Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jan 1.
Published in final edited form as: Biochem Pharmacol. 2021 Dec 2;195:114866. doi: 10.1016/j.bcp.2021.114866

Epoxylipids and Soluble Epoxide Hydrolase in Heart Diseases

John D Imig 1, Ludek Cervenka 2,3, Jan Neckar 2,4
PMCID: PMC8712413  NIHMSID: NIHMS1761489  PMID: 34863976

Abstract

Cardiovascular and heart diseases are leading causes of morbidity and mortality. Coronary artery endothelial and vascular dysfunction, inflammation, and mitochondrial dysfunction contribute to progression of heart diseases such as arrhythmias, congestive heart failure, and heart attacks. Classes of fatty acid epoxylipids and their enzymatic regulation by soluble epoxide hydrolase (sEH) have been implicated in coronary artery dysfunction, inflammation, and mitochondrial dysfunction in heart diseases. Likewise, genetic and pharmacological manipulations of epoxylipids have been demonstrated to have therapeutic benefits for heart diseases. Increasing epoxylipids reduce cardiac hypertrophy and fibrosis and improve cardiac function. Beneficial actions for epoxylipids have been demonstrated in cardiac ischemia reperfusion injury, electrical conductance abnormalities and arrhythmias, and ventricular tachycardia. This review discusses past and recent findings on the contribution of epoxylipids in heart diseases and the potential for their manipulation to treat heart attacks, arrhythmias, ventricular tachycardia, and heart failure.

Keywords: eicosanoids, cytochrome P450, soluble epoxide hydrolase, hypertension, heart failure, myocardial infarction, inflammation, mitochondrial function, coronary artery

Graphical Abstract

graphic file with name nihms-1761489-f0001.jpg

1. Introduction

Cardiovascular diseases are estimated to result in 18 million deaths globally each year which is projected to rise to 24 million deaths per year by 2030 [1,2]. This represents one third of all global deaths with over 80% of cardiovascular deaths being due to heart attacks and strokes [1,2]. Risk factors for cardiovascular diseases include a sedentary lifestyle, inappropriate diet, tobacco use, and alcohol intake [3,4]. These risk factors can result in an elevated blood pressure, hyperglycemia, increased plasma lipid levels, and obesity [3,4]. There have been advances in patient care that have led to improvements in cardiovascular outcomes [3,5]. Despite the progress, the decreases in cardiovascular mortality are dissipating [1,2]. Unlike other diseases areas such as cancer and neurological disorders, the pipeline for cardiovascular disease drugs is weak [6]. Therefore, identifying targets for cardiovascular drugs is desperately needed.

The pathology of cardiovascular diseases includes changes in vascular and cardiac function that can damage multiple organs resulting in heart failure [7,8]. Vascular endothelium dysfunction is a major contributor to cardiovascular diseases such as atherosclerosis, coronary artery disease, and heart disease [9,10]. Heart attacks are a consequence of these cardiovascular diseases [3,9]. Damage to the heart is due to inflammation, oxidative stress, apoptosis, necrosis, ionic disturbances, and alterations in mitochondrial bioenergetics [11,12]. Extensive deterioration of cardiac function occurs due to atrial or ventricular arrhythmias, myocardial fibrosis, and ventricular hypertrophy [7,8,9]. Eventually cardiac function declines and progresses to heart failure [7,8].

Although drugs for cardiovascular diseases can combat hypertension, dyslipidemia, and diabetes, there is an urgent need to identify therapeutic targets that will further decrease mortality. Manipulation of fatty acids provides opportunities to develop cardiovascular drugs [1316]. Epoxy fatty acids and soluble epoxide hydrolase (sEH) are promising targets treating cardiovascular diseases [13,15,17,18]. It is well recognized that polyunsaturated fatty acids have vascular and cardiac biological activities that could be beneficial in combating cardiovascular diseases [14,15,16]. In addition, there is significant evidence that regulation of enzymes that regulate polyunsaturated fatty acids can impact cardiovascular diseases [14,15,16]. Epoxy fatty acids of arachidonic acid, eicosapentaenoic acid (EPA), and docosohexaenoic acid (DHA) are epoxylipids that can improve mitochondrial function, decrease lipidemia, decrease inflammation, improve insulin sensitivity, lower blood pressure, and improve endothelial dysfunction [14,16]. These epoxy fatty acids can be converted to diols by sEH that decreases beneficial vascular and cardiac biological activities (Figure 1) [14,15,16]. These findings have led to development of drugs that can inhibit sEH to increase epoxy fatty acid levels or drugs that act as epoxy fatty acid mimetics [14,15,16].

Figure 1 – Epoxylipid Metabolites.

Figure 1 –

Fatty acids arachidonic acid, eicosapentaenoic acid, and docosohexaenoic acid are metabolized by CYP2C and CYP2J epoxygenase enzymes to epoxyeicosatrienoic acids (EET), epoxyeicosatetraenoic acids (EEQ), and epoxydocosapentaenoic acids (EDP). Epoxylipids are then metabolized to their corresponding diols, dihydroxyeicosatetraenoic acids (diHETE) and dihydroxydocosapentaenoic acids (diHDPA) by soluble epoxide hydrolase (sEH).

This review will focus on the contribution of epoxy fatty acids and sEH to myocardial infarction, arrythmias, ventricular tachycardia, and chronic heart failure. The protective actions and cell signaling mechanisms by which arachidonic acid, EPA, and DHA epoxy fatty acids improve cardiac function will be detailed. Finally, the development and testing of sEH inhibitors and epoxylipid mimics in cardiac experimental models will be provided. The progress to date demonstrates the potential for epoxylipid based drugs to treat heart attacks, arrhythmias, ventricular tachycardia, and heart failure.

2. Epoxylipids and Soluble Epoxide Hydrolase in Heart Diseases

Cardiovascular diseases have been associated with a decrease in arachidonic acid epoxylipids, epoxyeicosatrienoic acids (EETs) and an increased degradation by sEH to their corresponding diols [19,20]. Biological actions demonstrated by EETs can combat cardiovascular diseases. EETs demonstrate vasodilation, anti-inflammatory, anti-apoptotic, and anti-smooth muscle cell migration actions [15,17]. Human genetic studies have found associations between cytochrome P450 (CYP) epoxygenase and sEH enzymes with increased risk for cardiovascular diseases [19,2124]. In most of these studies genetic polymorphisms that result in decreased EETs or increased sEH activity associate with increased cardiovascular disease risk or progression [19,24]. Associations between genetic variations that decrease CYP epoxygenases or plasma EET levels and hypertension have been demonstrated in different cohorts [19,24,25,26]. On the other hand, increased plasma EET levels were found in patients with stable angiographically confirmed coronary artery disease [23]. Increased plasma EET levels in patients after stroke and decreased expression of EPHX2, the gene for sEH, in heart failure patients have also been observed [27,28]. These finding demonstrate that mechanisms to upregulate EETs can act to oppose cardiovascular disease in certain patients.

Experimental studies in animal disease models have provide greater mechanistic insight into epoxylipids and their ability to combat cardiovascular diseases [14,15,16]. Hypertension animal models have been the most extensively studied regarding EETs and sEH to blood pressure control and progression of end organ damage and heart disease [13,15]. Decreased EET levels and increased sEH expression were found to contribute to increased vascular resistance and electrolyte homeostasis in hypertension [29,30,31]. Genetic manipulation in mice that increased or decreased EET levels further demonstrated a contribution to regulation of blood pressure and progression of heart disease in hypertension [3236]. In addition to effects on vascular resistance and electrolyte homeostasis, EETs regulate inflammation and oxidative stress in hypertension [13,15]. These findings led to evaluating sEH inhibitors and EET analogs as therapeutics for hypertension [13,18,37].

Myocardial infarction studies in rodents have provided additional mechanistic evidence for EETs and other epoxylipids to cardiovascular diseases [15,16,38]. EETs have been demonstrated to improve cardiac mitochondrial function, decrease inflammation, and oppose apoptosis to reduce cardiac fibrosis and hypertrophy (Figure 2) [14,38]. Cardiomyocyte mitochondrial function is regulated by EETs which improve heart function following ischemia [39,40,41]. Cardioprotective mitochondrial actions were found in mice overexpressing the CYP2J2 epoxygenase or lacking sEH [38,39,41]. EET actions on vascular smooth muscle cell K+ channels vasodilate coronary arteries and EET actions on cardiomyocyte K+ channels oppose metabolic stress to decrease heart damage following ischemia [42]. EETs have also been clearly demonstrated to reduce cardiac cell death [14,38]. Cardiomyocytes exposed to hypoxic conditions and treated with EETs demonstrated increased cell viability, mitochondrial membrane stability, and decreased caspase-3 activity [43,44]. EET actions on phosphoinositide 3-kinase (PI3K) and KATP channels are critical signaling mechanisms for the anti-apoptotic cardiomyocyte effects. Lastly, EET anti-inflammatory actions on the heart provide protection from adverse cardiac remodeling and fibrosis that contribute to left ventricular dysfunction [14,15]. EETs can oppose vascular and heart monocyte and neutrophil infiltration and can inhibit nuclear factor-κB (NF-κB) activation to reduce proliferation of cardiac fibroblasts and development of heart fibrosis [34,35]. Cardiac dysfunction following lipopolysaccharide injection in mice is decreased by EET actions on peroxisome proliferator-activated receptor gamma (PPARγ) and heme oxygenase-1 (HO-1) pathways to reduce M1 macrophage polarization and infiltration resulting in decreased inflammatory cytokine production [45]. These cardioprotective EET actions ultimately protect from ischemic and non-ischemic heart diseases.

Figure 2 – Cardiomyocyte Actions for Epoxyeicosatrienoic Acids (EET), Epoxyeicosatetraenoic Acids (EEQ), and Epoxydocosapentaenoic Acids (EDP).

Figure 2 –

Arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosohexaenoic acid (DHA) are metabolized by CYP2C and CYP2J epoxygenase enzymes to EET, EEQ, and EDP. EET, EEQ, and EDP decrease endoplasmic reticulum (ER) stress, combat mitochondrial dysfunction, inhibit transforming growth factor β1 (TGF-β1), reduce the NLRP3 inflammasome, and inhibits nuclear factor-κB (NF-κB) activation to reduce proinflammatory cytokines, chemokines, and adhesion molecules. These cardiomyocyte EET, EEQ, and EDP actions reduce apoptosis and inflammation to decrease fibrosis and heart remodeling.

The cardioprotective actions for n-3 polyunsaturated fatty acids diets have been long recognized [16,46,47]. EPA and DHA rich diets result in increased plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP) [16]. These experimental studies have demonstrated cardiomyocyte cell signaling mechanisms responsible for improved heart function (Figure 2) [16]. Increased dietary EPA and DHA can improve cardiac mitochondrial function and decrease apoptosis [46,48,49]. In addition, cardiomyocytes in cell culture have demonstrated direct actions for EPA and DHA to decrease mitochondrial dysfunction, decrease oxidative stress, and oppose apoptosis [46,49]. EPA can attenuate H92C cardiac cell oxidative stress and apoptosis through activating an adaptive autophagic response [46,49]. EPA treatment protected against apoptosis via ERK activation in cultured cardiomyocytes exposed to hypoxia [50]. Likewise, the DHA epoxygenase metabolite 19,20-EDP can protect against lipopolysaccharide-induced cardiac cell toxicity by activation of sirtuin 1 (SIRT1) to improve mitochondrial function resulting in cell survival [51]. DHA also decreased cardiac damage in isolated heart rats exposed to ischemic reperfusion injury [52]. An experimental study that compared EPA, DHA, 17,18-EEQ, and 19,20-EDP on isolated mouse hearts exposed to ischemia reperfusion revealed that DHA and 19,20-EDP but not EPA and 17,18-EEQ provided cardioprotection [53]. 19,20-EDP provided the greatest protection against myocardial infarction through reduction in the NLPR3 inflammasome to preserve mitochondrial function [53]. These findings demonstrate the potential for EPA, DHA, and their epoxylipid metabolites to protect cardiomyocytes via cell signaling mechanisms that impact mitochondrial function, oxidative stress, and apoptosis.

Experiments conducted in rodents with cardiac ischemia reperfusion injury provide additional evidence for EPA, DHA, and their epoxylipid metabolites to decrease apoptosis and decrease mitochondrial dysfunction [14,16]. Mice fed a diet enriched with EPA and DHA protects against thoracic aortic constriction induced left ventricular dysfunction and fibrosis [54]. Cardiomyocyte studies demonstrate that the anti-fibrotic actions for EPA and DHA are due to inhibiting transforming growth factor β1 (TGF-β1) [54]. Dietary EPA was also found to oppose pressure overload cardiac fibrosis through decreasing TGF-β1 signaling [55]. Anti-inflammatory actions for dietary EPA have been demonstrated in rodents that decrease cardiac remodeling following myocardial infarction [46,56]. Clinical studies have provided additional evidence for cardiac benefits for diets enriched with EPA and DHA [57,58,59]. Consumption of EPA/DHA supplements reduces cardiac sudden death and the severity of ventricular arrhythmias [57,58]. The findings that EPA, DHA, and their epoxylipid cardioprotective actions has led to therapeutically targeting these fatty acids for various heart diseases.

3. Pharmacological Manipulation of sEH and Epoxylipids in Heart Diseases

The development of therapeutics to manipulate sEH and epoxylipids has provided promising results in animal models of hypertension, vascular diseases, and heart diseases [1317]. Inhibitors of sEH are orally active and have advanced to clinical trials for several diseases including cardiovascular diseases [15,18]. The primary action for sEH inhibitors under normal fatty acid diet conditions is to increase arachidonic acid epoxylipids, EETs [15,18]. Epoxylipid mimics (analogs) that are orally active are also under development by pharmaceutical companies and are in preclinical development and the early stages of clinical trials [13,37]. Epoxylipid mimics for arachidonic acid, EPA, and DHA epoxylipids have been developed and tested in cardiac diseases [13,14,16]. More recent animal studies are evaluating combinations of sEH inhibitors and epoxylipid mimics to treat cardiac diseases [13,37].

Cardioprotective action for sEH inhibitors have been well documented in animal models of left ventricular hypertrophy and ischemia reperfusion injury (Figure 3) [14,15]. The heart protective actions for sEH inhibition is due to effects on the coronary vasculature, inflammatory cells, and cardiomyocytes [14,15]. Left ventricular hypertrophy in mice subjected to pressure overload is prevented by sEH inhibitor actions to decrease cardiac myocyte inflammation [60]. Administration of sEH inhibitors to obese mice decreases inflammation to reduce cardiac fibrosis and hypertrophy [61]. These finding have provided strong evidence that sEH inhibitors have potential to combat the progression of cardiac fibrosis and left ventricular hypertrophy.

Figure 3 – Epoxylipid Drugs to Combat Heart Disease.

Figure 3 –

Soluble epoxide hydrolase inhibitors (sEH), epoxyeicosatrienoic acids (EET) analogs, epoxyeicosatetraenoic acids (EEQ) analogs, and epoxydocosapentaenoic acids (EDP) analogs act to combat cardiac inflammation and oxidative stress, decrease cardiac cell mitochondrial dysfunction and endoplasmic reticulum (ER) stress, and reduce cardiac hypertrophy and fibrosis. These positive cardiac actions of sEH inhibitors, EET analogs, EEQ analogs, and EDP analogs result in a decrease in arrythmias and heart failure

There is also ample evidence in animal models that sEH inhibitors improve cardiac function following an ischemic event [14,15]. Experimental evidence in rodents and canines using the Langendorff-perfused heart and in vivo studies have demonstrated that sEH inhibition decreased heart injury following ischemia [62,63,64]. Coronary artery administration of the sEH inhibitor, AUDA, reduced heart ischemia reperfusion injury in dogs [64]. Interestingly, AUDA also enhanced the cardioprotective actions of 14,15-EET by preventing conversion to the inactive diol [64]. In rodents, sEH inhibitors have been demonstrated to protect against cardiac ischemia reperfusion injury when administered prior to, during, or following reperfusion [62,63,65,66]. The recovery of postischemic left ventricular pressure has also been demonstrated to be improved by sEH inhibition [14,66]. Signaling mechanisms responsible for the sEH inhibitor cardiomyocyte actions include K+ channel activation, anti-apoptosis actions, and improved mitochondrial function [14,15]. Reduction in infarct size with sEH inhibitors is due to anti-apoptotic extracellular signal-regulated kinase (ERK) / mitogen-activated protein kinase (MAPK) and Janus kinase/signal transducer and activator of transcription (JAK/STAT) cell signaling pathways [67,68]. Cardiomyocyte activation of KATP channels to enhance PI3 kinase signaling and the subsequent protein kinase B (Akt) phosphorylation to increase phospho-glycogen kinase-3β (p-GSK-β) is critical for sEH to reduce myocardial ischemia reperfusion injury [65,66]. Activation of cardiac KATP channels also delays or inhibits mitochondrial permeability transition pore (mPTP) opening to prevent apoptosis and necrosis [69]. Taken together, sEH inhibitors have strong cardioprotective effects from ischemia reperfusion injury through activation of cardiomyocyte KATP channels that improves mitochondrial function and decreases apoptosis.

Cardiac protection from the long-term consequences of myocardial infarction have also been described for sEH inhibitors [14,15]. This is an important finding because following the acute phase recovery after a myocardial infarction there is extensive cardiac inflammation, apoptosis, and left ventricular remodeling [15,16,70]. The myocardial infarction induced cardiac hypertrophy and fibrosis can be diminished with sEH inhibition [16,70]. Long-term treatment with sEH inhibitors acts to reduce inflammation that occurs after ischemia reperfusion injury to improve cardiac outcomes [71]. Treatment of sEH inhibitors to rats subjected to permanent coronary artery occlusion reduced cardiac fibrosis [72]. Administration of sEH inhibitors days and weeks following coronary artery ligation also improved left ventricular function [72,73,74]. Mice subjected to acute left anterior descending artery and administered an sEH inhibitor for three weeks had reduced cardiac fibrosis and improved left ventricular function [72]. The improvement in cardiac function with sEH inhibition following myocardial infarction also reduces electrical conductance abnormalities and arrythmias [28,73,74]. Myocardial infarction studies in hypertensive rats have also demonstrated long-term cardioprotective actions for sEH inhibitors [75,76]. Chronic administration of the sEH inhibitor c-AUCB to TGR (Ren-2 renin transgenic rats) increased EET levels, reduced blood pressure, and significantly decreased infarct size following coronary artery occlusion [75]. Selective EET antagonism with 14,15-EEZE completely prevented c-AUCB cardioprotective actions confirming that cardiomyocyte EET actions are responsible for reducing cardiac injury [75]. These experimental findings demonstrate long-term protective sEH inhibitor actions following myocardial infarction that appear to be mediated by cardiomyocyte actions of the epoxylipid EETs.

The cardioprotective actions for sEH inhibitors extend to non-ischemic heart injury [15,16,70]. Atrial fibrillation, arrhythmias, and congestive heart failure are cardiac disorders where sEH inhibitors demonstrate potential as a therapeutic [15,16,70]. Treatment with sEH inhibitors reduced cardiac atrial and ventricular arrhythmias in cardiac hypertrophy rodent models [28,73,74]. Decreased inflammation in response to sEH inhibition is a key mechanism responsible for preventing atrial arrhythmias in cardiac hypertrophy [73,74]. Administration of sEH inhibitors to mice with thoracic aortic constriction resulted in inhibition of NF-κB activation and reduced inflammatory cytokine levels [74]. This reduction in inflammation led to decreased MAPK and endoplasmic reticulum stress to reduce adverse cardiac structural and electrical remodeling of the heart which reduced arrhythmias in mice [74]. Isoproterenol-induced cardiac hypertrophy is also decreased by sEH inhibition [77]. Likewise, inhibition of sEH in obese mice improves coronary artery endothelial function and prevents cardiac remodeling [61]. Mechanisms responsible for the decreased cardiac remodeling and reduced arrythmias include decreased inflammation, improved coronary artery endothelial function, and decreased cardiomyocyte AMP-activated protein kinase (AMPK) signaling resulting in decreased apoptosis and endoplasmic reticulum stress [61].

Less clear are the cardioprotective actions for sEH inhibitors in congestive heart failure. Evidence for a contribution for sEH to congestive heart failure comes from the spontaneously heart failure rat that has an alteration in the EPHX2 gene that facilitates progression of congestive heart failure [28]. Experimental studies that administered an sEH inhibitor to Fawn-hooded hypertensive rats with congestive heart failure and chronic kidney disease demonstrated beneficial actions [76]. Survival rates and urinary albumin levels were improved by sEH inhibition, and the effects were more pronounced in the hypertensive rats [76]. The protective actions of sEH inhibition in the hypertensive Fawn-hooded rats was a result of decreased cardiac hypertrophy and lung congestion [76]. On the other hand, sEH inhibition in aorto-caval fistula in Hannover Sprague-Dawley rats did not improve the course of congestive heart failure or chronic kidney disease [78]. The sEH inhibitor c-AUCB increased renal and cardiac EET availability in the aorto-caval fistula congestive heart failure rats; however, cardiac systolic and diastolic function failed to improve [78]. These findings in congestive heart failure rat models suggest that the experimental conditions and disease progression can influence the beneficial actions for sEH inhibitors. Additional studies are required in congestive heart failure to better understand the potential for sEH inhibitors to combat disease progression.

EET analogs represent a class of epoxylipid mimics that have been extensively evaluated in heart disease animal models [14,15,37]. These EET analogs mimic 11,12-EET and 14,15-EET and resist sEH metabolism [37]. Three generations of EET analogs have been developed with good water solubility, oral bioavailability, and favorable half-life to allow for chronic administration [37]. Initial studies in cardiac diseases with EET analogs have demonstrated excellent therapeutic potential [14,15,37].

The cardiac protective actions for EET analogs have been demonstrated in ischemic reperfusion injury studies (Figure 3) [14,37]. Administration of the 11,12-EET analog, NUDSA to mice following myocardial infarction resulted in improved left ventricular function [79]. Mice treated five days following left anterior descending coronary artery ligation resulted in improved left ventricular diastolic function and decreased cardiac fibrosis at one month [79]. Another 11,12-EET analog, UA-8 also improved cardiac ischemic tolerance in isolated mice hearts [80]. Like 11,12-EET analogs, the 14,15-EET analog, EET-A administered for two weeks can reduce cardiac hypertrophy [81]. Ventricular arrythmias following myocardial infarction in hypertensive rats are also reduced by EET-A treatment [81]. Cardiac hypertrophy in TGR rats was reduced by EET-A treatment which could be to a certain extent due to the anti-hypertensive actions [81]. More recently, TGR hypertensive rats with aorto-caval fistula induced congestive heart failure were treated with EET-A which improved survival [82]. The increased survival in this congestive heart failure was greatly improved by the combination of EET-A and angiotensin converting enzyme inhibition [82]. Another 14,15-EET analog, EET-B can reduce cardiac hypertrophy and fibrosis in the Dahl salt-sensitive hypertensive rat independent of blood pressure lowering [83]. EET-B also limited infarct size in rats subjected to regional ischemia in the heart in vivo [84]. The cardioprotective actions for EET-B extends to spontaneously hypertensive rats (SHR) subjected to 30 minutes of left coronary artery occlusion [84]. EET-B administered for several weeks did not lower blood pressure in SHR but improved cardiac fractional shortening, reduced cardiac inflammation, and decreased cardiac fibrosis [84]. These experimental findings demonstrate that 11,12-EET and 14,15-EET mimics can combat heart disease.

The mechanisms by which EET analogs decrease fibrosis and improve heart function has been investigated in cardiac ischemia reperfusion and other cardiac disease animal models [14,15,37]. Cardioprotective actions for EET analogs include improved mitochondrial function, anti-inflammatory, and anti-apoptotic actions [14,15]. EET analogs increase OPA1 oligomers and activate mitochondrial KATP channels in cardiac myocytes [39]. Anti-apoptotic actions for EET analogs are partially mediated by MAPK signaling activation to prevent ischemic cardiac cell stress [39]. Along these lines, EET-B was demonstrated to improve cardiac ischemic tolerance via hypoxia-inducible factor-1α (HIF-1α) [85]. These studies found that EET-B blunted the decrease in cadiomyocyte HIF-1α following reperfusion via downregulation of its degrading enzyme prolyl hydroxylase domain protein 3 (PHD3) [85]. This EET-B mediated change in HIF-α and PHD3 balance resulted in favoring the cardioprotective actions of HIF-1α [85]. Anti-inflammatory actions for EET-B are also responsible for improved cardiac function and attenuation of congestive heart failure in SHR following myocardial infarction [84]. Decreased cardiac CD68 positive macrophage/monocyte levels in SHR following myocardial infarction and treated with EET-B [84]. The EET-B anti-inflammatory actions could be partly attributed to the increase in cardiac HO-1 levels [84]. Future studies with EET analogs are needed to further examine cardiac cellular mechanisms responsible for improved cardiac function.

More recently combinations of sEH inhibitors, EET analogs, and renin-angiotensin system inhibitors have been evaluated for their ability to combat cardiac disease. TGR hypertensive rats were evaluated for cardiac hypertrophy and a subset subjected to acute ischemia reperfusion injury and treated with an sEH inhibitor, EET-A, or the combination [81]. Chronic administration of c-AUCB, EET-A or the combination decreased blood pressure and reduced ventricular fibrillation induced by myocardial infarction in the TGR hypertensive rats [81]. Importantly, the combination of c-AUCB and EET-A did not provide additive anti-hypertensive or cardioprotective actions [81]. Likewise, these treatments alone or in combination had similar actions to improve left ventricular function and slow the progression of heart failure in Hannover Sprague-Dawley but not in TGR hypertensive rats subjected to myocardial infarction [86]. Experimental findings in aorto-caval fistula induced congestive heart failure rats demonstrated differences between the combination of sEH inhibition and angiotensin converting enzyme inhibition and the combination of an EET analog and angiotensin converting enzyme inhibition [87]. The sEH inhibitor c-AUCB was given alone or in combination with the angiotensin converting enzyme inhibitor, trandolapril to TGR hypertensive rats with aorto-caval fistula induced congestive heart failure [87]. Intriguingly, c-AUCB in combination with trandolapril worsened the survival rate when compared to trandolapril alone in TGR hypertensive congestive heart failure rats [87]. In contrast, EET-A in combination with an angiotensin converting enzyme inhibitor improved survival to a greater extent than either treatment alone in TGR hypertensive congestive heart failure rats [87]. This combined EET-A and angiotensin converting enzyme inhibitor treatment had beneficial actions on cardiac morphology and left ventricular function [87]. Regrettably, these findings with combination treatments of sEH inhibitor, EET analog, and angiotensin converting enzyme inhibitor in hypertensive congestive heart failure rats do not provide strong evidence for additive actions to improve cardiac function.

Analogs mimicking EPA and DHA epoxy fatty acids have recently been synthesized and tested for their ability to decrease inflammation and combat cardiac diseases (Figure 3) [16,88,89]. The initial focus has been synthesizing EPA epoxylipid, 17,18-EEQ analogs [16,88]. Analogs of 17,18-EEQ were demonstrated to decrease ventricular tachyarrhythmias following coronary ligation in rats [90]. Additional studies demonstrated that 17,18-EEQ analogs can reduce atrial fibrillation in mice [91]. Further preclinical development of 17,18-EEQ analogs has led to clinical phase 1 trials in 2017 [16]. A third generation of 17,18-EEQ analogs have been synthesized and tested for cardioprotective actions [88]. This generation of 17,18-EEQ analogs demonstrated a similar action as previous generations to decrease ventricular tachyarrhythmias following coronary ligation [88]. Development of 16,17-EDP and 19,20-EDP analogs are in the initial stages and there has been initial testing for their ability to provide cardioprotection. Three novel 16,17-EDP and 19,20-EDP analogs were tested for their ability to improve cardiac function in isolated mice hearts exposed to ischemia reperfusion injury [53,89]. The 16,17-EDP analog, SA-26 improved postischemic cardiac function, reduced oxidative stress, preserved mitochondrial SIRT3 activity, and attenuated the NLPR3 inflammasome response [89]. Therefore, the cardiac therapeutic potential for EEQ and EDP analogs is promising; however, there remains much to be explored to fully understand the cardiac mechanisms of action to reach the ability to treat cardiac disease in humans.

4. Conclusions and Future Directions

The investigation of epoxylipids and their regulation by sEH in cardiac diseases has advanced from determining broad cardioprotective actions in myocardial infarction and progressive heart failure to determining cardiomyocyte cell signaling mechanisms that improve mitochondrial function, decrease oxidative stress, reduce inflammation, and oppose apoptosis. Subsequently, development of therapeutics to inhibit sEH and mimic epoxylipids have advanced to human clinical trials. Despite this outstanding progress, there is still much to be explored.

Novel pharmacological and genetic approaches to manipulate epoxylipids to combat heart disease are needed. Development of multi-target drugs that manipulate these epoxylipids could be beneficial for cardiac diseases. The concept for multi-target drugs to combat heart diseases is intriguing given the success of a combined neprilysin and angiotensin type 1 receptor blocker for heart failure [92]. Multi-target drugs that include sEH inhibitor activity have demonstrated to decrease inflammation and organ fibrosis in hypertension, kidney diseases, and metabolic diseases [9196]. These multi-target drugs have sEH inhibitor activity combined with inhibition against other fatty acid metabolizing enzymes or transcription factor agonists [93,95,96]. Positive cardiovascular actions for these sEH inhibitor multi-target include lowering blood pressure and decreasing plasma lipids [95,96]. In addition, sEH inhibitors with EET mimic activity have been developed but these compounds have not been extensively tested in cardiac diseases [37,97]. Thus, multi-target drugs that act to manipulate epoxylipids combined with another activity could provide a therapeutic approach for heart diseases.

Likewise, although significant progress has been made with 17,18-EEQ mimics, 16,17-EDP mimics and 19,20-EDP mimics are in their infancy. The precise molecular and protein target for EETs, 17,18-EEQ, 16,17- EDP, and 19,20-EDP have yet to be identified. Attractively, a G-protein receptor for another CYP hydroxylase metabolite, 20-HETE has been identified as GPR75 [98,99]. This 20-HETE receptor has been implicated in vascular function and cardiovascular disease [98]. There has been extensive characterization for EETs to identify G-protein coupled EET receptors [37]. EETs have been demonstrated to act through G-proteins and bind to cells and cell membranes with low and high affinity binding [100,101,102]. However, a protein receptor for EETs has yet to be identified. Receptor or target protein identification for epoxylipids would allow for target-based screening drug design approaches.

Combining genetic and metabolomic data in humans is another approach that could guide treatment by sEH inhibitors and epoxylipid mimics. There are known genetic EPHX2 polymorphisms in humans that associate with endothelial dysfunction and cardiovascular diseases [19,2124]. Testing for these genetic variants combined with measurements of epoxylipids could determine cardiac disease patients that would benefit from sEH inhibitors and epoxylipid mimics.

Ultimately, there has been significant progress with evaluating sEH and epoxylipids in heart diseases and the prospects that manipulation of epoxylipids can treat several heart diseases in humans is eminent.

Acknowledgment

The National Institute of Diabetes and Digestive and Kidney Diseases grant DK103616 provided support to John D. Imig. Jan Neckar was supported by grant of Czech Science Foundation (grant no. 18-03207S). Servier Medical Art was used to generate Figures 2 and 3 is licensed by Servier under a Creative Commons Attribution 3.0 Unported License.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest Statement

JDI has multiple patents and patent applications that cover the composition of matter for multi-target sEH inhibitors and epoxylipid mimics.

Declaration of Interests

John D. Imig – Several patents for soluble epoxide hydrolase inhibitors, EET analogs, and multi-target drugs. Licensed patents to Nephraegis Therapeutics and MetaSyn Therapeutics.

Ludek Cervenka – no interest to declare

Jan Neckar – no interest to declare

REFERENCES

  • 1.Townsend N, Kazakiewicz D, Lucy Wright F, Timmis A, Huculeci R, Torbica A, Gale CP, Achenbach S, Weidinger F, Vardas P. Epidemiology of cardiovascular disease in Europe. Nat Rev Cardiol. 2021. September 8. doi: 10.1038/s41569-021-00607-3. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 2.Saglietto A, Manfredi R, Elia E, D’Ascenzo F, DE Ferrari GM, Biondi-Zoccai G, Munzel T. Cardiovascular disease burden: Italian and global perspectives. Minerva Cardiol Angiol. 2021. June;69(3):231–240. doi: 10.23736/S2724-5683.21.05538-9. Epub 2021 Mar 11. [DOI] [PubMed] [Google Scholar]
  • 3.Francula-Zaninovic S, Nola IA. Management of Measurable Variable Cardiovascular Disease’ Risk Factors. Curr Cardiol Rev. 2018;14(3):153–163. doi: 10.2174/1573403X14666180222102312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.van Trier TJ, Mohammadnia N, Snaterse M, Peters RJG, Jørstad HT, Bax WA, Mackenbach JD. An appeal to our government for nationwide policies in the prevention of cardiovascular disease. Neth Heart J. 2021. October 4:1–5. doi: 10.1007/s12471-021-01628-w. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Garcia RA, Spertus JA. Using Patient-Reported Outcomes to Assess Healthcare Quality: Toward Better Measurement of Patient-Centered Care in Cardiovascular Disease. Methodist Debakey Cardiovasc J. 2021. April 5;17(1):e1–e9. doi: 10.14797/VUWD7697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Figtree GA, Broadfoot K, Casadei B, Califf R, Crea F, Drummond GR, Freedman JE, Guzik TJ, Harrison D, Hausenloy DJ, Hill JA, Januzzi JL, Kingwell BA, Lam CSP, MacRae CA, Misselwitz F, Miura T, Ritchie RH, Tomaszewski M, Wu JC, Xiao J, Zannad F. A Call to Action for New Global Approaches to Cardiovascular Disease Drug Solutions. Circulation. 2021. July 13;144(2):159–169. doi: 10.1161/CIR.0000000000000981. Epub 2021 Apr 20. [DOI] [PubMed] [Google Scholar]
  • 7.Paz Landim M, Cosenso-Martin LN, Polegati Santos A, Roma Uyemura JR, Barufi Fernandes L, da Silva Lopes V, Yugar-Toledo JC, Vilela-Martin JF. Predictive Factors for Target Organ Injuries in Hypertensive Individuals. Integr Blood Press Control. 2021. August 14;14:113–121. doi: 10.2147/IBPC.S324151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dahlöf B Cardiovascular disease risk factors: epidemiology and risk assessment. Am J Cardiol. 2010. January 4;105(1 Suppl):3A–9A. doi: 10.1016/j.amjcard.2009.10.007. [DOI] [PubMed] [Google Scholar]
  • 9.Versari D, Daghini E, Virdis A, Ghiadoni L, Taddei S. Endothelial dysfunction as a target for prevention of cardiovascular disease. Diabetes Care. 2009. November;32 Suppl 2(Suppl 2):S314–21. doi: 10.2337/dc09-S330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Heitzer T, Schlinzig T, Krohn K, Meinertz T, Münzel T. Endothelial dysfunction, oxidative stress, and risk of cardiovascular events in patients with coronary artery disease. Circulation. 2001. November 27;104(22):2673–8. doi: 10.1161/hc4601.099485. Erratum in: Circulation. 2003 Jul 29;108(4):500. [DOI] [PubMed] [Google Scholar]
  • 11.Tsutsui H, Kinugawa S, Matsushima S. Oxidative stress and heart failure. Am J Physiol Heart Circ Physiol. 2011. December;301(6):H2181–90. doi: 10.1152/ajpheart.00554.2011. Epub 2011 Sep 23. [DOI] [PubMed] [Google Scholar]
  • 12.Mei Y, Thompson MD, Cohen RA, Tong X. Autophagy and oxidative stress in cardiovascular diseases. Biochim Biophys Acta. 2015. February;1852(2):243–51. doi: 10.1016/j.bbadis.2014.05.005. Epub 2014 May 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Imig JD, Jankiewicz WK, Khan AH. Epoxy Fatty Acids: From Salt Regulation to Kidney and Cardiovascular Therapeutics: 2019 Lewis K. Dahl Memorial Lecture. Hypertension. 2020. July;76(1):3–15. doi: 10.1161/HYPERTENSIONAHA.120.13898. Epub 2020 Jun 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Jamieson KL, Endo T, Darwesh AM, Samokhvalov V, Seubert JM. Cytochrome P450-derived eicosanoids and heart function. Pharmacol Ther. 2017. November;179:47–83. doi: 10.1016/j.pharmthera.2017.05.005. Epub 2017 May 25. [DOI] [PubMed] [Google Scholar]
  • 15.Imig JD. Prospective for cytochrome P450 epoxygenase cardiovascular and renal therapeutics. Pharmacol Ther. 2018. December;192:1–19. doi: 10.1016/j.pharmthera.2018.06.015. Epub 2018 Jun 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Schunck WH, Konkel A, Fischer R, Weylandt KH. Therapeutic potential of omega-3 fatty acid-derived epoxyeicosanoids in cardiovascular and inflammatory diseases. Pharmacol Ther. 2018. March;183:177–204. doi: 10.1016/j.pharmthera.2017.10.016. Epub 2017 Nov 7. [DOI] [PubMed] [Google Scholar]
  • 17.Imig JD. Epoxides and soluble epoxide hydrolase in cardiovascular physiology. Physiol Rev. 2012. January;92(1):101–30. doi: 10.1152/physrev.00021.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Imig JD, Hammock BD. Soluble epoxide hydrolase as a therapeutic target for cardiovascular diseases. Nat Rev Drug Discov. 2009. October;8(10):794–805. doi: 10.1038/nrd2875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bellien J, Joannides R. Epoxyeicosatrienoic acid pathway in human health and diseases. J Cardiovasc Pharmacol. 2013. March;61(3):188–96. doi: 10.1097/FJC.0b013e318273b007. [DOI] [PubMed] [Google Scholar]
  • 20.Bellien J, Iacob M, Remy-Jouet I, Lucas D, Monteil C, Gutierrez L, Vendeville C, Dreano Y, Mercier A, Thuillez C, Joannides R. Epoxyeicosatrienoic acids contribute with altered nitric oxide and endothelin-1 pathways to conduit artery endothelial dysfunction in essential hypertension. Circulation. 2012. March 13;125(10):1266–75. doi: 10.1161/CIRCULATIONAHA.111.070680. [DOI] [PubMed] [Google Scholar]
  • 21.Aliwarga T, Evangelista EA, Sotoodehnia N, Lemaitre RN, Totah RA. Regulation of CYP2J2 and EET Levels in Cardiac Disease and Diabetes. Int J Mol Sci. 2018. June 29;19(7):1916. doi: 10.3390/ijms19071916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Akasaka T, Sueta D, Arima Y, Tabata N, Takashio S, Izumiya Y, Yamamoto E, Tsujita K, Kojima S, Kaikita K, Kajiwara A, Morita K, Oniki K, Saruwatari J, Nakagawa K, Hokimoto S. CYP2C19 variants and epoxyeicosatrienoic acids in patients with microvascular angina. Int J Cardiol Heart Vasc. 2017. April 12;15:15–20. doi: 10.1016/j.ijcha.2017.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Theken KN, Schuck RN, Edin ML, Tran B, Ellis K, Bass A, Lih FB, Tomer KB, Poloyac SM, Wu MC, Hinderliter AL, Zeldin DC, Stouffer GA, Lee CR. Evaluation of cytochrome P450-derived eicosanoids in humans with stable atherosclerotic cardiovascular disease. Atherosclerosis. 2012. June;222(2):530–6. doi: 10.1016/j.atherosclerosis.2012.03.022. Epub 2012 Mar 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Xu Y, Ding H, Peng J, Cui G, Liu L, Cianflone K, Wang DW. Association between polymorphisms of CYP2J2 and EPHX2 genes and risk of coronary artery disease. Pharmacogenet Genomics. 2011. August;21(8):489–94. doi: 10.1097/FPC.0b013e3283485eb2. [DOI] [PubMed] [Google Scholar]
  • 25.Polonikov A, Bykanova M, Ponomarenko I, Sirotina S, Bocharova A, Vagaytseva K, Stepanov V, Churnosov M, Bushueva O, Solodilova M, Shvetsov Y, Ivanov V. The contribution of CYP2C gene subfamily involved in epoxygenase pathway of arachidonic acids metabolism to hypertension susceptibility in Russian population. Clin Exp Hypertens. 2017;39(4):306–311. doi: 10.1080/10641963.2016.1246562. Epub 2017 May 17. [DOI] [PubMed] [Google Scholar]
  • 26.Wu SN, Zhang Y, Gardner CO, Chen Q, Li Y, Wang GL, Gao PJ, Zhu DL. Evidence for association of polymorphisms in CYP2J2 and susceptibility to essential hypertension. Ann Hum Genet. 2007. July;71(Pt 4):519–25. doi: 10.1111/j.1469-1809.2007.00346.x. Epub 2007 Feb 5. [DOI] [PubMed] [Google Scholar]
  • 27.Ward NC, Croft KD, Blacker D, Hankey GJ, Barden A, Mori TA, Puddey IB, Beer CD. Cytochrome P450 metabolites of arachidonic acid are elevated in stroke patients compared with healthy controls. Clin Sci (Lond). 2011. December;121(11):501–7. doi: 10.1042/CS20110215. [DOI] [PubMed] [Google Scholar]
  • 28.Monti J, Fischer J, Paskas S, Heinig M, Schulz H, Gösele C, Heuser A, Fischer R, Schmidt C, Schirdewan A, Gross V, Hummel O, Maatz H, Patone G, Saar K, Vingron M, Weldon SM, Lindpaintner K, Hammock BD, Rohde K, Dietz R, Cook SA, Schunck WH, Luft FC, Hubner N. Soluble epoxide hydrolase is a susceptibility factor for heart failure in a rat model of human disease. Nat Genet. 2008. May;40(5):529–37. doi: 10.1038/ng.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhao X, Pollock DM, Inscho EW, Zeldin DC, Imig JD. Decreased renal cytochrome P450 2C enzymes and impaired vasodilation are associated with angiotensin salt-sensitive hypertension. Hypertension. 2003. March;41(3 Pt 2):709–14. doi: 10.1161/01.HYP.0000047877.36743.FA. Epub 2002 Dec 16. [DOI] [PubMed] [Google Scholar]
  • 30.Imig JD, Zhao X, Capdevila JH, Morisseau C, Hammock BD. Soluble epoxide hydrolase inhibition lowers arterial blood pressure in angiotensin II hypertension. Hypertension. 2002. February;39(2 Pt 2):690–4. doi: 10.1161/hy0202.103788. [DOI] [PubMed] [Google Scholar]
  • 31.Jung O, Brandes RP, Kim IH, Schweda F, Schmidt R, Hammock BD, Busse R, Fleming I. Soluble epoxide hydrolase is a main effector of angiotensin II-induced hypertension. Hypertension. 2005. April;45(4):759–65. doi: 10.1161/01.HYP.0000153792.29478.1d. Epub 2005 Feb 7. [DOI] [PubMed] [Google Scholar]
  • 32.Lee CR, Imig JD, Edin ML, Foley J, DeGraff LM, Bradbury JA, Graves JP, Lih FB, Clark J, Myers P, Perrow AL, Lepp AN, Kannon MA, Ronnekleiv OK, Alkayed NJ, Falck JR, Tomer KB, Zeldin DC. Endothelial expression of human cytochrome P450 epoxygenases lowers blood pressure and attenuates hypertension-induced renal injury in mice. FASEB J. 2010. October;24(10):3770–81. doi: 10.1096/fj.10-160119. Epub 2010 May 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Seubert JM, Sinal CJ, Graves J, DeGraff LM, Bradbury JA, Lee CR, Goralski K, Carey MA, Luria A, Newman JW, Hammock BD, Falck JR, Roberts H, Rockman HA, Murphy E, Zeldin DC. Role of soluble epoxide hydrolase in postischemic recovery of heart contractile function. Circ Res. 2006. August 18;99(4):442–50. doi: 10.1161/01.RES.0000237390.92932.37. Epub 2006 Jul 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zhou C, Huang J, Li Q, Zhan C, Xu X, Zhang X, Ai D, Zhu Y, Wen Z, Wang DW. CYP2J2-derived EETs attenuated ethanol-induced myocardial dysfunction through inducing autophagy and reducing apoptosis. Free Radic Biol Med. 2018. March;117:168–179. doi: 10.1016/j.freeradbiomed.2018.02.009. Epub 2018 Feb 8. [DOI] [PubMed] [Google Scholar]
  • 35.Yang L, Ni L, Duan Q, Wang X, Chen C, Chen S, Chaugai S, Zeldin DC, Tang JR, Wang DW. CYP epoxygenase 2J2 prevents cardiac fibrosis by suppression of transmission of pro-inflammation from cardiomyocytes to macrophages. Prostaglandins Other Lipid Mediat. 2015. Jan-Mar;116–117:64–75. doi: 10.1016/j.prostaglandins.2015.01.004. Epub 2015 Feb 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Manhiani M, Quigley JE, Knight SF, Tasoobshirazi S, Moore T, Brands MW, Hammock BD, Imig JD. Soluble epoxide hydrolase gene deletion attenuates renal injury and inflammation with DOCA-salt hypertension. Am J Physiol Renal Physiol. 2009. September;297(3):F740–8. doi: 10.1152/ajprenal.00098.2009. Epub 2009 Jun 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Campbell WB, Imig JD, Schmitz JM, Falck JR. Orally Active Epoxyeicosatrienoic Acid Analogs. J Cardiovasc Pharmacol. 2017. October;70(4):211–224. doi: 10.1097/FJC.0000000000000523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Seubert JM, Zeldin DC, Nithipatikom K, Gross GJ. Role of epoxyeicosatrienoic acids in protecting the myocardium following ischemia/reperfusion injury. Prostaglandins Other Lipid Mediat. 2007. January;82(1–4):50–9. doi: 10.1016/j.prostaglandins.2006.05.017. Epub 2006 Jul 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.El-Sikhry HE, Alsaleh N, Dakarapu R, Falck JR, Seubert JM. Novel Roles of Epoxyeicosanoids in Regulating Cardiac Mitochondria. PLoS One. 2016. August 5;11(8):e0160380. doi: 10.1371/journal.pone.0160380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gross GJ, Hsu A, Pfeiffer AW, Nithipatikom K. Roles of endothelial nitric oxide synthase (eNOS) and mitochondrial permeability transition pore (MPTP) in epoxyeicosatrienoic acid (EET)-induced cardioprotection against infarction in intact rat hearts. J Mol Cell Cardiol. 2013. June;59:20–9. doi: 10.1016/j.yjmcc.2013.02.003. Epub 2013 Feb 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chaudhary KR, Cho WJ, Yang F, Samokhvalov V, El-Sikhry HE, Daniel EE, Seubert JM. Effect of ischemia reperfusion injury and epoxyeicosatrienoic acids on caveolin expression in mouse myocardium. J Cardiovasc Pharmacol. 2013. March;61(3):258–63. doi 10.1097/FJC.0b013e31827afcee. [DOI] [PubMed] [Google Scholar]
  • 42.Katragadda D, Batchu SN, Cho WJ, Chaudhary KR, Falck JR, Seubert JM. Epoxyeicosatrienoic acids limit damage to mitochondrial function following stress in cardiac cells. J Mol Cell Cardiol. 2009. June;46(6):867–75. doi: 10.1016/j.yjmcc.2009.02.028. Epub 2009 Mar 12. [DOI] [PubMed] [Google Scholar]
  • 43.Batchu SN, Chaudhary KR, El-Sikhry H, Yang W, Light PE, Oudit GY, Seubert JM. Role of PI3Kα and sarcolemmal ATP-sensitive potassium channels in epoxyeicosatrienoic acid mediated cardioprotection. J Mol Cell Cardiol. 2012. July;53(1):43–52. doi: 10.1016/j.yjmcc.2012.04.008. Epub 2012 Apr 27. [DOI] [PubMed] [Google Scholar]
  • 44.Dhanasekaran A, Gruenloh SK, Buonaccorsi JN, Zhang R, Gross GJ, Falck JR, Patel PK, Jacobs ER, Medhora M. Multiple antiapoptotic targets of the PI3K/Akt survival pathway are activated by epoxyeicosatrienoic acids to protect cardiomyocytes from hypoxia/anoxia. Am J Physiol Heart Circ Physiol. 2008. February;294(2):H724–35. doi: 10.1152/ajpheart.00979.2007. Epub 2007 Nov 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dai M, Wu L, He Z, Zhang S, Chen C, Xu X, Wang P, Gruzdev A, Zeldin DC, Wang DW. Epoxyeicosatrienoic acids regulate macrophage polarization and prevent LPS-induced cardiac dysfunction. J Cell Physiol. 2015. September;230(9):2108–19. doi: 10.1002/jcp.24939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Oppedisano F, Macrì R, Gliozzi M, Musolino V, Carresi C, Maiuolo J, Bosco F, Nucera S, Caterina Zito M, Guarnieri L, Scarano F, Nicita C, Coppoletta AR, Ruga S, Scicchitano M, Mollace R, Palma E, Mollace V. The Anti-Inflammatory and Antioxidant Properties of n-3 PUFAs: Their Role in Cardiovascular Protection. Biomedicines. 2020. August 25;8(9):306. doi: 10.3390/biomedicines8090306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Breslow JL. n-3 fatty acids and cardiovascular disease. Am J Clin Nutr. 2006. June;83(6 Suppl):1477S–1482S. doi: 10.1093/ajcn/83.6.1477S. [DOI] [PubMed] [Google Scholar]
  • 48.Macartney MJ, Peoples GE, McLennan PL. Cardiac contractile dysfunction, during and following ischaemia, is attenuated by low-dose dietary fish oil in rats. Eur J Nutr. 2021. June 13. doi: 10.1007/s00394-021-02608-x. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 49.Hsu HC, Chen CY, Chen MF. N-3 polyunsaturated fatty acids decrease levels of doxorubicin-induced reactive oxygen species in cardiomyocytes -- involvement of uncoupling protein UCP2. J Biomed Sci. 2014. November 18;21(1):101. doi: 10.1186/s12929-014-0101-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Engelbrecht AM, Engelbrecht P, Genade S, Niesler C, Page C, Smuts M, Lochner A. Long-chain polyunsaturated fatty acids protect the heart against ischemia/reperfusion-induced injury via a MAPK dependent pathway. J Mol Cell Cardiol. 2005. December;39(6):940–54. doi: 10.1016/j.yjmcc.2005.08.004. Epub 2005 Oct 10. [DOI] [PubMed] [Google Scholar]
  • 51.Samokhvalov V, Jamieson KL, Vriend J, Quan S, Seubert JM. CYP-epoxygenase metabolites of docosahexaenoic acid protect HL-1 cardiac cells against LPS-induced cytotoxicity Through SIRT1. Cell Death Discov. 2015. November 23;1:15054–. doi: 10.1038/cddiscovery.2015.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sergiel JP, Martine L, Raederstorff D, Grynberg A, Demaison L. Individual effects of dietary EPA and DHA on the functioning of the isolated working rat heart. Can J Physiol Pharmacol. 1998. Jul-Aug;76(7–8):728–36. doi: 10.1139/cjpp-76-7-8-728. [DOI] [PubMed] [Google Scholar]
  • 53.Darwesh AM, Jamieson KL, Wang C, Samokhvalov V, Seubert JM. Cardioprotective effects of CYP-derived epoxy metabolites of docosahexaenoic acid involve limiting NLRP3 inflammasome activation 1. Can J Physiol Pharmacol. 2019. June;97(6):544–556. doi: 10.1139/cjpp-2018-0480. Epub 2018 Oct 16. [DOI] [PubMed] [Google Scholar]
  • 54.Chen J, Shearer GC, Chen Q, Healy CL, Beyer AJ, Nareddy VB, Gerdes AM, Harris WS, O’Connell TD, Wang D. Omega-3 fatty acids prevent pressure overload-induced cardiac fibrosis through activation of cyclic GMP/protein kinase G signaling in cardiac fibroblasts. Circulation. 2011. February 15;123(6):584–93. doi: 10.1161/CIRCULATIONAHA.110.971853. Epub 2011 Jan 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Eclov JA, Qian Q, Redetzke R, Chen Q, Wu SC, Healy CL, Ortmeier SB, Harmon E, Shearer GC, O’Connell TD. EPA, not DHA, prevents fibrosis in pressure overload-induced heart failure: potential role of free fatty acid receptor 4. J Lipid Res. 2015. December;56(12):2297–308. doi: 10.1194/jlr.M062034. Epub 2015 Oct 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Takamura M, Kurokawa K, Ootsuji H, Inoue O, Okada H, Nomura A, Kaneko S, Usui S. Long-Term Administration of Eicosapentaenoic Acid Improves Post-Myocardial Infarction Cardiac Remodeling in Mice by Regulating Macrophage Polarization. J Am Heart Assoc. 2017. February 21;6(2):e004560. doi: 10.1161/JAHA.116.004560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tułowiecka N, Kotlęga D, Prowans P, Szczuko M. The Role of Resolvins: EPA and DHA Derivatives Can Be Useful in the Prevention and Treatment of Ischemic Stroke. Int J Mol Sci. 2020. October 15;21(20):7628. doi: 10.3390/ijms21207628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Nelson JR, Raskin S. The eicosapentaenoic acid:arachidonic acid ratio and its clinical utility in cardiovascular disease. Postgrad Med. 2019 May;131(4):268–277. doi: 10.1080/00325481.2019.1607414. Epub 2019 May 7. [DOI] [PubMed] [Google Scholar]
  • 59.Takahashi M, Ando J, Shimada K, Nishizaki Y, Tani S, Ogawa T, Yamamoto M, Nagao K, Hirayama A, Yoshimura M, Daida H, Nagai R, Komuro I. The ratio of serum n-3 to n-6 polyunsaturated fatty acids is associated with diabetes mellitus in patients with prior myocardial infarction: a multicenter cross-sectional study. BMC Cardiovasc Disord. 2017. January 26;17(1):41. doi: 10.1186/s12872-017-0479-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Xu D, Li N, He Y, Timofeyev V, Lu L, Tsai HJ, Kim IH, Tuteja D, Mateo RK, Singapuri A, Davis BB, Low R, Hammock BD, Chiamvimonvat N. Prevention and reversal of cardiac hypertrophy by soluble epoxide hydrolase inhibitors. Proc Natl Acad Sci U S A. 2006. December 5;103(49):18733–8. doi: 10.1073/pnas.0609158103. Epub 2006 Nov 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Roche C, Besnier M, Cassel R, Harouki N, Coquerel D, Guerrot D, Nicol L, Loizon E, Remy-Jouet I, Morisseau C, Mulder P, Ouvrard-Pascaud A, Madec AM, Richard V, Bellien J. Soluble epoxide hydrolase inhibition improves coronary endothelial function and prevents the development of cardiac alterations in obese insulin-resistant mice. Am J Physiol Heart Circ Physiol. 2015. May 1;308(9):H1020–9. doi: 10.1152/ajpheart.00465.2014. Epub 2015 Feb 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Darwesh AM, Keshavarz-Bahaghighat H, Jamieson KL, Seubert JM. Genetic Deletion or Pharmacological Inhibition of Soluble Epoxide Hydrolase Ameliorates Cardiac Ischemia/Reperfusion Injury by Attenuating NLRP3 Inflammasome Activation. Int J Mol Sci. 2019. July 17;20(14):3502. doi: 10.3390/ijms20143502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Islam O, Patil P, Goswami SK, Razdan R, Inamdar MN, Rizwan M, Mathew J, Inceoglu B, Stephen Lee KS, Hwang SH, Hammock BD. Inhibitors of soluble epoxide hydrolase minimize ischemiareperfusion-induced cardiac damage in normal, hypertensive, and diabetic rats. Cardiovasc Ther. 2017. June;35(3):10.1111/1755–5922.12259. doi: 10.1111/1755-5922.12259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gross GJ, Gauthier KM, Moore J, Falck JR, Hammock BD, Campbell WB, Nithipatikom K. Effects of the selective EET antagonist, 14,15-EEZE, on cardioprotection produced by exogenous or endogenous EETs in the canine heart. Am J Physiol Heart Circ Physiol. 2008. June;294(6):H2838–44. doi: 10.1152/ajpheart.00186.2008. Epub 2008 Apr 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Akhnokh MK, Yang FH, Samokhvalov V, Jamieson KL, Cho WJ, Wagg C, Takawale A, Wang X, Lopaschuk GD, Hammock BD, Kassiri Z, Seubert JM. Inhibition of Soluble Epoxide Hydrolase Limits Mitochondrial Damage and Preserves Function Following Ischemic Injury. Front Pharmacol. 2016. June 7;7:133. doi: 10.3389/fphar.2016.00133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chaudhary KR, Abukhashim M, Hwang SH, Hammock BD, Seubert JM. Inhibition of soluble epoxide hydrolase by trans-4- [4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid is protective against ischemia-reperfusion injury. J Cardiovasc Pharmacol. 2010. January;55(1):67–73. doi: 10.1097/FJC.0b013e3181c37d69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Merkel MJ, Liu L, Cao Z, Packwood W, Young J, Alkayed NJ, Van Winkle DM. Inhibition of soluble epoxide hydrolase preserves cardiomyocytes: role of STAT3 signaling. Am J Physiol Heart Circ Physiol. 2010. February;298(2):H679–87. doi: 10.1152/ajpheart.00533.2009. Epub 2009 Dec 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gui Y, Chen J, Hu J, Liao C, Ouyang M, Deng L, Yang J, Xu D. Soluble epoxide hydrolase inhibitors improve angiogenic function of endothelial progenitor cells via ERK/p38-mediated miR-126 upregulation in myocardial infarction mice after exercise. Exp Cell Res. 2020. December 15;397(2):112360. doi: 10.1016/j.yexcr.2020.112360. Epub 2020 Nov 12. [DOI] [PubMed] [Google Scholar]
  • 69.Batchu SN, Lee SB, Samokhvalov V, Chaudhary KR, El-Sikhry H, Weldon SM, Seubert JM. Novel soluble epoxide hydrolase inhibitor protects mitochondrial function following stress. Can J Physiol Pharmacol. 2012. June;90(6):811–23. doi: 10.1139/y2012-082. Epub 2012 May 24. [DOI] [PubMed] [Google Scholar]
  • 70.Oni-Orisan A, Alsaleh N, Lee CR, Seubert JM. Epoxyeicosatrienoic acids and cardioprotection: the road to translation. J Mol Cell Cardiol. 2014. September;74:199–208. doi: 10.1016/j.yjmcc.2014.05.016. Epub 2014 Jun 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Li N, Liu JY, Timofeyev V, Qiu H, Hwang SH, Tuteja D, Lu L, Yang J, Mochida H, Low R, Hammock BD, Chiamvimonvat N. Beneficial effects of soluble epoxide hydrolase inhibitors in myocardial infarction model: Insight gained using metabolomic approaches. J Mol Cell Cardiol. 2009. December;47(6):835–45. doi: 10.1016/j.yjmcc.2009.08.017. Epub 2009 Aug 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kompa AR, Wang BH, Xu G, Zhang Y, Ho PY, Eisennagel S, Thalji RK, Marino JP Jr, Kelly DJ, Behm DJ, Krum H. Soluble epoxide hydrolase inhibition exerts beneficial anti-remodeling actions post-myocardial infarction. Int J Cardiol. 2013. July 15;167(1):210–9. doi: 10.1016/j.ijcard.2011.12.062. Epub 2012 Jan 10. [DOI] [PubMed] [Google Scholar]
  • 73.Sirish P, Li N, Liu JY, Lee KS, Hwang SH, Qiu H, Zhao C, Ma SM, López JE, Hammock BD, Chiamvimonvat N. Unique mechanistic insights into the beneficial effects of soluble epoxide hydrolase inhibitors in the prevention of cardiac fibrosis. Proc Natl Acad Sci U S A. 2013. April 2;110(14):5618–23. doi: 10.1073/pnas.1221972110. Epub 2013 Mar 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sirish P, Li N, Timofeyev V, Zhang XD, Wang L, Yang J, Lee KS, Bettaieb A, Ma SM, Lee JH, Su D, Lau VC, Myers RE, Lieu DK, López JE, Young JN, Yamoah EN, Haj F, Ripplinger CM, Hammock BD, Chiamvimonvat N. Molecular Mechanisms and New Treatment Paradigm for Atrial Fibrillation. Circ Arrhythm Electrophysiol. 2016. May;9(5):10.1161/CIRCEP.115.003721 e003721. doi: 10.1161/CIRCEP.115.003721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Neckář J, Kopkan L, Husková Z, Kolář F, Papoušek F, Kramer HJ, Hwang SH, Hammock BD, Imig JD, Malý J, Netuka I, Ošťádal B, Červenka L. Inhibition of soluble epoxide hydrolase by cis-4-[4-(3-adamantan-1-ylureido)cyclohexyl-oxy]benzoic acid exhibits antihypertensive and cardioprotective actions in transgenic rats with angiotensin II-dependent hypertension. Clin Sci (Lond). 2012. June;122(11):513–25. doi: 10.1042/CS20110622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Vacková Š, Kopkan L, Kikerlová S, Husková Z, Sadowski J, Kompanowska-Jezierska E, Hammock BD, Imig JD, Táborský M, Melenovský V, Červenka L. Pharmacological Blockade of Soluble Epoxide Hydrolase Attenuates the Progression of Congestive Heart Failure Combined With Chronic Kidney Disease: Insights From Studies With Fawn-Hooded Hypertensive Rats. Front Pharmacol. 2019. January 23;10:18. doi: 10.3389/fphar.2019.00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zhang H, Zhang K, Liang J, Yan W, Wu F, Xu W, Wu Z, Chen Y, Pan R, Wu G. Soluble epoxide hydrolase inhibitor, TUPS, attenuates isoproterenol/angiotensin II-induced cardiac hypertrophy through mammalian target of rapamycin-mediated autophagy inhibition. J Pharm Pharmacol. 2019. August;71(8):1291–1300. doi: 10.1111/jphp.13113. Epub 2019 Jun 18. [DOI] [PubMed] [Google Scholar]
  • 78.Červenka L, Melenovský V, Husková Z, Sporková A, Bürgelová M, Škaroupková P, Hwang SH, Hammock BD, Imig JD, Sadowski J. Inhibition of soluble epoxide hydrolase does not improve the course of congestive heart failure and the development of renal dysfunction in rats with volume overload induced by aorto-caval fistula. Physiol Res. 2015;64(6):857–73. doi: 10.33549/physiolres.932977. Epub 2015 Jun 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cao J, Tsenovoy PL, Thompson EA, Falck JR, Touchon R, Sodhi K, Rezzani R, Shapiro JI, Abraham NG. Agonists of epoxyeicosatrienoic acids reduce infarct size and ameliorate cardiac dysfunction via activation of HO-1 and Wnt1 canonical pathway. Prostaglandins Other Lipid Mediat. 2015. Jan-Mar;116–117:76–86. doi: 10.1016/j.prostaglandins.2015.01.002. Epub 2015 Feb 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Batchu SN, Lee SB, Qadhi RS, Chaudhary KR, El-Sikhry H, Kodela R, Falck JR, Seubert JM. Cardioprotective effect of a dual acting epoxyeicosatrienoic acid analogue towards ischaemia reperfusion injury. Br J Pharmacol. 2011. February;162(4):897–907. doi 10.1111/j.1476-5381.2010.01093.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Červenka L, Husková Z, Kopkan L, Kikerlová S, Sedláková L, Vaňourková Z, Alánová P, Kolář F, Hammock BD, Hwang SH, Imig JD, Falck JR, Sadowski J, Kompanowska-Jezierska E, Neckář J. Two pharmacological epoxyeicosatrienoic acid-enhancing therapies are effectively antihypertensive and reduce the severity of ischemic arrhythmias in rats with angiotensin II-dependent hypertension. J Hypertens. 2018. June;36(6):1326–1341. doi: 10.1097/HJH.0000000000001708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kala P, Miklovič M, Jíchová Š, Škaroupková P, Vaňourková Z, Maxová H, Gawrys O, Kompanowska-Jezierska E, Sadowski J, Imig JD, Falck JR, Veselka J, Červenka L, Aiglová R, Vícha M, Gloger V, Táborský M. Effects of Epoxyeicosatrienoic Acid-Enhancing Therapy on the Course of Congestive Heart Failure in Angiotensin II-Dependent Rat Hypertension: From mRNA Analysis towards Functional In Vivo Evaluation. Biomedicines. 2021. August 20;9(8):1053. doi: 10.3390/biomedicines9081053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hye Khan MA, Neckár J, Manthati V, Errabelli R, Pavlov TS, Staruschenko A, Falck JR, Imig JD. Orally active epoxyeicosatrienoic acid analog attenuates kidney injury in hypertensive Dahl salt-sensitive rat. Hypertension. 2013. November;62(5):905–13. doi: 10.1161/HYPERTENSIONAHA.113.01949. Epub 2013 Aug 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Neckář J, Hye Khan MA, Gross GJ, Cyprová M, Hrdlička J, Kvasilová A, Falck JR, Campbell WB, Sedláková L, Škutová Š, Olejníčková V, Gregorovičová M, Sedmera D, Kolář F, Imig JD. Epoxyeicosatrienoic acid analog EET-B attenuates post-myocardial infarction remodeling in spontaneously hypertensive rats. Clin Sci (Lond). 2019. April 29;133(8):939–951. doi: 10.1042/CS20180728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.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. November 1;315(5):H1148–H1158. doi: 10.1152/ajpheart.00726.2017. Epub 2018 Aug 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.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. March 1;10:159. doi: 10.3389/fphar.2019.00159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kala P, Sedláková L, Škaroupková P, Kopkan L, Vaňourková Z, Táborský M, Nishiyama A, Hwang SH, Hammock BD, Sadowski J, Melenovský V, Imig JD, Červenka L. Effect of angiotensin-converting enzyme blockade, alone or combined with blockade of soluble epoxide hydrolase, on the course of congestive heart failure and occurrence of renal dysfunction in Ren-2 transgenic hypertensive rats with aorto-caval fistula. Physiol Res. 2018. July 17;67(3):401–415. doi: 10.33549/physiolres.933757. Epub 2018 Mar 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Adebesin AM, Wesser T, Vijaykumar J, Konkel A, Paudyal MP, Lossie J, Zhu C, Westphal C, Puli N, Fischer R, Schunck WH, Falck JR. Development of Robust 17(R),18(S)-Epoxyeicosatetraenoic Acid (17,18-EEQ) Analogues as Potential Clinical Antiarrhythmic Agents. J Med Chem. 2019. November 27;62(22):10124–10143. doi: 10.1021/acs.jmedchem.9b00952. Epub 2019 Nov 19. [DOI] [PubMed] [Google Scholar]
  • 89.Darwesh AM, Bassiouni W, Adebesin AM, Mohammad AS, Falck JR, Seubert JM. A Synthetic Epoxydocosapentaenoic Acid Analogue Ameliorates Cardiac Ischemia/Reperfusion Injury: The Involvement of the Sirtuin 3-NLRP3 Pathway. Int J Mol Sci. 2020. July 24;21(15):5261. doi: 10.3390/ijms21155261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Schunck WH, Wallukat G, Fischer R, Schmidt C, Müller DN, Puli N, Falck JR. Novel eicosanoid derivatives. 2010. (WO 2010/081683).
  • 91.Fischer R, Konkel A, Wesser T, Westphal P, Schunck WH, Westphal C, Falck JR. Metabolically robust analogs of Cyp-eicosanoids for the treatment of cardiac disease. 2017. (WO 2017/013265).
  • 92.Gallo G, Volpe M, Battistoni A, Russo D, Tocci G, Musumeci MB. Sacubitril/Valsartan as a Therapeutic Tool Across the Range of Heart Failure Phenotypes and Ejection Fraction Spectrum. Front Physiol. 2021. August 23;12:652163. doi: 10.3389/fphys.2021.652163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Lillich FF, Imig JD, Proschak E. Multi-Target Approaches in Metabolic Syndrome. Front Pharmacol. 2021. March 12;11:554961. doi: 10.3389/fphar.2020.554961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hye Khan MA, Hwang SH, Sharma A, Corbett JA, Hammock BD, Imig JD. A dual COX-2/sEH inhibitor improves the metabolic profile and reduces kidney injury in Zucker diabetic fatty rat. Prostaglandins Other Lipid Mediat. 2016. September;125:40–7. doi: 10.1016/j.prostaglandins.2016.07.003. Epub 2016 Jul 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Hye Khan MA, Kolb L, Skibba M, Hartmann M, Blöcher R, Proschak E, Imig JD. A novel dual PPAR-γ agonist/sEH inhibitor treats diabetic complications in a rat model of type 2 diabetes. Diabetologia. 2018. October;61(10):2235–2246. doi: 10.1007/s00125-018-4685-0. Epub 2018 Jul 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Stavniichuk A, Hye Khan MA, Yeboah MM, Chesnik MA, Jankiewicz WK, Hartmann M, Blöcher R, Kircher T, Savchuk O, Proschak E, Imig JD. Dual soluble epoxide hydrolase inhibitor/PPAR-γ agonist attenuates renal fibrosis. Prostaglandins Other Lipid Mediat. 2020. October;150:106472. doi: 10.1016/j.prostaglandins.2020.106472. Epub 2020 Jun 20. [DOI] [PubMed] [Google Scholar]
  • 97.Falck JR, Koduru SR, Mohapatra S, Manne R, Atcha KR, Atcha R, Manthati VL, Capdevila JH, Christian S, Imig JD, Campbell WB. 14,15-Epoxyeicosa-5,8,11-trienoic Acid (14,15-EET) surrogates: carboxylate modifications. J Med Chem. 2014. August 28;57(16):6965–72. doi: 10.1021/jm500262m. Epub 2014 Aug 19. Erratum in: J Med Chem. 2014 Nov 13;57(21):9218. Atcha, Krishnam Raju [added]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Garcia V, Gilani A, Shkolnik B, Pandey V, Zhang FF, Dakarapu R, Gandham SK, Reddy NR, Graves JP, Gruzdev A, Zeldin DC, Capdevila JH, Falck JR, Schwartzman ML. 20-HETE Signals Through G-Protein-Coupled Receptor GPR75 (Gq) to Affect Vascular Function and Trigger Hypertension. Circ Res. 2017. May 26;120(11):1776–1788. doi: 10.1161/CIRCRESAHA.116.310525. Epub 2017 Mar 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Pascale JV, Park EJ, Adebesin AM, Falck JR, Schwartzman ML, Garcia V. Uncovering the signalling, structure and function of the 20-HETE-GPR75 pairing: Identifying the chemokine CCL5 as a negative regulator of GPR75. Br J Pharmacol. 2021. September;178(18):3813–3828. doi: 10.1111/bph.15525. Epub 2021 Jun 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wong PY, Lai PS, Falck JR. Mechanism and signal transduction of 14 (R), 15 (S)-epoxyeicosatrienoic acid (14,15-EET) binding in guinea pig monocytes. Prostaglandins Other Lipid Mediat. 2000. October;62(4):321–33. doi: 10.1016/s0090-6980(00)00079-4. [DOI] [PubMed] [Google Scholar]
  • 101.Yang W, Holmes BB, Gopal VR, Kishore RV, Sangras B, Yi XY, Falck JR, Campbell WB. Characterization of 14,15-epoxyeicosatrienoyl-sulfonamides as 14,15-epoxyeicosatrienoic acid agonists: use for studies of metabolism and ligand binding. J Pharmacol Exp Ther. 2007. June;321(3):1023–31. doi: 10.1124/jpet.107.119651. Epub 2007 Feb 27. [DOI] [PubMed] [Google Scholar]
  • 102.Yang W, Tuniki VR, Anjaiah S, Falck JR, Hillard CJ, Campbell WB. Characterization of epoxyeicosatrienoic acid binding site in U937 membranes using a novel radiolabeled agonist, 20–125i-14,15-epoxyeicosa-8(Z)-enoic acid. J Pharmacol Exp Ther. 2008. March;324(3):1019–27. doi: 10.1124/jpet.107.129577. Epub 2008 Jan 2. [DOI] [PubMed] [Google Scholar]

RESOURCES