1
|
Qian SH, Liu S, Wang M, Wang Q, Hu CP, Huang JH, Zhang Z. Deficiency of endothelial TRPV4 cation channels ameliorates experimental abdominal aortic aneurysm. Eur J Pharmacol 2025; 986:177150. [PMID: 39577553 DOI: 10.1016/j.ejphar.2024.177150] [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: 04/18/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA), albeit usually asymptomatic, is highly lethal if ruptured. The 28-member transient receptor potential (TRP) ion channel superfamily, most of which are present in the aortic cells, is understudied in AAA. We aim to identify single TRP channel that could represent a novel therapeutic target, and dissect dysfunctional ionic signaling that drives AAA. METHODS AAA was developed in mice by perfusing porcine pancreatic elastase into the infrarenal abdominal aorta. AAA was assessed by measurement of external diameter with a digital caliper, or internal diameter with ultrasonography. Aortic pathohistology was evaluated via histological and immunohistochemical staining. The TRP channel family was analyzed in the GSE17901 dataset. TRPC6, TRPC1/4/5 and TRPC3 channels were blocked in aneurysmal mice by BI749327, Pico145 and Pyr3, respectively. Endothelial cell-selective Trpv4 knockout mice were generated and leveraged for AAA analysis. TRPV4 channel was activated indirectly by TPPU or directly opened by GSK1016790A. RESULTS RNA-seq data mining revealed altered expression profiles of Trpc3/Trpc6, Trpv4. Pharmacological block of TRPC6, TRPC1/4/5 or TRPC3 did not influence AAA, whereas selective deletion of endothelial TRPV4 protected against AAA in endothelial cell-selective Trpv4 knockout mice. Indirect activation of TRPV4 by TPPU exacerbated AAA, but TRPV4-mediated nitric oxide signaling contributed minimally to AAA. TRPV4 activation promoted endothelial cell apoptosis in a Ca2+-dependent manner, a relevant mechanism underlying AAA. CONCLUSIONS Our data underscore the pathogenic importance of Ca2+ perturbation in AAA and illuminate that endothelial TRPV4 cation channel could be harnessed for AAA treatment.
Collapse
MESH Headings
- Animals
- TRPV Cation Channels/genetics
- TRPV Cation Channels/metabolism
- TRPV Cation Channels/deficiency
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Mice
- Mice, Knockout
- Disease Models, Animal
- Male
- Mice, Inbred C57BL
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Endothelial Cells/pathology
- Humans
- Calcium/metabolism
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/drug effects
- Apoptosis/drug effects
Collapse
Affiliation(s)
- She-Hua Qian
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Shuai Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qing Wang
- Department of the Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410013, Hunan, China
| | - Jun-Hao Huang
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou, 510050, Guangdong, China.
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
2
|
Yousef A, Fang L, Heidari M, Kranrod J, Seubert JM. The role of CYP-sEH derived lipid mediators in regulating mitochondrial biology and cellular senescence: implications for the aging heart. Front Pharmacol 2024; 15:1486717. [PMID: 39703395 PMCID: PMC11655241 DOI: 10.3389/fphar.2024.1486717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
Cellular senescence is a condition characterized by stable, irreversible cell cycle arrest linked to the aging process. The accumulation of senescent cells in the cardiac muscle can contribute to various cardiovascular diseases (CVD). Telomere shortening, epigenetic modifications, DNA damage, mitochondrial dysfunction, and oxidative stress are known contributors to the onset of cellular senescence in the heart. The link between mitochondrial processes and cellular senescence contributed to the age-related decline in cardiac function. These include changes in mitochondrial functions and behaviours that arise from various factors, including impaired dynamics, dysregulated biogenesis, mitophagy, mitochondrial DNA (mtDNA), reduced respiratory capacity, and mitochondrial structural changes. Thus, regulation of mitochondrial biology has a role in cellular senescence and cardiac function in aging hearts. Targeting senescent cells may provide a novel therapeutic approach for treating and preventing CVD associated with aging. CYP epoxygenases metabolize N-3 and N-6 polyunsaturated fatty acids (PUFA) into epoxylipids that are readily hydrolyzed to diol products by soluble epoxide hydrolase (sEH). Increasing epoxylipids levels or inhibition of sEH has demonstrated protective effects in the aging heart. Evidence suggests they may play a role in cellular senescence by regulating mitochondria, thus reducing adverse effects of aging in the heart. In this review, we discuss how mitochondria induce cellular senescence and how epoxylipids affect the senescence process in the aged heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mobina Heidari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Joshua Kranrod
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
3
|
Huang P. Research progress on the protective mechanism of a novel soluble epoxide hydrolase inhibitor TPPU on ischemic stroke. Front Neurol 2023; 14:1083972. [PMID: 36846137 PMCID: PMC9945277 DOI: 10.3389/fneur.2023.1083972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/20/2023] [Indexed: 02/11/2023] Open
Abstract
Arachidonic Acid (AA) is the precursor of cerebrovascular active substances in the human body, and its metabolites are closely associated with the pathogenesis of cerebrovascular diseases. In recent years, the cytochrome P450 (CYP) metabolic pathway of AA has become a research hotspot. Furthermore, the CYP metabolic pathway of AA is regulated by soluble epoxide hydrolase (sEH). 1-trifluoromethoxyphenyl-3(1-propionylpiperidin-4-yl) urea (TPPU) is a novel sEH inhibitor that exerts cerebrovascular protective activity. This article reviews the mechanism of TPPU's protective effect on ischemic stroke disease.
Collapse
|
4
|
Thai PN, Ren L, Xu W, Overton J, Timofeyev V, Nader CE, Haddad M, Yang J, Gomes AV, Hammock BD, Chiamvimonvat N, Sirish P. Chronic Diclofenac Exposure Increases Mitochondrial Oxidative Stress, Inflammatory Mediators, and Cardiac Dysfunction. Cardiovasc Drugs Ther 2023; 37:25-37. [PMID: 34499283 PMCID: PMC8904649 DOI: 10.1007/s10557-021-07253-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 01/16/2023]
Abstract
PURPOSE Nonsteroidal anti-inflammatory drugs (NSAIDs) are among one of the most commonly prescribed medications for pain and inflammation. Diclofenac (DIC) is a commonly prescribed NSAID that is known to increase the risk of cardiovascular diseases. However, the mechanisms underlying its cardiotoxic effects remain largely unknown. In this study, we tested the hypothesis that chronic exposure to DIC increases oxidative stress, which ultimately impairs cardiovascular function. METHODS AND RESULTS Mice were treated with DIC for 4 weeks and subsequently subjected to in vivo and in vitro functional assessments. Chronic DIC exposure resulted in not only systolic but also diastolic dysfunction. DIC treatment, however, did not alter blood pressure or electrocardiographic recordings. Importantly, treatment with DIC significantly increased inflammatory cytokines and chemokines as well as cardiac fibroblast activation and proliferation. There was increased reactive oxygen species (ROS) production in cardiomyocytes from DIC-treated mice, which may contribute to the more depolarized mitochondrial membrane potential and reduced energy production, leading to a significant decrease in sarcoplasmic reticulum (SR) Ca2+ load, Ca2+ transients, and sarcomere shortening. Using unbiased metabolomic analyses, we demonstrated significant alterations in oxylipin profiles towards inflammatory features in chronic DIC treatment. CONCLUSIONS Together, chronic treatment with DIC resulted in severe cardiotoxicity, which was mediated, in part, by an increase in mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA
| | - Lu Ren
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA
| | - Wilson Xu
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA
| | - James Overton
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA
| | - Valeriy Timofeyev
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA
| | - Carol E Nader
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA
| | - Michael Haddad
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA
| | - Jun Yang
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Aldrin V Gomes
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA.
- Department of Pharmacology, University of California, Davis, CA, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
| | - Padmini Sirish
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, 451 Health Science Drive, CA, 95616, Davis, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
| |
Collapse
|
5
|
Qian J, Liang S, Wang Q, Xu J, Huang W, Wu G, Liang G. Toll-like receptor-2 in cardiomyocytes and macrophages mediates isoproterenol-induced cardiac inflammation and remodeling. FASEB J 2023; 37:e22740. [PMID: 36583707 DOI: 10.1096/fj.202201345r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/28/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality worldwide. Activation of the innate immune system initiates an inflammatory response during cardiac remodeling induced by isoproterenol (ISO). Here, we investigated whether Toll-like receptor-2 (TLR2) mediates ISO-induced inflammation, hypertrophy, and fibrosis. TLR2 was found to be increased in the heart tissues of mouse with HF under ISO challenge. Further, cardiomyocytes and macrophages were identified as the main cellular sources of the increased TLR2 levels in the model under ISO stimulation. The effect of TLR2 deficiency on ISO-induced cardiac remodeling was determined using TLR2 knockout mice and bone marrow transplantation models. In vitro studies involving ISO-treated cultured cardiomyocytes and macrophages showed that TLR2 knockdown significantly decreased ISO-induced cell inflammation and remodeling via MAPKs/NF-κB signaling. Mechanistically, ISO significantly increased the TLR2-MyD88 interaction in the above cells in a TLR1-dependent manner. Finally, DAMPs, such as HSP70 and fibronectin 1 (FN1), were found to be released from the cells under ISO stimulation, which further activated TLR1/2-Myd88 signaling and subsequently activated pro-inflammatory cytokine expression and cardiac remodeling. In summary, our findings suggest that TLR2 may be a target for the alleviation of chronic adrenergic stimulation-associated HF. In addition, this paper points out the possibility of TLR2 as a new target for heart failure under ISO stimulation.
Collapse
Affiliation(s)
- Jinfu Qian
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shiqi Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qinyan Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiachen Xu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Exploration of the Mechanism of Linoleic Acid Metabolism Dysregulation in Metabolic Syndrome. Genet Res (Camb) 2022; 2022:6793346. [PMID: 36518097 PMCID: PMC9722286 DOI: 10.1155/2022/6793346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/11/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
We aimed to explore the mechanism of the linoleic acid metabolism in metabolic syndrome (MetS). RNA-seq data for 16 samples with or without MetS from the GSE145412 dataset were collected. Gene set variation analysis (GSVA), gene set enrichment analysis (GSEA), and gene differential expression analysis were performed. Expression data of differentially expressed genes (DEGs) involved in the linoleic acid metabolism pathway were mapped to the pathway by using Pathview for visualization. There were 19 and 10 differentially expressed biological processes in the disease group and healthy group, respectively. 9 KEGG pathways were differentially expressed in the disease group. Linoleic acid metabolism was the only differentially expressed pathway in the healthy group. The GSVA enrichment score of the linoleic acid metabolism pathway in the disease group was markedly lower than that in the healthy group. The GSEA result showed that the linoleic acid metabolism pathway was significantly downregulated in the disease group. JMJD7-PLA2G4B, PLA2G1B, PLA2G2D, CYP2C8, and CYP2J2 involved in the pathway were significantly downregulated in the disease group. This study may provide novel insight into MetS from the point of linoleic acid metabolism dysregulation.
Collapse
|
7
|
Angelotti A, Snoke DB, Ormiston K, Cole RM, Borkowski K, Newman JW, Orchard TS, Belury MA. Potential Cardioprotective Effects and Lipid Mediator Differences in Long-Chain Omega-3 Polyunsaturated Fatty Acid Supplemented Mice Given Chemotherapy. Metabolites 2022; 12:metabo12090782. [PMID: 36144189 PMCID: PMC9505633 DOI: 10.3390/metabo12090782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Many commonly used chemotherapies induce mitochondrial dysfunction in cardiac muscle, which leads to cardiotoxicity and heart failure later in life. Dietary long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFA) have demonstrated cardioprotective function in non-chemotherapy models of heart failure, potentially through the formation of LC n-3 PUFA-derived bioactive lipid metabolites. However, it is unknown whether dietary supplementation with LC n-3 PUFA can protect against chemotherapy-induced cardiotoxicity. To test this, 36 female ovariectomized C57BL/6J mice were randomized in a two-by-two factorial design to either a low (0 g/kg EPA + DHA) or high (12.2 g/kg EPA + DHA) LC n-3 PUFA diet, and received either two vehicle or two chemotherapy (9 mg/kg anthracycline + 90 mg/kg cyclophosphamide) tail vein injections separated by two weeks. Body weight and food intake were measured as well as heart gene expression and fatty acid composition. Heart mitochondria were isolated using differential centrifugation. Mitochondrial isolate oxylipin and N-acylethanolamide levels were measured by mass spectrometry after alkaline hydrolysis. LC n-3 PUFA supplementation attenuated some chemotherapy-induced differences (Myh7, Col3a1) in heart gene expression, and significantly altered various lipid species in cardiac mitochondrial preparations including several epoxy fatty acids [17(18)-EpETE] and N-acylethanolamines (arachidonoylethanolamine, AEA), suggesting a possible functional link between heart lipids and cardiotoxicity.
Collapse
Affiliation(s)
- Austin Angelotti
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Deena B. Snoke
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05405, USA
| | - Kate Ormiston
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Rachel M. Cole
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - John W. Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
- Western Human Nutrition Research Center, United States Department of Agriculture-Agriculture Research Service, Davis, CA 95616, USA
- Department of Nutrition, University of California-Davis, Davis, CA 95616, USA
| | - Tonya S. Orchard
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Martha A. Belury
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Correspondence:
| |
Collapse
|
8
|
Maherinia H, Peeri M, Azarbayjani M, Delfan M. Aerobic exercise training combined with probiotic supplement improves antioxidant defence of cardiomyocytes by regulating Nrf2 and caspase3 gene expression in type 2 diabetic rats. COMPARATIVE EXERCISE PHYSIOLOGY 2022; 18:255-264. [DOI: 10.3920/cep200089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2023]
Abstract
This study aimed to evaluate the effect of four weeks of aerobic exercise training combined with probiotic supplementation on mRNA levels of Nrf-2 and caspase-3 genes, superoxide dismutase (SOD), and serum total antioxidant capacity (TAC) in rats with type 2 diabetes. 40 male Wistar diabetic rats were divided into five groups: healthy placebo control group (NC), diabetic control group without supplement (DC), diabetic control group with supplement (SDC), diabetic aerobic training group without supplement (DT), and diabetic aerobic training group with probiotic supplement (SDT). Each training group performed training five days per week for four weeks and each session of training consisted of 30 min running on a treadmill with an intensity of 65-60% of maximum speed. Simultaneously, rats were fed probiotic supplements. Serum glucose, SOD, and TAC were analysed. The real-time PCR technique was used to determine the gene expression of Nrf-2 and caspase-3. Both aerobic exercise training and probiotic supplementation interactively reduced caspase 3 gene expression, increased Nrf-2 gene expression and enhanced TAC in the left ventricle of diabetic rats. Also, the reduction of caspase-3 mRNA in the left ventricle was more effective in the SDT group than in other diabetic groups. There was no interaction effect on SOD. However, a simultaneous effect of training and supplementation was observed on increasing TAC levels when compared to the DC group. Pearson’s correlation showed that the heart weight gain in the SDT group occurred only by decreasing the expression of the caspase-3 gene. Based on these results, probiotics combined with exercise training can be a strategy for improving the antioxidant defence system and preventing risk factors of diabetic cardiomyocytes, especially cell death and myocardial ischemia.
Collapse
Affiliation(s)
- H. Maherinia
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - M. Peeri
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - M.A. Azarbayjani
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - M. Delfan
- Department of Exercise Physiology, Faculty of Sport Sciences, Alzahra University, Tehran, Iran
| |
Collapse
|
9
|
Charles R, Eaton P. Redox Regulation of Soluble Epoxide Hydrolase-Implications for Cardiovascular Health and Disease. Cells 2022; 11:cells11121932. [PMID: 35741062 PMCID: PMC9221603 DOI: 10.3390/cells11121932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/25/2022] Open
Abstract
Cell responses to changes in their redox state are significantly mediated by reversible oxido-reductive post-translational modifications of proteins, potentially altering their activities or interactions. These modifications are important for the homeostatic responses of cells to environmental changes that alter their redox state. Such redox regulatory mechanisms not only operate to maintain health, but can become dysregulated and contribute to pathophysiology. In this review, we focus on the redox control of soluble epoxide hydrolase (sEH), which is widely expressed, including in blood vessels and cardiomyocytes. We review the different types of oxidative modifications that regulate sEH and how they may alter cardiovascular physiology and affect disease progression during stress.
Collapse
|
10
|
A hypothesis-driven study to comprehensively investigate the association between genetic polymorphisms in EPHX2 gene and cardiovascular diseases: Findings from the UK Biobank. Gene X 2022; 822:146340. [PMID: 35183688 DOI: 10.1016/j.gene.2022.146340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Epoxyeicosatrienoic acids (EETs) are protective factors against cardiovascular diseases (CVDs) because of their vasodilatory, cholesterol-lowering, and anti-inflammatory effects. Soluble epoxide hydrolase (sEH), encoded by the EPHX2 gene, degrades EETs into less biologically active metabolites. EPHX2 is highly polymorphic, and genetic polymorphisms in EPHX2 have been linked to various types of CVDs, such as coronary heart disease, essential hypertension, and atrial fibrillation recurrence. METHODS Based on a priori hypothesis that EPHX2 genetic polymorphisms play an important role in the pathogenesis of CVDs, we comprehensively investigated the associations between 210 genetic polymorphisms in the EPHX2 gene and an array of 118 diseases in the circulatory system using a large sample from the UK Biobank (N = 307,516). The diseases in electronic health records were mapped to the phecode system, which was more representative of independent phenotypes. Survival analyses were employed to examine the effects of EPHX2 variants on CVD incidence, and a phenome-wide association study was conducted to study the impact of EPHX2 polymorphisms on 62 traits, including blood pressure, blood lipid levels, and inflammatory indicators. RESULTS A novel association between the intronic variant rs116932590 and the phenotype "aneurysm and dissection of heart" was identified. In addition, the rs149467044 and rs200286838 variants showed nominal evidence of association with arterial aneurysm and cerebrovascular disease, respectively. Furthermore, the variant rs751141, which was linked with a lower hydrolase activity of sEH, was significantly associated with metabolic traits, including blood levels of triglycerides, creatinine, and urate. CONCLUSIONS Multiple novel associations observed in the present study highlight the important role of EPHX2 genetic variation in the pathogenesis of CVDs.
Collapse
|
11
|
Sirish P, Diloretto DA, Thai PN, Chiamvimonvat N. The Critical Roles of Proteostasis and Endoplasmic Reticulum Stress in Atrial Fibrillation. Front Physiol 2022; 12:793171. [PMID: 35058801 PMCID: PMC8764384 DOI: 10.3389/fphys.2021.793171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) remains the most common arrhythmia seen clinically. The incidence of AF is increasing due to the aging population. AF is associated with a significant increase in morbidity and mortality, yet current treatment paradigms have proven largely inadequate. Therefore, there is an urgent need to develop new effective therapeutic strategies for AF. The endoplasmic reticulum (ER) in the heart plays critical roles in the regulation of excitation-contraction coupling and cardiac function. Perturbation in the ER homeostasis due to intrinsic and extrinsic factors, such as inflammation, oxidative stress, and ischemia, leads to ER stress that has been linked to multiple conditions including diabetes mellitus, neurodegeneration, cancer, heart disease, and cardiac arrhythmias. Recent studies have documented the critical roles of ER stress in the pathophysiological basis of AF. Using an animal model of chronic pressure overload, we demonstrate a significant increase in ER stress in atrial tissues. Moreover, we demonstrate that treatment with a small molecule inhibitor to inhibit the soluble epoxide hydrolase enzyme in the arachidonic acid metabolism significantly reduces ER stress as well as atrial electrical and structural remodeling. The current review article will attempt to provide a perspective on our recent understandings and current knowledge gaps on the critical roles of proteostasis and ER stress in AF progression.
Collapse
Affiliation(s)
- Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | - Daphne A Diloretto
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States.,Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
12
|
Reddy GR, Ren L, Thai PN, Caldwell JL, Zaccolo M, Bossuyt J, Ripplinger CM, Xiang YK, Nieves-Cintrón M, Chiamvimonvat N, Navedo MF. Deciphering cellular signals in adult mouse sinoatrial node cells. iScience 2022; 25:103693. [PMID: 35036877 PMCID: PMC8749457 DOI: 10.1016/j.isci.2021.103693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/30/2021] [Accepted: 12/22/2021] [Indexed: 01/27/2023] Open
Abstract
Sinoatrial node (SAN) cells are the pacemakers of the heart. This study describes a method for culturing and infection of adult mouse SAN cells with FRET-based biosensors that can be exploited to examine signaling events. SAN cells cultured in media with blebbistatin or (S)-nitro-blebbistatin retain their morphology, protein distribution, action potential (AP) waveform, and cAMP dynamics for at least 40 h. SAN cells expressing targeted cAMP sensors show distinct β-adrenergic-mediated cAMP pools. Cyclic GMP, protein kinase A, Ca2+/CaM kinase II, and protein kinase D in SAN cells also show unique dynamics to different stimuli. Heart failure SAN cells show a decrease in cAMP and cGMP levels. In summary, a reliable method for maintaining adult mouse SAN cells in culture is presented, which facilitates studies of signaling networks and regulatory mechanisms during physiological and pathological conditions.
Collapse
Affiliation(s)
- Gopireddy R. Reddy
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Lu Ren
- Department of Internal Medicine, University of California Davis, 451 Health Science Drive, GBSF 6315, Davis, CA 95616, USA
| | - Phung N. Thai
- Department of Internal Medicine, University of California Davis, 451 Health Science Drive, GBSF 6315, Davis, CA 95616, USA
| | - Jessica L. Caldwell
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Crystal M. Ripplinger
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
- VA Northern California Healthcare System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Madeline Nieves-Cintrón
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California Davis, 451 Health Science Drive, GBSF 6315, Davis, CA 95616, USA
- VA Northern California Healthcare System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| |
Collapse
|
13
|
He Z, Wang DW. The roles of eicosanoids in myocardial diseases. ADVANCES IN PHARMACOLOGY 2022; 97:167-200. [DOI: 10.1016/bs.apha.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
14
|
Imig JD, Cervenka L, Neckar J. Epoxylipids and soluble epoxide hydrolase in heart diseases. Biochem Pharmacol 2022; 195:114866. [PMID: 34863976 PMCID: PMC8712413 DOI: 10.1016/j.bcp.2021.114866] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular and heart diseases are leading causes of morbidity and mortality. Coronary artery endothelial and vascular dysfunction, inflammation, and mitochondrial dysfunction contribute to progression of heart diseases such as arrhythmias, congestive heart failure, and heart attacks. Classes of fatty acid epoxylipids and their enzymatic regulation by soluble epoxide hydrolase (sEH) have been implicated in coronary artery dysfunction, inflammation, and mitochondrial dysfunction in heart diseases. Likewise, genetic and pharmacological manipulations of epoxylipids have been demonstrated to have therapeutic benefits for heart diseases. Increasing epoxylipids reduce cardiac hypertrophy and fibrosis and improve cardiac function. Beneficial actions for epoxylipids have been demonstrated in cardiac ischemia reperfusion injury, electrical conductance abnormalities and arrhythmias, and ventricular tachycardia. This review discusses past and recent findings on the contribution of epoxylipids in heart diseases and the potential for their manipulation to treat heart attacks, arrhythmias, ventricular tachycardia, and heart failure.
Collapse
Affiliation(s)
- John D Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ludek Cervenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.,Department of Pathophysiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Neckar
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.,Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
15
|
Charles RL, Abis G, Fernandez BF, Guttzeit S, Buccafusca R, Conte MR, Eaton P. A thiol redox sensor in soluble epoxide hydrolase enables oxidative activation by intra-protein disulfide bond formation. Redox Biol 2021; 46:102107. [PMID: 34509915 PMCID: PMC8436062 DOI: 10.1016/j.redox.2021.102107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022] Open
Abstract
Soluble epoxide hydrolase (sEH), an enzyme that broadly regulates the cardiovascular system, hydrolyses epoxyeicosatrienoic acids (EETs) to their corresponding dihydroxyeicosatrienoic acids (DHETs). We previously showed that endogenous lipid electrophiles adduct within the catalytic domain, inhibiting sEH to lower blood pressure in angiotensin II-induced hypertensive mice. As angiotensin II increases vascular H2O2, we explored sEH redox regulation by this oxidant and how this integrates with inhibition by lipid electrophiles to regulate vasotone. Kinetics analyses revealed that H2O2 not only increased the specific activity of sEH but increased its affinity for substrate and increased its catalytic efficiency. This oxidative activation was mediated by formation of an intra-disulfide bond between C262 and C264, as determined by mass spectrometry and substantiated by biotin-phenylarsinate and thioredoxin-trapping mutant assays. C262S/264S sEH mutants were resistant to peroxide-induced activation, corroborating the disulfide-activation mechanism. The physiological impact of sEH redox state was determined in isolated arteries and the effect of the pro-oxidant vasopressor angiotensin II on arterial sEH redox state and vasodilatory EETs indexed in mice. Angiotensin II induced the activating intra-disulfide in sEH, causing a decrease in plasma EET/DHET ratios that is consistent with the pressor response to this hormone. Although sEH C262-C264 disulfide formation enhances hydrolysis of vasodilatory EETs, this modification also sensitized sEH to inhibition by lipid electrophiles. This explains why angiotensin II decreases EETs and increases blood pressure, but when lipid electrophiles are also present, that EETs are increased and blood pressure lowered.
Collapse
Affiliation(s)
- Rebecca L Charles
- Queen Mary University of London, William Harvey Research Institute, Charterhouse Square, London, EC1M 6BQ, UK
| | - Giancarlo Abis
- King's College London, Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, London, SE1 1UL, UK
| | - Beatriz F Fernandez
- King's College London, The British Heart Foundation Centre of Excellence, The Rayne Institute, St Thomas' Hospital, London, SE1 7EH, UK
| | - Sebastian Guttzeit
- King's College London, The British Heart Foundation Centre of Excellence, The Rayne Institute, St Thomas' Hospital, London, SE1 7EH, UK
| | - Roberto Buccafusca
- Queen Mary University of London, School of Biological and Chemical Sciences, Mile End Road, London, E1 4NS, UK
| | - Maria R Conte
- King's College London, Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, London, SE1 1UL, UK.
| | - Philip Eaton
- Queen Mary University of London, William Harvey Research Institute, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
16
|
Annexin A1 attenuates cardiac diastolic dysfunction in mice with inflammatory arthritis. Proc Natl Acad Sci U S A 2021; 118:2020385118. [PMID: 34526398 PMCID: PMC8463875 DOI: 10.1073/pnas.2020385118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 12/16/2022] Open
Abstract
Rheumatoid arthritis (RA) carries a twofold increased incidence of heart failure with preserved ejection fraction, accompanied by diastolic dysfunction, which can lead to death. The causes of diastolic dysfunction are unknown, and there are currently no well-characterized animal models for studying these mechanisms. Current medications for RA do not have marked beneficial cardio-protective effects. K/BxN F1 progeny and KRN control mice were analyzed over time for arthritis development, monitoring left ventricular diastolic and systolic function using echocardiography. Excised hearts were analyzed by flow cytometry, qPCR, and histology. In pharmacological experiments, K/BxN F1 mice were treated with human recombinant AnxA1 (hrAnxA1, 1 μg/mouse) or vehicle daily. K/BxN F1 mice exhibited fully developed arthritis with normal cardiac function at 4 wk; however, by week 8, all mice displayed left ventricular diastolic dysfunction with preserved ejection fraction. This dysfunction was associated with cardiac hypertrophy, myocardial inflammation and fibrosis, and inflammatory markers. Daily treatment of K/BxN F1 mice with hrAnxA1 from weeks 4 to 8 halted progression of the diastolic dysfunction. The treatment reduced cardiac transcripts of proinflammatory cytokines and profibrotic markers. At the cellular level, hrAnxA1 decreased activated T cells and increased MHC IIlow macrophage infiltration in K/BxN F1 hearts. Similar effects were obtained when hrAnxA1 was administered from week 8 to week 15. We describe an animal model of inflammatory arthritis that recapitulates the cardiomyopathy of RA. Treatment with hrAnxA1 after disease onset corrected the diastolic dysfunction through modulation of both fibroblast and inflammatory cell phenotype within the heart.
Collapse
|
17
|
Abstract
Background: Sepsis is a life-threatening organ dysfunction initiated by a dysregulated response to infection, with imbalanced inflammation and immune homeostasis. Macrophages play a pivotal role in sepsis. N-[1-(1-oxopropyl)-4-piperidinyl]-N’-[4-(trifluoromethoxy)phenyl)-urea (TPPU) is an inhibitor of soluble epoxide hydrolase (sEH), which can rapidly hydrolyze epoxyeicosatrienoic acids (EETs) to the bio-inactive dihydroxyeicosatrienoic acids. TPPU was linked with the regulation of macrophages and inflammation. Here, we hypothesized that sEH inhibitor TPPU ameliorates cecal ligation and puncture (CLP)-induced sepsis by regulating macrophage functions. Methods: A polymicrobial sepsis model induced by CLP was used in our study. C57BL/6 mice were divided into four groups: sham+ phosphate buffer saline (PBS), sham+TPPU, CLP+PBS, CLP+TPPU. Mice were observed 48 h after surgery to assess the survival rate. For other histological examinations, mice were sacrificed 6 h after surgery. Macrophage cell line RAW264.7 was used for in vitro studies. Results: TPPU treatment, accompanied with increased EETs levels, markedly improved the survival of septic mice induced by CLP surgery, which was associated with alleviated organ damage and dysfunction triggered by systemic inflammatory response. Moreover, TPPU treatment significantly inhibited systemic inflammatory response via EETs-induced inactivation of mitogen-activated protein kinase signaling due to enhanced macrophage phagocytic ability and subsequently reduced bacterial proliferation and dissemination, and decreased inflammatory factors release. Conclusion: sEH inhibitor TPPU ameliorates cecal ligation and puncture-induced sepsis by regulating macrophage functions, including improved phagocytosis and reduced inflammatory response. Our data indicate that sEH inhibition has potential therapeutic effects on polymicrobial-induced sepsis.
Collapse
|
18
|
Cizkova K, Koubova K, Foltynkova T, Jiravova J, Tauber Z. Soluble Epoxide Hydrolase as an Important Player in Intestinal Cell Differentiation. Cells Tissues Organs 2021; 209:177-188. [PMID: 33588415 DOI: 10.1159/000512807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/05/2020] [Indexed: 11/19/2022] Open
Abstract
There is growing evidence that soluble epoxide hydrolase (sEH) may play a role in cell differentiation. sEH metabolizes biologically highly active and generally cytoprotective epoxyeicosatrienoic acids (EETs), generated from arachidonic acid metabolism by CYP epoxygenases (CYP2C and CYP2J subfamilies), to less active corresponding diols. We investigated the effect of sEH inhibitor (TPPU) on the expression of villin, CYP2C8, CYP2C9, CYP2J2, and sEH in undifferentiated and in vitro differentiated HT-29 and Caco2 cell lines. The administration of 10 μM TPPU on differentiated HT-29 and Caco2 cells resulted in a significant decrease in expression of villin, a marker for intestinal cell differentiation. It was accompanied by a disruption of the brush border when microvilli appeared sparse and short in atomic force microscope scans of HT-29 cells. Although inhibition of sEH in differentiated HT-29 and Caco2 cells led to an increase in sEH expression in both cell lines, this treatment had an opposite effect on CYP2J2 expression in HT-29 and Caco2 cells. In addition, tissue samples of colorectal carcinoma and adjacent normal tissues from 45 patients were immunostained for sEH and villin. We detected a significant decrease in the expression of both proteins in colorectal carcinoma in comparison to adjacent normal tissue, and the decrease in both sEH and villin expression revealed a moderate positive association. Taken together, our results showed that sEH is an important player in intestinal cell differentiation.
Collapse
Affiliation(s)
- Katerina Cizkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Katerina Koubova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Tereza Foltynkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Jana Jiravova
- Department of Medical Biophysics, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Zdenek Tauber
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia,
| |
Collapse
|
19
|
Hildreth K, Kodani SD, Hammock BD, Zhao L. Cytochrome P450-derived linoleic acid metabolites EpOMEs and DiHOMEs: a review of recent studies. J Nutr Biochem 2020; 86:108484. [PMID: 32827665 PMCID: PMC7606796 DOI: 10.1016/j.jnutbio.2020.108484] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/24/2020] [Accepted: 08/13/2020] [Indexed: 12/29/2022]
Abstract
Linoleic acid (LA) is the most abundant polyunsaturated fatty acid found in the Western diet. Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. Previous studies of the EpOMEs and DiHOMEs have focused on their roles in cytotoxic processes, primarily in the inhibition of the neutrophil respiratory burst. More recent research has suggested the DiHOMEs may be important lipid mediators in pain perception, altered immune response and brown adipose tissue activation by cold and exercise. The purpose of this review is to summarize the current understanding of the physiological and pathophysiological roles and modes of action of the EpOMEs and DiHOMEs in health and disease.
Collapse
Affiliation(s)
- Kelsey Hildreth
- Department of Nutrition, University of Tennessee, Knoxville, TN
| | - Sean D Kodani
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA
| | - Ling Zhao
- Department of Nutrition, University of Tennessee, Knoxville, TN.
| |
Collapse
|
20
|
Zhang Y, Bai Y, Bai J, Li L, Gao L, Wang F. Targeting Soluble Epoxide Hydrolase with TPPU Alleviates Irradiation‐Induced Hyposalivation in Mice via Preventing Apoptosis and Microcirculation Disturbance. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yaoyang Zhang
- School of Stomatology Dalian Medical University No.9 West Section Lvshun South Road Dalian Liaoning Province 116044 P. R. China
| | - Yuwen Bai
- School of Stomatology Dalian Medical University No.9 West Section Lvshun South Road Dalian Liaoning Province 116044 P. R. China
| | - Jie Bai
- School of Stomatology Dalian Medical University No.9 West Section Lvshun South Road Dalian Liaoning Province 116044 P. R. China
| | - Lijun Li
- School of Stomatology Dalian Medical University No.9 West Section Lvshun South Road Dalian Liaoning Province 116044 P. R. China
| | - Lu Gao
- School of Stomatology Dalian Medical University No.9 West Section Lvshun South Road Dalian Liaoning Province 116044 P. R. China
| | - Fu Wang
- School of Stomatology Dalian Medical University No.9 West Section Lvshun South Road Dalian Liaoning Province 116044 P. R. China
| |
Collapse
|
21
|
Li Y, Chen X, Li P, Xiao Q, Hou D, Kong X. CD47 antibody suppresses isoproterenol-induced cardiac hypertrophy through activation of autophagy. Am J Transl Res 2020; 12:5908-5923. [PMID: 33042468 PMCID: PMC7540123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 06/02/2020] [Indexed: 06/11/2023]
Abstract
In this study, we investigated whether CD47 antibody could attenuate isoproterenol (ISO)-induced cardiac hypertrophy in mice and H9C2 cells. Cardiac hypertrophy was induced by intraperitoneal (i.p.) injection of ISO (60 mg.kg-1.d-1 in 100 µl of sterile normal saline) daily for 14 days, and cell hypertrophy was induced by ISO (10-5 mol/l) for 48 h. The injury was confirmed by increased levels of lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB), increased heart-to-body weight (HW/BW) ratio and visible cardiac fibrosis. Apoptosis was evaluated by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. Autophagic flux in H9c2 cells was monitored by TEM and mRFP-GFP-LC3 virus transfection. The expression levels of Cleaved caspase-3, Cleaved caspase-9 and autophagy-related proteins were detected by Western blotting. CD47 antibody significantly limited ISO-induced increases in LDH, CK-MB, HW/BW ratio and attenuated cardiac fibrosis, oxidative stress, and apoptosis in the heart; CD47 antibody promoted autophagy flow and decreased cell apoptosis in cardiac tissues. Moreover, autophagy inhibitor chloroquine (CQ) reversed the effect of CD47 antibody treatment. In conclusion, CD47 antibody reduced ISO-induced cardiac hypertrophy by improving autophagy flux and rescuing autophagic clearance.
Collapse
Affiliation(s)
- Yong Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, P. R. China
| | - Xuguan Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, P. R. China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, P. R. China
| | - Qianxuan Xiao
- Department of Stomatology, Nanjing Medical UniversityNanjing 211166, Jiangsu Province, P. R. China
| | - Daorong Hou
- Key Laboratory of Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical UniversityNanjing 211166, Jiangsu Province, P. R. China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, P. R. China
| |
Collapse
|
22
|
Sirish P, Thai PN, Lee JH, Yang J, Zhang X, Ren L, Li N, Timofeyev V, Lee KSS, Nader CE, Rowland DJ, Yechikov S, Ganaga S, Young N, Lieu DK, Yamoah EN, Hammock BD, Chiamvimonvat N. Suppression of inflammation and fibrosis using soluble epoxide hydrolase inhibitors enhances cardiac stem cell-based therapy. Stem Cells Transl Med 2020; 9:1570-1584. [PMID: 32790136 PMCID: PMC7695637 DOI: 10.1002/sctm.20-0143] [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: 04/05/2020] [Revised: 05/03/2020] [Accepted: 05/30/2020] [Indexed: 01/29/2023] Open
Abstract
Stem cell replacement offers a great potential for cardiac regenerative therapy. However, one of the critical barriers to stem cell therapy is a significant loss of transplanted stem cells from ischemia and inflammation in the host environment. Here, we tested the hypothesis that inhibition of the soluble epoxide hydrolase (sEH) enzyme using sEH inhibitors (sEHIs) to decrease inflammation and fibrosis in the host myocardium may increase the survival of the transplanted human induced pluripotent stem cell derived-cardiomyocytes (hiPSC-CMs) in a murine postmyocardial infarction model. A specific sEHI (1-trifluoromethoxyphenyl-3-(1-propionylpiperidine-4-yl)urea [TPPU]) and CRISPR/Cas9 gene editing were used to test the hypothesis. TPPU results in a significant increase in the retention of transplanted cells compared with cell treatment alone. The increase in the retention of hiPSC-CMs translates into an improvement in the fractional shortening and a decrease in adverse remodeling. Mechanistically, we demonstrate a significant decrease in oxidative stress and apoptosis not only in transplanted hiPSC-CMs but also in the host environment. CRISPR/Cas9-mediated gene silencing of the sEH enzyme reduces cleaved caspase-3 in hiPSC-CMs challenged with angiotensin II, suggesting that knockdown of the sEH enzyme protects the hiPSC-CMs from undergoing apoptosis. Our findings demonstrate that suppression of inflammation and fibrosis using an sEHI represents a promising adjuvant to cardiac stem cell-based therapy. Very little is known regarding the role of this class of compounds in stem cell-based therapy. There is consequently an enormous opportunity to uncover a potentially powerful class of compounds, which may be used effectively in the clinical setting.
Collapse
Affiliation(s)
- Padmini Sirish
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA,Department of Veterans AffairsNorthern California Health Care SystemMatherCaliforniaUSA
| | - Phung N. Thai
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Jeong Han Lee
- Department of Physiology and Cell BiologyUniversity of Nevada, RenoRenoNevadaUSA
| | - Jun Yang
- Department of Entomology and Nematology and Comprehensive Cancer CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Xiao‐Dong Zhang
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA,Department of Veterans AffairsNorthern California Health Care SystemMatherCaliforniaUSA
| | - Lu Ren
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Ning Li
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Valeriy Timofeyev
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Kin Sing Stephen Lee
- Department of Entomology and Nematology and Comprehensive Cancer CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Carol E. Nader
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Douglas J. Rowland
- Center for Molecular and Genomic ImagingUniversity of CaliforniaDavisCaliforniaUSA
| | - Sergey Yechikov
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Svetlana Ganaga
- Department of SurgeryUniversity of CaliforniaDavisCaliforniaUSA
| | - Nilas Young
- Department of SurgeryUniversity of CaliforniaDavisCaliforniaUSA
| | - Deborah K. Lieu
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell BiologyUniversity of Nevada, RenoRenoNevadaUSA
| | - Bruce D. Hammock
- Department of Entomology and Nematology and Comprehensive Cancer CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular MedicineUniversity of CaliforniaDavisCaliforniaUSA,Department of Veterans AffairsNorthern California Health Care SystemMatherCaliforniaUSA,Department of PharmacologyUniversity of CaliforniaDavisCaliforniaUSA
| |
Collapse
|
23
|
Yi X, Xu C, Huang P, Zhang L, Qing T, Li J, Wang C, Zeng T, Lu J, Han Z. 1-Trifluoromethoxyphenyl-3-(1-Propionylpiperidin-4-yl) Urea Protects the Blood-Brain Barrier Against Ischemic Injury by Upregulating Tight Junction Protein Expression, Mitigating Apoptosis and Inflammation In Vivo and In Vitro Model. Front Pharmacol 2020; 11:1197. [PMID: 32848796 PMCID: PMC7427473 DOI: 10.3389/fphar.2020.01197] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/23/2020] [Indexed: 02/05/2023] Open
Abstract
We previously have revealed that 1-trifluoromethoxyphenyl-3-(1- propionylpiperidin-4-yl) urea (TPPU), as a soluble epoxide hydrolase (sEH) inhibitor can reduce infarct volume, protect blood-brain barrier (BBB) and brain against ischemic injury in rats. Here, we investigated the potential mechanisms of TPPU on BBB integrity in both in permanent middle cerebral artery occlusion (pMCAO) rat model and in oxygen-glucose deprivation/reperfusion (OGD/R)-induced human brain microvascular endothelial cells (HBMVECs) model. In pMCAO rat, TPPU administration decreased brain edema and Evans blue content, increased tight junction proteins (TJs) expression of claudin-5, occludin, and zonula occludens-1 (ZO-1). In OGD/R model, OGD/R significantly increased permeability and cell apoptosis, downregulated the expression of claudin-5, ZO-1, occludin, and lymphoma (Bcl)-2. Notably, TPPU pretreatment effectively protected the BBB integrity by reducing the permeability, promoting expression of claudin-5, ZO-1, occluding and Bcl-2, mitigating reactive oxygen species (ROS) injury and release of interleukin-1β (IL-1β), IL-6β, and tumor necrosis factor-α (TNF-α), downregulating expression of matrix metalloproteinase-9 (MMP-9), MMP-2, bcl-2-associated X protein (Bax), IL-1β, IL-6β, and TNF-α. Moreover, OGD/R induced the up-regulation of p-p65, p-IκB, and p-p38, which were effectively decreased after TPPU pretreatment in comparison with that of the OGD/R group. Furthermore, pyrrolidinedithiocarbamate (PDTC, a selective inhibitor of NF-κB p65) not only alleviated the OGD/R-induced HBMVECs injury and permeability, but also reduced the expression of TNF-α, IL-6, IL-1β, p-p65, and p-IκB, and the protective effect of PDTC was equivalent to that of TPPU. These results indicate that TPPU protects BBB integrity against ischemic injury by multiple protective mechanisms, at least in part, by reducing ROS, inflammation, apoptosis, and suppressing the nuclear factor-κB (NF-κB) and p38 signaling pathways.
Collapse
Affiliation(s)
- Xingyang Yi
- Department of Neurology, People's Hospital of Deyang City, Deyang, China.,Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chongxi Xu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chendu, China
| | - Pan Huang
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Linlei Zhang
- Department of General Intensive Care Unit, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Qing
- Department of Neurology, People's Hospital of Deyang City, Deyang, China.,Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Li
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Chun Wang
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Tao Zeng
- Department of Neurology, People's Hospital of Deyang City, Deyang, China.,Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jing Lu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Neurology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Zhao Han
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
Imig JD, Jankiewicz WK, Khan AH. Epoxy Fatty Acids: From Salt Regulation to Kidney and Cardiovascular Therapeutics: 2019 Lewis K. Dahl Memorial Lecture. Hypertension 2020; 76:3-15. [PMID: 32475311 PMCID: PMC7448548 DOI: 10.1161/hypertensionaha.120.13898] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epoxyeicosatrienoic acids (EETs) are epoxy fatty acids that have biological actions that are essential for maintaining water and electrolyte homeostasis. An inability to increase EETs in response to a high-salt diet results in salt-sensitive hypertension. Vasodilation, inhibition of epithelial sodium channel, and inhibition of inflammation are the major EET actions that are beneficial to the heart, resistance arteries, and kidneys. Genetic and pharmacological means to elevate EETs demonstrated antihypertensive, anti-inflammatory, and organ protective actions. Therapeutic approaches to increase EETs were then developed for cardiovascular diseases. sEH (soluble epoxide hydrolase) inhibitors were developed and progressed to clinical trials for hypertension, diabetes mellitus, and other diseases. EET analogs were another therapeutic approach taken and these drugs are entering the early phases of clinical development. Even with the promise for these therapeutic approaches, there are still several challenges, unexplored areas, and opportunities for epoxy fatty acids.
Collapse
Affiliation(s)
- John D Imig
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| | - Wojciech K Jankiewicz
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| | - Abdul H Khan
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
25
|
Zhang L, Xu S, Wu X, Muse FM, Chen J, Cao Y, Yan J, Cheng Z, Yi X, Han Z. Protective Effects of the Soluble Epoxide Hydrolase Inhibitor 1-Trifluoromethoxyphenyl-3-(1-Propionylpiperidin-4-yl) Urea in a Rat Model of Permanent Middle Cerebral Artery Occlusion. Front Pharmacol 2020; 11:182. [PMID: 32184732 PMCID: PMC7058996 DOI: 10.3389/fphar.2020.00182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Acute ischemic stroke is a serious disease that endangers human health. In our efforts to develop an effective therapy, we previously showed that the potent, highly selective inhibitor of soluble epoxide hydrolase called 1-trifuoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) protects the brain against focal ischemia in rats. Here we explored the mechanism of TPPU action by assessing whether it could preserve blood-brain barrier integrity and reduce apoptosis in the brain during permanent middle cerebral artery occlusion in male Sprague-Dawley rats. TPPU administration at the onset of stroke and once daily thereafter led to smaller infarct volume and brain edema as well as milder neurological deficits. TPPU significantly inhibited the activity of soluble epoxide hydrolase and matrix metalloproteases 2 and 9, reducing 14,15-DHET levels, while increasing expression of tight junction proteins. TPPU decreased numbers of apoptotic cells by down-regulating the pro-apoptotic proteins BAX and Caspase-3, while up-regulating the anti-apoptotic protein BCL-2. Our results suggest that TPPU can protect the blood-brain barrier and reduce the apoptosis of brain tissue caused by ischemia.
Collapse
Affiliation(s)
- Linlei Zhang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of General Intensive Care Unit, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shasha Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoxiao Wu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Farah Mohamed Muse
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaou Chen
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yungang Cao
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jueyue Yan
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zicheng Cheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xingyang Yi
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Zhao Han
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
26
|
In vitro aged, hiPSC-origin engineered heart tissue models with age-dependent functional deterioration to study myocardial infarction. Acta Biomater 2019; 94:372-391. [PMID: 31146032 DOI: 10.1016/j.actbio.2019.05.064] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 01/29/2023]
Abstract
Deaths attributed to ischemic heart disease increased by 41.7% from 1990 to 2013. This is primarily due to an increase in the aged population, however, research on cardiovascular disease (CVD) has been overlooking aging, a well-documented contributor to CVD. The use of young animals is heavily preferred due to lower costs and ready availability, despite the prominent differences between young and aged heart structure and function. Here we present the first human induced pluripotent stem cell (hiPSC)-derived cardiomyocyte (iCM)-based, in vitro aged myocardial tissue model as an alternative research platform. Within 4 months, iCMs go through accelerated senescence and show cellular characteristics of aging. Furthermore, the model tissues fabricated using aged iCMs, with stiffness resembling that of aged human heart, show functional and pharmacological deterioration specific to aged myocardium. Our novel tissue model with age-appropriate physiology and pathology presents a promising new platform for investigating CVD or other age-related diseases. STATEMENT OF SIGNIFICANCE: In vitro and in vivo models of cardiovascular disease are aimed to provide crucial insight on the pathology and treatment of these diseases. However, the contribution of age-dependent cardiovascular changes is greatly underestimated through the use of young animals and premature cardiomyocytes. Here, we developed in vitro aged cardiac tissue models that mimic the aged heart tissue microenvironment and cellular phenotype and present the first evidence that age-appropriate in vitro disease models can be developed to gain more physiologically-relevant insight on development, progression, and amelioration of cardiovascular diseases.
Collapse
|
27
|
Li R, Shan Y, Gao L, Wang X, Wang X, Wang F. The Glp-1 Analog Liraglutide Protects Against Angiotensin II and Pressure Overload-Induced Cardiac Hypertrophy via PI3K/Akt1 and AMPKa Signaling. Front Pharmacol 2019; 10:537. [PMID: 31231210 PMCID: PMC6560159 DOI: 10.3389/fphar.2019.00537] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 04/29/2019] [Indexed: 12/17/2022] Open
Abstract
The Glp-1 analog, liraglutide (Lir), has been shown to reduce infarct size and improve cardiac function after myocardial ischemia in rodents with or without diabetes. However, the effect of Lir on angiotensin II (AngII) and pressure overload induced cardiac hypertrophy in nondiabetic mice and the underlying mechanisms are unclear. The aim of this study was to investigate the effect of Lir on cardiac hypertrophy induced by AngII infusion and pressure overload and to explore its possible mechanism. Mice were subjected to AngII as well as thoracic aorta coarctation (TAC) to induce a cardiac hypertrophy model. Mice were daily injected with either liraglutide or saline for 2 weeks after AngII infusion. Mice were also subjected to either liraglutide or saline for 25 days after TAC surgery. Neonatal rat cardiomyocytes and human AC cell lines were stimulated with AngII to induce a cardiomyocytes hypertrophy model. The results indicated Lir significantly inhibited cardiac hypertrophy and fibrosis and improved cardiac function in both the AngII and pressure overload induced model. The in vitro study showed that Lir inhibits AngII induced cell hypertrophy. Mechanistically, Lir directly suppressing the activation of PI3K/Akt1 and stimulated AMPKα signaling pathways in cardiomyocytes, which was confirmed by use of an mTOR activator (MHY1485), overexpression of constitutively active Akt, and the knockdown of AMPKa2 expression. Moreover, the protective effects of Lir were lost in AMPKa2 knockout mice. Taken together, Lir inhibits AngII and pressure overload induced cardiac remodeling via regulating PI3K/Akt1 and AMPKα signaling.
Collapse
Affiliation(s)
- Ran Li
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingguang Shan
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Gao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xi Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xule Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Wang
- Department of Endocrinology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Epoxyeicosatrienoic acid analog EET-B attenuates post-myocardial infarction remodeling in spontaneously hypertensive rats. Clin Sci (Lond) 2019; 133:939-951. [PMID: 30979784 PMCID: PMC6492034 DOI: 10.1042/cs20180728] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 03/31/2019] [Accepted: 04/11/2019] [Indexed: 12/30/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) and their synthetic analogs have cardiovascular protective effects. Here, we investigated the action of a novel EET analog EET-B on the progression of post-myocardial infarction (MI) heart failure in spontaneously hypertensive rats (SHR). Adult male SHR were divided into vehicle- and EET-B (10 mg/kg/day; p.o., 9 weeks)-treated groups. After 2 weeks of treatment, rats were subjected to 30-min left coronary artery occlusion or sham operation. Systolic blood pressure (SBP) and echocardiography (ECHO) measurements were performed at the beginning of study, 4 days before, and 7 weeks after MI. At the end of the study, tissue samples were collected for histological and biochemical analyses. We demonstrated that EET-B treatment did not affect blood pressure and cardiac parameters in SHR prior to MI. Fractional shortening (FS) was decreased to 18.4 ± 1.0% in vehicle-treated MI rats compared with corresponding sham (30.6 ± 1.0%) 7 weeks following MI induction. In infarcted SHR hearts, EET-B treatment improved FS (23.7 ± 0.7%), markedly increased heme oxygenase-1 (HO-1) immunopositivity in cardiomyocytes and reduced cardiac inflammation and fibrosis (by 13 and 19%, respectively). In conclusion, these findings suggest that EET analog EET-B has beneficial therapeutic actions to reduce cardiac remodeling in SHR subjected to MI.
Collapse
|
29
|
Yao L, Cao B, Cheng Q, Cai W, Ye C, Liang J, Liu W, Tan L, Yan M, Li B, He J, Hwang SH, Zhang X, Wang C, Ai D, Hammock BD, Zhu Y. Inhibition of soluble epoxide hydrolase ameliorates hyperhomocysteinemia-induced hepatic steatosis by enhancing β-oxidation of fatty acid in mice. Am J Physiol Gastrointest Liver Physiol 2019; 316:G527-G538. [PMID: 30789748 PMCID: PMC6483021 DOI: 10.1152/ajpgi.00148.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatic steatosis is the beginning phase of nonalcoholic fatty liver disease, and hyperhomocysteinemia (HHcy) is a significant risk factor. Soluble epoxide hydrolase (sEH) hydrolyzes epoxyeicosatrienoic acids (EETs) and other epoxy fatty acids, attenuating their cardiovascular protective effects. However, the involvement of sEH in HHcy-induced hepatic steatosis is unknown. The current study aimed to explore the role of sEH in HHcy-induced lipid disorder. We fed 6-wk-old male mice a chow diet or 2% (wt/wt) high-metnionine diet for 8 wk to establish the HHcy model. A high level of homocysteine induced lipid accumulation in vivo and in vitro, which was concomitant with the increased activity and expression of sEH. Treatment with a highly selective specific sEH inhibitor (0.8 mg·kg-1·day-1 for the animal model and 1 μM for cells) prevented HHcy-induced lipid accumulation in vivo and in vitro. Inhibition of sEH activated the peroxisome proliferator-activated receptor-α (PPAR-α), as evidenced by elevated β-oxidation of fatty acids and the expression of PPAR-α target genes in HHcy-induced hepatic steatosis. In primary cultured hepatocytes, the effect of sEH inhibition on PPAR-α activation was further confirmed by a marked increase in PPAR-response element luciferase activity, which was reversed by knock down of PPAR-α. Of note, 11,12-EET ligand dependently activated PPAR-α. Thus increased sEH activity is a key determinant in the pathogenesis of HHcy-induced hepatic steatosis, and sEH inhibition could be an effective treatment for HHcy-induced hepatic steatosis. NEW & NOTEWORTHY In the current study, we demonstrated that upregulation of soluble epoxide hydrolase (sEH) is involved in the hyperhomocysteinemia (HHcy)-caused hepatic steatosis in an HHcy mouse model and in murine primary hepatocytes. Improving hepatic steatosis in HHcy mice by pharmacological inhibition of sEH to activate peroxisome proliferator-activated receptor-α was ligand dependent, and sEH could be a potential therapeutic target for the treatment of nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Liu Yao
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Boyang Cao
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Qian Cheng
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Wenbin Cai
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chenji Ye
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jing Liang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Wenli Liu
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Lu Tan
- 2Department of Laboratory Animal Science and Technology, Tianjin Medical University, Tianjin, China
| | - Meng Yan
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Bochuan Li
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jinlong He
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Sung Hee Hwang
- 3Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center, Davis, California
| | - Xu Zhang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunjiong Wang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Ding Ai
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Bruce D. Hammock
- 3Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center, Davis, California
| | - Yi Zhu
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
30
|
Wang X, Lu L, Tan Y, Jiang L, Zhao M, Gao E, Yu S, Liu J. GPR 30 reduces myocardial infarct area and fibrosis in female ovariectomized mice by activating the PI3K/AKT pathway. Life Sci 2019; 226:22-32. [PMID: 30905784 DOI: 10.1016/j.lfs.2019.03.049] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/14/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022]
Abstract
AIMS Estrogen plays an important role in cardioprotection. Animal experiments showed that the G-protein coupled estrogen receptor 30 (GPR30) specific agonist G1 could reduce post-ischemic dysfunction and inhibit cardiac fibroblast proliferation. However, the underlying mechanism of action is not clear. The current study tests the hypothesis that GPR30 reduces myocardial infarct area and fibrosis in female ovariectomized (OVX) mice by activating the PI3K/AKT pathway. MAIN METHODS In this study, we established a myocardial infarction (MI) animal model derived from OVX C57BL/6 female mice, and investigated the effect of G1 on cardiac function by echocardiography and Hemodynamics, morphology and expression of fibrosis-related and apoptosis-related proteins by Masson's trichrome and H&E, Immunofluorescence, Western blotting and TUNEL. KEY FINDINGS Combination with OVX significantly increased myocardial fibrosis and MI area compared to MI treatment alone, as determined by echocardiography and hemodynamics. Further addition of G1 changed the expression of apoptosis-related proteins, decreased the levels of tumor necrosis factor-α and interleukin-10, and reduced the degree of myocardial fibrosis and myocardial infarct area. Primary cultured cardiac fibroblasts (CFs) were subjected to hypoxia/serum deprivation (H/SD) simulating the in vivo ischemia model. When the PI3K/AKT pathway was inhibited by wortmanin in H/SD CFs, G1 failed to induce significant changes in the expression of apoptosis-related proteins. SIGNIFICANCE It suggested that GPR30 may improve cardiac function in female OVX mice by activating the PI3K/AKT pathway and reducing myocardial infarct size and fibrosis.
Collapse
Affiliation(s)
- Xiaowu Wang
- Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Linhe Lu
- Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Yanzhen Tan
- Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Liqing Jiang
- Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Minggao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, PR China
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Shiqiang Yu
- Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Jincheng Liu
- Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
31
|
Hrdlička J, Neckář J, Papoušek F, Husková Z, Kikerlová S, Vaňourková Z, Vernerová Z, Akat F, Vašinová J, Hammock BD, Hwang SH, Imig JD, Falck JR, Červenka L, Kolář F. Epoxyeicosatrienoic Acid-Based Therapy Attenuates the Progression of Postischemic Heart Failure in Normotensive Sprague-Dawley but Not in Hypertensive Ren-2 Transgenic Rats. Front Pharmacol 2019; 10:159. [PMID: 30881303 PMCID: PMC6406051 DOI: 10.3389/fphar.2019.00159] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) and their analogs have been identified as potent antihypertensive compounds with cardio- and renoprotective actions. Here, we examined the effect of EET-A, an orally active EET analog, and c-AUCB, an inhibitor of the EETs degrading enzyme soluble epoxide hydrolase, on the progression of post-myocardial infarction (MI) heart failure (HF) in normotensive Hannover Sprague-Dawley (HanSD) and in heterozygous Ren-2 transgenic rats (TGR) with angiotensin II-dependent hypertension. Adult male rats (12 weeks old) were subjected to 60-min left anterior descending (LAD) coronary artery occlusion or sham (non-MI) operation. Animals were treated with EET-A and c-AUCB (10 and 1 mg/kg/day, respectively) in drinking water, given alone or combined for 5 weeks starting 24 h after MI induction. Left ventricle (LV) function and geometry were assessed by echocardiography before MI and during the progression of HF. At the end of the study, LV function was determined by catheterization and tissue samples were collected. Ischemic mortality due to the incidence of sustained ventricular fibrillation was significantly higher in TGR than in HanSD rats (35.4 and 17.7%, respectively). MI-induced HF markedly increased LV end-diastolic pressure (Ped) and reduced fractional shortening (FS) and the peak rate of pressure development [+(dP/dt)max] in untreated HanSD compared to sham (non-MI) group [Ped: 30.5 ± 3.3 vs. 9.7 ± 1.3 mmHg; FS: 11.1 ± 1.0 vs. 40.8 ± 0.5%; +(dP/dt)max: 3890 ± 291 vs. 5947 ± 309 mmHg/s]. EET-A and c-AUCB, given alone, tended to improve LV function parameters in HanSD rats. Their combination amplified the cardioprotective effect of single therapy and reached significant differences compared to untreated HanSD controls [Ped: 19.4 ± 2.2 mmHg; FS: 14.9 ± 1.0%; +(dP/dt)max: 5278 ± 255 mmHg/s]. In TGR, MI resulted in the impairment of LV function like HanSD rats. All treatments reduced the increased level of albuminuria in TGR compared to untreated MI group, but neither single nor combined EET-based therapy improved LV function. Our results indicate that EET-based therapy attenuates the progression of post-MI HF in HanSD, but not in TGR, even though they exhibited renoprotective action in TGR hypertensive rats.
Collapse
Affiliation(s)
- Jaroslav Hrdlička
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia.,Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Jan Neckář
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia.,Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - František Papoušek
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Zuzana Husková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Soňa Kikerlová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Zdenka Vaňourková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Zdenka Vernerová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Firat Akat
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia.,Department of Physiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Jana Vašinová
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern, Dallas, TX, United States
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - František Kolář
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
32
|
Liu JY. Inhibition of Soluble Epoxide Hydrolase for Renal Health. Front Pharmacol 2019; 9:1551. [PMID: 30687105 PMCID: PMC6335332 DOI: 10.3389/fphar.2018.01551] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 12/19/2018] [Indexed: 12/31/2022] Open
Abstract
A soluble epoxide hydrolase (sEH) mediates the metabolism of epoxy fatty acids to form the corresponding vicinal diols, which are usually inactive or less active than the epoxide substrates. The sEH enzyme presents in many organs, including but not limited to the liver, heart, spleen, lung, and kidney. Here we summarized the changes in the expression and activity of sEH in multiple renal diseases, such as acute kidney injury (AKI), diabetic nephrology (DN), chronic kidney diseases (CKD), hypertension-mediated renal damage, and other renal dysfunctions. We also discussed the pharmacologic effects and the underlying mechanisms of sEH inhibition by using an inhibitor of sEH and/or the generic deletion of sEH on multiple renal diseases. We believe that sEH is a potential therapeutic target for renal dysfunction although the target disease needs further investigation.
Collapse
Affiliation(s)
- Jun-Yan Liu
- Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, China
- Division of Nephrology, Shanghai Tenth Peoples Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Abstract
Therapeutics for arachidonic acid pathways began with the development of non-steroidal anti-inflammatory drugs that inhibit cyclooxygenase (COX). The enzymatic pathways and arachidonic acid metabolites and respective receptors have been successfully targeted and therapeutics developed for pain, inflammation, pulmonary and cardiovascular diseases. These drugs target the COX and lipoxygenase pathways but not the third branch for arachidonic acid metabolism, the cytochrome P450 (CYP) pathway. Small molecule compounds targeting enzymes and CYP epoxy-fatty acid metabolites have evolved rapidly over the last two decades. These therapeutics have primarily focused on inhibiting soluble epoxide hydrolase (sEH) or agonist mimetics for epoxyeicosatrienoic acids (EET). Based on preclinical animal model studies and human studies, major therapeutic indications for these sEH inhibitors and EET mimics/analogs are renal and cardiovascular diseases. Novel small molecules that inhibit sEH have advanced to human clinical trials and demonstrate promise for cardiovascular diseases. Challenges remain for sEH inhibitor and EET analog drug development; however, there is a high likelihood that a drug that acts on this third branch of arachidonic acid metabolism will be utilized to treat a cardiovascular or kidney disease in the next decade.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
34
|
Zarriello S, Tuazon JP, Corey S, Schimmel S, Rajani M, Gorsky A, Incontri D, Hammock BD, Borlongan CV. Humble beginnings with big goals: Small molecule soluble epoxide hydrolase inhibitors for treating CNS disorders. Prog Neurobiol 2018; 172:23-39. [PMID: 30447256 DOI: 10.1016/j.pneurobio.2018.11.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/06/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
Soluble epoxide hydrolase (sEH) degrades epoxides of fatty acids including epoxyeicosatrienoic acid isomers (EETs), which are produced as metabolites of the cytochrome P450 branch of the arachidonic acid pathway. EETs exert a variety of largely beneficial effects in the context of inflammation and vascular regulation. sEH inhibition is shown to be therapeutic in several cardiovascular and renal disorders, as well as in peripheral analgesia, via the increased availability of anti-inflammatory EETs. The success of sEH inhibitors in peripheral systems suggests their potential in targeting inflammation in the central nervous system (CNS) disorders. Here, we describe the current roles of sEH in the pathology and treatment of CNS disorders such as stroke, traumatic brain injury, Parkinson's disease, epilepsy, cognitive impairment, dementia and depression. In view of the robust anti-inflammatory effects of stem cells, we also outlined the potency of stem cell treatment and sEH inhibitors as a combination therapy for these CNS disorders. This review highlights the gaps in current knowledge about the pathologic and therapeutic roles of sEH in CNS disorders, which should guide future basic science research towards translational and clinical applications of sEH inhibitors for treatment of neurological diseases.
Collapse
Affiliation(s)
- Sydney Zarriello
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States
| | - Julian P Tuazon
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States
| | - Sydney Corey
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States
| | - Samantha Schimmel
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States
| | - Mira Rajani
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States
| | - Anna Gorsky
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States
| | - Diego Incontri
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States
| | - Bruce D Hammock
- Department of Entomology & UCD Comprehensive Cancer Center, NIEHS-UCD Superfund Research Program, University of California - Davis, United States.
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, United States.
| |
Collapse
|
35
|
Oni-Orisan A, Cresci S, Jones PG, Theken KN, Spertus JA, Lee CR. Association between the EPHX2 p.Lys55Arg polymorphism and prognosis following an acute coronary syndrome. Prostaglandins Other Lipid Mediat 2018; 138:15-22. [PMID: 30096423 PMCID: PMC6162147 DOI: 10.1016/j.prostaglandins.2018.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/06/2018] [Accepted: 07/31/2018] [Indexed: 01/14/2023]
Abstract
Inhibition of soluble epoxide hydrolase (sEH, EPHX2) elicits potent cardiovascular protective effects in preclinical models of ischemic cardiovascular disease (CVD), and genetic polymorphisms in EPHX2 have been associated with developing ischemic CVD in humans. However, it remains unknown whether EPHX2 variants are associated with prognosis following an ischemic CVD event. We evaluated the association between EPHX2 p.Lys55Arg and p.Arg287Gln genotype with survival in 667 acute coronary syndrome (ACS) patients. No association with p.Arg287Gln genotype was observed (P = 0.598). Caucasian EPHX2 Arg55 carriers (Lys/Arg or Arg/Arg) had a significantly higher risk of 5-year mortality (adjusted hazard ratio [HR] 1.61, 95% confidence interval [CI] 1.01-2.55, P = 0.045). In an independent population of 2712 ACS patients, this association was not replicated (adjusted HR 0.92, 95% CI 0.70-1.21, P = 0.559). In a secondary analysis, Caucasian homozygous Arg55 allele carriers (Arg/Arg) appeared to exhibit a higher risk of cardiovascular mortality (adjusted HR 2.60, 95% CI 1.09-6.17). These results demonstrate that EPHX2 p.Lys55Arg and p.Arg287Gln polymorphisms do not significantly modify survival after an ACS event. Investigation of other sEH metabolism biomarkers in ischemic CVD appears warranted.
Collapse
Affiliation(s)
- Akinyemi Oni-Orisan
- Department of Clinical Pharmacy, UCSF School of Pharmacy, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Sharon Cresci
- Department of Medicine and Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip G Jones
- Department of Cardiovascular Research, Saint Luke's Mid America Heart Institute, Kansas City, MO, USA
| | - Katherine N Theken
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John A Spertus
- Department of Cardiovascular Research, Saint Luke's Mid America Heart Institute, Kansas City, MO, USA; University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
36
|
Zhou C, Huang J, Li Q, Zhan C, He Y, Liu J, Wen Z, Wang DW. Pharmacological Inhibition of Soluble Epoxide Hydrolase Ameliorates Chronic Ethanol-Induced Cardiac Fibrosis by Restoring Autophagic Flux. Alcohol Clin Exp Res 2018; 42:1970-1978. [PMID: 30047995 DOI: 10.1111/acer.13847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Chronic drinking leads to myocardial contractile dysfunction and dilated cardiomyopathy, and cardiac fibrosis is a consequence of these alcoholic injuries. Soluble epoxide hydrolase (sEH) hydrolyzes epoxyeicosatrienoic acids (EETs) to less bioactive diols, and EETs have cardioprotective properties. However, the effects of sEH inhibition in ethanol (EtOH)-induced cardiac fibrosis are unknown. METHODS This study was designed to investigate the role and underlying mechanisms of sEH inhibition in chronic EtOH feeding-induced cardiac fibrosis. C57BL/6J mice were fed a 4% Lieber-DeCarli EtOH diet for 8 weeks, and the sEH inhibitor 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) was administered throughout the experimental period. RESULTS The results showed that chronic EtOH intake led to cardiac dilatation, collagen deposition, and autophagosome accumulation, while TPPU administration ameliorated these effects. In vitro, treating primary cardiac fibroblasts (CFs) with EtOH resulted in CF activation, including alpha smooth muscle actin overexpression, collagen synthesis, and cell migration. Moreover, EtOH disturbed CF autophagic flux, as evidenced by the increased LC3 II/I ratio and SQSTM1 expression, and by the enhanced autophagosome accumulation. TPPU treatment prevented the activation of CF induced by EtOH and restored the impaired autophagic flux by suppressing mTOR activation. CONCLUSIONS Taken together, these findings suggest that sEH pharmacological inhibition may be a unique therapeutic strategy for treating EtOH-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Chi Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jin Huang
- Division of Hematology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Qing Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Chenao Zhan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ying He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jinyan Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
37
|
Harris TR, Kodani S, Rand AA, Yang J, Imai DM, Hwang SH, Hammock BD. Celecoxib Does Not Protect against Fibrosis and Inflammation in a Carbon Tetrachloride-Induced Model of Liver Injury. Mol Pharmacol 2018; 94:834-841. [PMID: 29844231 PMCID: PMC6022802 DOI: 10.1124/mol.118.111831] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/18/2018] [Indexed: 12/11/2022] Open
Abstract
The cyclooxygenase-2 (COX-2) selective inhibitor celecoxib is widely used in the treatment of pain and inflammation. Celecoxib has been explored as a possible treatment of liver fibrosis with contradictory results, depending on the model. The present study reports the effect of celecoxib in a 5-week carbon tetrachloride (CCl4)-induced liver fibrosis mouse model. Celecoxib alone and in combination with inhibitors of the enzyme-soluble epoxide hydrolase (sEH), as well as a dual inhibitor that targets both COX-2 and sEH, were administered via osmotic minipump to mice receiving intraperitoneal injections of CCl4 Collagen deposition was elevated in the mice treated with both celecoxib and CCl4 compared with the control or CCl4-only groups, as assessed by trichrome staining. Histopathology revealed more extensive fibrosis and cell death in the animals treated with both celecoxib and CCl4 compared with all other experimental groups. Although some markers of fibrosis, such as matrix metalloprotease, were unchanged or lowered in the animals treated with both celecoxib and CCl4, overall, hepatic fibrosis was more severe in this group. Cotreatment with celecoxib and an inhibitor of sEH or treatment with a dual inhibitor of COX-2 and sEH decreased the elevated levels of fibrotic markers observed in the group that received both celecoxib and CCl4 Oxylipid analysis revealed that celecoxib reduced the level of prostaglandin E2 relative to the CCl4 only group. Overall, celecoxib treatment did not decrease liver fibrosis in CCl4-treated mice.
Collapse
Affiliation(s)
- Todd R Harris
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center (T.R.H., S.K., A.A.R., J.Y., S.H.H., B.D.H.), and Comparative Pathology Laboratory, School of Veterinary Medicine (D.M.I.), University of California, Davis, California
| | - Sean Kodani
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center (T.R.H., S.K., A.A.R., J.Y., S.H.H., B.D.H.), and Comparative Pathology Laboratory, School of Veterinary Medicine (D.M.I.), University of California, Davis, California
| | - Amy A Rand
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center (T.R.H., S.K., A.A.R., J.Y., S.H.H., B.D.H.), and Comparative Pathology Laboratory, School of Veterinary Medicine (D.M.I.), University of California, Davis, California
| | - Jun Yang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center (T.R.H., S.K., A.A.R., J.Y., S.H.H., B.D.H.), and Comparative Pathology Laboratory, School of Veterinary Medicine (D.M.I.), University of California, Davis, California
| | - Denise M Imai
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center (T.R.H., S.K., A.A.R., J.Y., S.H.H., B.D.H.), and Comparative Pathology Laboratory, School of Veterinary Medicine (D.M.I.), University of California, Davis, California
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center (T.R.H., S.K., A.A.R., J.Y., S.H.H., B.D.H.), and Comparative Pathology Laboratory, School of Veterinary Medicine (D.M.I.), University of California, Davis, California
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center (T.R.H., S.K., A.A.R., J.Y., S.H.H., B.D.H.), and Comparative Pathology Laboratory, School of Veterinary Medicine (D.M.I.), University of California, Davis, California
| |
Collapse
|
38
|
Gui Y, Li D, Chen J, Wang Y, Hu J, Liao C, Deng L, Xiang Q, Yang T, Du X, Zhang S, Xu D. Soluble epoxide hydrolase inhibitors, t-AUCB, downregulated miR-133 in a mouse model of myocardial infarction. Lipids Health Dis 2018; 17:129. [PMID: 29843720 PMCID: PMC5975509 DOI: 10.1186/s12944-018-0780-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 05/15/2018] [Indexed: 12/26/2022] Open
Abstract
Background It has been demonstrated that soluble epoxide hydrolase inhibitors (sEHIs) are protective against ischemia-induced lethal arrhythmias, but the mechanisms involved are unknown. Previously, we showed that sEHIs might reduce the incidence of ischemic arrhythmias by suppressing microRNA-1 (miR-1) in the myocardium. As miR-1 and miR-133 have the same proarrhythmic effects in the heart, we assumed that the beneficial effects of sEHIs might also relate to the regulation of miR-133. Methods A mouse model of myocardial infarction (MI) was established by ligating the coronary artery. The sEHI t-AUCB (trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid) was administered daily for 7 days before MI. Myocardial infarct size and cardiac function was assessed at 24 h post-MI. The miRNA expression profiles of sham and MI mice treated with or without t-AUCB were determined by microarray and verified by real-time PCR. The incidence of arrhythmias was assessed by in vivo electrophysiologic studies. The mRNA levels of miR-133, its target genes (KCNQ1 [potassium voltage-gated channel subfamily Q member 1] and KCNH2 [potassium voltage-gated channel subfamily H member 2]), and serum response factor (SRF) were measured by real-time PCR; KCNQ1, KCNH2, and SRF protein levels were assessed by western blotting. Results We demonstrated that the treatment with sEHIs could reduce infarct size, improve cardia function, and prevent the development of cardiac arrhythmias in MI mice. The expression levels of 14 miRNAs differed between the sham and MI groups. t-AUCB treatment altered the expression of eight miRNAs: two were upregulated and six were downregulated. Of these, the muscle-specific miR-133 was downregulated in the ischemic myocardium. In line with this, up-regulation of miR-133 and down-regulation of KCNQ1 and KCNH2 mRNA/protein were observed in ischemic myocaridum, whereas administration of sEHIs produced an opposite effect. In addition, miR-133 overexpression inhibited expression of the target mRNA, whereas t-AUCB reversed the effects. Furthermore, SRF might participate in the negative regulation of miR-133 by t-AUCB. Conclusions In MI mice, sEHI t-AUCB can repress miR-133, consequently stimulating KCNQ1 and KCNH2 mRNA and protein expression, suggesting a possible mechanism for its potential therapeutic application in ischemic arrhythmias. Electronic supplementary material The online version of this article (10.1186/s12944-018-0780-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yajun Gui
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Da Li
- Department of Geratology, Internal Medicine, the Third Hospital of Changsha, Changsha, Hunan, 410011, China
| | - Jingyuan Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yating Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Caixiu Liao
- Department of Geratology, Internal Medicine, the Third Hospital of Changsha, Changsha, Hunan, 410011, China
| | - Limin Deng
- Center for Pulmonary Vascular Disease, FuWai Hospital & Cardiovascular Institute Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qunyan Xiang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Tao Yang
- Department of Cardiology, Internal Medicine, Changsha Central Hospital, Changsha, Hunan, 410011, China
| | - Xiao Du
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shilan Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
39
|
Guo Y, Luo F, Zhang X, Chen J, Shen L, Zhu Y, Xu D. TPPU enhanced exercise-induced epoxyeicosatrienoic acid concentrations to exert cardioprotection in mice after myocardial infarction. J Cell Mol Med 2018; 22:1489-1500. [PMID: 29265525 PMCID: PMC5824362 DOI: 10.1111/jcmm.13412] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/26/2017] [Indexed: 12/14/2022] Open
Abstract
Exercise training (ET) is a safe and efficacious therapeutic approach for myocardial infarction (MI). Given the numerous benefits of exercise, exercise-induced mediators may be promising treatment targets for MI. C57BL/6 mice were fed 1-trifluoromethoxyphenyl-3-(1-propionylpiperidine-4-yl) urea (TPPU), a novel soluble epoxide hydrolase inhibitor (sEHI), to increase epoxyeicosatrienoic acid (EET) levels, for 1 week before undergoing MI surgery. After 1-week recovery, the mice followed a prescribed exercise programme. Bone marrow-derived endothelial progenitor cells (EPCs) were isolated from the mice after 4 weeks of exercise and cultured for 7 days. Angiogenesis around the ischaemic area, EPC functions, and the expression of microRNA-126 (miR-126) and its target gene Spred1 were measured. The results were confirmed in vitro by adding TPPU to EPC culture medium. ET significantly increased serum EET levels and promoted angiogenesis after MI. TPPU enhanced the effects of ET to reduce the infarct area and improve cardiac function after MI. ET increased EPC function and miR-126 expression, which were further enhanced by TPPU, while Spred1 expression was significantly down-regulated. Additionally, the protein kinase B/glycogen synthase kinase 3β (AKT/GSK3β) signalling pathway was activated after the administration of TPPU. EETs are a potential mediator of exercise-induced cardioprotection in mice after MI. TPPU enhances exercise-induced cardiac recovery in mice after MI by increasing EET levels and promoting angiogenesis around the ischaemic area.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/metabolism
- 8,11,14-Eicosatrienoic Acid/pharmacology
- Adaptor Proteins, Signal Transducing
- Animals
- Bone Marrow Cells/cytology
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/metabolism
- Cardiotonic Agents/metabolism
- Cardiotonic Agents/pharmacology
- Coronary Vessels/surgery
- Disease Models, Animal
- Endothelial Progenitor Cells/cytology
- Endothelial Progenitor Cells/drug effects
- Endothelial Progenitor Cells/metabolism
- Enzyme Inhibitors/pharmacology
- Epoxide Hydrolases/antagonists & inhibitors
- Epoxide Hydrolases/genetics
- Epoxide Hydrolases/metabolism
- Gene Expression Regulation
- Glycogen Synthase Kinase 3 beta/genetics
- Glycogen Synthase Kinase 3 beta/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Myocardial Infarction/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/physiopathology
- Myocardial Infarction/therapy
- Neovascularization, Physiologic
- Phenylurea Compounds/pharmacology
- Physical Conditioning, Animal
- Piperidines/pharmacology
- Primary Cell Culture
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Yuan Guo
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Fei Luo
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xv Zhang
- Department of Physiology and PathophysiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsTianjin Medical UniversityTianjinChina
| | - Jingyuan Chen
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Li Shen
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yi Zhu
- Department of Physiology and PathophysiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsTianjin Medical UniversityTianjinChina
| | - Danyan Xu
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
40
|
Moon SH, Liu X, Cedars AM, Yang K, Kiebish MA, Joseph SM, Kelley J, Jenkins CM, Gross RW. Heart failure-induced activation of phospholipase iPLA 2γ generates hydroxyeicosatetraenoic acids opening the mitochondrial permeability transition pore. J Biol Chem 2017; 293:115-129. [PMID: 29158256 DOI: 10.1074/jbc.ra117.000405] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/08/2017] [Indexed: 11/06/2022] Open
Abstract
Congestive heart failure typically arises from cardiac myocyte necrosis/apoptosis, associated with the pathological opening of the mitochondrial permeability transition pore (mPTP). mPTP opening decreases the mitochondrial membrane potential leading to the activation of Ca2+-independent phospholipase A2γ (iPLA2γ) and the production of downstream toxic metabolites. However, the array of enzymatic mediators and the exact chemical mechanisms responsible for modulating myocardial mPTP opening remain unclear. Herein, we demonstrate that human heart failure activates specific myocardial mitochondrial phospholipases that increase Ca2+-dependent production of toxic hydroxyeicosatetraenoic acids (HETEs) and attenuate the activity of phospholipases that promote the synthesis of protective epoxyeicosatrienoic acids (EETs). Mechanistically, HETEs activated the Ca2+-induced opening of the mPTP in failing human myocardium, and the highly selective pharmacological blockade of either iPLA2γ or lipoxygenases attenuated mPTP opening in failing hearts. In contrast, pharmacological inhibition of cytochrome P450 epoxygenases opened the myocardial mPTP in human heart mitochondria. Remarkably, the major mitochondrial phospholipase responsible for Ca2+-activated release of arachidonic acid (AA) in mitochondria from non-failing hearts was calcium-dependent phospholipase A2ζ (cPLA2ζ) identified by sequential column chromatographies and activity-based protein profiling. In contrast, iPLA2γ predominated in failing human myocardium. Stable isotope kinetics revealed that in non-failing human hearts, cPLA2ζ metabolically channels arachidonic acid into EETs, whereas in failing hearts, increased iPLA2γ activity channels AA into toxic HETEs. These results mechanistically identify the sequelae of pathological remodeling of human mitochondrial phospholipases in failing myocardium. This remodeling metabolically channels AA into toxic HETEs promoting mPTP opening, which induces necrosis/apoptosis leading to further progression of heart failure.
Collapse
Affiliation(s)
- Sung Ho Moon
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xinping Liu
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Ari M Cedars
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kui Yang
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Michael A Kiebish
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Susan M Joseph
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - John Kelley
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Christopher M Jenkins
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Richard W Gross
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110; Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Chemistry, Washington University, St. Louis, Missouri 63130.
| |
Collapse
|
41
|
Jamieson KL, Endo T, Darwesh AM, Samokhvalov V, Seubert JM. Cytochrome P450-derived eicosanoids and heart function. Pharmacol Ther 2017; 179:47-83. [PMID: 28551025 DOI: 10.1016/j.pharmthera.2017.05.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
42
|
López JE, Jaradeh K, Silva E, Aminololama-Shakeri S, Simpson PC. A method to increase reproducibility in adult ventricular myocyte sizing and flow cytometry: Avoiding cell size bias in single cell preparations. PLoS One 2017; 12:e0186792. [PMID: 29084228 PMCID: PMC5662089 DOI: 10.1371/journal.pone.0186792] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/07/2017] [Indexed: 12/24/2022] Open
Abstract
RATIONALE Flow cytometry (FCM) of ventricular myocytes (VMs) is an emerging technology in adult cardiac research that is challenged by the wide variety of VM shapes and sizes. Cellular variability and cytometer flow cell size can affect cytometer performance. These two factors of variance limit assay validity and reproducibility across laboratories. Washing and filtering of ventricular cells in suspension are routinely done to prevent cell clumping and minimize data variability without the appropriate standardization. We hypothesize that washing and filtering arbitrarily biases towards sampling smaller VMs than what actually exist in the adult heart. OBJECTIVE To determine the impact of washing and filtering on adult ventricular cells for cell sizing and FCM. METHODS AND RESULTS Left ventricular cardiac cells in single-cell suspension were harvested from New Zealand White rabbits and fixed prior to analysis. Each ventricular sample was aliquoted before washing or filtering through a 40, 70, 100 or 200μm mesh. The outcomes of the study are VM volume by Coulter Multisizer and light-scatter signatures by FCM. Data are presented as mean±SD. Myocyte volumes without washing or filtering (NF) served as the "gold standard" within the sample and ranged from 11,017 to 46,926μm3. Filtering each animal sample through a 200μm mesh caused no variation in the post-filtration volume (1.01+0.01 fold vs. NF, n = 4 rabbits, p = 0.999) with an intra-assay coefficient of variation (%CV) of <5% for all 4 samples. Filtering each sample through a 40, 70 or 100μm mesh invariably reduced the post-filtration volume by 41±10%, 9.0±0.8% and 8.8±0.8% respectively (n = 4 rabbits, p<0.0001), and increased the %CV (18% to 1.3%). The high light-scatter signature by FCM, a simple parameter for the identification of ventricular myocytes, was measured after washing and filtering. Washing discarded VMs and filtering cells through a 40 or 100μm mesh reduced larger VM by 46% or 11% respectively (n = 6 from 2 rabbits, p<0.001). CONCLUSION Washing and filtering VM suspensions through meshes 100μm or less biases myocyte volumes to smaller sizes, excludes larger cells, and increases VM variability. These findings indicate that validity and reproducibility across laboratories can be compromised unless cell preparation is standardized. We propose no wash prior to fixation and a 200μm mesh for filtrations to provide a reproducible standard for VM studies using FCM.
Collapse
Affiliation(s)
- Javier E. López
- University of California, Davis, CA, United States of America
| | - Katrin Jaradeh
- University of California, Davis, CA, United States of America
| | - Emmanuel Silva
- University of California, Davis, CA, United States of America
| | | | - Paul C. Simpson
- VA Medical Center and University of California, San Francisco, CA, United States of America
| |
Collapse
|
43
|
Gui YJ, Yang T, Liu Q, Liao CX, Chen JY, Wang YT, Hu JH, Xu DY. Soluble epoxide hydrolase inhibitors, t-AUCB, regulated microRNA-1 and its target genes in myocardial infarction mice. Oncotarget 2017; 8:94635-94649. [PMID: 29212255 PMCID: PMC5706901 DOI: 10.18632/oncotarget.21831] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/20/2017] [Indexed: 02/04/2023] Open
Abstract
Purpose Soluble epoxide hydrolase inhibitors (sEHIs) had been demonstrated to produce cardioprotective effects against ischemia-induced lethal arrhythmias, but the exact mechanisms remain unknown. The present study was designed to investigate whether the beneficial effects of sEHIs are related to regulation of microRNA-1, which was a proarrhythmic factor in the ischemic heart. Methods A mousemyocardial infarction (MI) model was established by ligating the coronary artery. sEHI t-AUCB (0.2, 1, 5 mg/L in drinking-water) was administered daily seven days before MI. The incidence of arrhythmias was assessed by in vivo electrophysiologic studies. miR-1, KCNJ2 (encoding the K+ channel subunit Kir2.1), and GJA1 (encoding connexin 43 [Cx43]) mRNA were measured by real-time PCR; Kir2.1 and Cx43 protein were assessed by western blotting and immunohistochemistry. Results We demonstrated that sEHIs reduced the myocardium infarct size and incidence of inducible arrhythmias in MI mice. Up-regulation of miR-1 and down-regulation of KCNJ2/Kir2.1 and GJA1/Cx43 mRNA/protein were observed in ischemic myocaridum, whereas administration of sEHIs produced an opposite effect. In addition, miR-1 overexpression inhibited expression of the target mRNA and their corresponding proteins, whereas t-AUCB reversed the effects. Our results further revealed that PI3K/Akt signaling pathway might participate in the negatively regulation of miR-1 by sEHi. Conclusions We conclude that sEHIs can repress miR-1, thus stimulate expression of KCNJ2/Kir2.1 and GJA1/Cx43 mRNA/protein in MI mice, suggesting a possible mechanism for its potential therapeutic application in ischemic arrhythmias.
Collapse
Affiliation(s)
- Ya-Jun Gui
- Department of Cardiology, Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Tao Yang
- Department of Cardiology, Internal Medicine, Changsha Central Hospital, Changsha, Hunan 410011, China
| | - Qiong Liu
- Department of Cardiology, Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Cai-Xiu Liao
- Department of Geratology, Internal Medicine, The Third Hospital of Changsha, Changsha, Hunan 410011, China
| | - Jing-Yuan Chen
- Department of Cardiology, Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ya-Ting Wang
- Department of Cardiology, Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jia-Hui Hu
- Department of Cardiology, Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Dan-Yan Xu
- Department of Cardiology, Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
44
|
Jamieson KL, Samokhvalov V, Akhnokh MK, Lee K, Cho WJ, Takawale A, Wang X, Kassiri Z, Seubert JM. Genetic deletion of soluble epoxide hydrolase provides cardioprotective responses following myocardial infarction in aged mice. Prostaglandins Other Lipid Mediat 2017; 132:47-58. [DOI: 10.1016/j.prostaglandins.2017.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/24/2016] [Accepted: 01/03/2017] [Indexed: 01/17/2023]
|
45
|
Soluble Epoxide Hydrolase Inhibition Protected against Angiotensin II-induced Adventitial Remodeling. Sci Rep 2017; 7:6926. [PMID: 28761179 PMCID: PMC5537243 DOI: 10.1038/s41598-017-07512-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/29/2017] [Indexed: 01/15/2023] Open
Abstract
Epoxyeicosatrienoic acids (EETs), the metabolites of cytochrome P450 epoxygenases derived from arachidonic acid, exert important biological activities in maintaining cardiovascular homeostasis. Soluble epoxide hydrolase (sEH) hydrolyzes EETs to less biologically active dihydroxyeicosatrienoic acids. However, the effects of sEH inhibition on adventitial remodeling remain inconclusive. In this study, the adventitial remodeling model was established by continuous Ang II infusion for 2 weeks in C57BL/6 J mice, before which sEH inhibitor 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) was administered by gavage. Adventitial remodeling was evaluated by histological analysis, western blot, immunofluorescent staining, calcium imaging, CCK-8 and transwell assay. Results showed that Ang II infusion significantly induced vessel wall thickening, collagen deposition, and overexpression of α-SMA and PCNA in aortic adventitia, respectively. Interestingly, these injuries were attenuated by TPPU administration. Additionally, TPPU pretreatment overtly prevented Ang II-induced primary adventitial fibroblasts activation, characterized by differentiation, proliferation, migration, and collagen synthesis via Ca2+-calcineurin/NFATc3 signaling pathway in vitro. In summary, our results suggest that inhibition of sEH could be considered as a novel therapeutic strategy to treat adventitial remodeling related disorders.
Collapse
|
46
|
Frederich BJ, Timofeyev V, Thai PN, Haddad MJ, Poe AJ, Lau VC, Moshref M, Knowlton AA, Sirish P, Chiamvimonvat N. Electrotaxis of cardiac progenitor cells, cardiac fibroblasts, and induced pluripotent stem cell-derived cardiac progenitor cells requires serum and is directed via PI3'K pathways. Heart Rhythm 2017; 14:1685-1692. [PMID: 28668623 DOI: 10.1016/j.hrthm.2017.06.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND The limited regenerative capacity of cardiac tissue has long been an obstacle to treating damaged myocardium. Cell-based therapy offers an enormous potential to the current treatment paradigms. However, the efficacy of regenerative therapies remains limited by inefficient delivery and engraftment. Electrotaxis (electrically guided cell movement) has been clinically used to improve recovery in a number of tissues but has not been investigated for treating myocardial damage. OBJECTIVE The purpose of this study was to test the electrotactic behaviors of several types of cardiac cells. METHODS Cardiac progenitor cells (CPCs), cardiac fibroblasts (CFs), and human induced pluripotent stem cell-derived cardiac progenitor cells (hiPSC-CPCs) were used. RESULTS CPCs and CFs electrotax toward the anode of a direct current electric field, whereas hiPSC-CPCs electrotax toward the cathode. The voltage-dependent electrotaxis of CPCs and CFs requires the presence of serum in the media. Addition of soluble vascular cell adhesion molecule to serum-free media restores directed migration. We provide evidence that CPC and CF electrotaxis is mediated through phosphatidylinositide 3-kinase signaling. In addition, very late antigen-4, an integrin and growth factor receptor, is required for electrotaxis and localizes to the anodal edge of CPCs in response to direct current electric field. The hiPSC-derived CPCs do not express very late antigen-4, migrate toward the cathode in a voltage-dependent manner, and, similar to CPCs and CFs, require media serum and phosphatidylinositide 3-kinase activity for electrotaxis. CONCLUSION The electrotactic behaviors of these therapeutic cardiac cells may be used to improve cell-based therapy for recovering function in damaged myocardium.
Collapse
Affiliation(s)
- Bert J Frederich
- Division of Cardiovascular Medicine, University of California, Davis, California
| | - Valeriy Timofeyev
- Division of Cardiovascular Medicine, University of California, Davis, California
| | - Phung N Thai
- Division of Cardiovascular Medicine, University of California, Davis, California
| | - Michael J Haddad
- Division of Cardiovascular Medicine, University of California, Davis, California
| | - Adam J Poe
- Division of Cardiovascular Medicine, University of California, Davis, California
| | - Victor C Lau
- Division of Cardiovascular Medicine, University of California, Davis, California
| | - Maryam Moshref
- Division of Cardiovascular Medicine, University of California, Davis, California
| | - Anne A Knowlton
- Division of Cardiovascular Medicine, University of California, Davis, California; US Department of Veterans Affairs, Northern California Health Care System, Mather, California
| | - Padmini Sirish
- Division of Cardiovascular Medicine, University of California, Davis, California.
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, University of California, Davis, California; US Department of Veterans Affairs, Northern California Health Care System, Mather, California.
| |
Collapse
|
47
|
Halade GV, Kain V, Ingle KA, Prabhu SD. Interaction of 12/15-lipoxygenase with fatty acids alters the leukocyte kinetics leading to improved postmyocardial infarction healing. Am J Physiol Heart Circ Physiol 2017; 313:H89-H102. [PMID: 28411230 PMCID: PMC5538863 DOI: 10.1152/ajpheart.00040.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/11/2017] [Accepted: 04/11/2017] [Indexed: 12/22/2022]
Abstract
The metabolic transformation of fatty acids to form oxylipids using 12/15-lipoxygenase (LOX) can promote either resolving or nonresolving inflammation. However, the mechanism of how 12/15-LOX interacts with polyunsaturated fatty acids (PUFA) in postmyocardial infarction (post-MI) healing is unclear. Here, we reported the role of 12/15-LOX in post-MI cardiac remodeling in a PUFA [10% (wt/wt), 22 kcal]-enriched environment. Wild-type (WT; C57BL/6J) and 12/15-LOX-null (12/15-LOX-/-) male mice of 8-12 wk of age were fed a PUFA-enriched diet for 1 mo and subjected to permanent coronary artery ligation. Post-MI mice were monitored for day 1 or until day 5 along with standard diet-fed MI controls. No-MI surgery mice served as naïve controls. PUFA-fed WT and 12/15-LOX-/- mice improved ejection fraction and reduced lung edema greater than WT mice at day 5 post-MI (P < 0.05). Post-MI, neutrophil density was decreased in PUFA-fed WT and 12/15-LOX-/- mice at day 1 (P < 0.05). Deletion of 12/15-LOX in mice led to increased cytochrome P-450-derived bioactive lipid mediator epoxyeicosatrienoic acids (EETs), i.e., 11,12-EpETrE and 14,15-EpETrE, which were further enhanced by acute PUFA intake post-MI. Macrophage density was decreased in WT + PUFA and 12/15-LOX-/- mice compared with their respective standard diet-fed WT controls at day 5 post-MI. 12/15-LOX-/- + PUFA mice displayed an increased expression of chemokine (C-C motif) ligand 2 and reparative macrophages markers (Ym-1, Mrc-1, and Arg-1, all P < 0.05) in the infarcted area. Furthermore, 12/15-LOX-/- mice, with or without PUFA, showed reduced collagen deposition at day 5 post-MI compared with WT mice. In conclusion, deletion of 12/15-LOX and short-term exposure of PUFA promoted leukocyte clearance, thereby limiting cardiac remodeling and promoting an effective resolution of inflammation.NEW & NOTEWORTHY This study determined that 1) deletion of 12/15-lipoxygenase (LOX) promotes the generation of epoxyeicosatrienoic acids, the cytochrome P-450-derived metabolites in postmyocardial infarction (post-MI) healing; 2) acute exposure of fatty acids to 12/15-LOX-/- mice drives leukocyte (neutrophils and macrophages) clearance post-MI; and 3) metabolic transformation of fats is the significant contributor in leukocyte clearance to drive either resolving or nonresolving inflammation post-MI.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Kevin A Ingle
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Sumanth D Prabhu
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
48
|
Zhang S, Lin X, Li G, Shen X, Niu D, Lu G, Fu X, Chen Y, Cui M, Bai Y. Knockout of Eva1a leads to rapid development of heart failure by impairing autophagy. Cell Death Dis 2017; 8:e2586. [PMID: 28151473 PMCID: PMC5386466 DOI: 10.1038/cddis.2017.17] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 01/13/2023]
Abstract
EVA1A (Eva-1 homologue A) is a novel lysosome and endoplasmic reticulum-associated protein that can regulate cell autophagy and apoptosis. Eva1a is expressed in the myocardium, but its function in myocytes has not yet been investigated. Therefore, we generated inducible, cardiomyocyte-specific Eva1a knockout mice with an aim to determine the role of Eva1a in cardiac remodelling in the adult heart. Data from experiments showed that loss of Eva1a in the adult heart increased cardiac fibrosis, promoted cardiac hypertrophy, and led to cardiomyopathy and death. Further investigation suggested that this effect was associated with impaired autophagy and increased apoptosis in Eva1a knockout hearts. Moreover, knockout of Eva1a activated Mtor signalling and the subsequent inhibition of autophagy. In addition, Eva1a knockout hearts showed disorganized sarcomere structure and mitochondrial misalignment and aggregation, leading to the lack of ATP generation. Collectively, these data demonstrated that Eva1a improves cardiac function and inhibits cardiac hypertrophy and fibrosis by increasing autophagy. In conclusion, our results demonstrated that Eva1a may have an important role in maintaining cardiac homeostasis.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China
| | - Xin Lin
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Ge Li
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Xue Shen
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Di Niu
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Guang Lu
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Xin Fu
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China
| | - Yingyu Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Ming Cui
- Department of Cardiology, Peking University Third Hospital, Beijing 100191, China
| | - Yun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
49
|
Liu Q, Zhao X, Peng R, Wang M, Zhao W, Gui YJ, Liao CX, Xu DY. Soluble epoxide hydrolase inhibitors might prevent ischemic arrhythmias via microRNA-1 repression in primary neonatal mouse ventricular myocytes. MOLECULAR BIOSYSTEMS 2017; 13:556-564. [PMID: 28112313 DOI: 10.1039/c6mb00824k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ischemic arrhythmias are the main causes of sudden cardiac death.
Collapse
Affiliation(s)
- Qiong Liu
- Department of Cardiovascular Medicine
- The Second Xiangya Hospital
- Central South University
- Changsha
- China
| | - Xuan Zhao
- Department of Cardiovascular Medicine
- People's Hospital of Dongying
- Dongying
- China
| | - Ran Peng
- Department of Cardiovascular Medicine
- The Second Xiangya Hospital
- Central South University
- Changsha
- China
| | - Mi Wang
- Department of Cardiovascular Medicine
- The Second Xiangya Hospital
- Central South University
- Changsha
- China
| | - Wang Zhao
- Department of Cardiovascular Medicine
- The Second Xiangya Hospital
- Central South University
- Changsha
- China
| | - Ya-jun Gui
- Department of Cardiovascular Medicine
- The Second Xiangya Hospital
- Central South University
- Changsha
- China
| | - Cai-xiu Liao
- Department of Cardiovascular Medicine
- The Second Xiangya Hospital
- Central South University
- Changsha
- China
| | - Dan-yan Xu
- Department of Cardiovascular Medicine
- The Second Xiangya Hospital
- Central South University
- Changsha
- China
| |
Collapse
|
50
|
Liu JY, Morisseau C, Huang H, Hammock BD. Screening of soluble epoxide hydrolase inhibitory ingredients from traditional Chinese medicines for anti-inflammatory use. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:475-482. [PMID: 27702689 PMCID: PMC5584568 DOI: 10.1016/j.jep.2016.09.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/12/2016] [Accepted: 09/21/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inhibition of soluble epoxide hydrolase (sEH) has been extensively reported to be anti-inflammatory in multiple animal models. Some anti-inflammatory traditional Chinese medicines (TCMs) and a few natural compounds were also found to be inhibitory to sEH in vitro. AIM OF THE STUDY To determine whether the active intergradient (AI) against sEH of anti-inflammatory TCMs in vitro is anti-inflammatory in vivo and the sEH inhibitory action of the AI contributes to its anti-inflammatory effect in vivo. MATERIALS AND METHODS In vitro inhibition assay of the sEH was conducted for the methanol and ethanol extracts of 27 anti-inflammatory TCMs. Two potent extracts were subject to further separation guided by bioassay to afford promising AI against sEH in vitro [Fr.5 of the crude ethanol extract of Rhizoma coptidis (FFCERC)]. Finally, the in vivo anti-inflammatory effect and sEH inhibitory potency of FFCERC was evaluated in a lipopolysacchride (LPS)-challenged murine model of acute systemic inflammation. The inflammatory status was characterized by the inflammatory cytokines TNF-α and interleukin-6 (IL-6) and sEH inhibitory function was evaluated by the plasma levels of epoxyeicosantrienoic acids (EETs) and dihydroxyeicosatrienoic acids (DHETs), which are the sEH mediated substrates and products, respectively. RESULTS At the concentration of 25µg/mL, the crude ethanol extracts of 6 TCMs including Herba Asari, Radix Polygalae, Fructus Amomi, Radix Astragali, Radix Scutellariae, and Rhizoma Coptidis were potent against sEH. The crude extracts of Herba Asari and Rhizoma Coptidis were selected for further separation to afford FFCERC as the most promising AI for in vivo evaluation. Oral administration of FFCERC attenuated the significant increase in TNF-α and IL-6 caused by LPS challenge in a dose-dependent manner. In parallel, oral administration of FFCERC shifted the changes in plasma levels of EETs and DHETs caused by LPS-challenge like a synthetic sEH inhibitor. CONCLUSIONS A sEH inhibitory AI from Rhizoma Coptidis is anti-inflammatory and the inhibition of sEH contributes to this biological effect, indicating that sEH may be at least one of multiple therapeutic targets for relevant TCMs.
Collapse
Affiliation(s)
- Jun-Yan Liu
- Center for Nephrology and Clinical Metabolomics, Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Mid Yanchang Rd, Shanghai 200072, PR China; Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California, One Shields Avenue, Davis, CA 95616, United States.
| | - Christophe Morisseau
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California, One Shields Avenue, Davis, CA 95616, United States
| | - Huazhang Huang
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California, One Shields Avenue, Davis, CA 95616, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California, One Shields Avenue, Davis, CA 95616, United States
| |
Collapse
|