps. Blood pressure, Hcy level, renal vascular density, oxidative stress, inflammation and fibrosis markers, and angiogenic- and anti-angiogenic factors were measured. Ang II hypertension increased plasma Hcy levels and reduced renal cortical blood flow and microvascular density. Elevated Hcy in Ang II hypertension was associated with decreased 4, 5-Diaminofluorescein (DAF-2DA) staining suggesting impaired endothelial function. Increased expression of Nox-2, -4 and dihydroethidium stain revealed oxidative stress. Excess collagen IV deposition in the peri-glomerular area and increased MMP-2, and -9 expression and activity indicated renal remodeling. The mRNA and protein expression of asymmetric dimethylarginine (ADMA) was increased and eNOS protein was decreased suggesting the involvement of this pathway in Hcy mediated hypertension. Decreased expressions of VEGF and increased anti-angiogenic factors, angiostatin and endostatin indicated impaired vasculogenesis. FA treatment partially reduced hypertension by mitigating HHcy in Ang II-treated animals and alleviated pro-inflammatory, pro-fibrotic and anti-angiogenic factors. These results suggest that renovascular remodeling in Ang II-induced hypertension is, in part, due to HHcy.
Collapse
Affiliation(s)
- Sathnur B. Pushpakumar
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Sourav Kundu
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Naira Metreveli
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Utpal Sen
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
47
|
Tilly P, Charles AL, Ludwig S, Slimani F, Gross S, Meilhac O, Geny B, Stefansson K, Gurney ME, Fabre JE. Blocking the EP3 receptor for PGE2 with DG-041 decreases thrombosis without impairing haemostatic competence. Cardiovasc Res 2013; 101:482-91. [PMID: 24323317 DOI: 10.1093/cvr/cvt276] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Haemostasis interrupts bleeding from disrupted blood vessels by activating platelet aggregation and coagulation. A similar mechanism termed thrombosis generates obstructive thrombi inside diseased arteries. As a consequence of this similarity, current anti-thrombotic agents increase the risk of bleeding. Atherosclerotic plaques produce significant amounts of prostaglandin E2 (PGE2), which activates its receptor EP3 on platelets and aggravates atherothrombosis. We investigated whether blocking EP3 could dissociate atherothrombosis from haemostasis. METHODS AND RESULTS Inhibiting in vivo the receptor EP3 for PGE2 with the blocking agent DG-041 reduced murine thrombosis triggered by local delivery of arachidonic acid or ferric chloride on healthy arteries. Importantly, it also reduced thrombosis triggered by scratching murine atherosclerotic plaques. PGE2 was not produced at the bleeding site after tail clipping. Consistently, blocking EP3 did not alter murine tail, liver, or cerebral haemostasis. Furthermore, blocking EP3 reduced murine pulmonary embolism and intensified platelet inhibition by clopidogrel leaving tail bleeding times unchanged. Human atherosclerotic plaques produced PGE2, which facilitated platelet aggregation in human blood and rescued the function of P2Y12-blocked platelets. Finally, in healthy patients, DG-041 reduced platelet aggregation, but did not significantly alter the cutaneous bleeding time at doses up to eight times the dose that inhibited the facilitating effect of PGE2 on platelets. CONCLUSION In mice, blocking EP3 inhibited atherothrombosis without affecting haemostasis and intensified efficiency of conventional anti-platelet treatment without aggravating the bleeding risk. In patients, blocking EP3 should improve the prevention of cardiovascular diseases, which is currently limited by the risk of bleeding.
Collapse
Affiliation(s)
- Peggy Tilly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U596, Centre National de la Recherche Scientifique UMR7104, Université Louis Pasteur, 67400 Illkirch, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
New roles for old pathways? A circuitous relationship between reactive oxygen species and cyclo-oxygenase in hypertension. Clin Sci (Lond) 2013; 126:111-21. [PMID: 24059588 DOI: 10.1042/cs20120651] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elevated production of prostanoids from the constitutive (COX-1) or inducible (COX-2) cyclo-oxygenases has been involved in the alterations in vascular function, structure and mechanical properties observed in cardiovascular diseases, including hypertension. In addition, it is well known that production of ROS (reactive oxygen species) plays an important role in the impaired contractile and vasodilator responses, vascular remodelling and altered vascular mechanics of hypertension. Of particular interest is the cross-talk between NADPH oxidase and mitochondria, the main ROS sources in hypertension, which may represent a vicious feed-forward cycle of ROS production. In recent years, there is experimental evidence showing a relationship between ROS and COX-derived products. Thus ROS can activate COX and the COX/PG (prostaglandin) synthase pathways can induce ROS production through effects on different ROS generating enzymes. Additionally, recent evidence suggests that the COX-ROS axis might constitute a vicious circle of self-perpetuating vasoactive products that have a pathophysiological role in altered vascular contractile and dilator responses and hypertension development. The present review discusses the current knowledge on the role of oxidative stress and COX-derived prostanoids in the vascular alterations observed in hypertension, highlighting new findings indicating that these two pathways act in concert to induce vascular dysfunction.
Collapse
|
49
|
PTGER1 deletion attenuates renal injury in diabetic mouse models. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1789-1802. [PMID: 24113456 DOI: 10.1016/j.ajpath.2013.08.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 07/23/2013] [Accepted: 08/22/2013] [Indexed: 01/11/2023]
Abstract
We hypothesized that the EP1 receptor promotes renal damage in diabetic nephropathy. We rendered EP1 (PTGER1, official symbol) knockout mice (EP1(-/-)) diabetic using the streptozotocin and OVE26 models. Albuminuria, mesangial matrix expansion, and glomerular hypertrophy were each blunted in EP1(-/-) streptozotocin and OVE26 cohorts compared with wild-type counterparts. Although diabetes-associated podocyte depletion was unaffected by EP1 deletion, EP1 antagonism with ONO-8711 in cultured podocytes decreased angiotensin II-mediated superoxide generation, suggesting that EP1-associated injury of remaining podocytes in vivo could contribute to filtration barrier dysfunction. Accordingly, EP1 deletion in OVE26 mice prevented nephrin mRNA expression down-regulation and ameliorated glomerular basement membrane thickening and foot process effacement. Moreover, EP1 deletion reduced diabetes-induced expression of fibrotic markers fibronectin and α-actin, whereas EP1 antagonism decreased fibronectin in cultured proximal tubule cells. Similarly, proximal tubule megalin expression was reduced by diabetes but was preserved in EP1(-/-) mice. Finally, the diabetes-associated increase in angiotensin II-mediated constriction of isolated mesenteric arteries was blunted in OVE26EP1(-/-) mice, demonstrating a role for EP1 receptors in the diabetic vasculature. These data suggest that EP1 activation contributes to diabetic nephropathy progression at several locations, including podocytes, proximal tubule, and the vasculature. The EP1 receptor facilitates the actions of angiotensin II, thereby suggesting that targeting of both the renin-angiotensin system and the EP1 receptor could be beneficial in diabetic nephropathy.
Collapse
|
50
|
Neuman JC, Kimple ME. The EP3 Receptor: Exploring a New Target for Type 2 Diabetes Therapeutics. JOURNAL OF ENDOCRINOLOGY, DIABETES & OBESITY 2013; 1:1002. [PMID: 26322320 PMCID: PMC4551503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Affiliation(s)
- Joshua C. Neuman
- Department of Nutritional Sciences, University of Wisconsin-Madison, Wisconsin, USA
| | | |
Collapse
|