1
|
Zhu X, Soh YK, Wan M, Pang JKS, Leow WL, Tian C, Soh BS, Chan ECY. Development of human embryonic stem cell-derived cardiomyocytes and application of fluorescence probe substrate for characterization of cytochrome P450 enzyme 2J2. Drug Metab Dispos 2025; 53:100053. [PMID: 40153898 DOI: 10.1016/j.dmd.2025.100053] [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: 09/23/2024] [Accepted: 02/07/2025] [Indexed: 04/01/2025] Open
Abstract
Cardiac cytochrome P450 2J2 (CYP2J2) plays a significant role in cardiovascular homeostasis due to its dual functions in drug metabolism and the epoxidation of polyunsaturated fatty acids. Additionally, the inhibition of CYP2J2 by xenobiotics has been linked to drug-induced cardiotoxicity, warranting further investigation of this critical enzyme in cardiac systems. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) are physiologically relevant in vitro models that recapitulate relevant phenotypes important for cardiovascular research. However, no studies have so far characterized CYP2J2 expression and activities in these models. Here, we developed and validated H7 hESC-CMs as suitable in vitro models for investigating CYP2J2 in drug metabolism and cardiotoxicity. We first performed the genotyping and confirmed the presence of wild-type CYP2J2∗1/∗1 alleles in wild-type hESCs. Our optimized cardiomyocyte differentiation protocols yielded virtually pure (93.3% ± 6.8%) hESC-CMs, which exhibited P450 epoxygenase mRNA-expression profiles consistent with human cardiomyocytes, with CYP2J2 as the dominant isozyme and minor contributions from CYP2C8 and CYP2C9. By employing a CYP2J2-selective fluorescent substrate, ER-BnXPI, and astemizole as probe substrates, CYP2J2-mediated demethylation of both substrates exhibited typical Michaelis-Menten kinetics, which confirms functional CYP2J2 activities in vitro. Additionally, we demonstrated the capacity of CYP2J2 for arachidonic acid epoxidation, validating its ability to metabolize polyunsaturated fatty acid substrates. Finally, CYP2J2 activity in hESC-CMs was significantly inhibited by danazol and dronedarone, which are established CYP2J2 inhibitors known to cause cardiotoxicity. Ultimately, our study sheds novel insights on hESC-CMs as a suitable model for investigating CYP2J2-mediated metabolism and its inhibition in vitro. SIGNIFICANCE STATEMENT: H7 human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were developed and validated as an in vitro model for investigating CYP2J2-mediated drug metabolism and its inhibition. By characterizing CYP2J2 transcriptional expression, catalytic activity, and inhibition response to established CYP2J2 inhibitors, our study confirmed functional CYP2J2 in hESC-CMs and ascertained that the model recapitulates the physiology of primary cardiomyocytes. This pioneering research highlights the potential of hESC-CMs in advancing our understanding of CYP2J2-mediated metabolism, its inhibition, and implications in drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xingyu Zhu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yee Kiat Soh
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Mingxin Wan
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Jeremy Kah Sheng Pang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wei Liang Leow
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Chong Tian
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore.
| | - Boon Seng Soh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.
| | - Eric Chun Yong Chan
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore.
| |
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
|
Gerges SH, El-Kadi AOS. Changes in cardiovascular arachidonic acid metabolism in experimental models of menopause and implications on postmenopausal cardiac hypertrophy. Prostaglandins Other Lipid Mediat 2024; 173:106851. [PMID: 38740361 DOI: 10.1016/j.prostaglandins.2024.106851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Menopause is a normal stage in the human female aging process characterized by the cessation of menstruation and the ovarian production of estrogen and progesterone hormones. Menopause is associated with an increased risk of several different diseases. Cardiovascular diseases are generally less common in females than in age-matched males. However, this female advantage is lost after menopause. Cardiac hypertrophy is a disease characterized by increased cardiac size that develops as a response to chronic overload or stress. Similar to other cardiovascular diseases, the risk of cardiac hypertrophy significantly increases after menopause. However, the exact underlying mechanisms are not yet fully elucidated. Several studies have shown that surgical or chemical induction of menopause in experimental animals is associated with cardiac hypertrophy, or aggravates cardiac hypertrophy induced by other stressors. Arachidonic acid (AA) released from the myocardial phospholipids is metabolized by cardiac cytochrome P450 (CYP), cyclooxygenase (COX), and lipoxygenase (LOX) enzymes to produce several eicosanoids. AA-metabolizing enzymes and their respective metabolites play an important role in the pathogenesis of cardiac hypertrophy. Menopause is associated with changes in the cardiovascular levels of CYP, COX, and LOX enzymes and the levels of their metabolites. It is possible that these changes might play a role in the increased risk of cardiac hypertrophy after menopause.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
4
|
Jiang S, Han S, Wang DW. The involvement of soluble epoxide hydrolase in the development of cardiovascular diseases through epoxyeicosatrienoic acids. Front Pharmacol 2024; 15:1358256. [PMID: 38628644 PMCID: PMC11019020 DOI: 10.3389/fphar.2024.1358256] [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: 01/16/2024] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Arachidonic acid (AA) has three main metabolic pathways: the cycloxygenases (COXs) pathway, the lipoxygenases (LOXs) pathway, and the cytochrome P450s (CYPs) pathway. AA produces epoxyeicosatrienoic acids (EETs) through the CYPs pathway. EETs are very unstable in vivo and can be degraded in seconds to minutes. EETs have multiple degradation pathways, but are mainly degraded in the presence of soluble epoxide hydrolase (sEH). sEH is an enzyme of bifunctional nature, and current research focuses on the activity of its C-terminal epoxide hydrolase (sEH-H), which hydrolyzes the EETs to the corresponding inactive or low activity diol. Previous studies have reported that EETs have cardiovascular protective effects, and the activity of sEH-H plays a role by degrading EETs and inhibiting their protective effects. The activity of sEH-H plays a different role in different cells, such as inhibiting endothelial cell proliferation and migration, but promoting vascular smooth muscle cell proliferation and migration. Therefore, it is of interest whether the activity of sEH-H is involved in the initiation and progression of cardiovascular diseases by affecting the function of different cells through EETs.
Collapse
Affiliation(s)
- Shan Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Siyi Han
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
5
|
Dos Santos JM, Joiakim A, Putt DA, Scherrer-Crosbie M, Kim H. 14,15-Dihydroxyeicosatrienoic acid, a soluble epoxide hydrolase metabolite in blood, is a predictor of anthracycline-induced cardiotoxicity - a hypothesis generating study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:47. [PMID: 38102716 PMCID: PMC10722875 DOI: 10.1186/s40959-023-00198-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND Early identification of patients susceptible to chemotherapy-induced cardiotoxicity could lead to targeted treatment to reduce cardiac dysfunction. Rats treated with doxorubicin (DOX), a chemotherapeutic agent, have increased cardiac expression of 14,15-dihydroxyeicosatrienoic acid (14,15-DHET), a bioactive lipid implicated in hypertension and coronary artery disease. However, the utility of 14,15-DHET as plasma biomarkers was not defined. The aim of this study is to investigate if levels of 14,15-DHET are an early blood biomarker to predict the subsequent occurrence of cardiotoxicity in cancer patients after chemotherapy. METHODS H9c2 rat cardiomyocytes were treated with DOX (1 μM) for 2 h and levels of 14,15-DHET in cell media was quantified at 2, 6 or 24 h in media after DOX treatment. Similarly, female Sprague-Dawley rats were treated with DOX for two weeks and levels of 14,15-DHET was assessed in plasma at 48 h and 2 weeks after DOX treatment. Changes in brain natriuretic peptide (BNP) mRNA, an early cardiac hypertrophy process, were determined in the H9c2 cells and rat cardiac tissue. Results were confirmed in human subjects by assessment of levels of 14,15-DHET in plasma of breast cancer patients before and after DOX treatment and left ventricular ejection fraction (LVEF), a clinical marker of cardiotoxicity. RESULTS Levels of 14,15-DHET in cell media and rat plasma increased ~ 3-fold and was accompanied with increase in BNP mRNA in H9c2 cells and rat cardiac tissue after DOX treatment. In matched plasma samples from breast cancer patients, levels of 14,15-DHET were increased in patients that developed cardiotoxicity at 3 months before occurrence of LVEF decrease. CONCLUSIONS Together, these results indicate that levels of 14,15-DHET are elevated prior to major changes in cardiac structure and function after exposure to anthracyclines. Increased levels of 14,15-DHET in plasma may be an important clinical biomarker for early detection of anthracycline-induced cardiotoxicity in cancer patients.
Collapse
Affiliation(s)
- Julia Matzenbacher Dos Santos
- Detroit R&D, Inc., 2727 2nd Ave, Suite 4113, Detroit, MI, 48201, USA
- Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Aby Joiakim
- Detroit R&D, Inc., 2727 2nd Ave, Suite 4113, Detroit, MI, 48201, USA
| | - David A Putt
- Detroit R&D, Inc., 2727 2nd Ave, Suite 4113, Detroit, MI, 48201, USA
| | - Marielle Scherrer-Crosbie
- Cardiac Ultrasound Laboratory, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hyesook Kim
- Detroit R&D, Inc., 2727 2nd Ave, Suite 4113, Detroit, MI, 48201, USA.
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
6
|
Zhao C, Jiang X, Peng L, Zhang Y, Li H, Zhang Q, Wang Y, Yang F, Wu J, Wen Z, He Z, Shen J, Chen C, Wang DW. Glimepiride, a novel soluble epoxide hydrolase inhibitor, protects against heart failure via increasing epoxyeicosatrienoic acids. J Mol Cell Cardiol 2023; 185:13-25. [PMID: 37871528 DOI: 10.1016/j.yjmcc.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Epoxyeicosatrienoic acids (EETs), which exert multiple endogenous protective effects, are hydrolyzed into less active dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). However, commercial drugs related to EETs or sEH are not yet in clinical use. METHODS Firstly, the plasma concentration of EETs and DHETs of 316 patients with heart failure (HF) were detected and quantitated by liquid chromatography-tandem mass spectrometry. Then, transverse aortic constriction (TAC)-induced HF was introduced in cardiomyocyte-specific Ephx2-/- mice. Moreover, Western blot, real-time PCR, luciferase reporter, ChIP assays were employed to explore the underlying mechanism. Finally, multiple sEH inhibitors were designed, synthesized, and validated in vitro and in vivo. RESULTS The ratios of DHETs/EETs were increased in the plasma from patients with HF. Meanwhile, the expression of sEH was upregulated in the heart of patients and mice with HF, especially in cardiomyocytes. Cardiomyocyte-specific Ephx2-/- mice ameliorated cardiac dysfunction induced by TAC. Consistently, Ephx2 knockdown protected Angiotensin II (AngII)-treated cardiomyocytes via increasing EETs in vitro. Mechanistically, AngII could enhance the expression of transcript factor Krüppel-like factor 15 (KLF15), which in turn upregulated sEH. Importantly, glimepiride was identified as a novel sEH inhibitor, which benefited from the elevated EETs during HF. CONCLUSIONS Glimepiride attenuates HF in mice in part by increasing EETs. CLINICAL TRIAL IDENTIFIER NCT03461107 (https://clinicaltrials.gov).
Collapse
Affiliation(s)
- Chengcheng Zhao
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiangrui Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China.
| | - Liyuan Peng
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Yan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huihui Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Qiumeng Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yinhui Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Feipu Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Junfang Wu
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Zuowen He
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jingshan Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| |
Collapse
|
7
|
Pan S, Karey E, Nieves-Cintron M, Chen YJ, Hwang SH, Hammock BD, Pinkerton KE, Chen CY. Effects of chronic secondhand smoke exposure on cardiovascular regulation and the role of soluble epoxide hydrolase in mice. Front Physiol 2023; 14:1185744. [PMID: 37362438 PMCID: PMC10285070 DOI: 10.3389/fphys.2023.1185744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Background: Secondhand smoke (SHS) is a significant risk factor for cardiovascular morbidity and mortality with an estimated 80% of SHS-related deaths attributed to cardiovascular causes. Public health measures and smoking bans have been successful both in reducing SHS exposure and improving cardiovascular outcomes in non-smokers. Soluble epoxide hydrolase (sEH) inhibitors have been shown to attenuate tobacco exposure-induced lung inflammatory responses, making them a promising target for mitigating SHS exposure-induced cardiovascular outcomes. Objectives: The objectives of this study were to determine 1) effects of environmentally relevant SHS exposure on cardiac autonomic function and blood pressure (BP) regulation and 2) whether prophylactic administration of an sEH inhibitor (TPPU) can reduce the adverse cardiovascular effects of SHS exposure. Methods: Male C57BL/6J mice (11 weeks old) implanted with BP/electrocardiogram (ECG) telemetry devices were exposed to filtered air or 3 mg/m3 of SHS (6 hr/d, 5 d/wk) for 12 weeks, followed by 4 weeks of recovery in filtered air. Some mice received TPPU in drinking water (15 mg/L) throughout SHS exposure. BP, heart rate (HR), HR variability (HRV), baroreflex sensitivity (BRS), and BP variability were determined monthly. Results: SHS exposure significantly decreased 1) short-term HRV by ∼20% (p < 0.05) within 4 weeks; 2) overall HRV with maximum effect at 12 weeks (-15%, p < 0.05); 3) pulse pressure (-8%, p < 0.05) as early as week 4; and 4) BRS with maximum effect at 12 weeks (-11%, p < 0.05). Four weeks of recovery following 12 weeks of SHS ameliorated all SHS-induced cardiovascular detriments. Importantly, mice exposed to TPPU in drinking water during SHS-related exposure were protected from SHS cardiovascular consequences. Discussion: The data suggest that 1) environmental relevant SHS exposure significantly alters cardiac autonomic function and BP regulation; 2) cardiovascular consequences from SHS can be reversed by discontinuing SHS exposure; and 3) inhibiting sEH can prevent SHS-induced cardiovascular consequences.
Collapse
Affiliation(s)
- Shiyue Pan
- Department of Pharmacology, University of California Davis, Davis, CA, United States
| | - Emma Karey
- Department of Pharmacology, University of California Davis, Davis, CA, United States
| | | | - Yi-Je Chen
- Department of Pharmacology, 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
| | - Bruce D. Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California Davis, Davis, CA, United States
| | - Chao-Yin Chen
- Department of Pharmacology, University of California Davis, Davis, CA, United States
| |
Collapse
|
8
|
Chauhan PK, Sowdhamini R. Transcriptome data analysis of primary cardiomyopathies reveals perturbations in arachidonic acid metabolism. Front Cardiovasc Med 2023; 10:1110119. [PMID: 37288265 PMCID: PMC10242083 DOI: 10.3389/fcvm.2023.1110119] [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: 11/28/2022] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction Cardiomyopathies are complex heart diseases with significant prevalence around the world. Among these, primary forms are the major contributors to heart failure and sudden cardiac death. As a high-energy demanding engine, the heart utilizes fatty acids, glucose, amino acid, lactate and ketone bodies for energy to meet its requirement. However, continuous myocardial stress and cardiomyopathies drive towards metabolic impairment that advances heart failure (HF) pathogenesis. So far, metabolic profile correlation across different cardiomyopathies remains poorly understood. Methods In this study, we systematically explore metabolic differences amongst primary cardiomyopathies. By assessing the metabolic gene expression of all primary cardiomyopathies, we highlight the significantly shared and distinct metabolic pathways that may represent specialized adaptations to unique cellular demands. We utilized publicly available RNA-seq datasets to profile global changes in the above diseases (|log2FC| ≥ 0.28 and BH adjusted p-val 0.1) and performed gene set analysis (GSA) using the PAGE statistics on KEGG pathways. Results Our analysis demonstrates that genes in arachidonic acid metabolism (AA) are significantly perturbed across cardiomyopathies. In particular, the arachidonic acid metabolism gene PLA2G2A interacts with fibroblast marker genes and can potentially influence fibrosis during cardiomyopathy. Conclusion The profound significance of AA metabolism within the cardiovascular system renders it a key player in modulating the phenotypes of cardiomyopathies.
Collapse
Affiliation(s)
- Pankaj Kumar Chauhan
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bangalore, India
| | - Ramanathan Sowdhamini
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bangalore, India
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| |
Collapse
|
9
|
Favor OK, Chauhan PS, Pourmand E, Edwards AM, Wagner JG, Lewandowski RP, Heine LK, Harkema JR, Lee KSS, Pestka JJ. Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice. Front Immunol 2023; 14:1124910. [PMID: 36875087 PMCID: PMC9978350 DOI: 10.3389/fimmu.2023.1124910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/17/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Lipopolysaccharide (LPS)-accelerated autoimmune glomerulonephritis (GN) in NZBWF1 mice is a preclinical model potentially applicable for investigating lipidome-modulating interventions against lupus. LPS can be expressed as one of two chemotypes: smooth LPS (S-LPS) or rough LPS (R-LPS) which is devoid of O-antigen polysaccharide sidechain. Since these chemotypes differentially affect toll-like receptor 4 (TLR4)-mediated immune cell responses, these differences may influence GN induction. Methods We initially compared the effects of subchronic intraperitoneal (i.p.) injection for 5 wk with 1) Salmonella S-LPS, 2) Salmonella R-LPS, or 3) saline vehicle (VEH) (Study 1) in female NZBWF1 mice. Based on the efficacy of R-LPS in inducing GN, we next used it to compare the impact of two lipidome-modulating interventions, ω-3 polyunsaturated fatty acid (PUFA) supplementation and soluble epoxide hydrolase (sEH) inhibition, on GN (Study 2). Specifically, effects of consuming ω-3 docosahexaenoic acid (DHA) (10 g/kg diet) and/or the sEH inhibitor 1-(4-trifluoro-methoxy-phenyl)-3-(1-propionylpiperidin-4-yl) urea (TPPU) (22.5 mg/kg diet ≈ 3 mg/kg/day) on R-LPS triggering were compared. Results In Study 1, R-LPS induced robust elevations in blood urea nitrogen, proteinuria, and hematuria that were not evident in VEH- or S-LPS-treated mice. R-LPS-treated mice further exhibited kidney histopathology including robust hypertrophy, hyperplasia, thickened membranes, lymphocytic accumulation containing B and T cells, and glomerular IgG deposition consistent with GN that was not evident in VEH- or SLPS-treated groups. R-LPS but not S-LPS induced spleen enlargement with lymphoid hyperplasia and inflammatory cell recruitment in the liver. In Study 2, resultant blood fatty acid profiles and epoxy fatty acid concentrations reflected the anticipated DHA- and TPPU-mediated lipidome changes, respectively. The relative rank order of R-LPS-induced GN severity among groups fed experimental diets based on proteinuria, hematuria, histopathologic scoring, and glomerular IgG deposition was: VEH/CON< R-LPS/DHA ≈ R-LPS/TPPU<<< R-LPS/TPPU+DHA ≈ R-LPS/CON. In contrast, these interventions had modest-to- negligible effects on R-LPS-induced splenomegaly, plasma antibody responses, liver inflammation, and inflammation-associated kidney gene expression. Discussion We show for the first time that absence of O-antigenic polysaccharide in R-LPS is critical to accelerated GN in lupus-prone mice. Furthermore, intervention by lipidome modulation through DHA feeding or sEH inhibition suppressed R-LPS-induced GN; however, these ameliorative effects were greatly diminished upon combining the treatments.
Collapse
Affiliation(s)
- Olivia K. Favor
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Preeti S. Chauhan
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Elham Pourmand
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Angel M. Edwards
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - James G. Wagner
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Ryan P. Lewandowski
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Lauren K. Heine
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jack R. Harkema
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Kin Sing Stephen Lee
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
10
|
Peng L, Song Z, Zhao C, Abuduwufuer K, Wang Y, Wen Z, Ni L, Li C, Yu Y, Zhu Y, Jiang H, Shen J, Jiang X, Chen C, Zhang X, Wang DW. Increased Soluble Epoxide Hydrolase Activity Positively Correlates with Mortality in Heart Failure Patients with Preserved Ejection Fraction: Evidence from Metabolomics. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:34-49. [PMID: 36939801 PMCID: PMC9883375 DOI: 10.1007/s43657-022-00069-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) have pleiotropic endogenous cardiovascular protective effects and can be hydrolyzed to the corresponding dihydroxyeicosatrienoic acids by soluble epoxide hydrolase (sEH). Heart failure with preserved ejection fraction (HFpEF) has shown an increased prevalence and worse prognosis over the decades. However, the role of sEH activity in HFpEF remains unclear. We enrolled 500 patients with HFpEF and 500 healthy controls between February 2010 and March 2016. Eight types of sEH-related eicosanoids were measured according to target metabolomics, and their correlation with clinical endpoints was also analyzed. The primary endpoint was cardiac mortality, and the secondary endpoint was a composite of cardiac events, including heart failure (HF) readmission, cardiogenic hospitalization, and all-cause mortality. Furthermore, the effect of sEH inhibitors on cardiac diastolic function in HFpEF was investigated in vivo and in vitro. Patients with HFpEF showed significantly enhanced EET degradation by the sEH enzyme compared with healthy controls. More importantly, sEH activity was positively correlated with cardiac mortality in patients with HFpEF, especially in older patients with arrhythmia. A consistent result was obtained in the multiple adjusted models. Decreased sEH activity by the sEH inhibitor showed a significant effective effect on the improvement of cardiac diastolic function by ameliorating lipid disorders in cardiomyocytes of HFpEF mouse model. This study demonstrated that increased sEH activity was associated with cardiac mortality in patients with HFpEF and suggested that sEH inhibition could be a promising therapeutic strategy to improve diastolic cardiac function. Clinical trial identifier: NCT03461107 (https://clinicaltrials.gov). Supplementary Information The online version contains supplementary material available at 10.1007/s43657-022-00069-8.
Collapse
Affiliation(s)
- Liyuan Peng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Ziping Song
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Chengcheng Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Kudusi Abuduwufuer
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Yanwen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Li Ni
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Chenze Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030 China
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070 China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Center for Cardiovascular Diseases, Department of Physiology and Pathophysiology, Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070 China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Jinshan Shen
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Xiangrui Jiang
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Center for Cardiovascular Diseases, Department of Physiology and Pathophysiology, Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070 China
- Tianjin Key Laboratory of Metabolic Diseases, Key Laboratory of Immune Microenvironment and Disease-Ministry of Education, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070 China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
11
|
Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis. Int J Mol Sci 2023; 24:ijms24021637. [PMID: 36675152 PMCID: PMC9863938 DOI: 10.3390/ijms24021637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
It is commonly believed that the inactivation of inflammation is mainly due to the decay or cessation of inducers. In reality, in connection with the development of atherosclerosis, spontaneous decay of inducers is not observed. It is now known that lipid mediators originating from polyunsaturated fatty acids (PUFAs), which are important constituents of all cell membranes, can act in the inflamed tissue and bring it to resolution. In fact, PUFAs, such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are precursors to both pro-inflammatory and anti-inflammatory compounds. In this review, we describe the lipid mediators of vascular inflammation and resolution, and their biochemical activity. In addition, we highlight data from the literature that often show a worsening of atherosclerotic disease in subjects deficient in lipid mediators of inflammation resolution, and we also report on the anti-proteasic and anti-thrombotic properties of these same lipid mediators. It should be noted that despite promising data observed in both animal and in vitro studies, contradictory clinical results have been observed for omega-3 PUFAs. Many further studies will be required in order to clarify the observed conflicts, although lifestyle habits such as smoking or other biochemical factors may often influence the normal synthesis of lipid mediators of inflammation resolution.
Collapse
|
12
|
Cho C, Aliwarga T, Wiley AM, Totah RA. Cardioprotective mechanisms of cytochrome P450 derived oxylipins from ω-3 and ω-6 PUFAs. ADVANCES IN PHARMACOLOGY 2023; 97:201-227. [DOI: 10.1016/bs.apha.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
13
|
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
|
14
|
A novel soluble epoxide hydrolase vaccine protects murine cardiac muscle against myocardial infarction. Sci Rep 2022; 12:6923. [PMID: 35484372 PMCID: PMC9051153 DOI: 10.1038/s41598-022-10641-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 04/05/2022] [Indexed: 11/08/2022] Open
Abstract
Myocardial infarction is still a life-threatening disease, even though its prognosis has been improved through the development of percutaneous coronary intervention and pharmacotherapy. In addition, heart failure due to remodeling after myocardial infarction requires lifelong management. The aim of this study was to develop a novel treatment suppressing the myocardial damage done by myocardial infarction. We focused on inhibition of soluble epoxide hydrolase to prolong the activation of epoxyeicosatrienoic acids, which have vasodilatory and anti-inflammatory properties. We successfully made a new vaccine to inactivate soluble epoxide hydrolase, and we have evaluated the effect of the vaccine in a rat myocardial infarction model. In the vaccinated group, the ischemic area was significantly reduced, and cardiac function was significantly preserved. Vaccine treatment clearly increased microvessels in the border area and suppressed fibrosis secondary to myocardial infarction. This soluble epoxide hydrolase vaccine is a novel treatment for improving cardiac function following myocardial infarction.
Collapse
|
15
|
Ma K, Yang J, Shao Y, Li P, Guo H, Wu J, Zhu Y, Zhang H, Zhang X, Du J, Li Y. Therapeutic and Prognostic Significance of Arachidonic Acid in Heart Failure. Circ Res 2022; 130:1056-1071. [PMID: 35255710 DOI: 10.1161/circresaha.121.320548] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Accurate prediction of death is an unmet need in patients with acute decompensated heart failure (HF). Arachidonic acid (AA) metabolites play an important role in the multiple pathophysiological processes. We aimed to develop an AA score to accurately predict mortality in patients with acute decompensated HF and explore the causal relationship between the AA predictors and HF. METHODS The serum AA metabolites was measured in patients with acute decompensated HF (discovery cohort n=419; validation cohort n=386) by mass spectroscopy. We assessed the prognostic importance of AA metabolites for 1-year death using Cox regression and machine learning approaches. An machine learning-based AA score for predicting 1-year death was created and validated. We explored the mechanisms using transcriptome and functional experiments in a mouse model of early ischemic cardiomyopathy. RESULTS Among the 27 AA metabolites, elevated 14,15-DHET/14,15-EET ratio was the strongest predictor of 1-year death (hazard ratio, 2.10, P=3.1×10-6). Machine learning-based AA score using a combination of the 14,15-DHET/14,15-EET ratio, 14,15-DHET, PGD2, and 9-HETE performed best (area under the curve [AUC]: 0.85). The machine learning-based AA score provided incremental information to predict mortality beyond BNP (B-type natriuretic peptide; ΔAUC: 0.19), clinical score (ΔAUC: 0.09), and preexisting ADHERE, Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients With Heart Failure, and Get With The Guidelines Heart Failure scores (ΔAUC: 0.17, 0.17, 0.15, respectively). In the validation cohort, the AA score accurately predicted mortality (AUC:0.81). False-negative and false-positive findings, as classified by the BNP threshold, were correctly reclassified by the AA score (46.2% of false-negative and 84.5% of false-positive). In a murine model, the expression and enzymatic activity of sEH (soluble epoxide hydrolase) increased after myocardial infarction. Genetic deletion of sEH improved HF and the blockade of 14,15-EET abolished this cardioprotection. We mechanistically revealed the beneficial effect of 14,15-EET by impairing the activation of monocytes/macrophages. CONCLUSIONS Our studies propose that the AA score predicts death in patients with acute decompensated HF and inhibiting sEH serves as a therapeutic target for treating HF. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT04108182.
Collapse
Affiliation(s)
- Ke Ma
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Jie Yang
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Yihui Shao
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Ping Li
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Hongchang Guo
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Jianing Wu
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Center for Cardiovascular Diseases, Research Center of Basic Medical Sciences, Department of Physiology and Pathophysiology, Tianjin Medical University, China (Y.Z., X.Z.)
| | - Hui Zhang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University (H.Z.)
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Center for Cardiovascular Diseases, Research Center of Basic Medical Sciences, Department of Physiology and Pathophysiology, Tianjin Medical University, China (Y.Z., X.Z.)
| | - Jie Du
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| | - Yulin Li
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (K.M., J.Y., Y.S., P.L., H.G., J.W., J.D., Y.L.)
| |
Collapse
|
16
|
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
|
17
|
Kumar G, Saini M, Kundu S. Therapeutic enzymes as non-conventional targets in cardiovascular impairments:A Comprehensive Review. Can J Physiol Pharmacol 2021; 100:197-209. [PMID: 34932415 DOI: 10.1139/cjpp-2020-0732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Over the last few decades, substantial progress has been made towards the understanding of cardiovascular diseases (CVDs). In-depth mechanistic insights have also provided opportunities to explore novel therapeutic targets and treatment regimens to be discovered. Therapeutic enzymes are an example of such opportunities. The balanced functioning of such enzymes protects against a variety of CVDs while on the other hand, even a small shift in the normal functioning of these enzymes may lead to deleterious outcomes. Owing to the great versatility of these enzymes, inhibition and activation are key regulatory approaches to counter the onset and progression of several cardiovascular impairments. While cardiovascular remedies are already available in excess and of course they are efficacious, a comprehensive description of novel therapeutic enzymes to combat CVDs is the need of the hour. In light of this, the regulation of the functional activity of these enzymes also opens a new avenue for the treatment approaches to be employed. This review describes the importance of non-conventional enzymes as potential candidates in several cardiovascular disorders while highlighting some of the recently targeted therapeutic enzymes in CVDs.
Collapse
Affiliation(s)
- Gaurav Kumar
- University of Delhi - South Campus, 93081, Biochemistry, New Delhi, Delhi, India;
| | - Manisha Saini
- University of Delhi - South Campus, 93081, Biochemistry, New Delhi, Delhi, India;
| | - Suman Kundu
- University of Delhi - South Campus, 93081, Biochemistry, New Delhi, Delhi, India;
| |
Collapse
|
18
|
Wang Y, Pan W, Bai X, Wang X, Wang Y, Yin Y. microRNA-454-mediated NEDD4-2/TrkA/cAMP axis in heart failure: Mechanisms and cardioprotective implications. J Cell Mol Med 2021; 25:5082-5098. [PMID: 33949117 PMCID: PMC8178253 DOI: 10.1111/jcmm.16491] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/23/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
The current study aimed to investigate the mechanism by which miR-454 influences the progression of heart failure (HF) in relation to the neural precursor cell expressed, developmentally downregulated 4-2 (NEDD4-2)/tropomyosin receptor kinase A (TrkA)/cyclic adenosine 3',5'-monophosphate (cAMP) axis. Sprague-Dawley rats were used to establish a HF animal model via ligation of the left anterior descending branch of the coronary artery. The cardiomyocyte H9c2 cells were treated with H2 O2 to stimulate oxidative stress injury in vitro. RT-qPCR and Western blot assay were subsequently performed to determine the expression patterns of miR-454, NEDD4-2, TrkA, apoptosis-related proteins and cAMP pathway markers. Dual-luciferase reporter gene assay coupled with co-immunoprecipitation was performed to elucidate the relationship between miR-454, NEDD4-2 and TrkA. Gain- or loss-of-function experiments as well as rescue experiments were conducted via transient transfection (in vitro) and adenovirus infection (in vivo) to examine their respective functions on H9c2 cell apoptosis and myocardial damage. Our results suggested that miR-454 was aberrantly downregulated in the context of HF, while evidence was obtained suggesting that it targeted NEDD4-2 to downregulate NEDD4-2 in cardiomyocytes. miR-454 exerted anti-apoptotic and protective effects on cardiomyocytes through inhibition of NEDD4-2, while NEDD4-2 stimulated ubiquitination and degradation of TrkA protein. Furthermore, miR-454 activated the cAMP pathway via the NEDD4-2/TrkA axis, which ultimately suppressed cardiomyocyte apoptosis and attenuated myocardial damage. Taken together, the key findings of the current study highlight the cardioprotective role of miR-454, which is achieved through activation of the cAMP pathway by impairing NEDD4-2-induced TrkA ubiquitination.
Collapse
Affiliation(s)
- Yaowen Wang
- Department of Cardiology, Chongqing Cardiac Arrhythmias Therapeutic Service Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Pan
- Department of Cardiology, Chongqing Cardiac Arrhythmias Therapeutic Service Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyu Bai
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Xukai Wang
- Department of Cardiology, Institute of Field Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yuehui Yin
- Department of Cardiology, Chongqing Cardiac Arrhythmias Therapeutic Service Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Lai J, Chen C. The Role of Epoxyeicosatrienoic Acids in Cardiac Remodeling. Front Physiol 2021; 12:642470. [PMID: 33716791 PMCID: PMC7943617 DOI: 10.3389/fphys.2021.642470] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid by cytochrome P450 (CYP) epoxygenases, which include four regioisomers: 5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET. Each of them possesses beneficial effects against inflammation, fibrosis, and apoptosis, which could combat cardiovascular diseases. Numerous studies have demonstrated that elevation of EETs by overexpression of CYP2J2, inhibition of sEH, or treatment with EET analogs showed protective effects in various cardiovascular diseases, including hypertension, myocardial infarction, and heart failure. As is known to all, cardiac remodeling is the major pathogenesis of cardiovascular diseases. This review will begin with the introduction of EETs and their protective effects in cardiovascular diseases. In the following, the roles of EETs in cardiac remodeling, with a particular emphasis on myocardial hypertrophy, apoptosis, fibrosis, inflammation, and angiogenesis, will be summarized. Finally, it is suggested that upregulation of EETs is a potential therapeutic strategy for cardiovascular diseases. The EET-related drug development against cardiac remodeling is also discussed, including the overexpression of CYP2J2, inhibition of sEH, and the analogs of EET.
Collapse
Affiliation(s)
- Jinsheng Lai
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, Hu J, Fleming I, Wang DW. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Target Ther 2021; 6:94. [PMID: 33637672 PMCID: PMC7910446 DOI: 10.1038/s41392-020-00443-w] [Citation(s) in RCA: 584] [Impact Index Per Article: 146.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/04/2020] [Accepted: 10/15/2020] [Indexed: 01/31/2023] Open
Abstract
The arachidonic acid (AA) pathway plays a key role in cardiovascular biology, carcinogenesis, and many inflammatory diseases, such as asthma, arthritis, etc. Esterified AA on the inner surface of the cell membrane is hydrolyzed to its free form by phospholipase A2 (PLA2), which is in turn further metabolized by cyclooxygenases (COXs) and lipoxygenases (LOXs) and cytochrome P450 (CYP) enzymes to a spectrum of bioactive mediators that includes prostanoids, leukotrienes (LTs), epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid (diHETEs), eicosatetraenoic acids (ETEs), and lipoxins (LXs). Many of the latter mediators are considered to be novel preventive and therapeutic targets for cardiovascular diseases (CVD), cancers, and inflammatory diseases. This review sets out to summarize the physiological and pathophysiological importance of the AA metabolizing pathways and outline the molecular mechanisms underlying the actions of AA related to its three main metabolic pathways in CVD and cancer progression will provide valuable insight for developing new therapeutic drugs for CVD and anti-cancer agents such as inhibitors of EETs or 2J2. Thus, we herein present a synopsis of AA metabolism in human health, cardiovascular and cancer biology, and the signaling pathways involved in these processes. To explore the role of the AA metabolism and potential therapies, we also introduce the current newly clinical studies targeting AA metabolisms in the different disease conditions.
Collapse
Affiliation(s)
- Bei Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Lujin Wu
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jing Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jiong Hu
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China.
| |
Collapse
|
21
|
|
22
|
Sun CP, Zhang XY, Morisseau C, Hwang SH, Zhang ZJ, Hammock BD, Ma XC. Discovery of Soluble Epoxide Hydrolase Inhibitors from Chemical Synthesis and Natural Products. J Med Chem 2020; 64:184-215. [PMID: 33369424 DOI: 10.1021/acs.jmedchem.0c01507] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Soluble epoxide hydrolase (sEH) is an α/β hydrolase fold protein and widely distributed in numerous organs including the liver, kidney, and brain. The inhibition of sEH can effectively maintain endogenous epoxyeicosatrienoic acids (EETs) levels and reduce dihydroxyeicosatrienoic acids (DHETs) levels, resulting in therapeutic potentials for cardiovascular, central nervous system, and metabolic diseases. Therefore, since the beginning of this century, the development of sEH inhibitors is a hot research topic. A variety of potent sEH inhibitors have been developed by chemical synthesis or isolated from natural sources. In this review, we mainly summarized the interconnected aspects of sEH with cardiovascular, central nervous system, and metabolic diseases and then focus on representative inhibitors, which would provide some useful guidance for the future development of potential sEH inhibitors.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xin-Yue Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Zhan-Jun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China.,College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou 311121, People's Republic of China
| |
Collapse
|
23
|
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
|
24
|
Martins GL, Duarte RCF, Mukhamedyarov MA, Palotás A, Ferreira CN, Reis HJ. Inflammatory and Infectious Processes Serve as Links between Atrial Fibrillation and Alzheimer's Disease. Int J Mol Sci 2020; 21:E3226. [PMID: 32370194 PMCID: PMC7247326 DOI: 10.3390/ijms21093226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 04/25/2020] [Accepted: 04/30/2020] [Indexed: 11/16/2022] Open
Abstract
Atrial fibrillation (AF) is one of the most prevalent forms of arrhythmia that carries an increased risk of stroke which, in turn, is strongly associated with cognitive decline. The majority of dementia cases are caused by Alzheimer's disease (AD) with obscure pathogenesis. While the exact mechanisms are unknown, the role of inflammatory processes and infectious agents have recently been implicated in both AD and AF, suggesting a common link between these maladies. Here, we present the main shared pathways underlying arrhythmia and memory loss. The overlapping predictive biomarkers and emerging joint pharmacological approaches are also discussed.
Collapse
Affiliation(s)
- Gabriela Lopes Martins
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| | - Rita Carolina Figueiredo Duarte
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| | | | - András Palotás
- Asklepios-Med (Private Medical Practice and Research Center), H-6722 Szeged, Hungary
- Institute of Fundamental Medicine and Biology, Kazan Federal University, R-420008 Kazan, Russia
| | - Cláudia Natália Ferreira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| | - Helton José Reis
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| |
Collapse
|
25
|
Pallàs M, Vázquez S, Sanfeliu C, Galdeano C, Griñán-Ferré C. Soluble Epoxide Hydrolase Inhibition to Face Neuroinflammation in Parkinson's Disease: A New Therapeutic Strategy. Biomolecules 2020; 10:E703. [PMID: 32369955 PMCID: PMC7277900 DOI: 10.3390/biom10050703] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroinflammation is a crucial process associated with the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). Several pieces of evidence suggest an active role of lipid mediators, especially epoxy-fatty acids (EpFAs), in the genesis and control of neuroinflammation; 14,15-epoxyeicosatrienoic acid (14,15-EET) is one of the most commonly studied EpFAs, with anti-inflammatory properties. Soluble epoxide hydrolase (sEH) is implicated in the hydrolysis of 14,15-EET to its corresponding diol, which lacks anti-inflammatory properties. Preventing EET degradation thus increases its concentration in the brain through sEH inhibition, which represents a novel pharmacological approach to foster the reduction of neuroinflammation and by end neurodegeneration. Recently, it has been shown that sEH levels increase in brains of PD patients. Moreover, the pharmacological inhibition of the hydrolase domain of the enzyme or the use of sEH knockout mice reduced the deleterious effect of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration. This paper overviews the knowledge of sEH and EETs in PD and the importance of blocking its hydrolytic activity, degrading EETs in PD physiopathology. We focus on imperative neuroinflammation participation in the neurodegenerative process in PD and the putative therapeutic role for sEH inhibitors. In this review, we also describe highlights in the general knowledge of the role of sEH in the central nervous system (CNS) and its participation in neurodegeneration. We conclude that sEH is one of the most promising therapeutic strategies for PD and other neurodegenerative diseases with chronic inflammation process, providing new insights into the crucial role of sEH in PD pathophysiology as well as a singular opportunity for drug development.
Collapse
Affiliation(s)
- Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain;
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Department de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain;
| | - Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, IDIBAPS and CIBERESP, C/Roselló 161, 08036 Barcelona, Spain;
| | - Carles Galdeano
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain;
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain;
| |
Collapse
|
26
|
Shafi A, Timiri Sathyamurthy RD, Seetharaman J, Sambanthan M, Murugesan R, Sundaram S, Bhanumathy Ramarathinam R. Molecular docking, quantum chemical computational and vibrational studies on bicyclic heterocycle "6-nitro-2,3-dihydro-1,4-benzodioxine": Anti-cancer agent. Comput Biol Chem 2020; 86:107226. [PMID: 32142983 DOI: 10.1016/j.compbiolchem.2020.107226] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/15/2020] [Accepted: 01/26/2020] [Indexed: 10/24/2022]
Abstract
The heterocyclic aromatic compounds are primarily used to make pharmaceutical and agrochemicals. In addition, these compounds can be chosen as antioxidants, corrosion inhibitors, electro and opto-electronic devices, polymer material, dye stuff, developers, etc. On the account of this, the heterocyclic aromatic 6-nitro-2,3-dihydro-1,4-benzodioxine (6N3DB) was chosen and the structure is optimized to predict the important properties of it. The structural parameters such as bond length and bond angle have been obtained by DFT/B3LYP/6-311++G(d,p) basis set to know the geometry and orientation of 6N3DB. The molecule has been characterized by FT-IR and FT-Raman spectroscopic techniques to predict the functional groups, vibrational modes and aromatic nature of 6N3DB. The chemical shifts of 1H and 13C have been obtained experimentally and compared with the theoretical data. The parameters such as the band gap between HOMO-LUMO orbitals, λmax, and electron transition probability in frontier orbitals have been estimated to know the NLO and corrosion inhibition activity. HOMO-LUMO orbital diagram has been obtained for different energy levels and their band gap energies have been compared with UV-vis band gap values. The chemical significance of the molecule has been explained using ELF, LOL, and RDG. The binding energy and intermolecular energy values indicate that the title compound possesses anti-cancer property through hydrolase inhibition activity.
Collapse
Affiliation(s)
- Aayisha Shafi
- Research and Development Centre, Bharathiar University, Coimbatore, 641046, Tamilnadu, India; Department of Physics, Meenakshi College for Women, Chennai, 600024, Tamilnadu, India
| | | | - Janani Seetharaman
- Department of Physics, Queen Mary's College, Chennai, 600005, Tamilnadu, India
| | - Muthu Sambanthan
- Department of Physics, Arignar Anna Govt.Arts College, Cheyyar, 604407, Tamilnadu, India.
| | - Raja Murugesan
- Department of Physics, Govt. Thirumagal Mill's College, Gudiyattam, 632602, Tamilnadu, India
| | - Sevvanthi Sundaram
- Department of Physics, Arignar Anna Govt.Arts College, Cheyyar, 604407, Tamilnadu, India
| | | |
Collapse
|
27
|
Abis G, Charles RL, Kopec J, Yue WW, Atkinson RA, Bui TTT, Lynham S, Popova S, Sun YB, Fraternali F, Eaton P, Conte MR. 15-deoxy-Δ 12,14-Prostaglandin J 2 inhibits human soluble epoxide hydrolase by a dual orthosteric and allosteric mechanism. Commun Biol 2019; 2:188. [PMID: 31123712 PMCID: PMC6525171 DOI: 10.1038/s42003-019-0426-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/12/2019] [Indexed: 01/01/2023] Open
Abstract
Human soluble epoxide hydrolase (hsEH) is an enzyme responsible for the inactivation of bioactive epoxy fatty acids, and its inhibition is emerging as a promising therapeutical strategy to target hypertension, cardiovascular disease, pain and insulin sensitivity. Here, we uncover the molecular bases of hsEH inhibition mediated by the endogenous 15-deoxy-Δ12,14-Prostaglandin J2 (15d-PGJ2). Our data reveal a dual inhibitory mechanism, whereby hsEH can be inhibited by reversible docking of 15d-PGJ2 in the catalytic pocket, as well as by covalent locking of the same compound onto cysteine residues C423 and C522, remote to the active site. Biophysical characterisations allied with in silico investigations indicate that the covalent modification of the reactive cysteines may be part of a hitherto undiscovered allosteric regulatory mechanism of the enzyme. This study provides insights into the molecular modes of inhibition of hsEH epoxy-hydrolytic activity and paves the way for the development of new allosteric inhibitors.
Collapse
Affiliation(s)
- Giancarlo Abis
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, SE1 1UL UK
| | - Rebecca L. Charles
- School of Cardiovascular Medicine & Science, The Rayne Institute, Lambeth Wing, St Thomas’ Hospital, King’s College London, London, SE1 7EH UK
| | - Jolanta Kopec
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| | - Wyatt W. Yue
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| | - R. Andrew Atkinson
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, SE1 1UL UK
- Centre for Biomolecular Spectroscopy, King’s College London, London, SE1 1UL UK
| | - Tam T. T. Bui
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, SE1 1UL UK
- Centre for Biomolecular Spectroscopy, King’s College London, London, SE1 1UL UK
| | - Steven Lynham
- Proteomics Facility, Centre of Excellence for Mass Spectrometry, The James Black Centre, King’s College London, London, SE5 9NU UK
| | - Simona Popova
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, SE1 1UL UK
| | - Yin-Biao Sun
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, SE1 1UL UK
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, SE1 1UL UK
| | - Philip Eaton
- School of Cardiovascular Medicine & Science, The Rayne Institute, Lambeth Wing, St Thomas’ Hospital, King’s College London, London, SE1 7EH UK
| | - Maria R. Conte
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, SE1 1UL UK
- Centre for Biomolecular Spectroscopy, King’s College London, London, SE1 1UL UK
| |
Collapse
|
28
|
Wan D, Yang J, McReynolds CB, Barnych B, Wagner KM, Morisseau C, Hwang SH, Sun J, Blöcher R, Hammock BD. In vitro and in vivo Metabolism of a Potent Inhibitor of Soluble Epoxide Hydrolase, 1-(1-Propionylpiperidin-4-yl)-3-(4-(trifluoromethoxy)phenyl)urea. Front Pharmacol 2019; 10:464. [PMID: 31143115 PMCID: PMC6520522 DOI: 10.3389/fphar.2019.00464] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022] Open
Abstract
1-(1-Propionylpiperidin-4-yl)-3-(4-(trifluoromethoxy)phenyl)urea (TPPU) is a potent soluble epoxide hydrolase (sEH) inhibitor that is used extensively in research for modulating inflammation and protecting against hypertension, neuropathic pain, and neurodegeneration. Despite its wide use in various animal disease models, the metabolism of TPPU has not been well-studied. A broader understanding of its metabolism is critical for determining contributions of metabolites to the overall safety and effectiveness of TPPU. Herein, we describe the identification of TPPU metabolites using LC-MS/MS strategies. Four metabolites of TPPU (M1–M4) were identified from rat urine by a sensitive and specific LC-MS/MS method with double precursor ion scans. Their structures were further supported by LC-MS/MS comparison with synthesized standards. Metabolites M1 and M2 were formed from hydroxylation on a propionyl group of TPPU; M3 was formed by amide hydrolysis of the 1-propionylpiperdinyl group on TPPU; and M4 was formed by further oxidation of the hydroxylated metabolite M2. Interestingly, the predicted α-keto amide metabolite and 4-(trifluoromethoxy)aniline (metabolite from urea cleavage) were not detected by the LC-MRM-MS method. This indicates that if formed, the two potential metabolites represent <0.01% of TPPU metabolism. Species differences in the formation of these four identified metabolites was assessed using liver S9 fractions from dog, monkey, rat, mouse, and human. M1, M2, and M3 were generated in liver S9 fractions from all species, and higher amounts of M3 were generated in monkey S9 fractions compared to other species. In addition, rat and human S9 metabolism showed the highest species similarity based on the quantities of each metabolite. The presence of all four metabolites were confirmed in vivo in rats over 72-h post single oral dose of TPPU. Urine and feces were major routes for TPPU excretion. M1, M4 and parent drug were detected as major substances, and M2 and M3 were minor substances. In blood, M1 accounted for ~9.6% of the total TPPU-related exposure, while metabolites M2, M3, and M4 accounted for <0.4%. All four metabolites were potent inhibitors of human sEH but were less potent than the parent TPPU. In conclusion, TPPU is metabolized via oxidation and amide hydrolysis without apparent breakdown of the urea. The aniline metabolites were not observed either in vitro or in vivo. Our findings increase the confidence in the ability to translate preclinical PK of TPPU in rats to humans and facilitates the potential clinical development of TPPU and other sEH inhibitors.
Collapse
Affiliation(s)
- Debin Wan
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Jun Yang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Cindy B McReynolds
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Bogdan Barnych
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Karen M Wagner
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Christophe Morisseau
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Jia Sun
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States.,State Forestry Administration Key Open Laboratory, International Center for Bamboo and Rattan, Beijing, China
| | - René Blöcher
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| |
Collapse
|
29
|
Dai N, Zhao C, Kong Q, Li D, Cai Z, Wang M. Vascular repair and anti-inflammatory effects of soluble epoxide hydrolase inhibitor. Exp Ther Med 2019; 17:3580-3588. [PMID: 30988740 PMCID: PMC6447806 DOI: 10.3892/etm.2019.7396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022] Open
Abstract
Kawasaki disease (KD) is the leading cause of acquired heart disease in pediatric patients in developed countries. Coronary artery aneurysms and myocardial infarction may occur if the disease remains untreated. An estimated 10-20% of KD patients do not respond to intravenous gamma globulin (IVIG), and thus, alternative treatments are currently being investigated. Epoxyeicosatrienoic acids (EETs) are natural anti-inflammatory factors and angiogenic mediators degraded by soluble epoxide hydrolase (sEH). sEH inhibitory factors have been demonstrated to stabilize EET levels, inhibit inflammation and promote vascular repair in vivo. The present study aimed to determine whether an increase in EET levels induced by treatment with the sEH inhibitor 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA) promotes vascular repair in human coronary arterial endothelial cells (HCAECs) and reduces inflammation in a mouse model of KD induced by Lactobacillus casei cell wall extract. The effect of AUDA on vascular repair in HCAECs was assessed by using cell proliferation, migration, adhesion and tube formation assays, and the anti-inflammatory effect of AUDA in the mouse model of KD was determined by detecting the expression of matrix metalloproteinase (MMP)-9, tumor necrosis factor (TNF)-α and interleukin (IL)-1β at the protein level via ELISA. The results demonstrated that AUDA increased the proliferation, migration, adhesion and tube formation ability of HCAECs in a dose-dependent manner. Furthermore, in the mouse model of KD, AUDA reduced the protein expression of MMP-9, IL-1β and TNF-α, indicating that AUDA may alleviate inflammatory reactions in the coronary arteries of KD model mice. The present results also indicate that these effects may be exerted through the peroxisome proliferator activated receptor γ signaling pathway. Taken together, the present study supports the potential utility of AUDA in the treatment of KD.
Collapse
Affiliation(s)
- Na Dai
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, P.R. China.,Department of Pediatrics, Jinan Maternity and Child Care Hospital, Ji'nan, Shandong 250012, P.R. China
| | - Cuifen Zhao
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Qingyu Kong
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Dong Li
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Zhifeng Cai
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Minmin Wang
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, P.R. China
| |
Collapse
|
30
|
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
|
31
|
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: 62] [Impact Index Per Article: 8.9] [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
|
32
|
Abis G, Charles RL, Eaton P, Conte MR. Expression, purification, and characterisation of human soluble Epoxide Hydrolase (hsEH) and of its functional C-terminal domain. Protein Expr Purif 2018; 153:105-113. [PMID: 30218745 PMCID: PMC6189638 DOI: 10.1016/j.pep.2018.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/30/2018] [Accepted: 09/05/2018] [Indexed: 12/30/2022]
Abstract
The human soluble Epoxide Hydrolase (hsEH) is an enzyme involved in the hydrolysis of endogenous anti-inflammatory and cardio-protective signalling mediators known as epoxyeicosatrienoic acids (EETs). EETs’ conversion into the corresponding diols by hsEH generates non-bioactive molecules, thereby the enzyme inhibition would be expected to enhance the EETs bioavailability, and their beneficial properties. Numerous inhibitors have been developed to target the enzyme, some of which are showing promising antihypertensive and anti-inflammatory properties in vivo. Thus far, the preparation of the recombinant enzyme for enzymatic and structural in vitro studies has been performed mainly using a baculovirus expression system. More recently, it was reported that the enzyme could be exogenously expressed and isolated from E. coli, although limited amounts of active protein were obtained. We herein describe two novel methods to yield pure recombinant enzyme. The first describes the expression and purification of the full-length enzyme from eukaryotic cells HEK293-F, whilst the second concerns the C-terminal domain of hsEH obtained from the cost-effective and rapid E. coli prokaryotic system. The two methods successfully generated satisfactory amounts of functional enzyme, with virtually identical enzymatic activity. Overall, the protocols described in this paper can be employed for the recombinant expression and purification of active hsEH, to be used in future biomedical investigations and for high-throughput screening of inhibitors for potential use in the treatment of cardiovascular disease. hsEH is a key regulator of cardiovascular homeostasis. A HEK293-F mammalian expression system for hsEH full-length (FL) was developed. An E. coli expression system for the hsEH C-terminal Domain (CTD) was established. Both proteins exhibited the same enzymatic specific activity in vitro. The CTD preparation provides benefits of easy operation, and high yield and purity.
Collapse
Affiliation(s)
- Giancarlo Abis
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Excellence, School of Basic and Medical Biosciences, King's College London, London, SE1 1UL, United Kingdom.
| | - Rebecca L Charles
- Cardiovascular Division and British Heart Foundation Centre of Excellence, The Rayne Institute, King´s College London, St Thomas' Hospital, London, SE1 7EH, United Kingdom
| | - Philip Eaton
- Cardiovascular Division and British Heart Foundation Centre of Excellence, The Rayne Institute, King´s College London, St Thomas' Hospital, London, SE1 7EH, United Kingdom
| | - Maria R Conte
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Excellence, School of Basic and Medical Biosciences, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
33
|
|
34
|
Drug-Disease Interaction: Effect of Inflammation and Nonsteroidal Anti-Inflammatory Drugs on Cytochrome P450 Metabolites of Arachidonic Acid. J Pharm Sci 2018; 107:756-763. [DOI: 10.1016/j.xphs.2017.09.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/05/2017] [Accepted: 09/22/2017] [Indexed: 01/11/2023]
|
35
|
Bukhari IA, Almotrefi AA, Mohamed OY, Al-Masri AA, Sheikh SA. Protective effect of fenofibrate against ischemia-/reperfusion-induced cardiac arrhythmias in isolated rat hearts. Fundam Clin Pharmacol 2018; 32:141-146. [DOI: 10.1111/fcp.12342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/06/2017] [Accepted: 12/22/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Ishfaq A. Bukhari
- Department of Pharmacology; College of Medicine; King Saud University P. O. BOX 2454; Riyadh 11451 Saudi Arabia
| | - Abdulrahman A. Almotrefi
- Department of Pharmacology; College of Medicine; King Saud University P. O. BOX 2454; Riyadh 11451 Saudi Arabia
| | - Osama Y. Mohamed
- Department of Pharmacology; College of Medicine; King Saud University P. O. BOX 2454; Riyadh 11451 Saudi Arabia
| | - Abeer A. Al-Masri
- Department of Physiology; Cardiovascular Research Group; College of Medicine; King Saud University P. O. BOX 2454; Riyadh 11451 Saudi Arabia
| | - Saeed A. Sheikh
- Department of Pharmacology; College of Medicine; King Saud University P. O. BOX 2454; Riyadh 11451 Saudi Arabia
| |
Collapse
|
36
|
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]
|
37
|
Sari I, Pinarbasi H, Pinarbasi E, Yildiz C. Association between the soluble epoxide hydrolase gene and preeclampsia. Hypertens Pregnancy 2017; 36:315-325. [DOI: 10.1080/10641955.2017.1388390] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Ismail Sari
- Department of Medical Biochemistry, School of Medicine, Niğde Ömer Halisdemir University, Niğde, Turkey
| | - Hatice Pinarbasi
- Department of Biochemistry, School of Medicine, Cumhuriyet University, Sivas, Turkey
| | - Ergun Pinarbasi
- Department of Medical Biology, School of Medicine, Cumhuriyet University, Sivas, Turkey
| | - Caglar Yildiz
- Department of Gynecology and Obstetrics, School of Medicine, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
38
|
Redina OE, Abramova TO, Klimov LO, Ryazanova MA, Fedoseeva LA, Smolenskaya SE, Ershov NI, Dubinina AD, Markel AL. Soluble epoxide hydrolase (sEH) as a potential target for arterial hypertension therapy. RUSS J GENET+ 2017. [DOI: 10.1134/s1022795417080063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
39
|
Thao NP, Luyen BTT, Lee JS, Kim JH, Dat NT, Kim YH. Inhibition Potential of Cycloartane-Type Glycosides from the Roots of Cimicifuga dahurica against Soluble Epoxide Hydrolase. JOURNAL OF NATURAL PRODUCTS 2017; 80:1867-1875. [PMID: 28558206 DOI: 10.1021/acs.jnatprod.7b00166] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A phytochemical assay-guided fractionation of the 95% ethanol extract of Cimicifuga dahurica roots afforded 29 9,19-cycloartane triterpenoid glycosides, including the new cimiricasides A-F (1-6). The structures of 1-6 were established using contemporary NMR methods and from the HRESIMS data, and the sugar moiety in each case was confirmed by acid hydrolysis and subsequent GC/MS analysis. Compounds 2, 4, 5, 7-9, 18, 25, and 29 showed soluble epoxide hydrolase inhibitory effects with IC50 values of 0.4 ± 0.1 to 24.0 ± 0.2 μM. The compounds were analyzed by enzyme kinetic studies to explore the binding mode between the ligand and receptor. Compounds 4 (mixed type), 8, 18, and 29 (noncompetitive type) bound to a preferred allosteric site, while compounds 2, 5, 7, 9, and 25 had competitive interactions at the active site. The binding mechanism of selected inhibitors was investigated using molecular docking and dynamics simulations.
Collapse
Affiliation(s)
- Nguyen Phuong Thao
- College of Pharmacy, Chungnam National University , Daejeon 34134, Republic of Korea
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology , 18-Hoang Quoc Viet, Hanoi, Vietnam
| | - Bui Thi Thuy Luyen
- Department of Pharmaceutical Industry, Hanoi University of Pharmacy , 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Ji Sun Lee
- College of Pharmacy, Chungnam National University , Daejeon 34134, Republic of Korea
| | - Jang Hoon Kim
- Korea Atomic Energy Research Institute , Jeongeup, Jeollabuk-do 580-185, Republic of Korea
| | - Nguyen Tien Dat
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology , 18-Hoang Quoc Viet, Hanoi, Vietnam
| | - Young Ho Kim
- College of Pharmacy, Chungnam National University , Daejeon 34134, Republic of Korea
| |
Collapse
|
40
|
Abramova TO, Ryazanova MA, Antonov EV, Redina OE, Markel AL. Increase in the concentration of sEH protein in renal medulla of ISIAH rats with inherited stress-induced arterial hypertension. Mol Biol 2017. [DOI: 10.1134/s0026893317020029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Cuong NM, Van DT, Son NT, Cuong TD, Khanh PN, Binh NQ, Thanh TTV, Lee JS, Jo AR, Kim YH. Inhibitory Effects of Novel Diarylheptanoids and Other Constituents of the Rhizomes ofCurcuma singularison the Catalytic Activity of Soluble Epoxide Hydrolase. B KOREAN CHEM SOC 2017. [DOI: 10.1002/bkcs.11033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Nguyen Manh Cuong
- Department of Bioactive Products, Institute of Natural Products Chemistry; Vietnam Academy of Science and Technology (VAST); Hanoi Vietnam
| | - Doan Thi Van
- Department of Bioactive Products, Institute of Natural Products Chemistry; Vietnam Academy of Science and Technology (VAST); Hanoi Vietnam
| | - Ninh The Son
- Department of Bioactive Products, Institute of Natural Products Chemistry; Vietnam Academy of Science and Technology (VAST); Hanoi Vietnam
| | - To Dao Cuong
- Department of Bioactive Products, Institute of Natural Products Chemistry; Vietnam Academy of Science and Technology (VAST); Hanoi Vietnam
| | - Pham Ngoc Khanh
- Department of Bioactive Products, Institute of Natural Products Chemistry; Vietnam Academy of Science and Technology (VAST); Hanoi Vietnam
| | - Nguyen Quoc Binh
- Vietnam National Museum of Nature; Vietnam Academy of Science and Technology (VAST); Hanoi Vietnam
| | - Tran Thi Viet Thanh
- Vietnam National Museum of Nature; Vietnam Academy of Science and Technology (VAST); Hanoi Vietnam
| | - Ji Sun Lee
- College of Pharmacy; Chungnam National University; Daejeon 34134 Republic of Korea
| | - Ah Reum Jo
- College of Pharmacy; Chungnam National University; Daejeon 34134 Republic of Korea
| | - Young Ho Kim
- College of Pharmacy; Chungnam National University; Daejeon 34134 Republic of Korea
| |
Collapse
|
42
|
Karami L, Saboury AA, Rezaee E, Tabatabai SA. Investigation of the binding mode of 1, 3, 4-oxadiazole derivatives as amide-based inhibitors for soluble epoxide hydrolase (sEH) by molecular docking and MM-GBSA. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 46:445-459. [PMID: 27928588 DOI: 10.1007/s00249-016-1188-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/03/2016] [Accepted: 11/17/2016] [Indexed: 01/24/2023]
Abstract
The soluble epoxide hydrolase (sEH) enzyme plays an important role in the metabolism of endogenous chemical mediators involved in the regulation of blood pressure and inflammation. Inhibition of sEH provides a new approach to the treatment of inflammation, hypertension and atherosclerosis. In this study, the binding modes and inhibition mechanisms of the new oxadiazole-based amide inhibitors of the human soluble epoxide hydrolase were investigated by molecular docking and molecular dynamics (MD) simulation followed by the MM-GBSA method to calculate the binding free energy of each inhibitor to sEH. The results obtained from the binding free energy (ΔG binding) calculation and normal mode analysis indicate that the major favorable contributors are the van der Waals and electrostatic terms, whereas the polar solvation term opposes binding. In addition, a good agreement between the calculated ΔG binding and the experimental IC50 was obtained [correlation coefficient, r 2 = 0.89 (with) and 0.87 (without) entropy]. Besides, comparison of the enthalpy changes (ΔG MM-GBSA) with entropy changes (-TΔS) indicates that binding process of all inhibitors to sEH is enthalpy-driven. Based on the ΔG binding on per residue decomposition, Asp335 and Tyr383 residues from the active site and Trp336, Leu499 and His524 residues from hydrophobic pockets contribute the most to ΔG binding. Moreover, hydrogen bond analysis reveals that Tyr383, Tyr466 and Asp335 residues have an important role in the binding to inhibitors by forming hydrogen bonds with high occupancies. Our obtained results are useful for the understanding of the sEH-inhibitor interactions and may have great importance in the design of future sEH inhibitors.
Collapse
Affiliation(s)
- Leila Karami
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.,Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - Elham Rezaee
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayyed Abbas Tabatabai
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Yang HH, Chen Y, Gao CY. Associations ofP2Y12Rgene polymorphisms with susceptibility to coronary heart disease and clinical efficacy of antiplatelet treatment with clopidogrel. Cardiovasc Ther 2016; 34:460-467. [PMID: 27566695 DOI: 10.1111/1755-5922.12223] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Hong-Hui Yang
- Department of Cardiology; Henan Provincial People's Hospital; Zhengzhou Henan China
| | - Yan Chen
- Department of Cardiology; Henan Provincial People's Hospital; Zhengzhou Henan China
| | - Chuan-Yu Gao
- Department of Cardiology; Henan Provincial People's Hospital; Zhengzhou Henan China
| |
Collapse
|
44
|
Lakkappa N, Krishnamurthy PT, Hammock BD, Velmurugan D, Bharath MMS. Possible role of Epoxyeicosatrienoic acid in prevention of oxidative stress mediated neuroinflammation in Parkinson disorders. Med Hypotheses 2016; 93:161-5. [PMID: 27372879 PMCID: PMC4985172 DOI: 10.1016/j.mehy.2016.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/24/2016] [Accepted: 06/04/2016] [Indexed: 11/19/2022]
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disease involving oxidative stress, neuroinflammation and apoptosis. Epoxyeicosatrienoic acids (EETs) are arachidonic acid metabolites and they play a role in cytoprotection by modulating various cell signaling pathways. This cytoprotective role of EETs are well established in cerebral stroke, cardiac failure, and hypertension, and it is due to their ability to attenuate oxidative stress, endoplasmic reticulum stress, inflammation, caspase activation and apoptosis. The actions of EETs in brain closely parallel the effects which is observed in the peripheral tissues. Since many of these effects could potentially contribute to neuroprotection, EETs are, therefore, one of the potential therapeutic candidates in PD. Therefore, by increasing the half life of endogenous EETs in vivo via inhibition of sEH, its metabolizing enzyme can, therefore, constitutes an important therapeutic strategy in PD.
Collapse
Affiliation(s)
- Navya Lakkappa
- Department of Pharmacology, JSS College of Pharmacy (A Constituent College of JSS University, Mysore), Ootacamund, Tamilnadu, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (A Constituent College of JSS University, Mysore), Ootacamund, Tamilnadu, India.
| | - Bruce D Hammock
- Department of Entomology and Nematology, and Comprehensive Cancer Research Center, University of California, Davis, CA, USA
| | - D Velmurugan
- Department of Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, India
| | - M M Srinivas Bharath
- Department of Neurochemistry, National Institute of Mental Health & Neuro Sciences, Bangalore, India
| |
Collapse
|
45
|
He J, Wang C, Zhu Y, Ai D. Soluble epoxide hydrolase: A potential target for metabolic diseases. J Diabetes 2016; 8:305-13. [PMID: 26621325 DOI: 10.1111/1753-0407.12358] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/16/2015] [Accepted: 11/22/2015] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs), important lipid mediators derived from arachidonic acid, have many beneficial effects in metabolic diseases, including atherosclerosis, hypertension, cardiac hypertrophy, diabetes, non-alcoholic fatty liver disease, and kidney disease. Epoxyeicosatrienoic acids can be further hydrolyzed to less active diols by the enzyme soluble epoxide hydrolase (sEH). Increasing evidence suggests that inhibition of sEH increases levels of EETs, which have anti-inflammatory effects and can prevent the development of hypertension, atherosclerosis, heart failure, fatty liver, and multiple organ fibrosis. Arachidonic acid is the most abundant omega-6 polyunsaturated fatty acid (PUFA) and shares the same set of enzymes with omega-3 PUFAs, such as docosahexaenoic acid and eicosapentaenoic acid. The omega-3 PUFAs and metabolites, such as regioisomeric epoxyeicosatetraenoic acids and epoxydocosapentaenoic acids, have been reported to have strong vasodilatory and anti-inflammatory effects. Therefore, sEH may be a potential therapeutic target for metabolic disorders. In this review, we focus on our and other recent studies of the functions of sEH, including the effects of its eicosanoid products from both omega-3 and omega-6 PUFAs, in various metabolic diseases. We also discuss the possible cellular and molecular mechanisms underlying the regulation of sEH.
Collapse
Affiliation(s)
- Jinlong He
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunjiong Wang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yi Zhu
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Ding Ai
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
46
|
Qin J, Sun D, Jiang H, Kandhi S, Froogh G, Hwang SH, Hammock BD, Wolin MS, Thompson CI, Hintze TH, Huang A. Inhibition of soluble epoxide hydrolase increases coronary perfusion in mice. Physiol Rep 2015; 3:3/6/e12427. [PMID: 26071213 PMCID: PMC4510629 DOI: 10.14814/phy2.12427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Roles of soluble epoxide hydrolase (sEH), the enzyme responsible for hydrolysis of epoxyeicosatrienoic acids (EETs) to their diols (DHETs), in the coronary circulation and cardiac function remain unknown. We tested the hypothesis that compromising EET hydrolysis/degradation, via sEH deficiency, lowers the coronary resistance to promote cardiac perfusion and function. Hearts were isolated from wild type (WT), sEH knockout (KO) mice and WT mice chronically treated with t-TUCB (sEH inhibitor), and perfused with constant flow at different pre-loads. Compared to WT controls, sEH-deficient hearts required significantly greater basal coronary flow to maintain the perfusion pressure at 100 mmHg and exhibited a greater reduction in vascular resistance during tension-induced heart work, implying a better coronary perfusion during cardiac performance. Cardiac contractility, characterized by developed tension in response to changes in preload, was potentially increased in sEH-KO hearts, manifested by an enlarged magnitude at each step-wise increase in end-diastolic to peak-systolic tension. 14,15-EEZE (EET antagonist) prevented the adaptation of coronary circulation in sEH null hearts whereas responses in WT hearts were sensitive to the inhibition of NO. Cardiac expression of EET synthases (CYP2J2/2C29) was comparable in both genotypic mice whereas, levels of 14,15-, 11,12- and 8,9-EETs were significantly higher in sEH-KO hearts, accompanied with lower levels of DHETs. In conclusion, the elevation of cardiac EETs, as a function of sEH deficiency, plays key roles in the adaptation of coronary flow and cardiac function.
Collapse
Affiliation(s)
- Jun Qin
- Department of Physiology, New York Medical College, Valhalla, New York Department of GI Surgery, Renji Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Houli Jiang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Ghezal Froogh
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Sung Hee Hwang
- Department of Entomology, University of California Davis Comprehensive Cancer Center University of California, Davis, California
| | - Bruce D Hammock
- Department of Entomology, University of California Davis Comprehensive Cancer Center University of California, Davis, California
| | - Michael S Wolin
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Carl I Thompson
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Thomas H Hintze
- Department of Physiology, New York Medical College, Valhalla, New York
| | - An Huang
- Department of Physiology, New York Medical College, Valhalla, New York
| |
Collapse
|
47
|
Fetal-adult cardiac transcriptome analysis in rats with contrasting left ventricular mass reveals new candidates for cardiac hypertrophy. PLoS One 2015; 10:e0116807. [PMID: 25646840 PMCID: PMC4315412 DOI: 10.1371/journal.pone.0116807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 12/15/2014] [Indexed: 01/20/2023] Open
Abstract
Reactivation of fetal gene expression patterns has been implicated in common cardiac diseases in adult life including left ventricular (LV) hypertrophy (LVH) in arterial hypertension. Thus, increased wall stress and neurohumoral activation are discussed to induce the return to expression of fetal genes after birth in LVH. We therefore aimed to identify novel potential candidates for LVH by analyzing fetal-adult cardiac gene expression in a genetic rat model of hypertension, i.e. the stroke-prone spontaneously hypertensive rat (SHRSP). To this end we performed genome-wide transcriptome analysis in SHRSP to identify differences in expression patterns between day 20 of fetal development (E20) and adult animals in week 14 in comparison to a normotensive rat strain with contrasting low LV mass, i.e. Fischer (F344). 15232 probes were detected as expressed in LV tissue obtained from rats at E20 and week 14 (p < 0.05) and subsequently screened for differential expression. We identified 24 genes with SHRSP specific up-regulation and 21 genes with down-regulation as compared to F344. Further bioinformatic analysis presented Efcab6 as a new candidate for LVH that showed only in the hypertensive SHRSP rat differential expression during development (logFC = 2.41, p < 0.001) and was significantly higher expressed in adult SHRSP rats compared with adult F344 (+ 76%) and adult normotensive Wistar-Kyoto rats (+ 82%). Thus, it represents an interesting new target for further functional analyses and the elucidation of mechanisms leading to LVH. Here we report a new approach to identify candidate genes for cardiac hypertrophy by combining the analysis of gene expression differences between strains with a contrasting cardiac phenotype with a comparison of fetal-adult cardiac expression patterns.
Collapse
|
48
|
Matsumoto N, Suzuki E, Ishikawa M, Shirafuji T, Hasumi K. Soluble epoxide hydrolase as an anti-inflammatory target of the thrombolytic stroke drug SMTP-7. J Biol Chem 2014; 289:35826-38. [PMID: 25361765 DOI: 10.1074/jbc.m114.588087] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Although ischemic stroke is a major cause of death and disability worldwide, only a small fraction of patients benefit from the current thrombolytic therapy due to a risk of cerebral hemorrhage caused by inflammation. Thus, the development of a new strategy to combat inflammation during thrombolysis is an urgent demand. The small molecule thrombolytic SMTP-7 effectively treats ischemic stroke in several animal models with reducing cerebral hemorrhage. Here we revealed that SMTP-7 targeted soluble epoxide hydrolase (sEH) to suppress inflammation. SMTP-7 inhibited both of the two sEH enzyme activities: epoxide hydrolase (which inactivates anti-inflammatory epoxy-fatty acids) and lipid phosphate phosphatase. SMTP-7 suppressed epoxy-fatty acid hydrolysis in HepG2 cells in culture, implicating the sEH inhibition in the anti-inflammatory mechanism. The sEH inhibition by SMTP-7 was independent of its thrombolytic activity. The simultaneous targeting of thrombolysis and sEH by a single molecule is a promising strategy to revolutionize the current stroke therapy.
Collapse
Affiliation(s)
- Naoki Matsumoto
- From the Department of Applied Biological Science, Tokyo Noko University, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan
| | - Eriko Suzuki
- From the Department of Applied Biological Science, Tokyo Noko University, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan
| | - Makoto Ishikawa
- Pharmaceutical Research Laboratory, Nihon Pharmaceutical, 34 Shin-izumi, Narita, Chiba 286-0825, Japan, and
| | - Takumi Shirafuji
- Pharmaceutical Research Laboratory, Nihon Pharmaceutical, 34 Shin-izumi, Narita, Chiba 286-0825, Japan, and
| | - Keiji Hasumi
- From the Department of Applied Biological Science, Tokyo Noko University, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan, TMS Co., Ltd., 1-32-1-102 Fuchucho, Fuchu, Tokyo 183-0055, Japan
| |
Collapse
|
49
|
Sharkey LC, Radin MJ, Heller L, Rogers LK, Tobias A, Matise I, Wang Q, Apple FS, McCune SA. Differential cardiotoxicity in response to chronic doxorubicin treatment in male spontaneous hypertension-heart failure (SHHF), spontaneously hypertensive (SHR), and Wistar Kyoto (WKY) rats. Toxicol Appl Pharmacol 2013; 273:47-57. [DOI: 10.1016/j.taap.2013.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 07/28/2013] [Accepted: 08/10/2013] [Indexed: 12/20/2022]
|
50
|
Westphal C, Spallek B, Konkel A, Marko L, Qadri F, DeGraff LM, Schubert C, Bradbury JA, Regitz-Zagrosek V, Falck JR, Zeldin DC, Müller DN, Schunck WH, Fischer R. CYP2J2 overexpression protects against arrhythmia susceptibility in cardiac hypertrophy. PLoS One 2013; 8:e73490. [PMID: 24023684 PMCID: PMC3758319 DOI: 10.1371/journal.pone.0073490] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 07/29/2013] [Indexed: 01/04/2023] Open
Abstract
Maladaptive cardiac hypertrophy predisposes one to arrhythmia and sudden death. Cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) promote anti-inflammatory and antiapoptotic mechanisms, and are involved in the regulation of cardiac Ca(2+)-, K(+)- and Na(+)-channels. To test the hypothesis that enhanced cardiac EET biosynthesis counteracts hypertrophy-induced electrical remodeling, male transgenic mice with cardiomyocyte-specific overexpression of the human epoxygenase CYP2J2 (CYP2J2-TG) and wildtype littermates (WT) were subjected to chronic pressure overload (transverse aortic constriction, TAC) or β-adrenergic stimulation (isoproterenol infusion, ISO). TAC caused progressive mortality that was higher in WT (42% over 8 weeks after TAC), compared to CYP2J2-TG mice (6%). In vivo electrophysiological studies, 4 weeks after TAC, revealed high ventricular tachyarrhythmia inducibility in WT (47% of the stimulation protocols), but not in CYP2J2-TG mice (0%). CYP2J2 overexpression also enhanced ventricular refractoriness and protected against TAC-induced QRS prolongation and delocalization of left ventricular connexin-43. ISO for 14 days induced high vulnerability for atrial fibrillation in WT mice (54%) that was reduced in CYP-TG mice (17%). CYP2J2 overexpression also protected against ISO-induced reduction of atrial refractoriness and development of atrial fibrosis. In contrast to these profound effects on electrical remodeling, CYP2J2 overexpression only moderately reduced TAC-induced cardiac hypertrophy and did not affect the hypertrophic response to β-adrenergic stimulation. These results demonstrate that enhanced cardiac EET biosynthesis protects against electrical remodeling, ventricular tachyarrhythmia, and atrial fibrillation susceptibility during maladaptive cardiac hypertrophy.
Collapse
Affiliation(s)
| | - Bastian Spallek
- Experimental and Clinical Research Center, a joint cooperation between the Charité Universitätsmedizin and the MDC, Berlin, Germany
| | - Anne Konkel
- Max-Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Lajos Marko
- Experimental and Clinical Research Center, a joint cooperation between the Charité Universitätsmedizin and the MDC, Berlin, Germany
| | - Fatimunnisa Qadri
- Experimental and Clinical Research Center, a joint cooperation between the Charité Universitätsmedizin and the MDC, Berlin, Germany
| | - Laura M. DeGraff
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, United States of America
| | - Carola Schubert
- Institute of Gender in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - J. Alyce Bradbury
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, United States of America
| | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - John R. Falck
- University of Texas Southwestern Medical Center, Dallas, United States of America
| | - Darryl C. Zeldin
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, United States of America
| | - Dominik N. Müller
- Max-Delbrueck Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité Universitätsmedizin and the MDC, Berlin, Germany
- Department of Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Germany
| | | | - Robert Fischer
- Clinic for Cardiology and Pulmonology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|