1
|
Gladkikh BP, Danilov DV, D’yachenko VS, Butov GM. 1,3-Dichloroadamantyl-Containing Ureas as Potential Triple Inhibitors of Soluble Epoxide Hydrolase, p38 MAPK and c-Raf. Int J Mol Sci 2023; 25:338. [PMID: 38203510 PMCID: PMC10779153 DOI: 10.3390/ijms25010338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Soluble epoxide hydrolase (sEH) is an enzyme involved in the metabolism of bioactive lipid signaling molecules. sEH converts epoxyeicosatrienoic acids (EET) to virtually inactive dihydroxyeicosatrienoic acids (DHET). The first acids are "medicinal" molecules, the second increase the inflammatory infiltration of cells. Mitogen-activated protein kinases (p38 MAPKs) are key protein kinases involved in the production of inflammatory mediators, including tumor necrosis factor-α (TNF-α) and cyclooxygenase-2 (COX-2). p38 MAPK signaling plays an important role in the regulation of cellular processes, especially inflammation. The proto-oncogenic serine/threonine protein kinase Raf (c-Raf) is a major component of the mitogen-activated protein kinase (MAPK) pathway: ERK1/2 signaling. Normal cellular Raf genes can also mutate and become oncogenes, overloading the activity of MEK1/2 and ERK1/2. The development of multitarget inhibitors is a promising strategy for the treatment of socially dangerous diseases. We synthesized 1,3-disubstituted ureas and diureas containing a dichloroadamantyl moiety. The results of computational methods show that soluble epoxide hydrolase inhibitors can act on two more targets in different signaling pathways of mitogen-activated protein kinases p38 MAPK and c-Raf. The two chlorine atoms in the adamantyl moiety may provide additional Cl-π interactions in the active site of human sEH. Molecular dynamics studies have shown that the stability of ligand-protein complexes largely depends on the "spacer effect." The compound containing a bridge between the chloroadamantyl fragment and the ureide group forms more stable ligand-protein complexes with sEH and p38 MAPK, which indicates a better conformational ability of the molecule in the active sites of these targets. In turn, a compound containing two chlorine atoms forms a more stable complex with c-Raf, probably due to the presence of additional halogen bonds of chlorine atoms with amino acid residues.
Collapse
Affiliation(s)
- Boris P. Gladkikh
- Department of Technology of Organic and Petrochemical Synthesis, Volgograd State Technical University, Volgograd 400005, Russia; (B.P.G.); (D.V.D.); (G.M.B.)
| | - Dmitry V. Danilov
- Department of Technology of Organic and Petrochemical Synthesis, Volgograd State Technical University, Volgograd 400005, Russia; (B.P.G.); (D.V.D.); (G.M.B.)
| | - Vladimir S. D’yachenko
- Department of Technology of Organic and Petrochemical Synthesis, Volgograd State Technical University, Volgograd 400005, Russia; (B.P.G.); (D.V.D.); (G.M.B.)
- Department of Chemistry, Technology and Equipment of Chemical Industry, Volzhsky Polytechnic Institute (Branch), Volgograd State Technical University (VSTU), Volzhsky 404121, Russia
| | - Gennady M. Butov
- Department of Technology of Organic and Petrochemical Synthesis, Volgograd State Technical University, Volgograd 400005, Russia; (B.P.G.); (D.V.D.); (G.M.B.)
- Department of Chemistry, Technology and Equipment of Chemical Industry, Volzhsky Polytechnic Institute (Branch), Volgograd State Technical University (VSTU), Volzhsky 404121, Russia
| |
Collapse
|
2
|
Gur Maz T, Koc B, Jordan PM, İbiş K, Çalışkan B, Werz O, Banoglu E. Benzoxazolone-5-Urea Derivatives as Human Soluble Epoxide Hydrolase (sEH) Inhibitors. ACS OMEGA 2023; 8:2445-2454. [PMID: 36687110 PMCID: PMC9850727 DOI: 10.1021/acsomega.2c06936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
Inhibition of soluble epoxide hydrolase (sEH) is indicated as a new therapeutic modality against a variety of inflammatory diseases, including metabolic, renal, and cardiovascular disorders. In our ongoing research on sEH inhibitors, we synthesized novel benzoxazolone-5-urea analogues with highly potent sEH inhibitory properties inspired by the crystallographic fragment scaffolds incorporating a single H-bond donor/acceptor pair. The tractable SAR results indicated that the aryl or benzyl fragments flanking the benzoxazolone-urea scaffold conferred potent sEH inhibition, and compounds 31-39 inhibited the sEH activity with IC50 values in the range of 0.39-570 nM. Docking studies and molecular dynamics simulations with the most potent analogue 33 provided valuable insights into potential binding interactions of the inhibitor in the sEH binding region. In conclusion, benzoxazolone-5-ureas furnished with benzyl groups on the urea function can be regarded as novel lead structures, which allow the development of advanced analogues with enhanced properties against sEH.
Collapse
Affiliation(s)
- Tugce Gur Maz
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Beyzanur Koc
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Paul M. Jordan
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Kübra İbiş
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Burcu Çalışkan
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Oliver Werz
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Erden Banoglu
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| |
Collapse
|
3
|
Turanlı S, Ergül AG, Jordan PM, Olğaç A, Çalışkan B, Werz O, Banoglu E. Quinazoline-4(3 H)-one-7-carboxamide Derivatives as Human Soluble Epoxide Hydrolase Inhibitors with Developable 5-Lipoxygenase Activating Protein Inhibition. ACS OMEGA 2022; 7:36354-36365. [PMID: 36278102 PMCID: PMC9583330 DOI: 10.1021/acsomega.2c04039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
Soluble epoxide hydrolase (sEH) metabolizes epoxyeicosatrienoic acids (EETs), which are endowed with beneficial biological activities as they reduce inflammation, regulate endothelial tone, improve mitochondrial function, and decrease oxidative stress. Therefore, inhibition of sEH for maintaining high EET levels is implicated as a new therapeutic modality with broad clinical applications for metabolic, renal, and cardiovascular disorders. In our search for new sEH inhibitors, we designed and synthesized novel amide analogues of the quinazolinone-7-carboxylic acid derivative 5, a previously discovered 5-lipoxygenase-activating protein (FLAP) inhibitor, to evaluate their potential for inhibiting sEH. As a result, we identified new quinazolinone-7-carboxamides that demonstrated selective sEH inhibition with decreased FLAP inhibitor properties. The tractable SAR results indicated that the amide and thiobenzyl fragments flanking the quinazolinone nucleus are critical features governing the potent sEH inhibition, and compounds 34, 35, 37, and 43 inhibited the sEH activity with IC50 values of 0.30-0.66 μM. Compound 34 also inhibited the FLAP-mediated leukotriene biosynthesis (IC50 = 2.91 μM). In conclusion, quinazolinone-7-carboxamides can be regarded as novel lead structures, and newer analogues with improved efficiency against sEH along with or without FLAP inhibition can be generated.
Collapse
Affiliation(s)
- Sümeyye Turanlı
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Azize Gizem Ergül
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Paul M. Jordan
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Abdurrahman Olğaç
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Burcu Çalışkan
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Oliver Werz
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Erden Banoglu
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| |
Collapse
|
4
|
Han WW, Wang XR, He YF, Zhang HS, Cong X, Xiang RL, Wu LL, Yu GY, Liu LM, Zhang Y. Soluble epoxide hydrolase inhibitor, t-AUCB, improves salivary gland function by ameliorating endothelial injury. Life Sci 2022; 308:120942. [PMID: 36096247 DOI: 10.1016/j.lfs.2022.120942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 10/31/2022]
Abstract
AIMS Inhibitor of soluble epoxide hydrolase (t-AUCB) has been used in the experimental therapy of hypertension. This study aimed to investigate whether the secretion of submandibular glands (SMGs) altered in renal hypertensive rats, and to explore whether t-AUCB could improve the salivary secretion. MAIN METHODS 2-kidney 1-clip Sprague-Dawley rats were used as renal hypertensive animals. t-AUCB treatment was given for 1 week after 8 weeks modeling. Blood pressure, blood perfusion and the secretion of SMGs, and endothelium-dependent relaxation of external maxillary artery were measured to investigate the effects of t-AUCB on the vascular tone and the secretion of SMGs in renal hypertensive rats. SMGs were collected for histological evaluation and the internal arteries were dissected for primary endothelial cells culture. KEY FINDINGS The blood perfusion and flow rate of SMGs in the renal hypertensive rats were significantly lower than those in the controls. Endothelium-dependent relaxation of the external maxillary artery and AMPK/Akt/eNOS signaling was impaired in hypertensive rats. The glandular morphology and the concentration of salivary ions did not change obviously. t-AUCB treatment ameliorated the secretion of SMGs, the blood perfusion, and the dysfunction of endothelium-dependent relaxation of the external maxillary artery by activating the AMPK/Akt/eNOS pathway in hypertensive rats. SIGNIFICANCE t-AUCB increases the blood perfusion through ameliorating dysfunction of endothelium-dependent relaxation of SMGs arteries and thus improves the hyposecretion of SMGs in hypertensive rats.
Collapse
Affiliation(s)
- Wen-Wen Han
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Xiao-Rui Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yu-Feng He
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Han-Shu Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.
| | - Guang-Yan Yu
- Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Li-Mei Liu
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.
| |
Collapse
|
5
|
Soluble Epoxide Hydrolase Inhibitor t-AUCB Ameliorates Vascular Endothelial Dysfunction by Influencing the NF-κB/miR-155-5p/eNOS/NO/IκB Cycle in Hypertensive Rats. Antioxidants (Basel) 2022; 11:antiox11071372. [PMID: 35883863 PMCID: PMC9311992 DOI: 10.3390/antiox11071372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs), angiogenic mediators degraded by soluble epoxide hydrolase (sEH), have been shown to exert beneficial effects on the cardiovascular system. The current study assessed the impact of increased EETs with an sEH inhibitor, t-AUCB, on two-kidney-one-clip (2K1C)-induced renovascular endothelial dysfunction, associated with hypertension, in rats. The hypertensive rats exhibited increased systolic blood pressure, reduced renal blood flow, impaired endothelium-dependent relaxation and eNOS phosphorylation in the renal arteries, elevated ROS production in the endothelium of the renal arteries, and decreased EET levels in plasma, the renal arteries, and endothelial cells; however, t-AUCB reversed all the deleterious effects. Moreover, we found that the stimulation of AMPK/UCP2 scavenged ROS and restored endothelial function in the renal arteries of hypertensive rats undergoing therapy with t-AUCB. In addition, we were the first to reveal the potential role of miR-155-5p in the occurrence and development of vascular endothelial dysfunction in hypertension. Importantly, t-AUCB recovered NO bioavailability by regulating the NF-κB/miR-155-5p/eNOS/NO/IκB cycle after the activation of AMPK/UCP2 and the subsequent inhibition of ROS in hypertensive rat renal artery endothelial cells. This study will provide evidence for this additional new mechanism, underlying the benefits of EETs and the related agents against hypertensive vasculopathy.
Collapse
|
6
|
A hypothesis-driven study to comprehensively investigate the association between genetic polymorphisms in EPHX2 gene and cardiovascular diseases: Findings from the UK Biobank. Gene X 2022; 822:146340. [PMID: 35183688 DOI: 10.1016/j.gene.2022.146340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Epoxyeicosatrienoic acids (EETs) are protective factors against cardiovascular diseases (CVDs) because of their vasodilatory, cholesterol-lowering, and anti-inflammatory effects. Soluble epoxide hydrolase (sEH), encoded by the EPHX2 gene, degrades EETs into less biologically active metabolites. EPHX2 is highly polymorphic, and genetic polymorphisms in EPHX2 have been linked to various types of CVDs, such as coronary heart disease, essential hypertension, and atrial fibrillation recurrence. METHODS Based on a priori hypothesis that EPHX2 genetic polymorphisms play an important role in the pathogenesis of CVDs, we comprehensively investigated the associations between 210 genetic polymorphisms in the EPHX2 gene and an array of 118 diseases in the circulatory system using a large sample from the UK Biobank (N = 307,516). The diseases in electronic health records were mapped to the phecode system, which was more representative of independent phenotypes. Survival analyses were employed to examine the effects of EPHX2 variants on CVD incidence, and a phenome-wide association study was conducted to study the impact of EPHX2 polymorphisms on 62 traits, including blood pressure, blood lipid levels, and inflammatory indicators. RESULTS A novel association between the intronic variant rs116932590 and the phenotype "aneurysm and dissection of heart" was identified. In addition, the rs149467044 and rs200286838 variants showed nominal evidence of association with arterial aneurysm and cerebrovascular disease, respectively. Furthermore, the variant rs751141, which was linked with a lower hydrolase activity of sEH, was significantly associated with metabolic traits, including blood levels of triglycerides, creatinine, and urate. CONCLUSIONS Multiple novel associations observed in the present study highlight the important role of EPHX2 genetic variation in the pathogenesis of CVDs.
Collapse
|
7
|
Tian Y, Li S, Dong K, Su X, Fu S, Lv X, Duan M, Yang T, Han Y, Hu G, Liu J, Sun Y, Yue H, Sun Y, Zhang H, Du Z, Miao Z, Tong M, Liu Y, Qin M, Gong P, Hou Y, Gao Z, Zhao Y. Discovery of benzamide derivatives containing urea moiety as soluble epoxide hydrolase inhibitors. Bioorg Chem 2022; 127:105898. [DOI: 10.1016/j.bioorg.2022.105898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022]
|
8
|
Ma Y, Bhuiyan MS, Kim I, Tang X. Editorial: Metabolic Regulation in the Development of Cardiovascular Diseases. Front Cell Dev Biol 2021; 9:768689. [PMID: 34722552 PMCID: PMC8548466 DOI: 10.3389/fcell.2021.768689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/21/2021] [Indexed: 01/17/2023] Open
Affiliation(s)
- Yimei Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - InKyeom Kim
- Department of Pharmacology, Cardiovascular Research Institute, BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Abdelazeem AH, El-Din AGS, Arab HH, El-Saadi MT, El-Moghazy SM, Amin NH. Design, synthesis and anti-inflammatory/analgesic evaluation of novel di-substituted urea derivatives bearing diaryl-1,2,4-triazole with dual COX-2/sEH inhibitory activities. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
10
|
Sun CP, Zhang XY, Zhou JJ, Huo XK, Yu ZL, Morisseau C, Hammock BD, Ma XC. Inhibition of sEH via stabilizing the level of EETs alleviated Alzheimer's disease through GSK3β signaling pathway. Food Chem Toxicol 2021; 156:112516. [PMID: 34411643 DOI: 10.1016/j.fct.2021.112516] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/04/2021] [Accepted: 08/14/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by dementia. Inhibition of soluble epoxide hydrolase (sEH) regulates inflammation involving in central nervous system (CNS) diseases. However, the exactly mechanism of sEH in AD is still unclear. In this study, we evaluated the vital role of sEH in amyloid beta (Aβ)-induced AD mice, and revealed a possible molecular mechanism for inhibition of sEH in the treatment of AD. The results showed that the sEH expression and activity were remarkably increased in the hippocampus of Aβ-induced AD mice. Chemical inhibition of sEH by TPPU, a selective sEH inhibitor, alleviated spatial learning and memory deficits, and elevated levels of neurotransmitters in Aβ-induced AD mice. Furthermore, inhibition of sEH could ameliorate neuroinflammation, neuronal death, and oxidative stress via stabilizing the in vivo level of epoxyeicosatrienoic acids (EETs), especially 8,9-EET and 14,15-EET, further resulting in the anti-AD effect through the regulation of GSK3β-mediated NF-κB, p53, and Nrf2 signaling pathways. These findings revealed the underlying mechanism of sEH as a potential therapeutic target in treatment of AD.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xin-Yue Zhang
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Jun-Jun Zhou
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Zhen-Long Yu
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, United States.
| | - Xiao-Chi Ma
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China.
| |
Collapse
|
11
|
Wu T, Xi X, Chen Y, Jiang C, Zhang Q, Dai G, Bai Y, Zhang W, Ni T, Zou J, Ju W, Xu M. Absolute protein assay for the simultaneous quantification of two epoxide hydrolases in rats by mass spectrometry-based targeted proteomics. J Sep Sci 2021; 44:2754-2763. [PMID: 34008891 DOI: 10.1002/jssc.202100066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/30/2021] [Accepted: 05/16/2021] [Indexed: 11/07/2022]
Abstract
Epoxide hydrolases catalyze the hydrolysis of both exogenous and endogenous epoxides to the corresponding vicinal diols by adding water. Microsomal and soluble epoxide hydrolase are two main mammalian enzymes that have been intensely characterized. The purpose of this investigation was to develop and validate a proteomics assay allowing the simultaneous quantification of microsomal and soluble epoxide hydrolase in rats. Protein quantification was realized through targeted proteomics using liquid chromatography with tandem mass spectrometry for the determination of trypsin-specific surrogate peptides after digestion. Stable isotope-labeled peptides were used as the internal standards. The chromatography of the surrogate peptides was performed on an Agilent SB C18 column (100 mm × 4.6 mm, 1.8 µm) with gradient elution. Acetonitrile containing 0.1% formic acid and 0.1% formic acid aqueous solution were used as mobile phases. A multiple reaction monitoring method in a positive ionization mode was used for the simultaneous detection of the peptides. The method was validated concerning the specificity, linearity, within-day and between-day accuracy and precision, matrix effect, stability, and digestion efficiency. The developed assay was successfully used to quantify the protein levels of microsomal and soluble epoxide hydrolase in rat liver, kidney, and heart S9 samples.
Collapse
Affiliation(s)
- Ting Wu
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Xiaoyun Xi
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Ying Chen
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Chao Jiang
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Qian Zhang
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Guoliang Dai
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Yongtao Bai
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, P. R. China
| | - Weidong Zhang
- Department of Pharmacy, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, P. R. China
| | - Ting Ni
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Jiandong Zou
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Wenzheng Ju
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| | - Meijuan Xu
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, P. R. China
| |
Collapse
|
12
|
Ripon MAR, Bhowmik DR, Amin MT, Hossain MS. Role of arachidonic cascade in COVID-19 infection: A review. Prostaglandins Other Lipid Mediat 2021; 154:106539. [PMID: 33592322 PMCID: PMC7882227 DOI: 10.1016/j.prostaglandins.2021.106539] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
The World Health Organization has described the 2019 Coronavirus disease caused by an influenza-like virus called SARS-CoV-2 as a pandemic. Millions of people worldwide are already infected by this virus, and severe infection causes hyper inflammation, thus disrupting lung function, exacerbating breath difficulties, and death. Various inflammatory mediators bio-synthesized through the arachidonic acid pathway play roles in developing cytokine storms, injuring virus-infected cells. Since pro-inflammatory eicosanoids, including prostaglandins, and leukotrienes, are key brokers for physiological processes such as inflammation, fever, allergy, and pain but, their function in COVID-19 is not well defined. This study addresses eicosanoid's crucial role through the arachidonic pathway in inflammatory cascading and recommends using bioactive lipids, NSAIDs, steroids, cell phospholipase A2 (cPLA2) inhibitors, and specialized pro-resolving mediators (SPMs) to treat COVID-19 disease. The role of soluble epoxide hydrolase inhibitors (SEHIs) in promoting the activity of epoxyeicosatrienoic acids (EETs) and 17-hydroxide-docosahexaenoic acid (17-HDHA) is also discussed. Additional research that assesses the eicosanoid profile in COVID-19 patients or preclinical models generates novel insights into coronavirus-host interaction and inflammation regulation.
Collapse
Affiliation(s)
- Md Abdur Rahman Ripon
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Dipty Rani Bhowmik
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Mohammad Tohidul Amin
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Mohammad Salim Hossain
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali 3814, Bangladesh.
| |
Collapse
|
13
|
Association of rs11780592 Polymorphism in the Human Soluble Epoxide Hydrolase Gene (EPHX2) with Oxidized LDL and Mortality in Patients with Diabetic Chronic Kidney Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8817502. [PMID: 34040693 PMCID: PMC8121583 DOI: 10.1155/2021/8817502] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 04/01/2021] [Accepted: 04/24/2021] [Indexed: 01/30/2023]
Abstract
Soluble epoxide hydrolase 2 (EPHX2) is an enzyme promoting increased cellular apoptosis through induction of oxidative stress (OS) and inflammation. The EPHX2 gene which encodes soluble EPHX2 might be implicated in the pathogenesis and development of OS and atherosclerosis. We aimed to assess the possible association between two functional polymorphisms of the EPHX2 gene (rs2741335 and rs11780592) with oxidized LDL (ox-LDL), carotid atherosclerosis, mortality, and cardiovascular (CV) disease in 118 patients with diabetic chronic kidney disease (CKD). At baseline, ox-LDL and carotid intima-media thickness (cIMT) were evaluated and all patients were followed for seven years with outcomes all-cause mortality and CV events. rs11780592 EPHX2 polymorphism was associated with ox-LDL, cIMT, albuminuria, and hypertension. Compared to AG and GG, AA homozygotes had higher values of albuminuria, ox-LDL, and cIMT (p = 0.046, p = 0.003, and p = 0.038, respectively). These associations remained significant, even after grouping for the G allele. After the follow-up period, 42/118 patients died (30/60 with AA genotype, 11/42 with AG genotype, and 1/12 with GG genotype) and 49/118 experienced a new CV event (fatal or nonfatal). The Kaplan-Meier analysis revealed that patients with the AA genotype exhibited a significantly higher mortality risk, compared to patients with AG and GG genotypes (p = 0.006). This association became even stronger, when AG and GG genotypes were grouped (AA vs. AG/GG, p = 0.002). AA homozygotes were strongly associated with all-cause mortality in both univariate (hazard ratio (HR) = 2.74, confidence interval (CI) = 1.40-5.35, p = 0.003) and multivariate Cox regression analysis (HR = 2.61, CI = 1.32-5.17, p = 0.006). In conclusion, our study demonstrated that genetic variations of EPHX2 gene were associated with increased circulating ox-LDL, increased cIMT, and all-cause mortality in diabetic CKD. Since EPHX2 regulates the cholesterol efflux and the oxidation of LDL in foam cells and macrophages, our study suggests that a genetic basis to endothelial dysfunction and OS might be present in diabetic CKD.
Collapse
|
14
|
Hamzaoui M, Roche C, Coquerel D, Duflot T, Brunel V, Mulder P, Richard V, Bellien J, Guerrot D. Soluble Epoxide Hydrolase Inhibition Prevents Experimental Type 4 Cardiorenal Syndrome. Front Mol Biosci 2021; 7:604042. [PMID: 33777999 PMCID: PMC7991096 DOI: 10.3389/fmolb.2020.604042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/14/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives: Cardiovascular diseases (CVD) remain the leading cause of morbimortality in patients with chronic kidney disease (CKD). The aim of this study was to assess the cardiovascular impact of the pharmacological inhibition of soluble epoxide hydrolase (sEH), which metabolizes the endothelium-derived vasodilatory and anti-inflammatory epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acid (DHETs), in the 5/6 nephrectomy (Nx) mouse model. Methods and Results: Compared to sham-operated mice, there was decrease in EET-to-DHET ratio 3 months after surgery in vehicle-treated Nx mice but not in mice treated with the sEH inhibitor t-AUCB. Nx induced an increase in plasma creatinine and in urine albumin-to-creatinine ratio as well as the development of kidney histological lesions, all of which were not modified by t-AUCB. In addition, t-AUCB did not oppose Nx-induced blood pressure increase. However, t-AUCB prevented the development of cardiac hypertrophy and fibrosis induced by Nx, as well as normalized the echocardiographic indices of diastolic and systolic function. Moreover, the reduction in endothelium-dependent flow-mediated dilatation of isolated mesenteric arteries induced by Nx was blunted by t-AUCB without change in endothelium-independent dilatation to sodium nitroprusside. Conclusion: Inhibition of sEH reduces the cardiac remodelling, and the diastolic and systolic dysfunctions associated with CKD. These beneficial effects may be mediated by the prevention of endothelial dysfunction, independent from kidney preservation and antihypertensor effect. Thus, inhibition of sEH holds a therapeutic potential in preventing type 4 cardiorenal syndrome.
Collapse
Affiliation(s)
- Mouad Hamzaoui
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France.,Nephrology Department, Rouen University Hospital, Rouen, France
| | - Clothilde Roche
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | - David Coquerel
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | - Thomas Duflot
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France.,Pharmacology Department, Rouen University Hospital, Rouen, France
| | - Valery Brunel
- Biochemistry Department, Rouen University Hospital, Rouen, France
| | - Paul Mulder
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | - Vincent Richard
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | - Jérémy Bellien
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France.,Pharmacology Department, Rouen University Hospital, Rouen, France
| | - Dominique Guerrot
- Normandie University, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France.,Nephrology Department, Rouen University Hospital, Rouen, France
| |
Collapse
|
15
|
Duflot T, Laurent C, Soudey A, Fonrose X, Hamzaoui M, Iacob M, Bertrand D, Favre J, Etienne I, Roche C, Coquerel D, Le Besnerais M, Louhichi S, Tarlet T, Li D, Brunel V, Morisseau C, Richard V, Joannidès R, Stanke-Labesque F, Lamoureux F, Guerrot D, Bellien J. Preservation of epoxyeicosatrienoic acid bioavailability prevents renal allograft dysfunction and cardiovascular alterations in kidney transplant recipients. Sci Rep 2021; 11:3739. [PMID: 33580125 PMCID: PMC7881112 DOI: 10.1038/s41598-021-83274-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/27/2021] [Indexed: 12/25/2022] Open
Abstract
This study addressed the hypothesis that epoxyeicosatrienoic acids (EETs) synthesized by CYP450 and catabolized by soluble epoxide hydrolase (sEH) are involved in the maintenance of renal allograft function, either directly or through modulation of cardiovascular function. The impact of single nucleotide polymorphisms (SNPs) in the sEH gene EPHX2 and CYP450 on renal and vascular function, plasma levels of EETs and peripheral blood monuclear cell sEH activity was assessed in 79 kidney transplant recipients explored at least one year after transplantation. Additional experiments in a mouse model mimicking the ischemia–reperfusion (I/R) injury suffered by the transplanted kidney evaluated the cardiovascular and renal effects of the sEH inhibitor t-AUCB administered in drinking water (10 mg/l) during 28 days after surgery. There was a long-term protective effect of the sEH SNP rs6558004, which increased EET plasma levels, on renal allograft function and a deleterious effect of K55R, which increased sEH activity. Surprisingly, the loss-of-function CYP2C9*3 was associated with a better renal function without affecting EET levels. R287Q SNP, which decreased sEH activity, was protective against vascular dysfunction while CYP2C8*3 and 2C9*2 loss-of-function SNP, altered endothelial function by reducing flow-induced EET release. In I/R mice, sEH inhibition reduced kidney lesions, prevented cardiac fibrosis and dysfunction as well as preserved endothelial function. The preservation of EET bioavailability may prevent allograft dysfunction and improve cardiovascular disease in kidney transplant recipients. Inhibition of sEH appears thus as a novel therapeutic option but its impact on other epoxyfatty acids should be carefully evaluated.
Collapse
Affiliation(s)
- Thomas Duflot
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France.,UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France.,Laboratory of Pharmacokinetics, Toxicology and Pharmacogenetics, Rouen University Hospital, 76000, Rouen, France
| | - Charlotte Laurent
- Department of Nephrology, Rouen University Hospital, 76000, Rouen, France
| | - Anne Soudey
- UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - Xavier Fonrose
- Department of Pharmacology, Grenoble Alpes University Hospital, HP2, INSERM U1042, University of Grenoble Alpes, 38000, Grenoble, France
| | - Mouad Hamzaoui
- UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France.,Department of Nephrology, Rouen University Hospital, 76000, Rouen, France
| | - Michèle Iacob
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France
| | - Dominique Bertrand
- Department of Nephrology, Rouen University Hospital, 76000, Rouen, France
| | - Julie Favre
- UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - Isabelle Etienne
- Department of Nephrology, Rouen University Hospital, 76000, Rouen, France
| | - Clothilde Roche
- UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - David Coquerel
- UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - Maëlle Le Besnerais
- UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - Safa Louhichi
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France.,UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - Tracy Tarlet
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France.,UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - Dongyang Li
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, Davis, CA, 95616, USA
| | - Valéry Brunel
- Department of General Biochemistry, Rouen University Hospital, 76000, Rouen, France
| | - Christophe Morisseau
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, Davis, CA, 95616, USA
| | - Vincent Richard
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France.,UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France
| | - Robinson Joannidès
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France.,UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France.,Centre d'Investigation Clinique (CIC)-INSERM 1404, Rouen University Hospital, 76000, Rouen, France
| | - Françoise Stanke-Labesque
- Department of Pharmacology, Grenoble Alpes University Hospital, HP2, INSERM U1042, University of Grenoble Alpes, 38000, Grenoble, France
| | - Fabien Lamoureux
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France.,UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France.,Laboratory of Pharmacokinetics, Toxicology and Pharmacogenetics, Rouen University Hospital, 76000, Rouen, France
| | - Dominique Guerrot
- UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France.,Department of Nephrology, Rouen University Hospital, 76000, Rouen, France
| | - Jérémy Bellien
- Department of Pharmacology, Rouen University Hospital, 76000, Rouen, France. .,UNIROUEN, INSERM U1096, FHU CARNAVAL, Normandie University, 76000, Rouen, France. .,Centre d'Investigation Clinique (CIC)-INSERM 1404, Rouen University Hospital, 76000, Rouen, France. .,Department of Pharmacology, Rouen University Hospital, 76031, Rouen Cedex, France.
| |
Collapse
|
16
|
Walker RE, Savinova OV, Pedersen TL, Newman JW, Shearer GC. Effects of inflammation and soluble epoxide hydrolase inhibition on oxylipin composition of very low-density lipoproteins in isolated perfused rat livers. Physiol Rep 2021; 9:e14480. [PMID: 33625776 PMCID: PMC7903942 DOI: 10.14814/phy2.14480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Oxylipins are metabolites of polyunsaturated fatty acids that mediate cardiovascular health by attenuation of inflammation, vascular tone, hemostasis, and thrombosis. Very low-density lipoproteins (VLDL) contain oxylipins, but it is unknown whether the liver regulates their concentrations. In this study, we used a perfused liver model to observe the effect of inflammatory lipopolysaccharide (LPS) challenge and soluble epoxide hydrolase inhibition (sEHi) on VLDL oxylipins. A compartmental model of deuterium-labeled linoleic acid and palmitic acid incorporation into VLDL was also developed to assess the dependence of VLDL oxylipins on fatty acid incorporation rates. LPS decreased the total fatty acid VLDL content by 30% [6%,47%], and decreased final concentration of several oxylipins by a similar amount (13-HOTrE, 35% [4%,55%], -1.3 nM; 9(10)-EpODE, 29% [3%,49%], -2.0 nM; 15(16)-EpODE, 29% [2%,49%], -1.6 nM; AA-derived diols, 32% [5%,52%], -2.4 nM; 19(20)-DiHDPA, 31% [7%,50%], -1.0 nM). However, the EPA-derived epoxide, 17(18)-EpETE, was decreased by 75% [49%,88%], (-0.52 nM) with LPS, double the suppression of other oxylipins. sEHi increased final concentration of DHA epoxide, 16(17)-EpDPE, by 99% [35%,193%], (2.0 nM). Final VLDL-oxylipin concentrations with LPS treatment were not correlated with linoleic acid kinetics, suggesting they were independently regulated under inflammatory conditions. We conclude that the liver regulates oxylipin incorporation into VLDL, and the oxylipin content is altered by LPS challenge and by inhibition of the epoxide hydrolase pathway. This provides evidence for delivery of systemic oxylipin signals by VLDL transport.
Collapse
Affiliation(s)
- Rachel E. Walker
- Department of Nutritional SciencesThe Pennsylvania State UniversityUniversity ParkPAUSA
| | - Olga V. Savinova
- Department of Biomedical SciencesNew York Institute of Technology College of Osteopathic MedicineOld WestburyNYUSA
- Sanford ResearchUniversity of South DakotaSioux FallsSDUSA
| | - Theresa L. Pedersen
- Advanced AnalyticsDavisCAUSA
- Department of Food Science and TechnologyUniversity of CaliforniaDavisCAUSA
| | - John W. Newman
- Department of Food Science and TechnologyUniversity of CaliforniaDavisCAUSA
- Obesity and Metabolism Research UnitWestern Human Nutrition Research CenterAgricultural Research ServiceUS Department of AgricultureDavisCAUSA
| | - Gregory C. Shearer
- Department of Nutritional SciencesThe Pennsylvania State UniversityUniversity ParkPAUSA
- Sanford ResearchUniversity of South DakotaSioux FallsSDUSA
- Sanford School of MedicineUniversity of South DakotaSioux FallsSDUSA
| |
Collapse
|
17
|
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: 13.3] [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
|
18
|
Imig JD, Jankiewicz WK, Khan AH. Epoxy Fatty Acids: From Salt Regulation to Kidney and Cardiovascular Therapeutics: 2019 Lewis K. Dahl Memorial Lecture. Hypertension 2020; 76:3-15. [PMID: 32475311 PMCID: PMC7448548 DOI: 10.1161/hypertensionaha.120.13898] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epoxyeicosatrienoic acids (EETs) are epoxy fatty acids that have biological actions that are essential for maintaining water and electrolyte homeostasis. An inability to increase EETs in response to a high-salt diet results in salt-sensitive hypertension. Vasodilation, inhibition of epithelial sodium channel, and inhibition of inflammation are the major EET actions that are beneficial to the heart, resistance arteries, and kidneys. Genetic and pharmacological means to elevate EETs demonstrated antihypertensive, anti-inflammatory, and organ protective actions. Therapeutic approaches to increase EETs were then developed for cardiovascular diseases. sEH (soluble epoxide hydrolase) inhibitors were developed and progressed to clinical trials for hypertension, diabetes mellitus, and other diseases. EET analogs were another therapeutic approach taken and these drugs are entering the early phases of clinical development. Even with the promise for these therapeutic approaches, there are still several challenges, unexplored areas, and opportunities for epoxy fatty acids.
Collapse
Affiliation(s)
- John D Imig
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| | - Wojciech K Jankiewicz
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| | - Abdul H Khan
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
19
|
Inhibition of soluble epoxide hydrolase attenuates renal tubular mitochondrial dysfunction and ER stress by restoring autophagic flux in diabetic nephropathy. Cell Death Dis 2020; 11:385. [PMID: 32439839 PMCID: PMC7242354 DOI: 10.1038/s41419-020-2594-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022]
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease (ESRD), and renal tubular cell dysfunction contributes to the pathogenesis of DN. Soluble epoxide hydrolase (sEH) is an enzyme that can hydrolyze epoxyeicosatrienoic acids (EETs) and other epoxy fatty acids (EpFAs) into the less biologically active metabolites. Inhibition of sEH has multiple beneficial effects on renal function, however, the exact role of sEH in hyperglycemia-induced dysfunction of tubular cells is still not fully elucidated. In the present study, we showed that human proximal tubular epithelial (HK-2) cells revealed an upregulation of sEH expression accompanied by the impairment of autophagic flux, mitochondrial dysfunction, ubiquitinated protein accumulation and enhanced endoplasmic reticulum (ER) stress after high glucose (HG) treatment. Furthermore, dysfunctional mitochondria accumulated in the cytoplasm, which resulted in excessive reactive oxygen species (ROS) generation, Bax translocation, cytochrome c release, and apoptosis. However, t-AUCB, an inhibitor of sEH, partially reversed these negative outcomes. Moreover, we also observed increased sEH expression, impaired autophagy flux, mitochondrial dysfunction and enhanced ER stress in the renal proximal tubular cells of db/db diabetic mice. Notably, inhibition of sEH by treatment with t-AUCB attenuated renal injury and partially restored autophagic flux, improved mitochondrial function, and reduced ROS generation and ER stress in the kidneys of db/db mice. Taken together, these results suggest that inhibition of sEH by t-AUCB plays a protective role in hyperglycemia-induced proximal tubular injury and that the potential mechanism of t-AUCB-mediated protective autophagy is involved in modulating mitochondrial function and ER stress. Thus, we provide new evidence linking sEH to the autophagic response during proximal tubular injury in the pathogenesis of DN and suggest that inhibition of sEH can be considered a potential therapeutic strategy for the amelioration of DN.
Collapse
|
20
|
Arad M, Waldman M, Abraham NG, Hochhauser E. Therapeutic approaches to diabetic cardiomyopathy: Targeting the antioxidant pathway. Prostaglandins Other Lipid Mediat 2020; 150:106454. [PMID: 32413571 DOI: 10.1016/j.prostaglandins.2020.106454] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/23/2020] [Accepted: 05/06/2020] [Indexed: 12/25/2022]
Abstract
The global epidemic of cardiovascular disease continues unabated and remains the leading cause of death both in the US and worldwide. We hereby summarize the available therapies for diabetes and cardiovascular disease in diabetics. Clearly, the current approaches to diabetic heart disease often target the manifestations and certain mediators but not the specific pathways leading to myocardial injury, remodeling and dysfunction. Better understanding of the molecular events determining the evolution of diabetic cardiomyopathy will provide insight into the development of specific and targeted therapies. Recent studies largely increased our understanding of the role of enhanced inflammatory response, ROS production, as well as the contribution of Cyp-P450-epoxygenase-derived epoxyeicosatrienoic acid (EET), Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1α (PGC-1α), Heme Oxygenase (HO)-1 and 20-HETE in pathophysiology and therapy of cardiovascular disease. PGC-1α increases production of the HO-1 which has a major role in protecting the heart against oxidative stress, microcirculation and mitochondrial dysfunction. This review describes the potential drugs and their downstream targets, PGC-1α and HO-1, as major loci for developing therapeutic approaches beside diet and lifestyle modification for the treatment and prevention of heart disease associated with obesity and diabetes.
Collapse
Affiliation(s)
- Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Maayan Waldman
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Tel Aviv, Israel
| | - Nader G Abraham
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
21
|
Sonnweber T, Pizzini A, Nairz M, Weiss G, Tancevski I. Arachidonic Acid Metabolites in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2018; 19:ijms19113285. [PMID: 30360467 PMCID: PMC6274989 DOI: 10.3390/ijms19113285] [Citation(s) in RCA: 257] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022] Open
Abstract
Lipid and immune pathways are crucial in the pathophysiology of metabolic and cardiovascular disease. Arachidonic acid (AA) and its derivatives link nutrient metabolism to immunity and inflammation, thus holding a key role in the emergence and progression of frequent diseases such as obesity, diabetes, non-alcoholic fatty liver disease, and cardiovascular disease. We herein present a synopsis of AA metabolism in human health, tissue homeostasis, and immunity, and explore the role of the AA metabolome in diverse pathophysiological conditions and diseases.
Collapse
Affiliation(s)
- Thomas Sonnweber
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
22
|
[Cardiovascular consequences of chronic kidney disease, impact of modulation of epoxyeicosatrienoic acids]. Ann Cardiol Angeiol (Paris) 2018; 67:141-148. [PMID: 29793671 DOI: 10.1016/j.ancard.2018.04.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 11/22/2022]
Abstract
Cardiovascular events are more prevalent in chronic kidney disease than in the general population, being the main cause of morbi-mortality. The physiopathology explaining this association remains complex. Thus, research for new therapies to prevent cardiovascular events in chronic kidney disease is a major issue. Epoxyeicosatrienoic acids, products of the arachidonic acid metabolism, are endothelium-derived hyperpolarizing factors with vasodilatory, anti-inflammatory, thrombolytic, pro-angiogenic and anti-apoptotic properties. A decrease in the bioavailability of epoxyeicosatrienoic acids has been observed in many cardiovascular diseases such as hypertension, myocardial infarction or diabetes. Moreover, human studies of genetic polymorphisms of soluble epoxide hydrolase, the enzyme degrading epoxyeicoatrienoic acids, have shown that allelic variants related to an increase in its activity is associated with higher risk of cardiovascular events. Modulation of epoxyeicosatrienoic acids by soluble epoxide hydrolase inhibitors in some cardiovascular diseases induces structural improvements in the heart, vessels and kidneys, including decrease in cardiomyocyte hypertrophy, reduction in cardiac and renal interstitial fibrosis, improvement in renal hemodynamics, and prevention of endothelial dysfunction. In this context, increasing the bioavailability of epoxyeicosatrienoic acids appears to be an interesting therapeutic option in the prevention of cardiovascular events related to chronic kidney disease.
Collapse
|
23
|
Jamieson KL, Endo T, Darwesh AM, Samokhvalov V, Seubert JM. Cytochrome P450-derived eicosanoids and heart function. Pharmacol Ther 2017; 179:47-83. [DOI: 10.1016/j.pharmthera.2017.05.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
A sensitive LC-MS/MS method for the quantification of regioisomers of epoxyeicosatrienoic and dihydroxyeicosatrienoic acids in human plasma during endothelial stimulation. Anal Bioanal Chem 2016; 409:1845-1855. [PMID: 27981341 DOI: 10.1007/s00216-016-0129-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/16/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) are vasodilating lipid mediators metabolized into dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase. We aimed to develop a LC-MS/MS method to quantify EETs and DHETs in human plasma and monitored their levels during vascular endothelial stimulation. Plasma samples, collected from 14 healthy and five hypertensive subjects at baseline and during radial artery endothelium-dependent flow-mediated dilatation, were spiked with internal standards. Lipids were then extracted by a modified Bligh and Dyer method and saponified to release bound EETs and DHETs. Samples were purified by a second liquid-liquid extraction and analyzed by LC-MS/MS. The assay allowed identification of (±)8(9)-epoxy-5Z,11Z,14Z-eicosatrienoic acid (8,9-EET); (±)11(12)-epoxy-5Z,8Z,14Z-eicosatrienoic acid (11,12-EET); (±)14(15)-epoxy-5Z,8Z,11Z-eicosatrienoic acid (14,15-EET); (±)8,9-dihydroxy-5Z,11Z,14Z-eicosatrienoic acid (8,9-DHET); (±)11,12-dihydroxy-5Z,8Z,14Z-eicosatrienoic acid (11,12-DHET); and (±)14,15-dihydroxy-5Z,8Z,11Z-eicosatrienoic acid (14,15-DHET). (±)5(6)-epoxy-5Z,11Z,14Z-eicosatrienoic acid (5,6-EET) was virtually undetectable due to its chemical instability. The limits of quantification were 0.25 ng/mL for DHETs and 0.5 ng/mL for EETs. Intra- and inter-assay variations ranged from 1.6 to 13.2%. Heating induced a similar increase in 8,9-EET, 11,12-EET, and 14,15-EET levels and in corresponding DHET levels in healthy but not in hypertensive subjects. We validated a sensitive LC-MS/MS method for measuring simultaneously plasma EET and DHET regioisomers in human plasma and showed its interest for assessing endothelial function.
Collapse
|
25
|
Akhnokh MK, Yang FH, Samokhvalov V, Jamieson KL, Cho WJ, Wagg C, Takawale A, Wang X, Lopaschuk GD, Hammock BD, Kassiri Z, Seubert JM. Inhibition of Soluble Epoxide Hydrolase Limits Mitochondrial Damage and Preserves Function Following Ischemic Injury. Front Pharmacol 2016; 7:133. [PMID: 27375480 PMCID: PMC4896112 DOI: 10.3389/fphar.2016.00133] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022] Open
Abstract
Aims: Myocardial ischemia can result in marked mitochondrial damage leading to cardiac dysfunction, as such identifying novel mechanisms to limit mitochondrial injury is important. This study investigated the hypothesis that inhibiting soluble epoxide hydrolase (sEH), responsible for converting epoxyeicosatrienoic acids to dihydroxyeicosatrienoic acids protects mitochondrial from injury caused by myocardial infarction. Methods: sEH null and WT littermate mice were subjected to surgical occlusion of the left anterior descending (LAD) artery or sham operation. A parallel group of WT mice received an sEH inhibitor, trans-4-[4-(3-adamantan-1-y1-ureido)-cyclohexyloxy]-benzoic acid (tAUCB; 10 mg/L) or vehicle in the drinking water 4 days prior and 7 days post-MI. Cardiac function was assessed by echocardiography prior- and 7-days post-surgery. Heart tissues were dissected into infarct, peri-, and non-infarct regions to assess ultrastructure by electron microscopy. Complexes I, II, IV, citrate synthase, PI3K activities, and mitochondrial respiration were assessed in non-infarct regions. Isolated working hearts were used to measure the rates of glucose and palmitate oxidation. Results: Echocardiography revealed that tAUCB treatment or sEH deficiency significantly improved systolic and diastolic function post-MI compared to controls. Reduced infarct expansion and less adverse cardiac remodeling were observed in tAUCB-treated and sEH null groups. EM data demonstrated mitochondrial ultrastructure damage occurred in infarct and peri-infarct regions but not in non-infarct regions. Inhibition of sEH resulted in significant improvements in mitochondrial respiration, ATP content, mitochondrial enzymatic activities and restored insulin sensitivity and PI3K activity. Conclusion: Inhibition or genetic deletion of sEH protects against long-term ischemia by preserving cardiac function and maintaining mitochondrial efficiency.
Collapse
Affiliation(s)
- Maria K Akhnokh
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of Alberta Edmonton, AB, Canada
| | - Feng Hua Yang
- Guangdong Laboratory Animal Monitoring Institute Guangdong, China
| | - Victor Samokhvalov
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of Alberta Edmonton, AB, Canada
| | - Kristi L Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of Alberta Edmonton, AB, Canada
| | - Woo Jung Cho
- Imaging Core Facility, Faculty of Medicine and Dentistry, University of Alberta Edmonton, AB, Canada
| | - Cory Wagg
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Abhijit Takawale
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Xiuhua Wang
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Gary D Lopaschuk
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada; Department of Pediatrics, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - Bruce D Hammock
- Department of Entomology and Nematology Comprehensive Cancer Center, University of California, Davis Davis, CA, USA
| | - Zamaneh Kassiri
- Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, 2-020M Katz Group Centre for Pharmacy and Health Research, University of AlbertaEdmonton, AB, Canada; Mazankowski Alberta Heart Institute, University of AlbertaEdmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of AlbertaEdmonton, AB, Canada
| |
Collapse
|
26
|
Lazaar AL, Yang L, Boardley RL, Goyal NS, Robertson J, Baldwin SJ, Newby DE, Wilkinson IB, Tal‐Singer R, Mayer RJ, Cheriyan J. Pharmacokinetics, pharmacodynamics and adverse event profile of GSK2256294, a novel soluble epoxide hydrolase inhibitor. Br J Clin Pharmacol 2016; 81:971-9. [PMID: 26620151 PMCID: PMC4834590 DOI: 10.1111/bcp.12855] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/25/2022] Open
Abstract
AIMS Endothelial-derived epoxyeicosatrienoic acids may regulate vascular tone and are metabolized by soluble epoxide hydrolase enzymes (sEH). GSK2256294 is a potent and selective sEH inhibitor that was tested in two phase I studies. METHODS Single escalating doses of GSK2256294 2-20 mg or placebo were administered in a randomized crossover design to healthy male subjects or obese smokers. Once daily doses of 6 or 18 mg or placebo were administered for 14 days to obese smokers. Data were collected on safety, pharmacokinetics, sEH enzyme inhibition and blood biomarkers. Single doses of GSK2256294 10 mg were also administered to healthy younger males or healthy elderly males and females with and without food. Data on safety, pharmacokinetics and biliary metabolites were collected. RESULTS GSK2256294 was well-tolerated with no serious adverse events (AEs) attributable to the drug. The most frequent AEs were headache and contact dermatitis. Plasma concentrations of GSK2256294 increased with single doses, with a half-life averaging 25-43 h. There was no significant effect of age, food or gender on pharmacokinetic parameters. Inhibition of sEH enzyme activity was dose-dependent, from an average of 41.9% on 2 mg (95% confidence interval [CI] -51.8, 77.7) to 99.8% on 20 mg (95% CI 99.3, 100.0) and sustained for up to 24 h. There were no significant changes in serum VEGF or plasma fibrinogen. CONCLUSIONS GSK2256294 was well-tolerated and demonstrated sustained inhibition of sEH enzyme activity. These data support further investigation in patients with endothelial dysfunction or abnormal tissue repair, such as diabetes, wound healing or COPD.
Collapse
Affiliation(s)
| | - Lucy Yang
- Experimental Medicine & Immunotherapeutics, Department of MedicineUniversity of Cambridge, and Cambridge Clinical Trials UnitCambridge
| | | | | | | | | | - David E. Newby
- University Centre for Cardiovascular Science, University of EdinburghEdinburghUK
| | - Ian B. Wilkinson
- Experimental Medicine & Immunotherapeutics, Department of MedicineUniversity of Cambridge, and Cambridge Clinical Trials UnitCambridge
| | | | | | - Joseph Cheriyan
- Experimental Medicine & Immunotherapeutics, Department of MedicineUniversity of Cambridge, and Cambridge Clinical Trials UnitCambridge
- GSK R&DStevenageCambridge and Ware
- Cambridge University Hospitals NHS Foundation TrustCambridge and
| |
Collapse
|
27
|
Kim JH, Cho CW, Tai BH, Yang SY, Choi GS, Kang JS, Kim YH. Soluble Epoxide Hydrolase Inhibitory Activity of Selaginellin Derivatives from Selaginella tamariscina. Molecules 2015; 20:21405-14. [PMID: 26633335 PMCID: PMC6331899 DOI: 10.3390/molecules201219774] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/23/2015] [Accepted: 11/26/2015] [Indexed: 11/22/2022] Open
Abstract
Selaginellin derivatives 1–3 isolated from Selaginellatamariscina were evaluated for their inhibition of soluble epoxide hydrolase (sEH) to demonstrate their potential for the treatment of cardiovascular disease. All selaginellin derivatives (1–3) inhibited sEH enzymatic activity and PHOME hydrolysis, in a dose-dependent manner, with IC50 values of 3.1 ± 0.1, 8.2 ± 2.2, and 4.2 ± 0.2 μM, respectively. We further determined that the derivatives function as non-competitive inhibitors. Moreover, the predicted that binding sites and interaction between 1–3 and sEH were solved by docking simulations. According to quantitative analysis, 1–3 were confirmed to have high content in the roots of S. tamariscina; among them, selaginellin 3 exhibited the highest content of 189.3 ± 0.0 μg/g.
Collapse
Affiliation(s)
- Jang Hoon Kim
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea.
- Department of Horticultural Environment, National Institute of Horticultural and Herbal Science, RDA, Wanju-gun, Jeollabuk-do 595-890, Korea.
| | - Chong Woon Cho
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea.
| | - Bui Huu Tai
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea.
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Caugiay, Hanoi 364-545, Vietnam.
| | - Seo Young Yang
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea.
| | - Gug-Seoun Choi
- Department of Horticultural Environment, National Institute of Horticultural and Herbal Science, RDA, Wanju-gun, Jeollabuk-do 595-890, Korea.
| | - Jong Seong Kang
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea.
| | - Young Ho Kim
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea.
| |
Collapse
|
28
|
Valverde E, Seira C, McBride A, Binnie M, Luque FJ, Webster SP, Bidon-Chanal A, Vázquez S. Searching for novel applications of the benzohomoadamantane scaffold in medicinal chemistry: Synthesis of novel 11β-HSD1 inhibitors. Bioorg Med Chem 2015; 23:7607-17. [DOI: 10.1016/j.bmc.2015.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 12/28/2022]
|