101
|
Reisdorf WC, Xie Q, Zeng X, Xie W, Rajpal N, Hoang B, Burgert ME, Kumar V, Hurle MR, Rajpal DK, O’Donnell S, MacDonald TT, Vossenkämper A, Wang L, Reilly M, Votta BJ, Sanchez Y, Agarwal P. Preclinical evaluation of EPHX2 inhibition as a novel treatment for inflammatory bowel disease. PLoS One 2019; 14:e0215033. [PMID: 31002701 PMCID: PMC6474586 DOI: 10.1371/journal.pone.0215033] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are signaling lipids produced by cytochrome P450 epoxygenation of arachidonic acid, which are metabolized by EPHX2 (epoxide hydrolase 2, alias soluble epoxide hydrolase or sEH). EETs have pleiotropic effects, including anti-inflammatory activity. Using a Connectivity Map (CMAP) approach, we identified an inverse-correlation between an exemplar EPHX2 inhibitor (EPHX2i) compound response and an inflammatory bowel disease patient-derived signature. To validate the gene-disease link, we tested a pre-clinical tool EPHX2i (GSK1910364) in a mouse disease model, where it showed improved outcomes comparable to or better than the positive control Cyclosporin A. Up-regulation of cytoprotective genes and down-regulation of proinflammatory cytokine production were observed in colon samples obtained from EPHX2i-treated mice. Follow-up immunohistochemistry analysis verified the presence of EPHX2 protein in infiltrated immune cells from Crohn's patient tissue biopsies. We further demonstrated that GSK2256294, a clinical EPHX2i, reduced the production of IL2, IL12p70, IL10 and TNFα in both ulcerative colitis and Crohn's disease patient-derived explant cultures. Interestingly, GSK2256294 reduced IL4 and IFNγ in ulcerative colitis, and IL1β in Crohn's disease specifically, suggesting potential differential effects of GSK2256294 in these two diseases. Taken together, these findings suggest a novel therapeutic use of EPHX2 inhibition for IBD.
Collapse
Affiliation(s)
- William C. Reisdorf
- Computational Biology, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
- * E-mail:
| | - Qing Xie
- Computational Biology, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Xin Zeng
- Target & Pathway Validation, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Wensheng Xie
- Target & Pathway Validation, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Neetu Rajpal
- Computational Biology, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Bao Hoang
- Exploratory Biomarkers, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Mark E. Burgert
- Research Statistics, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Vinod Kumar
- Computational Biology, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Mark R. Hurle
- Computational Biology, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Deepak K. Rajpal
- Computational Biology, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Sarah O’Donnell
- Centre for Digestive Diseases, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | | | - Anna Vossenkämper
- Centre for Immunobiology, Blizard Institute, QMUL, London, United Kingdom
| | - Lin Wang
- Pattern Recognition Receptor DPU, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Mike Reilly
- Pattern Recognition Receptor DPU, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Bart J. Votta
- Pattern Recognition Receptor DPU, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Yolanda Sanchez
- Stress and Repair DPU, Respiratory Therapy Area, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Pankaj Agarwal
- Computational Biology, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| |
Collapse
|
102
|
Borkowski K, Yim SJ, Holt RR, Hackman RM, Keen CL, Newman JW, Shearer GC. Walnuts change lipoprotein composition suppressing TNFα-stimulated cytokine production by diabetic adipocyte. J Nutr Biochem 2019; 68:51-58. [PMID: 31030167 DOI: 10.1016/j.jnutbio.2019.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/31/2019] [Accepted: 03/12/2019] [Indexed: 12/15/2022]
Abstract
Walnut consumption can provide both vascular and metabolic health benefits, and walnut-induced changes in lipoprotein particle chemical payloads may be responsible for these health benefits. To explore this possibility with a focus on metabolic health, this study investigated the impact of walnut consumption on lipoprotein lipid composition and changes in LDL anti-inflammatory properties, as reported by inflamed adipocyte. Hypercholesterolemic, postmenopausal females were treated with 40 g/day (i.e., 1.6 servings/day; n=15) of walnuts for 4 weeks. Fatty acids and their oxygenated metabolites, i.e., oxylipins, were quantified in isolated lipoproteins. Human primary adipocytes were exposed to LDL and TNFα-stimulated adipokine production was measured. Walnut treatment elevated α-linolenic acid and its epoxides in all lipoproteins and depleted mid-chain alcohols in VLDL and LDL, but not HDL. Walnuts also reduced TNFα-induced diabetic adipocyte production of IL-6 (-48%, P=.0006) and IL-8 (-30%, P=.01), changes inversely correlated with levels of α-linolenic acid-derived epoxides but not α-linolenic acid itself. In conclusion, modest walnut consumption can alter lipoprotein lipid profiles and enhance their ability to inhibit TNFα-dependent pro-inflammatory responses in human diabetic primary adipocytes. Moreover, this study suggests the oxylipins, rather than the parent fatty acids, mediate LDL action of adipocytes.
Collapse
Affiliation(s)
- Kamil Borkowski
- Dept. of Nutritional Sciences, The Pennsylvania State University, University Park, PA; West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA; Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.
| | - Sun J Yim
- Department of Nutrition, University of California Davis, Davis, CA
| | - Roberta R Holt
- Department of Nutrition, University of California Davis, Davis, CA
| | - Robert M Hackman
- Department of Nutrition, University of California Davis, Davis, CA
| | - Carl L Keen
- Department of Nutrition, University of California Davis, Davis, CA
| | - John W Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA; Department of Nutrition, University of California Davis, Davis, CA; Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA
| | - Gregory C Shearer
- Dept. of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| |
Collapse
|
103
|
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: 14] [Impact Index Per Article: 2.3] [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
|
104
|
Hashimoto K. Role of Soluble Epoxide Hydrolase in Metabolism of PUFAs in Psychiatric and Neurological Disorders. Front Pharmacol 2019; 10:36. [PMID: 30761004 PMCID: PMC6363819 DOI: 10.3389/fphar.2019.00036] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/14/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammation plays a key role in the pathogenesis of a number of psychiatric and neurological disorders. Soluble epoxide hydrolases (sEH), enzymes present in all living organisms, metabolize epoxy fatty acids (EpFAs) to corresponding 1,2-diols by the addition of a molecule of water. Accumulating evidence suggests that sEH in the metabolism of polyunsaturated fatty acids (PUFAs) plays a key role in inflammation. Preclinical studies demonstrated that protein expression of sEH in the prefrontal cortex, striatum, and hippocampus from mice with depression-like phenotype was higher than control mice. Furthermore, protein expression of sEH in the parietal cortex from patients with major depressive disorder was higher than controls. Interestingly, Ephx2 knock-out (KO) mice exhibit stress resilience after chronic social defeat stress. Furthermore, the sEH inhibitors have antidepressant effects in animal models of depression. In addition, pharmacological inhibition or gene KO of sEH protected against dopaminergic neurotoxicity in the striatum after repeated administration of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) in an animal model of Parkinson’s disease (PD). Protein expression of sEH in the striatum from MPTP-treated mice was higher than control mice. A number of studies using postmortem brain samples showed that the deposition of protein aggregates of α-synuclein, termed Lewy bodies, is evident in multiple brain regions of patients from PD and dementia with Lewy bodies (DLB). Moreover, the expression of the sEH protein in the striatum from patients with DLB was significantly higher compared with controls. Interestingly, there was a positive correlation between sEH expression and the ratio of phosphorylated α-synuclein to α-synuclein in the striatum. In the review, the author discusses the role of sEH in the metabolism of PUFAs in inflammation-related psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Center for Forensic Mental Health, Chiba University, Chiba, Japan
| |
Collapse
|
105
|
Ning J, Liu T, Dong P, Wang W, Ge G, Wang B, Yu Z, Shi L, Tian X, Huo X, Feng L, Wang C, Sun C, Cui J, James TD, Ma X. Molecular Design Strategy to Construct the Near-Infrared Fluorescent Probe for Selectively Sensing Human Cytochrome P450 2J2. J Am Chem Soc 2019; 141:1126-1134. [PMID: 30525564 DOI: 10.1021/jacs.8b12136] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cytochrome P450 2J2 (CYP2J2), a key enzyme responsible for oxidative metabolism of various xenobiotics and endogenous compounds, participates in a diverse array of physiological and pathological processes in humans. Its biological role in tumorigenesis and cancer diagnosis remains poorly understood, owing to the lack of molecular tools suitable for real-time monitoring CYP2J2 in complex biological systems. Using molecular design principles, we were able to modify the distance between the catalytic unit and metabolic recognition moiety, allowing us to develop a CYP2J2 selective fluorescent probe using a near-infrared fluorophore ( E)-2-(2-(6-hydroxy-2, 3-dihydro-1 H-xanthen-4-yl)vinyl)-3,3-dimethyl-1-propyl-3 H-indol-1-ium iodide (HXPI). To improve the reactivity and isoform specificity, a self-immolative linker was introduced to the HXPI derivatives in order to better fit the narrow substrate channel of CYP2J2, the modification effectively shortened the spatial distance between the metabolic moiety ( O-alkyl group) and catalytic center of CYP2J2. After screening a panel of O-alkylated HXPI derivatives, BnXPI displayed the best combination of specificity, sensitivity and applicability for detecting CYP2J2 in vitro and in vivo. Upon O-demethylation by CYP2J2, a self-immolative reaction occurred spontaneously via 1,6-elimination of p-hydroxybenzyl resulting in the release of HXPI. Allowing BnXPI to be successfully used to monitor CYP2J2 activity in real-time for various living systems including cells, tumor tissues, and tumor-bearing animals. In summary, our practical strategy could help the development of a highly specific and broadly applicable tool for monitoring CYP2J2, which offers great promise for exploring the biological functions of CYP2J2 in tumorigenesis.
Collapse
Affiliation(s)
- Jing Ning
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China.,State Key Laboratory of Fine Chemicals, Dalian University of Technology , Dalian 116024 , China
| | - Tao Liu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology , Dalian 116024 , China
| | - Peipei Dong
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Sino-Pakistan TCM and Ethnomedicine Research 8 Center, School of Pharmacy , Hunan University of Chinese Medicine , Changsha 410208 , China
| | - Guangbo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Bo Wang
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Zhenlong Yu
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Lei Shi
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Xiangge Tian
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Xiaokui Huo
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Lei Feng
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China.,State Key Laboratory of Fine Chemicals, Dalian University of Technology , Dalian 116024 , China
| | - Chao Wang
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Chengpeng Sun
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| | - Jingnan Cui
- State Key Laboratory of Fine Chemicals, Dalian University of Technology , Dalian 116024 , China
| | - Tony D James
- Department of Chemistry , University of Bath , Bath BA2 7AY , United Kingdom
| | - Xiaochi Ma
- College of Integrative Medicine, The National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy , Dalian Medical University , Dalian 116044 , China
| |
Collapse
|
106
|
Yeh CF, Chuang TY, Hung YW, Lan MY, Tsai CH, Huang HX, Lin YY. Soluble epoxide hydrolase inhibition enhances anti-inflammatory and antioxidative processes, modulates microglia polarization, and promotes recovery after ischemic stroke. Neuropsychiatr Dis Treat 2019; 15:2927-2941. [PMID: 31686827 PMCID: PMC6800549 DOI: 10.2147/ndt.s210403] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/04/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Ischemic stroke triggers inflammatory responses and oxidative stress in the brain, and microglia polarization affects the degree of neuroinflammation. It has been reported that the inhibition of soluble epoxide hydrolase (sEH) activity protects brain tissue. However, the anti-inflammatory and antioxidative effects of sEH inhibition in the ischemic brain are not fully understood. This study aimed to investigate the effects of a selective sEH inhibitor, 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA), after ischemic stroke. METHODS Adult male rats with middle cerebral artery occlusion (MCAO) were administered with AUDA or a vehicle. Behavioral outcome, infarct volume, microglia polarization, and gene expression were assessed. RESULTS Rats treated with AUDA showed better behavioral outcomes and smaller infarct volumes after MCAO. After AUDA treatment, a reduction of M1 microglia and an increase of M2 microglia occurred at the ischemic cortex of rats. Additionally, there was an increase in the mRNA expressions of antioxidant enzymes and anti-inflammatory interleukin-10, and pro-inflammatory mediators were decreased after AUDA administration. Heme oxygenase-1 was mainly expressed by neurons, and AUDA was found to improve the survival of neurons. CONCLUSION The results of this study provided novel and significant insights into how AUDA can improve outcomes and modulate inflammation and oxidative stress after ischemic stroke.
Collapse
Affiliation(s)
- Chien-Fu Yeh
- Institute of Brain Science, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Otorhinolaryngology, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Tung-Yueh Chuang
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yu-Wen Hung
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Ming-Ying Lan
- Department of Otorhinolaryngology, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Ching-Han Tsai
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Hao-Xiang Huang
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yung-Yang Lin
- Institute of Brain Science, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,Institute of Physiology, National Yang-Ming University, Taipei 11221, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| |
Collapse
|
107
|
Subterminal hydroxyeicosatetraenoic acids: Crucial lipid mediators in normal physiology and disease states. Chem Biol Interact 2018; 299:140-150. [PMID: 30543782 DOI: 10.1016/j.cbi.2018.12.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 02/08/2023]
Abstract
Cytochrome P450 (P450) enzymes are superfamily of monooxygenases that hold the utmost diversity of substrate structures and catalytic reaction forms amongst all other enzymes. P450 enzymes metabolize arachidonic acid (AA) to a wide array of biologically active lipid mediators. P450-mediated AA metabolites have a significant role in normal physiological and pathophysiological conditions, hence they could be promising therapeutic targets in different disease states. P450 monooxygenases mediate the (ω-n)-hydroxylation reactions, which involve the introduction of a hydroxyl group to the carbon skeleton of AA, forming subterminal hydroxyeicosatetraenoic acids (HETEs). In the current review, we specified different P450 isozymes implicated in the formation of subterminal HETEs in varied tissues. In addition, we focused on the role of subterminal HETEs namely 19-HETE, 16-HETE, 17-HETE and 18-HETE in different organs, importantly the kidneys, heart, liver and brain. Furthermore, we highlighted their role in hypertension, acute coronary syndrome, diabetic retinopathy, non-alcoholic fatty liver disease, ischemic stroke as well as inflammatory diseases. Since each member of subterminal HETEs exist as R and S enantiomer, we addressed the issue of stereoselectivity related to the formation and differential effects of these enantiomers. In conclusion, elucidation of different roles of subterminal HETEs in normal and disease states leads to identification of novel therapeutic targets and development of new therapeutic modalities in different disease states.
Collapse
|
108
|
Sjödin MOD, Checa A, Yang M, Dahlén SE, Wheelock ÅM, Eklund A, Grunewald J, Wheelock CE. Soluble epoxide hydrolase derived lipid mediators are elevated in bronchoalveolar lavage fluid from patients with sarcoidosis: a cross-sectional study. Respir Res 2018; 19:236. [PMID: 30509266 PMCID: PMC6276236 DOI: 10.1186/s12931-018-0939-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/14/2018] [Indexed: 01/04/2023] Open
Abstract
Background Sarcoidosis is a systemic inflammatory multi-organ disease almost always affecting the lungs. The etiology remains unknown, but the hallmark of sarcoidosis is formation of non-caseating epithelioid cells granulomas in involved organs. In Scandinavia, > 30% of sarcoidosis patients have Löfgren’s syndrome (LS), an acute disease onset mostly indicating a favorable prognosis. The impact of dysregulation of lipid mediators, which has been investigated in other inflammatory disorders, is still unknown. Methods Using three different liquid chromatography coupled to tandem mass spectrometry targeted platforms (LC-MS/MS), we quantified a broad suite of lipid mediators including eicosanoids, sphingolipids and endocannabinoids in bronchoalveolar lavage (BAL) fluid from pulmonary sarcoidosis patients (n = 41) and healthy controls (n = 16). Results A total of 47 lipid mediators were consistently detected in BAL fluid of patients and controls. After false discovery rate adjustment, two products of the soluble epoxide hydrolase (sEH) enzyme, 11,12-dihydroxyeicosa-5,8,14-trienoic acid (11,12-DiHETrE, p = 4.4E-5, q = 1.2E-3, median fold change = 6.0) and its regioisomer 14,15-dihydroxyeicosa-5,8,11-trienoic acid (14,15-DiHETrE, p = 3.6E-3, q = 3.2E-2, median fold change = 1.8) increased in patients with sarcoidosis. Additional shifts were observed in sphingolipid metabolism, with a significant increase in palmitic acid-derived sphingomyelin (SM16:0, p = 1.3E-3, q = 1.7E-2, median fold change = 1.3). No associations were found between these 3 lipid mediators and LS, whereas levels of SM 16:0 and 11,12-DiHETrE associated with radiological stage (p < 0.05), and levels of 14,15-DiHETrE were associated with the BAL fluid CD4/CD8 ratio. Conclusions These observed shifts in lipid mediators provide new insights into the pathobiology of sarcoidosis and in particular highlight the sEH pathway to be dysregulated in disease. Electronic supplementary material The online version of this article (10.1186/s12931-018-0939-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marcus O D Sjödin
- Division of Physiological Chemistry II, Department of Medical Biochemistry & Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden.,Experimental Asthma & Allergy Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Checa
- Division of Physiological Chemistry II, Department of Medical Biochemistry & Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Mingxing Yang
- Respiratory Medicine Unit, Department of Medicine and Center for Molecular Medicine (CMM), Karolinska Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Sven-Erik Dahlén
- Experimental Asthma & Allergy Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine and Center for Molecular Medicine (CMM), Karolinska Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Anders Eklund
- Respiratory Medicine Unit, Department of Medicine and Center for Molecular Medicine (CMM), Karolinska Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine and Center for Molecular Medicine (CMM), Karolinska Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry & Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden.
| |
Collapse
|
109
|
Abstract
Therapeutics for arachidonic acid pathways began with the development of non-steroidal anti-inflammatory drugs that inhibit cyclooxygenase (COX). The enzymatic pathways and arachidonic acid metabolites and respective receptors have been successfully targeted and therapeutics developed for pain, inflammation, pulmonary and cardiovascular diseases. These drugs target the COX and lipoxygenase pathways but not the third branch for arachidonic acid metabolism, the cytochrome P450 (CYP) pathway. Small molecule compounds targeting enzymes and CYP epoxy-fatty acid metabolites have evolved rapidly over the last two decades. These therapeutics have primarily focused on inhibiting soluble epoxide hydrolase (sEH) or agonist mimetics for epoxyeicosatrienoic acids (EET). Based on preclinical animal model studies and human studies, major therapeutic indications for these sEH inhibitors and EET mimics/analogs are renal and cardiovascular diseases. Novel small molecules that inhibit sEH have advanced to human clinical trials and demonstrate promise for cardiovascular diseases. Challenges remain for sEH inhibitor and EET analog drug development; however, there is a high likelihood that a drug that acts on this third branch of arachidonic acid metabolism will be utilized to treat a cardiovascular or kidney disease in the next decade.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
110
|
Tantray JA, Reddy KP, Jamil K, Yerra SK. Role of cytochrome epoxygenase (CYP2J2) in the pathophysiology of coronary artery disease in South Indian population. Indian Heart J 2018; 71:60-64. [PMID: 31000184 PMCID: PMC6477124 DOI: 10.1016/j.ihj.2018.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/29/2018] [Accepted: 11/05/2018] [Indexed: 12/27/2022] Open
Abstract
Background The cytochrome P-450 2J2 (CYP2J2) is known to be one of the major enzymes of epoxygenase pathway of arachidonic acid in extrahepatic tissues, which produces series of regioisomeric cis-epoxyeicosatrienoic acids (EETs) such as 5,6-, 8,9-, 11,12-, and 14,15-EETs. In the present study, we analyzed the impact of a genetic variant in CYP2J2 on coronary artery disease (CAD) in the Telangana region of Indian population. Material and methods The case–control study consisted of 100 CAD cases and 110 healthy controls. The deoxyribonucleic acid was extracted using the salting out method. Genotyping and gene expression was performed by polymerase chain reaction (PCR)-restriction fragment length polymorphism and real-time-PCR methods. Results In the present study, the percentage of smokers, alcoholics, hypertensive patients, and diabetics was high. Increase in fasting glucose, urea, creatinine, fasting triglycerides, total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-C), total cholesterol/high-density lipoprotein (TC/HDL), LDL/HDL, homocysteine, and C-reactive protein levels were significantly higher in patients with CAD than in controls (p < 0.001). CYP2J2 G-50T was associated with CAD (p = 0.04). The mRNA expression of CYP2J2 showed altered gene expression in this study among CAD patients in comparison with control (p = 0.01). Conclusions A functionally relevant polymorphism of the CYP2J2 gene was independently associated with an increased risk of CAD.
Collapse
Affiliation(s)
- Javeed Ahmad Tantray
- Department of Zoology, Osmania University Hyderabad, Telangana, India; Dept. of Genetics, Bhagwan Mahavir Medical Research Centre, 10-1-1, Mahavir Marg, Hyderabad-500004, Telangana, India.
| | - K Pratap Reddy
- Department of Zoology, Osmania University Hyderabad, Telangana, India.
| | - Kaiser Jamil
- Dept. of Genetics, Bhagwan Mahavir Medical Research Centre, 10-1-1, Mahavir Marg, Hyderabad-500004, Telangana, India.
| | - Shiva Kumar Yerra
- Department of Cardiology, Mahavir Hospital and Research Centre Hyderabad, Telangana, India.
| |
Collapse
|
111
|
Neckář J, Hsu A, Hye Khan MA, Gross GJ, Nithipatikom K, Cyprová M, Benák D, Hlaváčková M, Sotáková-Kašparová D, Falck JR, Sedmera D, Kolář F, Imig JD. Infarct size-limiting effect of epoxyeicosatrienoic acid analog EET-B is mediated by hypoxia-inducible factor-1α via downregulation of prolyl hydroxylase 3. Am J Physiol Heart Circ Physiol 2018; 315:H1148-H1158. [PMID: 30074840 PMCID: PMC6734065 DOI: 10.1152/ajpheart.00726.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 07/06/2018] [Accepted: 07/18/2018] [Indexed: 12/27/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) decrease cardiac ischemia-reperfusion injury; however, the mechanism of their protective effect remains elusive. Here, we investigated the cardioprotective action of a novel EET analog, EET-B, in reperfusion and the role of hypoxia-inducible factor (HIF)-1α in such action of EET-B. Adult male rats were subjected to 30 min of left coronary artery occlusion followed by 2 h of reperfusion. Administration of 14,15-EET (2.5 mg/kg) or EET-B (2.5 mg/kg) 5 min before reperfusion reduced infarct size expressed as a percentage of the area at risk from 64.3 ± 1.3% in control to 42.6 ± 1.9% and 46.0 ± 1.6%, respectively, and their coadministration did not provide any stronger effect. The 14,15-EET antagonist 14,15-epoxyeicosa-5( Z)-enoic acid (2.5 mg/kg) inhibited the infarct size-limiting effect of EET-B (62.5 ± 1.1%). Similarly, the HIF-1α inhibitors 2-methoxyestradiol (2.5 mg/kg) and acriflavine (2 mg/kg) completely abolished the cardioprotective effect of EET-B. In a separate set of experiments, the immunoreactivity of HIF-1α and its degrading enzyme prolyl hydroxylase domain protein 3 (PHD3) were analyzed in the ischemic areas and nonischemic septa. At the end of ischemia, the HIF-1α immunogenic signal markedly increased in the ischemic area compared with the septum (10.31 ± 0.78% vs. 0.34 ± 0.08%). After 20 min and 2 h of reperfusion, HIF-1α immunoreactivity decreased to 2.40 ± 0.48% and 1.85 ± 0.43%, respectively, in the controls. EET-B blunted the decrease of HIF-1α immunoreactivity (7.80 ± 0.69% and 6.44 ± 1.37%, respectively) and significantly reduced PHD3 immunogenic signal in ischemic tissue after reperfusion. In conclusion, EET-B provides an infarct size-limiting effect at reperfusion that is mediated by HIF-1α and downregulation of its degrading enzyme PHD3. NEW & NOTEWORTHY The present study shows that EET-B is an effective agonistic 14,15-epoxyeicosatrienoic acid analog, and its administration before reperfusion markedly reduced myocardial infarction in rats. Most importantly, we demonstrate that increased hypoxia-inducible factor-1α levels play a role in cardioprotection mediated by EET-B in reperfusion likely by mechanisms including downregulation of the hypoxia-inducible factor -1α-degrading enzyme prolyl hydroxylase domain protein 3.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/pharmacology
- 8,11,14-Eicosatrienoic Acid/therapeutic use
- Animals
- Disease Models, Animal
- Down-Regulation
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor-Proline Dioxygenases/genetics
- Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism
- Male
- Myocardial Infarction/enzymology
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/enzymology
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/physiopathology
- Myocardial Reperfusion Injury/prevention & control
- Myocardium/enzymology
- Myocardium/pathology
- Proteolysis
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Jan Neckář
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine Physiology , Prague , Czech Republic
| | - Anna Hsu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Md Abdul Hye Khan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Garrett J Gross
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Kasem Nithipatikom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Michaela Cyprová
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Daniel Benák
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Markéta Hlaváčková
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Dita Sotáková-Kašparová
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas
| | - David Sedmera
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University , Prague , Czech Republic
| | - František Kolář
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
112
|
Huang A, Sun D. Sexually Dimorphic Regulation of EET Synthesis and Metabolism: Roles of Estrogen. Front Pharmacol 2018; 9:1222. [PMID: 30420806 PMCID: PMC6215857 DOI: 10.3389/fphar.2018.01222] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/08/2018] [Indexed: 01/03/2023] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid via cytochrome P450 (CYP)/epoxygenase and are hydrolyzed by soluble epoxide hydrolase (sEH). Circulating and tissue levels of EETs are controlled by CYP (EET synthesis) and sEH (EET degradation). Therefore, both increases in CYP activity and decreases in sEH expression potentiate EET bioavailability, responses that prevail in the female sex as a function of estrogen. This mini review, based on subtitles listed, briefly summarizes studies focusing specifically on (1) female-specific potentiation of CYP/epoxygenase activity to compensate for the endothelial dysfunction; and (2) estrogen-dependent downregulation of sEH expression, which yields divergent actions in both systemic and pulmonary circulation, respectively. Estrogen-Potentiating EET Synthesis in Response to Endothelial Dysfunction: This section summarizes the current understanding regarding the roles of estrogen in facilitating EET synthesis in response to endothelial dysfunction. In this regard, estrogen recruitment of EET-driven signaling serves as a back-up mechanism, which compensates for NO deficiency to preserve endothelium-dependent vasodilator responses and maintain normal blood pressure. Estrogen-Dependent Downregulation of Ephx2/sEH Expression: This section focuses on molecular mechanisms responsible for the female-specific downregulation of sEH expression. Roles of EETs in Systemic Circulation, as a Function of Estrogen-Dependent Downregulation of sEH: This section summarizes studies conducted on animals that are either deficient in the Ephx2 gene (sEH-KO) or have been treated with sEH inhibitors (sEHIs), and exhibit EET-mediated cardiovascular protections in the cerebral, coronary, skeletal, and splanchnic circulations. In particular, the estrogen-inherent silencing of the Ephx2 gene duplicates the action of sEH deficiency, yielding comparable adaptations in attenuated myogenic vasoconstriction, enhanced shear stress-induced vasodilation, and improved cardiac contractility among female WT mice, male sEH-KO and sEHI-treated mice. Roles of Estrogen-Driven EET Production in Pulmonary Circulation: This section reviews epidemiological and clinical studies that provide the correlation between the polymorphism, or mutation of gene(s) involving estrogen metabolism and female predisposition to pulmonary hypertension, and specifically addresses an intrinsic causation between the estrogen-dependent downregulation of Ephx2 gene/sEH expression and female-susceptibility of being pulmonary hypertensive, a topic that has never been explored before. Additionally, the issue of the “estrogen paradox” in the incidence and prognosis of pulmonary hypertension is discussed.
Collapse
Affiliation(s)
- An Huang
- Department of Physiology, New York Medical College, Valhalla, NY, United States
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
113
|
Shi X, Ohta Y, Shang J, Morihara R, Nakano Y, Fukui Y, Liu X, Feng T, Huang Y, Sato K, Takemoto M, Hishikawa N, Yamashita T, Suzuki E, Hasumi K, Abe K. Neuroprotective effects of SMTP-44D in mice stroke model in relation to neurovascular unit and trophic coupling. J Neurosci Res 2018; 96:1887-1899. [PMID: 30242877 DOI: 10.1002/jnr.24326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/26/2018] [Accepted: 08/27/2018] [Indexed: 12/21/2022]
Abstract
Stachybotrys microspora triprenyl phenol (SMTP)-44D has both anti-oxidative and anti-inflammatory activities, but its efficacy has not been proved in relation to the pathological changes of neurovascular unit (NVU) and neurovascular trophic coupling (NVTC) in ischemic stroke. Here, the present study was designed to assess the efficacies of SMTP-44D, moreover, compared with the standard neuroprotective reagent edaravone in ischemic brains. ICR mice were subjected to transient middle cerebral artery occlusion (tMCAO) for 60 min, SMTP-44D (10 mg/kg) or edaravone (3 mg/kg) was intravenously administrated through subclavian vein just after the reperfusion, and these mice were examined at 1, 3, and 7 d after reperfusion. Compared with the vehicle group, SMTP-44D treatment revealed obvious ameliorations in clinical scores and infarct volume, meanwhile, markedly suppressed the accumulations of 4-HNE, 8-OHdG, nitrotyrosine, RAGE, TNF-α, Iba-1, and cleaved caspase-3 after tMCAO. In addition, SMTP-44D significantly prevented the dissociation of NVU and improved the intensity of NAGO/BDNF and the number of BDNF/TrkB and BDNF/NeuN double positive cells. These effects of SMTP-44D in reducing oxidative and inflammatory stresses were similar to or stronger than those of edaravone. The present study demonstrated that SMTP-44D showed strong anti-oxidative, anti-inflammatory, and anti-apoptotic effects, moreover, the drug also significantly improved the NVU damage and NVTC in the ischemic brain.
Collapse
Affiliation(s)
- Xiaowen Shi
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jingwei Shang
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xia Liu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tian Feng
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yong Huang
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kota Sato
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Eriko Suzuki
- Department of Applied Biological Science, Tokyo Noko University, Fuchu, Japan
| | - Keiji Hasumi
- Department of Applied Biological Science, Tokyo Noko University, Fuchu, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
114
|
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
|
115
|
Oni-Orisan A, Cresci S, Jones PG, Theken KN, Spertus JA, Lee CR. Association between the EPHX2 p.Lys55Arg polymorphism and prognosis following an acute coronary syndrome. Prostaglandins Other Lipid Mediat 2018; 138:15-22. [PMID: 30096423 PMCID: PMC6162147 DOI: 10.1016/j.prostaglandins.2018.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/06/2018] [Accepted: 07/31/2018] [Indexed: 01/14/2023]
Abstract
Inhibition of soluble epoxide hydrolase (sEH, EPHX2) elicits potent cardiovascular protective effects in preclinical models of ischemic cardiovascular disease (CVD), and genetic polymorphisms in EPHX2 have been associated with developing ischemic CVD in humans. However, it remains unknown whether EPHX2 variants are associated with prognosis following an ischemic CVD event. We evaluated the association between EPHX2 p.Lys55Arg and p.Arg287Gln genotype with survival in 667 acute coronary syndrome (ACS) patients. No association with p.Arg287Gln genotype was observed (P = 0.598). Caucasian EPHX2 Arg55 carriers (Lys/Arg or Arg/Arg) had a significantly higher risk of 5-year mortality (adjusted hazard ratio [HR] 1.61, 95% confidence interval [CI] 1.01-2.55, P = 0.045). In an independent population of 2712 ACS patients, this association was not replicated (adjusted HR 0.92, 95% CI 0.70-1.21, P = 0.559). In a secondary analysis, Caucasian homozygous Arg55 allele carriers (Arg/Arg) appeared to exhibit a higher risk of cardiovascular mortality (adjusted HR 2.60, 95% CI 1.09-6.17). These results demonstrate that EPHX2 p.Lys55Arg and p.Arg287Gln polymorphisms do not significantly modify survival after an ACS event. Investigation of other sEH metabolism biomarkers in ischemic CVD appears warranted.
Collapse
Affiliation(s)
- Akinyemi Oni-Orisan
- Department of Clinical Pharmacy, UCSF School of Pharmacy, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Sharon Cresci
- Department of Medicine and Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip G Jones
- Department of Cardiovascular Research, Saint Luke's Mid America Heart Institute, Kansas City, MO, USA
| | - Katherine N Theken
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John A Spertus
- Department of Cardiovascular Research, Saint Luke's Mid America Heart Institute, Kansas City, MO, USA; University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
116
|
Neuroprotective effects of epoxyeicosatrienoic acids. Prostaglandins Other Lipid Mediat 2018; 138:9-14. [DOI: 10.1016/j.prostaglandins.2018.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 06/19/2018] [Accepted: 07/17/2018] [Indexed: 11/22/2022]
|
117
|
Allaqaband H, Gutterman DD, Kadlec AO. Physiological Consequences of Coronary Arteriolar Dysfunction and Its Influence on Cardiovascular Disease. Physiology (Bethesda) 2018; 33:338-347. [PMID: 30109826 PMCID: PMC6230549 DOI: 10.1152/physiol.00019.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
To date, the major focus of diagnostic modalities and interventions to treat coronary artery disease has been the large epicardial vessels. Despite substantial data showing that microcirculatory dysfunction is a strong predictor of future adverse cardiovascular events, very little research has gone into developing techniques for in vivo diagnosis and therapeutic interventions to improve microcirculatory function. In this review, we will discuss the pathophysiology of coronary arteriolar dysfunction, define its prognostic implications, evaluate the diagnostic modalities available, and provide speculation on current and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Hassan Allaqaband
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - David D Gutterman
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Medicine, Division of Cardiology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Veterans Administration Medical Center, Milwaukee, Wisconsin
| | - Andrew O Kadlec
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Medicine, Division of Cardiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
118
|
Gai Z, Visentin M, Gui T, Zhao L, Thasler WE, Häusler S, Hartling I, Cremonesi A, Hiller C, Kullak-Ublick GA. Effects of Farnesoid X Receptor Activation on Arachidonic Acid Metabolism, NF-kB Signaling, and Hepatic Inflammation. Mol Pharmacol 2018; 94:802-811. [PMID: 29743187 DOI: 10.1124/mol.117.111047] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/07/2018] [Indexed: 01/01/2023] Open
Abstract
Inflammation has a recognized role in nonalcoholic fatty liver disease (NAFLD) progression. In the present work, we studied the effect of high-fat diet (HFD) on arachidonic acid metabolism in the liver and investigated the role of the farnesoid X receptor (FXR, NR1H4) in eicosanoid biosynthetic pathways and nuclear factor κ light-chain enhancer of activated B cells (NF-kB) signaling, major modulators of the inflammatory cascade. Mice were fed an HFD to induce NAFLD and then treated with the FXR ligand obeticholic acid (OCA). Histology and gene expression analyses were performed on liver tissue. Eicosanoid levels were measured from serum and urine samples. The molecular mechanism underlying the effect of FXR activation on arachidonic acid metabolism and NF-kB signaling was studied in human liver Huh7 cells and primary cultured hepatocytes. NAFLD was characterized by higher (∼25%) proinflammatory [leukotrienes (LTB4)] and lower (∼3-fold) anti-inflammatory [epoxyeicosatrienoic acids (EETs)] eicosanoid levels than in chow mice. OCA induced the expression of several hepatic cytochrome P450 (P450) epoxygenases, the enzymes responsible for EET synthesis, and mitigated HFD-induced hepatic injury. In vitro, induction of CYP450 epoxygenases was sufficient to inhibit NF-kB signaling and cell migration. The CYP450 epoxygenase pan-inhibitor gemfibrozil fully abolished the protective effect of OCA, indicating that OCA-mediated inhibition of NF-kB signaling was EET-dependent. In summary, NAFLD was characterized by an imbalance in arachidonate metabolism. FXR activation reprogramed arachidonate metabolism by inducing P450 epoxygenase expression and EET production. In vitro, FXR-mediated NF-kB inhibition required active P450 epoxygenases.
Collapse
Affiliation(s)
- Zhibo Gai
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Michele Visentin
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Ting Gui
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Lin Zhao
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Wolfgang E Thasler
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Stephanie Häusler
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Ivan Hartling
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Alessio Cremonesi
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Christian Hiller
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (Z.G., M.V., S.H., C.H., G.A.K.-U.); Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (T.G.); Department of Endocrinology, Chinese PLA 309 Hospital, Peking, China (L.Z.); Department of General and Visceral Surgery, Rotkreuzklinikum Munich, Munich, Germany (W.E.T.); Department of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland (I.H., A.C.); and Mechanistic Safety, Novartis Global Drug Development, Basel, Switzerland (G.A.K.-U.)
| |
Collapse
|
119
|
Targeted metabolomic approach in men with carotid plaque. PLoS One 2018; 13:e0200547. [PMID: 30011297 PMCID: PMC6047792 DOI: 10.1371/journal.pone.0200547] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/28/2018] [Indexed: 12/22/2022] Open
Abstract
Background The aim of the study was to analyse the presence of several metabolites related to atherosclerosis in the plasma of patients with unstable carotid plaque and in the plasma of healthy subjects. Materials and methods We included 20 patients who had undergone carotid endarterectomy and 20 healthy subjects as a control group. All the subjects recruited were male. We used a metabolomic approach with liquid chromatography coupled to mass spectrometry to evaluate plasma metabolite levels in the metabolic pathway involved in the progression of atherosclerotic plaque. Results We observed that circulating levels of 20-HETE were significantly higher in patients with atheroma plaque than in healthy subjects (p = 0.018). No differences were found with regard to the other metabolites analysed. We also conducted a random forest analysis and found that 20-HETE was the main differentiator in the list of selected metabolites. In addition, plasma levels of 20-HETE correlated positively with body mass index (r = 0.427, p = 0.007) and diastolic blood pressure (r = 0.365, p = 0.028). Conclusion This study confirms that of all the molecules studied only 20-HETE is related to carotid plaque. Further studies are needed to compare patients with stable carotid plaque vs. patients with unstable carotid plaque in order to confirm that 20-HETE could be a potential factor related to carotid plaque.
Collapse
|
120
|
Valdes AM, Ravipati S, Pousinis P, Menni C, Mangino M, Abhishek A, Chapman V, Barrett DA, Doherty M. Omega-6 oxylipins generated by soluble epoxide hydrolase are associated with knee osteoarthritis. J Lipid Res 2018; 59:1763-1770. [PMID: 29986999 PMCID: PMC6121933 DOI: 10.1194/jlr.p085118] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 07/02/2018] [Indexed: 11/30/2022] Open
Abstract
Omega-6 FAs are inflammatory mediators that are increased in joints with osteoarthritis (OA), but their association with OA progression is not yet well defined. To investigate the relationship between omega-6 FAs and knee OA, we measured with LC-MS the levels of 22 omega-6 lipids (arachidonic acid, linoleic acid, and 20 oxylipins) in synovial fluid (SF) from 112 knees of 102 individuals (58 with knee OA; 44 controls). We hypothesized that oxylipin metabolites would increase in OA knee SF and with radiographically progressive disease. We validated results by comparing samples from affected and unaffected knees in 10 individuals with unilateral OA. In adjusted analysis, SF levels of three omega-6 oxylipins [prostaglandin D2, 11,12-dihydroxyeicosatrienoic acid (DHET), and 14,15-DHET] were associated with OA. Of these, 11,12-DHET and 14,15-DHET were higher in affected versus unaffected knees of people with unilateral disease (P < 0.014 and P < 0.003, respectively). Levels of these and 8,9-DHET were also associated with radiographic progression over 3.3 years in 87 individuals. Circulating levels of all three were associated with gene variants at the soluble epoxide hydrolase enzyme. Lipidomic profiling in SF identified an additional inflammatory pathway associated with knee OA and radiographic progression.
Collapse
Affiliation(s)
- Ana M Valdes
- Academic Rheumatology, Nottingham City Hospital, Nottingham, United Kingdom; National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom; Department of Twin Research, King's College London, St Thomas' Hospital, London, United Kingdom; Arthritis Research UK Centre of Excellence for Pain, University of Nottingham, Nottingham, United Kingdom.
| | - Srinivasarao Ravipati
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Petros Pousinis
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Cristina Menni
- Department of Twin Research, King's College London, St Thomas' Hospital, London, United Kingdom
| | - Massimo Mangino
- Department of Twin Research, King's College London, St Thomas' Hospital, London, United Kingdom
| | - Abhishek Abhishek
- Academic Rheumatology, Nottingham City Hospital, Nottingham, United Kingdom
| | - Victoria Chapman
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom; Arthritis Research UK Centre of Excellence for Pain, University of Nottingham, Nottingham, United Kingdom
| | - David A Barrett
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Michael Doherty
- Academic Rheumatology, Nottingham City Hospital, Nottingham, United Kingdom; National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom; Arthritis Research UK Centre of Excellence for Pain, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
121
|
Abstract
Globally, cardiovascular diseases (CVDs) are the number one cause of mortality. Approximately 18 million people died from CVDs in 2015, representing more than 30% of all global deaths. New diagnostic tools and therapies are eagerly required to decrease the prevalence of CVDs related to mortality and/or risk factors leading to CVDs. Oxylipins are a group of metabolites, generated via oxygenation of polyunsaturated fatty acids that are involved in inflammation, immunity, and vascular functions, etc. Thus far, over 100 oxylipins have been identified, and have overlapping and interconnected roles. Important CVD pathologies such as hyperlipidemia, hypertension, thrombosis, hemostasis and diabetes have been linked to abnormal oxylipin signaling. Oxylipins represent a new era of risk markers and/or therapeutic targets in several diseases including CVDs. The role of many oxylipins in the progression or regression in CVD, however, is still not fully understood. An increased knowledge of the role of these oxygenated polyunsaturated fatty acids in cardiovascular dysfunctions or CVDs including hypertension could possibly lead to the development of biomarkers for the detection and their treatment in the future.
Collapse
|
122
|
Su XD, Guo RH, Yang SY, Kim YH, Kim YR. Anti-bacterial effects of components from Sanguisorba officinalis L. on Vibrio vulnificus and their soluble epoxide hydrolase inhibitory activity. Nat Prod Res 2018; 33:3445-3449. [PMID: 29790361 DOI: 10.1080/14786419.2018.1478825] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sanguisorba officinalis L. is a traditional herbal medicine, which is prevailingly applied to cure hemorrhoids, wounds and ulcers in Eastern Asian countries. The purpose of this study was to investigate the antibacterial and soluble epoxide hydrolase (sEH) inhibitory effects of the extracts and components from S. officinalis. The methanol extract was divided into ethyl acetate (EtOAc), n-butanol (n-BuOH), and water layers. In our screening procedure, the EtOAc and n-BuOH extracts and compounds (1-2) remarkably suppressed the growth of V. vulnificus in a dose-dependent manner. In addition, the EtOAc extract and compound 1 exhibited significant inhibitory effect on the V. vulnificus induced cytotoxicity on HeLa cells. Furthermore, compound 4 displayed an inhibition against sEH with an IC50 value of 7.0 ± 0.5 μM. A kinetic analysis demonstrated that the inhibitory effect of compound 4 was a mixed type, with an inhibitory constant (Ki) 0.22 ± 0.0 μM.
Collapse
Affiliation(s)
- Xiang Dong Su
- College of Pharmacy, Chungnam National University , Daejeon , Republic of Korea
| | - Rui Hong Guo
- College of Pharmacy, Chungnam National University , Daejeon , Republic of Korea
| | - Seo Young Yang
- College of Pharmacy, Chungnam National University , Daejeon , Republic of Korea
| | - Young Ho Kim
- College of Pharmacy, Chungnam National University , Daejeon , Republic of Korea
| | - Young Ran Kim
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University , Gwangju , Republic of Korea
| |
Collapse
|
123
|
[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: 3] [Impact Index Per Article: 0.4] [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
|
124
|
Wang Q, Liang Y, Qiao Y, Zhao X, Yang Y, Yang S, Li B, Zhao Q, Dong L, Quan S, Tian R, Liu Z. Expression of soluble epoxide hydrolase in renal tubular epithelial cells regulates macrophage infiltration and polarization in IgA nephropathy. Am J Physiol Renal Physiol 2018; 315:F915-F926. [PMID: 29717935 DOI: 10.1152/ajprenal.00534.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tubulointerstitial inflammatory cell infiltration and activation contribute to kidney inflammation and fibrosis. Epoxyeicosatrienoic acids (EETs), which are rapidly metabolized to dihydroxyeicosatrienoic acids by the soluble epoxide hydrolase (sEH), have multiple biological functions, including vasodilation, anti-inflammatory action, and others. Inhibition of sEH has been demonstrated to attenuate inflammation in many renal disease models. However, the relationship between sEH expression and macrophage polarization in the kidney remains unknown. In this study, we investigated the relationships between the level of sEH and clinical and pathological parameters in IgA nephropathy. The level of sEH expression positively correlated with proteinuria and infiltration of macrophages. sEH-positive tubules were found to be surrounded by macrophages. Furthermore, we found that incubation of immortalized human proximal tubular HK-2 cells with total urinary protein and overexpression of sEH promoted inflammatory factor production, which was associated with M1 polarization. We also exposed RAW264.7 mouse leukemic monocytes/macrophages to different HK-2 cell culture media conditioned by incubation with various substances affecting sEH amount or activity. We found that the upregulation of sEH promoted M1 polarization. However, pharmacological inhibition of sEH and supplementation with EETs reversed the conditioning effects of urinary proteins by inhibiting M1 polarization through the NF-κB pathway and stimulating M2 polarization through the phosphatidylinositol 3-kinase pathway. These data suggest that inhibition of sEH could be a new strategy to prevent the progression of inflammation and to attenuate renal tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Qian Wang
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Nephrology, Zhengzhou University , Zhengzhou , China
| | - Yan Liang
- Institute of Nephrology, Zhengzhou University , Zhengzhou , China.,Department of Nephrology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Yingjin Qiao
- Institute of Nephrology, Zhengzhou University , Zhengzhou , China.,Blood Purification Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Xiangya Zhao
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Yang
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shengnan Yang
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Li
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qianru Zhao
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ling Dong
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songxia Quan
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Rui Tian
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhangsuo Liu
- Institute of Nephrology, Zhengzhou University , Zhengzhou , China.,Department of Nephrology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
125
|
Das UN. Arachidonic acid in health and disease with focus on hypertension and diabetes mellitus: A review. J Adv Res 2018; 11:43-55. [PMID: 30034875 PMCID: PMC6052660 DOI: 10.1016/j.jare.2018.01.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/01/2018] [Accepted: 01/02/2018] [Indexed: 02/06/2023] Open
Abstract
Arachidonic acid (AA 20:4n-6) is an essential component of cell membranes and modulates cell membrane fluidity. AA is metabolized by cyclo-oxygenase (COX), lipoxygenase (LOX) and cytochrome P450 enzymes to form several metabolites that have important biological actions. Of all the actions, role of AA in the regulation of blood pressure and its ability to prevent both type 1 and type 2 diabetes mellitus seems to be interesting. Studies showed that AA and its metabolites especially, lipoxin A4 (LXA4) and epoxyeicosatrienoic acids (EETs), potent anti-inflammatory metabolites, have a crucial role in the pathobiology of hypertension and diabetes mellitus. AA, LXA4 and EETs regulate smooth muscle function and proliferation, voltage gated ion channels, cell membrane fluidity, membrane receptors, G-coupled receptors, PPARs, free radical generation, nitric oxide formation, inflammation, and immune responses that, in turn, participate in the regulation blood pressure and pathogenesis of diabetes mellitus. In this review, role of AA and its metabolites LXA4 and EETs in the pathobiology of hypertension, pre-eclampsia and diabetes mellitus are discussed. Based on several lines of evidences, it is proposed that a combination of aspirin and AA could be of benefit in the prevention and management of hypertension, pre-eclampsia and diabetes mellitus.
Collapse
|
126
|
Soler A, Hunter I, Joseph G, Hutcheson R, Hutcheson B, Yang J, Zhang FF, Joshi SR, Bradford C, Gotlinger KH, Maniyar R, Falck JR, Proctor S, Schwartzman ML, Gupte SA, Rocic P. Elevated 20-HETE in metabolic syndrome regulates arterial stiffness and systolic hypertension via MMP12 activation. J Mol Cell Cardiol 2018; 117:88-99. [PMID: 29428638 PMCID: PMC5877315 DOI: 10.1016/j.yjmcc.2018.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/08/2018] [Accepted: 02/07/2018] [Indexed: 11/24/2022]
Abstract
Arterial stiffness plays a causal role in development of systolic hypertension. 20-hydroxyeicosatetraeonic acid (20-HETE), a cytochrome P450 (CYP450)-derived arachidonic acid metabolite, is known to be elevated in resistance arteries in hypertensive animal models and loosely associated with obesity in humans. However, the role of 20-HETE in the regulation of large artery remodeling in metabolic syndrome has not been investigated. We hypothesized that elevated 20-HETE in metabolic syndrome increases matrix metalloproteinase 12 (MMP12) activation leading to increased degradation of elastin, increased large artery stiffness and increased systolic blood pressure. 20-HETE production was increased ~7 fold in large, conduit arteries of metabolic syndrome (JCR:LA-cp, JCR) vs. normal Sprague-Dawley (SD) rats. This correlated with increased elastin degradation (~7 fold) and decreased arterial compliance (~75% JCR vs. SD). 20-HETE antagonists blocked elastin degradation in JCR rats concomitant with blocking MMP12 activation. 20-HETE antagonists normalized, and MMP12 inhibition (pharmacological and MMP12-shRNA-Lnv) significantly improved (~50% vs. untreated JCR) large artery compliance in JCR rats. 20-HETE antagonists also decreased systolic (182 ± 3 mmHg JCR, 145 ± 3 mmHg JCR + 20-HETE antagonists) but not diastolic blood pressure in JCR rats. Whereas diastolic pressure was fully angiotensin II (Ang II)-dependent, systolic pressure was only partially Ang II-dependent, and large artery stiffness was Ang II-independent. Thus, 20-HETE-dependent regulation of systolic blood pressure may be a unique feature of metabolic syndrome related to high 20-HETE production in large, conduit arteries, which results in increased large artery stiffness and systolic blood pressure. These findings may have implications for management of systolic hypertension in patients with metabolic syndrome.
Collapse
Affiliation(s)
- Amanda Soler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Ian Hunter
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Gregory Joseph
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Rebecca Hutcheson
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Brenda Hutcheson
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Jenny Yang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Frank Fan Zhang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Sachindra Raj Joshi
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Chastity Bradford
- Department of Biology, Tuskegee University, Tuskegee, AL 36088, United States
| | - Katherine H Gotlinger
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Rachana Maniyar
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - John R Falck
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Spencer Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Alberta Institute for Human Nutrition, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | | | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Petra Rocic
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States.
| |
Collapse
|
127
|
Abstract
A decade of research has established the phospholipase iPLA2γ as being involved in cardiomyocyte dysfunction and necrosis leading to heart failure, but the mechanisms by which iPLA2γ acts and its interaction with the mitochondrial permeability transition pore (mPTP) that is critical for cardiac homeostasis are unclear. New investigations by Moon et al. demonstrate that mitochondria in failing hearts undergo dynamic shifts in PLA2 isoform expression, leading to a redistribution of eicosanoid composition that contributes to pathologic mPTP opening.
Collapse
Affiliation(s)
- Matthew J Wolf
- Division of Cardiovascular Medicine, Department of Medicine, The University of Virginia School of Medicine, Charlottesville, Virginia 22908.
| |
Collapse
|
128
|
Falero-Perez J, Song YS, Sorenson CM, Sheibani N. CYP1B1: A key regulator of redox homeostasis. TRENDS IN CELL & MOLECULAR BIOLOGY 2018; 13:27-45. [PMID: 30894785 PMCID: PMC6422340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
CYP1B1 is a member of the CYP1 subfamily of CYP superfamily of enzymes, which contains three members, CYP1A1, CYP1A2, and CYP1B1. CYP1B1 is expressed in both adult and fetal human extrahepatic tissues, including the parenchymal and stromal cells of most organs. Mutations in the CYP1B1 gene are linked to the development of primary congenital glaucoma in humans. However, the underlying mechanisms remain unknown. Using Cyp1b1-deficient mice, we showed that CYP1B1 is constitutively expressed in retinal vascular cells with a significant role in retinal neovascularization during oxygen-induced ischemic retinopathy. We also showed CYP1B1 is constitutively expressed in trabecular meshwork (TM) cells and its expression plays a significant role in the normal development and function of the TM tissue. We have observed that germline deletion of Cyp1b1 is associated with increased oxidative stress in the retinal vascular and TM cells in culture, and retinal and TM tissue in vivo. We showed increased oxidative stress was responsible for altered production of the extracellular matrix proteins and had a significant impact on cellular integrity and function of these tissues. Collectively, our studies have established an important role for CYP1B1 expression in modulation of tissue integrity and function through the regulation of cellular redox homeostasis and extracellular microenvironment.
Collapse
Affiliation(s)
- Juliana Falero-Perez
- Departments of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine, Madison, WI USA
| | - Yong-Seok Song
- Departments of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine, Madison, WI USA
| | - Christine M. Sorenson
- Departments of Pediatrics, University of Wisconsin School of Medicine, Madison, WI USA
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine, Madison, WI USA
- Departments of Cell and Regenerative Biology, University of Wisconsin School of Medicine, Madison, WI USA
- Departments of Biomedical Engineering, University of Wisconsin School of Medicine, Madison, WI USA
| |
Collapse
|
129
|
Kharkwal H, Batool F, Koentgen F, Bell DR, Kendall DA, Ebling FJP, Duce IR. Generation and phenotypic characterisation of a cytochrome P450 4x1 knockout mouse. PLoS One 2017; 12:e0187959. [PMID: 29227996 PMCID: PMC5724839 DOI: 10.1371/journal.pone.0187959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/14/2017] [Indexed: 11/18/2022] Open
Abstract
Cytochrome P450 4x1 (Cyp4x1) is expressed at very high levels in the brain but the function of this protein is unknown. It has been hypothesised to regulate metabolism of fatty acids and to affect the activity of endocannabinoid signalling systems, which are known to influence appetite and energy metabolism. The objective of the present investigation was to determine the impact of Cyp4x1 on body weight and energy metabolism by developing a line of transgenic Cyp4x1-knock out mice. Mice were developed with a global knock-out of the gene; the full-length RNA was undetectable, and mice were viable and fertile. Both male and female Cyp4x1-knock out mice gained significantly more body weight on normal lab chow diet compared to control flox mice on the same genetic background. At necropsy, Cyp4x1-knock out male mice had significantly greater intra-abdominal fat deposits (P<0.01), and enlarged adipocytes. Metabolic rate and locomotor activity as inferred from VO2 measures and crossing of infrared beams in metabolic cages were not significantly affected by the mutation in either gender. The respiratory exchange ratio was significantly decreased in male knock out mice (P<0.05), suggesting a greater degree of fat oxidation, consistent with their higher adiposity. When mice were maintained on a high fat diet, VO2 was significantly decreased in both male and female Cyp4x1-knock out mice. We conclude that the Cyp4x1-knock out mouse strain demonstrates a mildly obese phenotype, consistent with the view that cytochrome P450 4x1 plays a role in regulating fat metabolism.
Collapse
Affiliation(s)
- Himanshu Kharkwal
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Farhat Batool
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Department of Biochemistry, University of Karachi, Karachi, Pakistan
| | - Frank Koentgen
- Ozgene Pty Ltd., Bentley DC, Western Australia, Australia
| | - David R. Bell
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- European Chemicals Agency, Helsinki, Finland
| | - David A. Kendall
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Ian R. Duce
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- * E-mail:
| |
Collapse
|
130
|
Effects of dronedarone, amiodarone and their active metabolites on sequential metabolism of arachidonic acid to epoxyeicosatrienoic and dihydroxyeicosatrienoic acids. Biochem Pharmacol 2017; 146:188-198. [DOI: 10.1016/j.bcp.2017.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/22/2017] [Indexed: 12/23/2022]
|
131
|
Zárate R, el Jaber-Vazdekis N, Tejera N, Pérez JA, Rodríguez C. Significance of long chain polyunsaturated fatty acids in human health. Clin Transl Med 2017; 6:25. [PMID: 28752333 PMCID: PMC5532176 DOI: 10.1186/s40169-017-0153-6] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
In the last decades, the development of new technologies applied to lipidomics has revitalized the analysis of lipid profile alterations and the understanding of the underlying molecular mechanisms of lipid metabolism, together with their involvement in the occurrence of human disease. Of particular interest is the study of omega-3 and omega-6 long chain polyunsaturated fatty acids (LC-PUFAs), notably EPA (eicosapentaenoic acid, 20:5n-3), DHA (docosahexaenoic acid, 22:6n-3), and ARA (arachidonic acid, 20:4n-6), and their transformation into bioactive lipid mediators. In this sense, new families of PUFA-derived lipid mediators, including resolvins derived from EPA and DHA, and protectins and maresins derived from DHA, are being increasingly investigated because of their active role in the "return to homeostasis" process and resolution of inflammation. Recent findings reviewed in the present study highlight that the omega-6 fatty acid ARA appears increased, and omega-3 EPA and DHA decreased in most cancer tissues compared to normal ones, and that increments in omega-3 LC-PUFAs consumption and an omega-6/omega-3 ratio of 2-4:1, are associated with a reduced risk of breast, prostate, colon and renal cancers. Along with their lipid-lowering properties, omega-3 LC-PUFAs also exert cardioprotective functions, such as reducing platelet aggregation and inflammation, and controlling the presence of DHA in our body, especially in our liver and brain, which is crucial for optimal brain functionality. Considering that DHA is the principal omega-3 FA in cortical gray matter, the importance of DHA intake and its derived lipid mediators have been recently reported in patients with major depressive and bipolar disorders, Alzheimer disease, Parkinson's disease, and amyotrophic lateral sclerosis. The present study reviews the relationships between major diseases occurring today in the Western world and LC-PUFAs. More specifically this review focuses on the dietary omega-3 LC-PUFAs and the omega-6/omega-3 balance, in a wide range of inflammation disorders, including autoimmune diseases. This review suggests that the current recommendations of consumption and/or supplementation of omega-3 FAs are specific to particular groups of age and physiological status, and still need more fine tuning for overall human health and well being.
Collapse
Affiliation(s)
- Rafael Zárate
- Canary Islands Cancer Research Institute (ICIC), Ave. La Trinidad 61, Torre A. Arévalo, 7th floor, 38204 La Laguna, Tenerife Spain
| | - Nabil el Jaber-Vazdekis
- Centre Algatech, Institute of Microbiology, Academy of Sciences of the Czech Republic, Třeboň, Czech Republic
| | - Noemi Tejera
- Department of Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia, Norwich, NR4 7UQ UK
| | - José A. Pérez
- Department of Animal Biology, Soil Science and Geology (Animal Physiology Unit), Faculty of Sciences, Universidad de La Laguna, Ave. Astrofísico Francisco Sánchez s/n, 38206 La Laguna, Tenerife Spain
| | - Covadonga Rodríguez
- Department of Animal Biology, Soil Science and Geology (Animal Physiology Unit), Faculty of Sciences, Universidad de La Laguna, Ave. Astrofísico Francisco Sánchez s/n, 38206 La Laguna, Tenerife Spain
- Institute of Biomedical Technologies (ITB), Universidad de La Laguna, Campus de Ofra, 38071 La Laguna, Tenerife Spain
| |
Collapse
|
132
|
Kingma JG, Simard D, Rouleau JR, Drolet B, Simard C. The Physiopathology of Cardiorenal Syndrome: A Review of the Potential Contributions of Inflammation. J Cardiovasc Dev Dis 2017; 4:E21. [PMID: 29367550 PMCID: PMC5753122 DOI: 10.3390/jcdd4040021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/25/2017] [Accepted: 11/26/2017] [Indexed: 12/12/2022] Open
Abstract
Inter-organ crosstalk plays an essential role in the physiological homeostasis of the heart and other organs, and requires a complex interaction between a host of cellular, molecular, and neural factors. Derangements in these interactions can initiate multi-organ dysfunction. This is the case, for instance, in the heart or kidneys where a pathological alteration in one organ can unfavorably affect function in another distant organ; attention is currently being paid to understanding the physiopathological consequences of kidney dysfunction on cardiac performance that lead to cardiorenal syndrome. Different cardiorenal connectors (renin-angiotensin or sympathetic nervous system activation, inflammation, uremia, etc.) and non-traditional risk factors potentially contribute to multi-organ failure. Of these, inflammation may be crucial as inflammatory cells contribute to over-production of eicosanoids and lipid second messengers that activate intracellular signaling pathways involved in pathogenesis. Indeed, inflammation biomarkers are often elevated in patients with cardiac or renal dysfunction. Epigenetics, a dynamic process that regulates gene expression and function, is also recognized as an important player in single-organ disease. Principal epigenetic modifications occur at the level of DNA (i.e., methylation) and histone proteins; aberrant DNA methylation is associated with pathogenesis of organ dysfunction through a number of mechanisms (inflammation, nitric oxide bioavailability, endothelin, etc.). Herein, we focus on the potential contribution of inflammation in pathogenesis of cardiorenal syndrome.
Collapse
Affiliation(s)
- John G Kingma
- Department of Medicine, Faculty of Medicine, Pavillon Ferdinand Vandry, 1050, Avenue de la Médecine, Université Laval, Quebec, QC G1V 0A6, Canada.
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, 2725, Chemin Sainte-Foy, Quebec, QC G1V 4G5, Canada.
| | - Denys Simard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, 2725, Chemin Sainte-Foy, Quebec, QC G1V 4G5, Canada.
| | - Jacques R Rouleau
- Department of Medicine, Faculty of Medicine, Pavillon Ferdinand Vandry, 1050, Avenue de la Médecine, Université Laval, Quebec, QC G1V 0A6, Canada.
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, 2725, Chemin Sainte-Foy, Quebec, QC G1V 4G5, Canada.
| | - Benoit Drolet
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, 2725, Chemin Sainte-Foy, Quebec, QC G1V 4G5, Canada.
- Faculty of Pharmacy, Pavillon Ferdinand Vandry, 1050, Avenue de la Médecine, Université Laval, Quebec, QC G1V 0A6, Canada.
| | - Chantale Simard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, 2725, Chemin Sainte-Foy, Quebec, QC G1V 4G5, Canada.
- Faculty of Pharmacy, Pavillon Ferdinand Vandry, 1050, Avenue de la Médecine, Université Laval, Quebec, QC G1V 0A6, Canada.
| |
Collapse
|
133
|
Moon SH, Liu X, Cedars AM, Yang K, Kiebish MA, Joseph SM, Kelley J, Jenkins CM, Gross RW. Heart failure-induced activation of phospholipase iPLA 2γ generates hydroxyeicosatetraenoic acids opening the mitochondrial permeability transition pore. J Biol Chem 2017; 293:115-129. [PMID: 29158256 DOI: 10.1074/jbc.ra117.000405] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/08/2017] [Indexed: 11/06/2022] Open
Abstract
Congestive heart failure typically arises from cardiac myocyte necrosis/apoptosis, associated with the pathological opening of the mitochondrial permeability transition pore (mPTP). mPTP opening decreases the mitochondrial membrane potential leading to the activation of Ca2+-independent phospholipase A2γ (iPLA2γ) and the production of downstream toxic metabolites. However, the array of enzymatic mediators and the exact chemical mechanisms responsible for modulating myocardial mPTP opening remain unclear. Herein, we demonstrate that human heart failure activates specific myocardial mitochondrial phospholipases that increase Ca2+-dependent production of toxic hydroxyeicosatetraenoic acids (HETEs) and attenuate the activity of phospholipases that promote the synthesis of protective epoxyeicosatrienoic acids (EETs). Mechanistically, HETEs activated the Ca2+-induced opening of the mPTP in failing human myocardium, and the highly selective pharmacological blockade of either iPLA2γ or lipoxygenases attenuated mPTP opening in failing hearts. In contrast, pharmacological inhibition of cytochrome P450 epoxygenases opened the myocardial mPTP in human heart mitochondria. Remarkably, the major mitochondrial phospholipase responsible for Ca2+-activated release of arachidonic acid (AA) in mitochondria from non-failing hearts was calcium-dependent phospholipase A2ζ (cPLA2ζ) identified by sequential column chromatographies and activity-based protein profiling. In contrast, iPLA2γ predominated in failing human myocardium. Stable isotope kinetics revealed that in non-failing human hearts, cPLA2ζ metabolically channels arachidonic acid into EETs, whereas in failing hearts, increased iPLA2γ activity channels AA into toxic HETEs. These results mechanistically identify the sequelae of pathological remodeling of human mitochondrial phospholipases in failing myocardium. This remodeling metabolically channels AA into toxic HETEs promoting mPTP opening, which induces necrosis/apoptosis leading to further progression of heart failure.
Collapse
Affiliation(s)
- Sung Ho Moon
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xinping Liu
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Ari M Cedars
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kui Yang
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Michael A Kiebish
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Susan M Joseph
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - John Kelley
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Christopher M Jenkins
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Richard W Gross
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110; Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Chemistry, Washington University, St. Louis, Missouri 63130.
| |
Collapse
|
134
|
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]
|
135
|
Abstract
Cytochrome P450 eicosanoids play important roles in brain function and disease through their complementary actions on cell-cell communications within the neurovascular unit (NVU) and mechanisms of brain injury. Epoxy- and hydroxyeicosanoids, respectively formed by cytochrome P450 epoxygenases and ω-hydroxylases, play opposing roles in cerebrovascular function and in pathological processes underlying neural injury, including ischemia, neuroinflammation and oxidative injury. P450 eicosanoids also contribute to cerebrovascular disease risk factors, including hypertension and diabetes. We summarize studies investigating the roles P450 eicosanoids in cerebrovascular physiology and disease to highlight the existing balance between these important lipid signaling molecules, as well as their roles in maintaining neurovascular homeostasis and in acute and chronic neurovascular and neurodegenerative disorders.
Collapse
Affiliation(s)
- Catherine M Davis
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239, United States; The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, United States
| | - Xuehong Liu
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, United States
| | - Nabil J Alkayed
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239, United States; The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|
136
|
Hammoud SH, Omar AG, Eid AA, El-Mas MM. CYP4A/CYP2C modulation of the interaction of calcium channel blockers with cyclosporine on EDHF-mediated renal vasodilations in rats. Toxicol Appl Pharmacol 2017; 334:110-119. [DOI: 10.1016/j.taap.2017.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/24/2017] [Accepted: 09/08/2017] [Indexed: 01/01/2023]
|
137
|
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
|
138
|
Santos JM, Park JA, Joiakim A, Putt DA, Taylor RN, Kim H. The role of soluble epoxide hydrolase in preeclampsia. Med Hypotheses 2017; 108:81-85. [DOI: 10.1016/j.mehy.2017.07.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/07/2017] [Accepted: 07/28/2017] [Indexed: 01/31/2023]
|
139
|
Abstract
Biologically active epoxyeicosatrienoic acid (EET) regioisomers are synthesized from arachidonic acid by cytochrome P450 epoxygenases of endothelial, myocardial, and renal tubular cells. EETs relax vascular smooth muscle and decrease inflammatory cell adhesion and cytokine release. Renal EETs promote sodium excretion and vasodilation to decrease hypertension. Cardiac EETs reduce infarct size after ischemia-reperfusion injury and decrease fibrosis and inflammation in heart failure. In diabetes, EETs improve insulin sensitivity, increase glucose tolerance, and reduce the renal injury. These actions of EETs emphasize their therapeutic potential. To minimize metabolic inactivation, 14,15-EET agonist analogs with stable epoxide bioisosteres and carboxyl surrogates were developed. In preclinical rat models, a subset of agonist analogs, termed EET-A, EET-B, and EET-C22, are orally active with good pharmacokinetic properties. These orally active EET agonists lower blood pressure and reduce cardiac and renal injury in spontaneous and angiotensin hypertension. Other beneficial cardiovascular actions include improved endothelial function and cardiac antiremodeling actions. In rats, EET analogs effectively combat acute and chronic kidney disease including drug- and radiation-induced kidney damage, hypertension and cardiorenal syndrome kidney damage, and metabolic syndrome and diabetes nephropathy. The compelling preclinical efficacy supports the prospect of advancing EET analogs to human clinical trials for kidney and cardiovascular diseases.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/administration & dosage
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/chemistry
- Administration, Oral
- Animals
- Blood Pressure/drug effects
- Blood Pressure/physiology
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/physiopathology
- Fatty Acids, Monounsaturated/administration & dosage
- Fatty Acids, Monounsaturated/chemistry
- Humans
- Hypertension/drug therapy
- Hypertension/physiopathology
- Kidney Diseases/drug therapy
- Kidney Diseases/physiopathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Structure-Activity Relationship
- Vasodilation/drug effects
- Vasodilation/physiology
Collapse
Affiliation(s)
- William B Campbell
- *Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI; and †Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | | | | | | |
Collapse
|
140
|
Schmidt J, Rotter M, Weiser T, Wittmann S, Weizel L, Kaiser A, Heering J, Goebel T, Angioni C, Wurglics M, Paulke A, Geisslinger G, Kahnt A, Steinhilber D, Proschak E, Merk D. A Dual Modulator of Farnesoid X Receptor and Soluble Epoxide Hydrolase To Counter Nonalcoholic Steatohepatitis. J Med Chem 2017; 60:7703-7724. [DOI: 10.1021/acs.jmedchem.7b00398] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Jurema Schmidt
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Marco Rotter
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Tim Weiser
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Sandra Wittmann
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Lilia Weizel
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Astrid Kaiser
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Jan Heering
- Project
Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60596 Frankfurt am Main, Germany
| | - Tamara Goebel
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Carlo Angioni
- Institute
of Clinical Pharmacology, Goethe University Frankfurt, Theodor-Stern-Kai
7, D-60590 Frankfurt, Germany
| | - Mario Wurglics
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Alexander Paulke
- Department
of Forensic Toxicology, Institute of Forensic Medicine, Goethe University Frankfurt, Kennedyallee 104, D-60596 Frankfurt, Germany
| | - Gerd Geisslinger
- Institute
of Clinical Pharmacology, Goethe University Frankfurt, Theodor-Stern-Kai
7, D-60590 Frankfurt, Germany
| | - Astrid Kahnt
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Dieter Steinhilber
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Ewgenij Proschak
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| | - Daniel Merk
- Institute
of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse
9, D-60438 Frankfurt, Germany
| |
Collapse
|
141
|
Reduced coronary reactive hyperemia in mice was reversed by the soluble epoxide hydrolase inhibitor (t-AUCB): Role of adenosine A 2A receptor and plasma oxylipins. Prostaglandins Other Lipid Mediat 2017; 131:83-95. [PMID: 28890385 DOI: 10.1016/j.prostaglandins.2017.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 12/24/2022]
Abstract
Coronary reactive hyperemia (CRH) protects the heart against ischemia. Adenosine A2AAR-deficient (A2AAR-/-) mice have increased expression of soluble epoxide hydrolase (sEH); the enzyme responsible for breaking down the cardioprotective epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids (DHETs). sEH-inhibition enhances CRH, increases EETs, and modulates oxylipin profiles. We investigated the changes of oxylipins and their impact on CRH in A2AAR-/- and wild type (WT) mice. We hypothesized that the attenuated CRH in A2AAR-/- mice is mediated by changes in oxylipin profiles, and that it can be reversed by either sEH- or ω-hydroxylases-inhibition. Compared to WT mice, A2AAR-/- mice had attenuated CRH and changed oxylipin profiles, which were consistent between plasma and heart perfusate samples, including decreased EET/DHET ratios, and increased hydroxyeicosatetraenoic acids (HETEs). Plasma oxylipns in A2AAR-/- mice indicated an increased proinflammatory state including increased ω-terminal HETEs, decreased epoxyoctadecaenoic/dihydroxyoctadecaenoic acids (EpOMEs/DiHOMEs) ratios, increased 9-hydroxyoctadecadienoic acid, and increased prostanoids. Inhibition of either sEH or ω-hydroxylases reversed the reduced CRH in A2AAR-/- mice. In WT and sEH-/- mice, blocking A2AAR decreased CRH. These data demonstrate that A2AAR-deletion was associated with changes in oxylipin profiles, which may contribute to the attenuated CRH. Also, inhibition of sEH and ω-hydroxylases reversed the reduction in CRH.
Collapse
|
142
|
Inceoglu B, Bettaieb A, Haj FG, Gomes AV, Hammock BD. Modulation of mitochondrial dysfunction and endoplasmic reticulum stress are key mechanisms for the wide-ranging actions of epoxy fatty acids and soluble epoxide hydrolase inhibitors. Prostaglandins Other Lipid Mediat 2017; 133:68-78. [PMID: 28847566 DOI: 10.1016/j.prostaglandins.2017.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022]
Abstract
The arachidonic acid cascade is arguably the most widely known biologic regulatory pathway. Decades after the seminal discoveries involving its cyclooxygenase and lipoxygenase branches, studies of this cascade remain an active area of research. The third and less widely known branch, the cytochrome P450 pathway leads to highly active oxygenated lipid mediators, epoxy fatty acids (EpFAs) and hydroxyeicosatetraenoic acids (HETEs), which are of similar potency to prostanoids and leukotrienes. Unlike the COX and LOX branches, no pharmaceuticals currently are marketed targeting the P450 branch. However, data support therapeutic benefits from modulating these regulatory lipid mediators. This is being approached by stabilizing or mimicking the EpFAs or even by altering the diet. These approaches lead to predominantly beneficial effects on a wide range of apparently unrelated states resulting in an enigma of how this small group of natural chemical mediators can have such diverse effects. EpFAs are degraded by soluble epoxide hydrolase (sEH) and stabilized by inhibiting this enzyme. In this review, we focus on interconnected aspects of reported mechanisms of action of EpFAs and inhibitors of soluble epoxide hydrolase (sEHI). The sEHI and EpFAs are commonly reported to maintain homeostasis under pathological conditions while remaining neutral under normal physiological conditions. Here we provide a conceptual framework for the unique and broad range of biological activities ascribed to epoxy fatty acids. We argue that their mechanism of action pivots on their ability to prevent mitochondrial dysfunction, to reduce subsequent ROS formation and to block resulting cellular signaling cascades, primarily the endoplasmic reticulum stress. By stabilizing the mitochondrial - ROS - ER stress axis, the range of activity of EpFAs and sEHI display an overlap with the disease conditions including diabetes, fibrosis, chronic pain, cardiovascular and neurodegenerative diseases, for which the above outlined mechanisms play key roles.
Collapse
Affiliation(s)
- Bora Inceoglu
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States.
| | - Ahmed Bettaieb
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996-0840, United States; Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN 37996-0840, United States.
| | - Fawaz G Haj
- Department of Nutrition, University of California Davis, CA 95616, United States; Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, United States
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, CA 95616, United States; Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States
| |
Collapse
|
143
|
Garscha U, Romp E, Pace S, Rossi A, Temml V, Schuster D, König S, Gerstmeier J, Liening S, Werner M, Atze H, Wittmann S, Weinigel C, Rummler S, Scriba GK, Sautebin L, Werz O. Pharmacological profile and efficiency in vivo of diflapolin, the first dual inhibitor of 5-lipoxygenase-activating protein and soluble epoxide hydrolase. Sci Rep 2017; 7:9398. [PMID: 28839250 PMCID: PMC5571211 DOI: 10.1038/s41598-017-09795-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022] Open
Abstract
Arachidonic acid (AA) is metabolized to diverse bioactive lipid mediators. Whereas the 5-lipoxygenase-activating protein (FLAP) facilitates AA conversion by 5-lipoxygenase (5-LOX) to pro-inflammatory leukotrienes (LTs), the soluble epoxide hydrolase (sEH) degrades anti-inflammatory epoxyeicosatrienoic acids (EETs). Accordingly, dual FLAP/sEH inhibition might be advantageous drugs for intervention of inflammation. We present the in vivo pharmacological profile and efficiency of N-[4-(benzothiazol-2-ylmethoxy)-2-methylphenyl]-N′-(3,4-dichlorophenyl)urea (diflapolin) that dually targets FLAP and sEH. Diflapolin inhibited 5-LOX product formation in intact human monocytes and neutrophils with IC50 = 30 and 170 nM, respectively, and suppressed the activity of isolated sEH (IC50 = 20 nM). Characteristic for FLAP inhibitors, diflapolin (I) failed to inhibit isolated 5-LOX, (II) blocked 5-LOX product formation in HEK cells only when 5-LOX/FLAP was co-expressed, (III) lost potency in intact cells when exogenous AA was supplied, and (IV) prevented 5-LOX/FLAP complex assembly in leukocytes. Diflapolin showed target specificity, as other enzymes related to AA metabolism (i.e., COX1/2, 12/15-LOX, LTA4H, LTC4S, mPGES1, and cPLA2) were not inhibited. In the zymosan-induced mouse peritonitis model, diflapolin impaired vascular permeability, inhibited cysteinyl-LTs and LTB4 formation, and suppressed neutrophil infiltration. Diflapolin is a highly active dual FLAP/sEH inhibitor in vitro and in vivo with target specificity to treat inflammation-related diseases.
Collapse
Affiliation(s)
- Ulrike Garscha
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany.
| | - Erik Romp
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Simona Pace
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Antonietta Rossi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131, Naples, Italy
| | - Veronika Temml
- Department of Pharmacy / Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Daniela Schuster
- Department of Pharmacy / Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Stefanie König
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Jana Gerstmeier
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Stefanie Liening
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Markus Werner
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Heiner Atze
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Sandra Wittmann
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438, Frankfurt, Germany
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, 07743, Jena, Germany
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, 07743, Jena, Germany
| | - Gerhard K Scriba
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Lidia Sautebin
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131, Naples, Italy
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743, Jena, Germany
| |
Collapse
|
144
|
Soluble epoxide hydrolase inhibitor AUDA decreases bleomycin-induced pulmonary toxicity in mice by inhibiting the p38/Smad3 pathways. Toxicology 2017; 389:31-41. [DOI: 10.1016/j.tox.2017.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/02/2017] [Accepted: 07/04/2017] [Indexed: 12/27/2022]
|
145
|
Gong Y, Fu Z, Liegl R, Chen J, Hellström A, Smith LEH. ω-3 and ω-6 long-chain PUFAs and their enzymatic metabolites in neovascular eye diseases. Am J Clin Nutr 2017; 106:16-26. [PMID: 28515072 PMCID: PMC5486202 DOI: 10.3945/ajcn.117.153825] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/18/2017] [Indexed: 01/01/2023] Open
Abstract
Neovascular eye diseases, including retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration, threaten the visual health of children and adults. Current treatment options, including anti-vascular endothelial growth factor therapy and laser retinal photocoagulation, have limitations and are associated with adverse effects; therefore, the identification of additional therapies is highly desirable. Both clinical and experimental studies show that dietary ω-3 (n-3) long-chain polyunsaturated fatty acids (LC-PUFAs) reduce retinal and choroidal angiogenesis. The ω-3 LC-PUFA metabolites from 2 groups of enzymes, cyclooxygenases and lipoxygenases, inhibit [and the ω-6 (n-6) LC-PUFA metabolites promote] inflammation and angiogenesis. However, both of the ω-3 and the ω-6 lipid products of cytochrome P450 oxidase 2C promote neovascularization in both the retina and choroid, which suggests that inhibition of this pathway might be beneficial. This review summarizes our current understanding of the roles of ω-3 and ω-6 LC-PUFAs and their enzymatic metabolites in neovascular eye diseases.
Collapse
Affiliation(s)
- Yan Gong
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA; and
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA; and
| | - Raffael Liegl
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA; and
| | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA; and
| | - Ann Hellström
- Department of Ophthalmology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lois EH Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA; and
| |
Collapse
|
146
|
Skibba M, Hye Khan MA, Kolb LL, Yeboah MM, Falck JR, Amaradhi R, Imig JD. Epoxyeicosatrienoic Acid Analog Decreases Renal Fibrosis by Reducing Epithelial-to-Mesenchymal Transition. Front Pharmacol 2017; 8:406. [PMID: 28713267 PMCID: PMC5491687 DOI: 10.3389/fphar.2017.00406] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/08/2017] [Indexed: 12/18/2022] Open
Abstract
Renal fibrosis, which is a critical pathophysiological event in chronic kidney diseases, is associated with renal epithelial-to-mesenchymal transition (EMT). Epoxyeicosatrienoic acids (EETs) are Cyp epoxygenase arachidonic acid metabolites that demonstrate biological actions that result in kidney protection. Herein, we investigated the ability of 14,15-EET and its synthetic analog, EET-A, to reduce kidney fibrosis induced by unilateral ureter obstruction (UUO). C57/BL6 male mice underwent sham or UUO surgical procedures and were treated with 14,15-EET or EET-A in osmotic pump (i.p.) for 10 days following UUO surgery. UUO mice demonstrated renal fibrosis with an 80% higher kidney-collagen positive area and 70% higher α-smooth muscle actin (SMA) positive renal areas compared to the sham group. As a measure of collagen content, kidney hydroxyproline content was also higher in UUO (6.4 ± 0.5 μg/10 mg) compared to sham group (2.5 ± 0.1 μg/10 mg). Along with marked renal fibrosis, UUO mice had reduced renal expression of EET producing Cyp epoxygenase enzymes. Endogenous 14,15-EET or EET-A demonstrated anti-fibrotic action in UUO by reducing kidney-collagen positive area (50–60%), hydroxyproline content (50%), and renal α-SMA positive area (85%). In UUO mice, renal expression of EMT inducers, Snail1 and ZEB1 were higher compared to sham group. Accordingly, renal epithelial marker E-cadherin expression was reduced and mesenchymal marker expression was elevated in the UUO compared to sham mice. Interestingly, EET-A reduced EMT in UUO mice by deceasing renal Snail1 and ZEB1 expression. EET-A treatment also opposed the decrease in renal E-cadherin expression and markedly reduced several prominent renal mesenchymal/myofibroblast markers in UUO mice. Overall, our results demonstrate that EET-A is a novel anti-fibrotic agent that reduces renal fibrosis by decreasing renal EMT.
Collapse
Affiliation(s)
- Melissa Skibba
- Department of Pharmacology and Toxicology, The Medical College of Wisconsin, MilwaukeeWI, United States
| | - Md Abdul Hye Khan
- Department of Pharmacology and Toxicology, The Medical College of Wisconsin, MilwaukeeWI, United States
| | - Lauren L Kolb
- Department of Pharmacology and Toxicology, The Medical College of Wisconsin, MilwaukeeWI, United States
| | - Michael M Yeboah
- Department of Medicine, The Medical College of Wisconsin, MilwaukeeWI, United States
| | - John R Falck
- Department of Biochemistry, UT Southwestern Medical Center, DallasTX, United States
| | - Radhika Amaradhi
- Department of Biochemistry, UT Southwestern Medical Center, DallasTX, United States
| | - John D Imig
- Department of Pharmacology and Toxicology, The Medical College of Wisconsin, MilwaukeeWI, United States
| |
Collapse
|
147
|
Cao J, Singh SP, McClung JA, Joseph G, Vanella L, Barbagallo I, Jiang H, Falck JR, Arad M, Shapiro JI, Abraham NG. EET intervention on Wnt1, NOV, and HO-1 signaling prevents obesity-induced cardiomyopathy in obese mice. Am J Physiol Heart Circ Physiol 2017; 313:H368-H380. [PMID: 28576832 PMCID: PMC5582926 DOI: 10.1152/ajpheart.00093.2017] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 01/15/2023]
Abstract
We have previously reported that epoxyeicosatrienoic acid (EET) has multiple beneficial effects on vascular function; in addition to its antiapoptotic action, it increases insulin sensitivity and inhibits inflammation. To uncover the signaling mechanisms by which EET reduces cardiomyopathy, we hypothesized that EET infusion might ameliorate obesity-induced cardiomyopathy by improving heme oxygenase (HO)-1, Wnt1, thermogenic gene levels, and mitochondrial integrity in cardiac tissues and improved pericardial fat phenotype. EET reduced levels of fasting blood glucose and proinflammatory adipokines, including nephroblastoma overexpressed (NOV) signaling, while increasing echocardiographic fractional shortening and O2 consumption. Of interest, we also noted a marked improvement in mitochondrial integrity, thermogenic genes, and Wnt 1 and HO-1 signaling mechanisms. Knockout of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) in EET-treated mice resulted in a reversal of these beneficial effects including a decrease in myocardial Wnt1 and HO-1 expression and an increase in NOV. To further elucidate the effects of EET on pericardial adipose tissues, we observed EET treatment increases in adiponectin, PGC-1α, phospho-AMP-activated protein kinase, insulin receptor phosphorylation, and thermogenic genes, resulting in a "browning" pericardial adipose phenotype under high-fat diets. Collectively, these experiments demonstrate that an EET agonist increased Wnt1 and HO-1 signaling while decreasing NOV pathways and the progression of cardiomyopathy. Furthermore, this report presents a portal into potential therapeutic approaches for the treatment of heart failure and metabolic syndrome.NEW & NOTEWORTHY The mechanism by which EET acts on obesity-induced cardiomyopathy is unknown. Here, we describe a previously unrecognized function of EET infusion that inhibits nephroblastoma overexpressed (NOV) levels and activates Wnt1, hence identifying NOV inhibition and enhanced Wnt1 expression as novel pharmacological targets for the prevention and treatment of cardiomyopathy and heart failure.Listen to this article's corresponding podcast at http://ajpheart.physiology.org/content/early/2017/05/31/ajpheart.00093.2017.
Collapse
Affiliation(s)
- Jian Cao
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, New York.,Chinese PLA General Hospital, Beijing, China
| | - Shailendra P Singh
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, New York
| | - John A McClung
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, New York
| | - Gregory Joseph
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, New York
| | - Luca Vanella
- Department of Drug Science/Section of Biochemistry, University of Catania, Catania, Italy
| | - Ignazio Barbagallo
- Department of Drug Science/Section of Biochemistry, University of Catania, Catania, Italy
| | - Houli Jiang
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, New York
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael Arad
- Leviev Heart Center, Tel Hashomer, Tel Aviv University, Tel Aviv, Israel; and
| | - Joseph I Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, New York; .,Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| |
Collapse
|
148
|
Trindade-da-Silva CA, Bettaieb A, Napimoga MH, Lee KSS, Inceoglu B, Ueira-Vieira C, Bruun D, Goswami SK, Haj FG, Hammock BD. Soluble Epoxide Hydrolase Pharmacological Inhibition Decreases Alveolar Bone Loss by Modulating Host Inflammatory Response, RANK-Related Signaling, Endoplasmic Reticulum Stress, and Apoptosis. J Pharmacol Exp Ther 2017; 361:408-416. [PMID: 28356494 PMCID: PMC5443319 DOI: 10.1124/jpet.116.238113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/24/2017] [Indexed: 12/14/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid derived from the cytochrome P450 enzymes, are mainly metabolized by soluble epoxide hydrolase (sEH) to their corresponding diols. EETs but not their diols, have anti-inflammatory properties, and inhibition of sEH might provide protective effects against inflammatory bone loss. Thus, in the present study, we tested the selective sEH inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), in a mouse model of periodontitis induced by infection with Aggregatibacter actinomycetemcomitans Oral treatment of wild-type mice with TPPU and sEH knockout (KO) animals showed reduced bone loss induced by A. actinomycetemcomitans This was associated with decreased expression of key osteoclastogenic molecules, receptor activator of nuclear factor-κB/RANK ligand/osteoprotegerin, and the chemokine monocyte chemotactic protein 1 in the gingival tissue without affecting bacterial counts. In addition, downstream kinases p38 and c-Jun N-terminal kinase known to be activated in response to inflammatory signals were abrogated after TPPU treatment or in sEH KO mice. Moreover, endoplasmic reticulum stress was elevated in periodontal disease but was abrogated after TPPU treatment and in sEH knockout mice. Together, these results demonstrated that sEH pharmacological inhibition may be of therapeutic value in periodontitis.
Collapse
Affiliation(s)
- Carlos Antonio Trindade-da-Silva
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Ahmed Bettaieb
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Marcelo Henrique Napimoga
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Kin Sing Stephen Lee
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Bora Inceoglu
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Carlos Ueira-Vieira
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Donald Bruun
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Sumanta Kumar Goswami
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Fawaz G Haj
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| | - Bruce D Hammock
- Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center (C.A.T.-S., K.S.S.L., B.I., S.K.G., B.D.H.), Nutrition Department (F.G.H.), and Department of Molecular Biosciences, School of Veterinary Medicine (D.B.), University of California, Davis, California; Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Brazil (C.A.T.-d.-S., C.U.-V.); Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil (M.H.N.); and Department of Nutrition, University of Tennessee-Knoxville, Knoxville, Tennessee (A.B.)
| |
Collapse
|
149
|
Gangadhariah MH, Dieckmann BW, Lantier L, Kang L, Wasserman DH, Chiusa M, Caskey CF, Dickerson J, Luo P, Gamboa JL, Capdevila JH, Imig JD, Yu C, Pozzi A, Luther JM. Cytochrome P450 epoxygenase-derived epoxyeicosatrienoic acids contribute to insulin sensitivity in mice and in humans. Diabetologia 2017; 60:1066-1075. [PMID: 28352940 PMCID: PMC5921930 DOI: 10.1007/s00125-017-4260-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/28/2017] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Insulin resistance is frequently associated with hypertension and type 2 diabetes. The cytochrome P450 (CYP) arachidonic acid epoxygenases (CYP2C, CYP2J) and their epoxyeicosatrienoic acid (EET) products lower blood pressure and may also improve glucose homeostasis. However, the direct contribution of endogenous EET production on insulin sensitivity has not been previously investigated. In this study, we tested the hypothesis that endogenous CYP2C-derived EETs alter insulin sensitivity by analysing mice lacking CYP2C44, a major EET producing enzyme, and by testing the association of plasma EETs with insulin sensitivity in humans. METHODS We assessed insulin sensitivity in wild-type (WT) and Cyp2c44 -/- mice using hyperinsulinaemic-euglycaemic clamps and isolated skeletal muscle. Insulin secretory function was assessed using hyperglycaemic clamps and isolated islets. Vascular function was tested in isolated perfused mesenteric vessels. Insulin sensitivity and secretion were assessed in humans using frequently sampled intravenous glucose tolerance tests and plasma EETs were measured by mass spectrometry. RESULTS Cyp2c44 -/- mice showed decreased glucose tolerance (639 ± 39.5 vs 808 ± 37.7 mmol/l × min for glucose tolerance tests, p = 0.004) and insulin sensitivity compared with WT controls (hyperinsulinaemic clamp glucose infusion rate average during terminal 30 min 0.22 ± 0.02 vs 0.33 ± 0.01 mmol kg-1 min-1 in WT and Cyp2c44 -/- mice respectively, p = 0.003). Although glucose uptake was diminished in Cyp2c44 -/- mice in vivo (gastrocnemius Rg 16.4 ± 2.0 vs 6.2 ± 1.7 μmol 100 g-1 min-1, p < 0.01) insulin-stimulated glucose uptake was unchanged ex vivo in isolated skeletal muscle. Capillary density was similar but vascular KATP-induced relaxation was impaired in isolated Cyp2c44 -/- vessels (maximal response 39.3 ± 6.5% of control, p < 0.001), suggesting that impaired vascular reactivity produces impaired insulin sensitivity in vivo. Similarly, plasma EETs positively correlated with insulin sensitivity in human participants. CONCLUSIONS/INTERPRETATION CYP2C-derived EETs contribute to insulin sensitivity in mice and in humans. Interventions to increase circulating EETs in humans could provide a novel approach to improve insulin sensitivity and treat hypertension.
Collapse
Affiliation(s)
- Mahesha H Gangadhariah
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Medical Center North B3109, Nashville, TN, 37232-6602, USA
| | - Blake W Dieckmann
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Medical Center North B3109, Nashville, TN, 37232-6602, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Li Kang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Manuel Chiusa
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Medical Center North B3109, Nashville, TN, 37232-6602, USA
| | - Charles F Caskey
- Department of Radiologic Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jaime Dickerson
- Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, USA
| | - Pengcheng Luo
- Huangshi Central Hospital, Hubei Province, People's Republic of China
| | - Jorge L Gamboa
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jorge H Capdevila
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Medical Center North B3109, Nashville, TN, 37232-6602, USA
| | - John D Imig
- Department of Pharmacology and Toxicology, Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Chang Yu
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Medical Center North B3109, Nashville, TN, 37232-6602, USA.
- Department of Veterans Affairs, Nashville, TN, USA.
| | - James M Luther
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Medical Center North B3109, Nashville, TN, 37232-6602, USA.
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
150
|
He Z, Yang Y, Wen Z, Chen C, Xu X, Zhu Y, Wang Y, Wang DW. CYP2J2 metabolites, epoxyeicosatrienoic acids, attenuate Ang II-induced cardiac fibrotic response by targeting Gα 12/13. J Lipid Res 2017; 58:1338-1353. [PMID: 28554983 DOI: 10.1194/jlr.m074229] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/23/2017] [Indexed: 12/23/2022] Open
Abstract
The arachidonic acid-cytochrome P450 2J2-epoxyeicosatrienoic acid (AA-CYP2J2-EET) metabolic pathway has been identified to be protective in the cardiovascular system. This study explored the effects of the AA-CYP2J2-EET metabolic pathway on cardiac fibrosis from the perspective of cardiac fibroblasts and underlying mechanisms. In in vivo studies, 8-week-old male CYP2J2 transgenic mice (aMHC-CYP2J2-Tr) and littermates were infused with angiotensin II (Ang II) or saline for 2 weeks. Results showed that CYP2J2 overexpression increased EET production. Meanwhile, impairment of cardiac function and fibrotic response were attenuated by CYP2J2 overexpression. The effects of CYP2J2 were associated with reduced activation of the α subunits of G12 family G proteins (Gα12/13)/RhoA/Rho kinase (ROCK) cascade and elevation of the NO/cyclic guanosine monophosphate (cGMP) level in cardiac tissue. In in vitro studies, cardiac fibroblast activation, proliferation, migration, and collagen production induced by Ang II were associated with activation of the Gα12/13/RhoA/ROCK pathway, which was inhibited by exogenous 11,12-EET. Moreover, silencing of Gα12/13 or RhoA exerted similar effects as 11,12-EET. Furthermore, inhibitory effects of 11,12-EET on Gα12/13 were blocked by NO/cGMP pathway inhibitors. Our findings indicate that enhancement of the AA-CYP2J2-EET metabolic pathway by CYP2J2 overexpression attenuates Ang II-induced cardiac dysfunction and fibrosis by reducing the fibrotic response of cardiac fibroblasts by targeting the Gα12/13/RhoA/ROCK pathway via NO/cGMP signaling.
Collapse
Affiliation(s)
- Zuowen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yong Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xizhen Xu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yanfang Zhu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| |
Collapse
|