Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Gene 526 (2013) 61–74

Contents lists available at SciVerse ScienceDirect

Gene
journal homepage: www.elsevier.com/locate/gene

Review

Soluble epoxide hydrolase: Gene structure, expression and deletion


Todd R. Harris, Bruce D. Hammock ⁎
Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA, USA

a r t i c l e i n f o a b s t r a c t

Article history: Mammalian soluble epoxide hydrolase (sEH) converts epoxides to their corresponding diols through the addi-
Accepted 9 May 2013 tion of a water molecule. sEH readily hydrolyzes lipid signaling molecules, including the epoxyeicosatrienoic
Available online 20 May 2013 acids (EETs), epoxidized lipids produced from arachidonic acid by the action of cytochrome p450s. Through its
metabolism of the EETs and other lipid mediators, sEH contributes to the regulation of vascular tone, nociception,
Keywords:
angiogenesis and the inflammatory response. Because of its central physiological role in disease states such as
EPHX2
Epoxyeicosatrienoic acid
cardiac hypertrophy, diabetes, hypertension, and pain sEH is being investigated as a therapeutic target. This
Lipid signaling review begins with a brief introduction to sEH protein structure and function. sEH evolution and gene structure
Inflammation are then discussed before human small nucleotide polymorphisms and mammalian gene expression are
Hypertension described in the context of several disease models. The review ends with an overview of studies that have
employed the sEH knockout mouse model.
© 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62
2. sEH protein structure and catalytic activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62
3. Gene evolution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
4. Mammalian gene structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
5. Physiological role of sEH in mammals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6. Single nucleotide polymorphisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
7. Mammalian gene expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
7.1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
7.2. Subcellular localization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
7.3. sEH, PPAR agonists, and angiotensin-II . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
7.4. Vasculature . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
7.5. Heart . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
7.6. Kidney . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
7.7. Liver . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
7.8. Brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
7.9. Lung and pulmonary arteries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
7.10. Fat . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
7.11. Inflammatory system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
7.12. Reproductive system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
7.13. Neoplasms and tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69

Abbreviations: 20-HETE, 20-hydroxyeicosatetraenoic acid; Ang-II, angiotensin II; AP-1, activator protein 1; ARIC, Atherosclerosis Risk in Communities; ATF-6, activating tran-
scription factor-6; CAC, coronary artery calcification; CARDIA, Coronary Artery Risk Development in Young Adults; ChIP, chromatin immunoprecipitation; CPR, cardiopulmonary
resuscitation; DHA, docosahexaenoic acid; DHETs, dihydroxyeicosatrienoic acids; EETs, epoxyeicosatrienoic acids; eNOS, endothelial nitric oxide synthase; EPA, eicosapentaenoic
acid; EpFAs, epoxy-fatty acids; FLAP, 5-lipoxygenase activation protein; GSIS, glucose-stimulated insulin secretion; Hcy, homocysteine; HUVECs, human umbilical vein endothelial
cells; IBD, inflammatory bowel disease; IL-10, interleukin-10; LPS, lipopolysaccharide; NSAID, nonsteroidal anti-inflammatory drug; OVX, ovariectomized; PPARs, peroxisome-proliferator
activated receptors; RAAS, renin–angiotensin aldosterone system; RBC, red blood cell; sEH, soluble epoxide hydrolase; SHR, spontaneously hypertensive rat; SNPs, single nucleotide
polymorphisms; SP-1, specificity protein 1; STZ, streptozotocin; THF, tetrahydrofuran; UPREs, unfolded protein response elements; UTR, untranslated region; VSM, vascular smooth
muscle; WKY, Wistar-Kyoto.
⁎ Corresponding author at: Department of Entomology and Comprehensive Cancer Center, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA. Tel.: +1 530 752
7519; fax: +1 530 752 1537.
E-mail address: bdhammock@ucdavis.edu (B.D. Hammock).

0378-1119/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.gene.2013.05.008
62 T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74

8. sEH genetic models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69


8.1. Acute inflammation and sepsis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
8.2. Ischemia/reperfusion injury in the heart and brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
8.3. Inflammatory bowel disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
8.4. Diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
8.5. Pulmonary hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
9. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71

1. Introduction hydrolyzes EpFA, including one important class of lipid signaling


molecules, the epoxyeicosatrienoic acids (EETs) (Spector and Norris,
This review first summarizes the role of sEH in the hydrolysis of 2007). EETs have vasoactive, anti-inflammatory and analgesic proper-
epoxy fatty acids (EpFAs), the proposed endogenous substrates of ties (Imig, 2012). Through metabolism of these molecules and other
the enzyme. We then provide a comprehensive evaluation of sEH EpFAs, sEH is implicated in several diseases, including hypertension,
gene expression and regulation, including its localization in mamma- cardiac hypertrophy, arteriosclerosis, brain and heart ischemia/reperfu-
lian tissues. Finally, we discuss sEH genetic models that have associat- sion injury, cancer and pain (Imig and Hammock, 2009; Morisseau and
ed sEH with diseases such as cardiovascular disease, cancer and Hammock, 2013; Wagner et al., 2011). Because of its possible role in
diabetes as well as pain, and have contributed to the identification cardiovascular and other diseases, sEH is being pursued as a pharmaco-
of the enzyme as a pharmaceutical target. logical target, and several potent small molecule inhibitors are available
(Shen and Hammock, 2012).
2. sEH protein structure and catalytic activities

Human sEH is a 62 kDa enzyme composed of two domains separated


by a short proline-rich linker (see Fig. 1) (Newman et al., 2005). In mam-
mals, sEH is a homodimer in the intracellular environment (Morisseau
and Hammock, 2013). The N-terminal domain exhibits a phosphatase
activity that hydrolyzes lipid phosphates, while the C-terminal domain
exhibits an epoxide hydrolase activity that converts epoxides to their
corresponding diols (see Fig. 2) (Morisseau and Hammock, 2013). sEH

Fig. 1. Crystal structure of the sEH dimer (PDB accession code 1S80 (Gomez et al.,
2004).) The sEH monomer is composed of two globular regions displaying alpha/beta Fig. 2. Substrates and products of sEH epoxide hydrolase activity. Through the addition
fold tertiary structure connected by a short proline-rich linker. The sEH dimer is of water, sEH converts lipid epoxides to diols (the oxygen of the epoxide moiety is in
anti-parallel, so that the N-terminal region of one monomer is in contact with the red). Potential substrates for sEH include the omega-three lipid epoxides formed
C-terminal region of the other. The catalytic site for the epoxide hydrolase activity is from DHA and EPA, the omega-six lipid epoxides formed from arachidonic acid
located within the C-terminal region, while the phosphatase activity is located within (ARA), linoleic acid, and stearic acid. The regioisomers preferred by the human sEH
the N-terminal region. are displayed (Morisseau et al., 2010).
T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74 63

Strongylocentrotus
Caenorhabditis Unknown purpuratus Gallus Homo
elegans Species SPEH1 SPEH2 gallus sapiens

Phosphatase Unknown No No No Yes

Epoxide Yes
Y Yes
Y No
N Yes
Y Yes
Y
Hydrolase

N
N SPEH1 N N
N2 N

C SPEH1
C C
C
C

SPEH2
N
C2
SPEH2
C

Fig. 3. sEH evolution. The mammalian sEH is the product of a gene fusion event and subsequent gene duplication that resulted in two full length sEH genes in Strongylocentrotus
purpuratus. One of these genes produces a protein with no phosphatase or epoxide hydrolase activity, while the other produces a protein with epoxide hydrolase but no phospha-
tase activity. In chicken there is a single sEH homolog with epoxide hydrolase but no phosphatase activity. The mammalian sEHs in mouse, rat, pig, and human exhibit both epoxide
hydrolase and phosphatase activities.

3. Gene evolution for minimum promoter expression. Methylation of these SP-1 sites is in-
volved in sEH gene silencing in HEPG2 cells (Zhang et al., 2010). AP-1
The two-domain, bi-functional mammalian sEH is the product of a sites are implicated in the upregulation of sEH by angiotensin-II (Ai et
gene fusion event (see Fig. 3) (Beetham et al., 1995). The C-terminal do- al., 2007). The sEH human promoter contains unfolded protein response
main exhibits homology with bacterial haloalkane dehalogenase, as well elements (UPREs) that upregulate sEH in human umbilical vein endo-
as single domain epoxide hydrolase enzymes in fungi, plants, and inverte- thelial cells (HUVECs) in response to homocysteine (Hcy) treatment
brates such as Caenorhabditis elegans (C. elegans) (Harris et al., 2008b; (Zhang et al., 2012a). Hcy significantly upregulates sEH mRNA expres-
Newman et al., 2005). The N-terminal domain of the mammalian enzyme sion, partially through the action of the activating transcription
is homologous to the bacterial haloacid dehalogenase (Beetham et al., factor-6 (ATF-6). The binding of ATF-6 is enhanced by Hcy-mediated
1995). Homologs of the phosphatase domain exist in the genome of ar- demethylation of the binding site.
chaea, bacteria and C. elegans (Cronin et al., 2003; Harris et al., 2008b). There are a number of physiologically relevant single nucleotide
Potential selective advantages of the fused gene include the proxim- polymorphisms (SNPs) in rat and human populations (Fornage et al.,
ity of phosphatase and epoxide hydrolase activities in the bi-functional 2002; Przybyla-Zawislak et al., 2003; Saito et al., 2001; Sandberg et al.,
enzyme that could allow shuttling of an unknown substrate (Argiriadi 2000). These SNPs fall within introns and exons. Two of these mutants
et al., 1999). However, the early fused homologs characterized in chicken impact sEH catalytic activities. R287Q, within the C-terminal domain
and sea urchin do not possess phosphatase activity (see Fig. 2) (Harris et of the enzyme, has decreased epoxide hydrolase and phosphatase activ-
al., 2008a,b). Interestingly, the sea urchin contains two two-domain ho- ities, while K55R, within the N-terminal of the enzyme, has reduced
mologs of sEH, and neither possesses N-terminal phosphatase activity, phosphatase activity when compared to the most frequent allele
indicating that the phosphatase activity is the result of evolutionarily re- (Przybyla-Zawislak et al., 2003; Srivastava et al., 2004).
cent mutations in the gene or was lost and subsequently regained (Harris
et al., 2008a). Other possible selective advantages of gene fusion include 5. Physiological role of sEH in mammals
the promotion of dimerization and enzyme stability (Argiriadi et al.,
1999; Beetham et al., 1995). sEH, through its epoxide hydrolase activity, metabolizes EpFA such as
the EETs. EETs are autocrine and paracrine lipid signaling molecules pro-
4. Mammalian gene structure duced from arachidonic acid (ARA) through the action of cytochrome
P450s (CYPs) that epoxidize the double bonds to form four regioisomers
The mouse gene is located on chromosome 14 and consists of 19 (see Fig. 3) (Imig, 2012; Spector and Norris, 2007). ARA is also the precur-
exons that encode for 554 amino acid residues (Grant et al., 1993, sor of the prostaglandins and the leukotrienes. These lipids play a role in
1994). The transcript was first cloned from mouse liver (Grant et al., asthma, pain, and inflammation (Haeggstrom and Funk, 2011; Smith et
1993, 1994). A reported mouse splice variant contains a sequence lo- al., 2011). The EET receptor or receptors are unknown, but the use of
cated in the second intron of the gene in place of the first two exons sEH inhibitors, EET mimics and the EETs themselves has demonstrated
(Hennebold et al., 2005). This splice variant lacks residues important the anti-inflammatory and vasoactive properties of the EETs in in vitro
for phosphatase activity, and is expressed in granulosa cells of pre- models such as isolated coronary rings and platelet aggregation assays,
ovulatory follicles. as well as in in vivo models such as angiotensin-II and dietary
The human gene is composed of 19 exons located on chromosome 8 salt-induced hypertension (Spector and Norris, 2007).
that encode for 555 amino acid residues (Beetham et al., 1993; Larsson A simple model of sEH function involves the regulation of blood
et al., 1995; Sandberg and Meijer, 1996). The human promoter lacks a pressure in the kidney (see Fig. 4). By hydrolyzing the vasodilatory
TATA box and contains several SP-1 sites (Tanaka et al., 2008). One EETs, sEH produces dihydroxyeicosatrienoic acids (DHETs), diol spe-
SP1 site immediately upstream of the transcription start site is required cies that are both less lipophilic and more readily conjugated and so
64 T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74

Fig. 4. Simplified model of the balance of lipid mediators in the kidney. The vasodilatory epoxyeicosatrienoic acids (EETs—represented by the single isomer 14,15-EET here) are
produced from arachidonic acid (ARA) through the action of cytochrome P450s (CYPs) 2J and 2C. This anti-hypertensive signal is offset by a second metabolite of ARA,
20-hydroxyeicosatetraenoic acid (20-HETE) produced by CYP 4A. The balance of antihypertensive and hypertensive signals is partially maintained through the action of sEH.
sEH metabolizes the EETs to diol species that are readily conjugated and removed from the site of action in the kidney vasculature. When sEH is inhibited there is an increase in
EET levels relative to 20-HETE levels. This tips the balance in favor of vasodilation, creating an anti-hypertensive effect. The process is highly simplified since the epoxy fatty
acids and their corresponding diols can be of multiple chain lengths in both the ω−3 and ω−6 series with varying regio, geometrical, and optical isomers involved.

more easily removed from the site of action and excreted than the Interestingly, with regard to angiogenesis, the EETs are mildly pro-
EETs (Morisseau and Hammock, 2013). In a simplified model, by re- angiogenic, resulting in enhanced wound repair but accelerated growth
moving this lipid mediator, sEH changes the balance of vasoconstric- of solid tumors (Panigrahy et al., 2012). In contrast, the DHA epoxides
tive and vasodilatory signals, thereby influencing vascular resistance are dramatically anti-angiogenic, acting in a non-VEGF mediated path-
to blood flow and blood pressure (Morisseau and Hammock, 2013). way. Stabilization of the ω−3 DHA epoxides results in a blockage of
Reduction of sEH epoxide hydrolase activity, either through a small tumor growth and metastasis in murine xenographs or breast and
molecular inhibitor, or through genetic knockout can stabilize or in- lung tumors (Zhang et al., 2013).
crease levels of EETs and other EpFAs and so reduce hypertension
(Morisseau and Hammock, 2013). The sEH inhibitors appear most ef- 6. Single nucleotide polymorphisms
fective in reducing angiotensin driven hypertension (Morisseau and
Hammock, 2013). Increasing endogenous EpFA chemical mediators sEH SNPs were first associated with coronary heart disease in the Cor-
by decreasing their rate of hydrolysis is the molecular basis of the onary Artery Risk Development in Young Adults (CARDIA) and the Ath-
therapeutic action of sEH inhibitors in many disease models. erosclerosis Risk in Communities (ARIC) studies (Fornage et al., 2004;
The situation in vivo is more complicated. EETs have displayed dif- Lee et al., 2006). In the CARDIA study, the R287Q allele was associated
ferent properties in different vascular beds, and the products of hydro- with coronary artery calcification (CAC) in African Americans, but not
lysis, the DHETs, are poorly characterized (Morisseau and Hammock, in Caucasians (Fornage et al., 2004). The risk for CAC associated with
2013). sEH hydrolyzes other lipid epoxides such as the ω−3 sEH SNPs was highest among smokers (Wei et al., 2007). R287Q was
docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) epox- not associated with hypertension in a smaller African American popula-
ides, and the 18-carbon leukotoxin and isoleukotoxin (see Fig. 2) tion (Dreisbach et al., 2005) Similarly, R287Q was not associated with is-
(Zheng et al., 2001). The 18:2 diol or the bisepoxide can be further me- chemic stroke, myocardial infarction, and ischemic heart disease in a
tabolized to tetrahydrofuran diols that display reproductive and behav- Danish population (Lee et al., 2010). The K55R allele is associated with
ioral properties in vivo (Markaverich et al., 2007; Moghaddam et al., the risk of developing coronary heart disease in Caucasians, but not in
1996). In addition, sEH hydrolyzes all other EpFAs tested at varying African Americans (Lee et al., 2006). A group examining the R287Q and
rates (Morisseau et al., 2010). In the linoleate epoxide series the K55R SNPs recapitulated these results in a Swedish population (Fava et
resulting diol is thought to be a potent chemical mediator needed for al., 2010). K55R was associated with a higher risk of hypertension and is-
normal development, acting to increase vascular permeability in some chemic stroke in male homozygotes. When forearm blood flow and vas-
tissues (Fromel et al., 2012), but when at high concentrations it is a cular resistance in black and white Americans was measured, the K55R
marker of acute respiratory distress syndrome (ARDS), promoting mutant was associated in white Americans with lower blood flow and
perivascular and pulmonary edema (Moghaddam et al., 1997). The high vascular resistance in response to bradykinin, but not in black
regioisomers of the ω−3 epoxides DHA and EPA are more rapidly hy- Americans (Lee et al., 2011). R287Q was associated with lower vascular
drolyzed than the corresponding ω−6 ARA epoxides, except for the resistance in response to bradykinin in black, but not in white Americans.
slowly hydrolyzed epoxide in the ω−3 position (Morisseau et al., Variations in the sEH gene are associated with other disease states,
2010). These ω−3 epoxides of DHA and EPA and possibly the poorly including ischemic stroke and diabetes. Two haplotypes in African
studied corresponding diols can be more potent than the EETs in American populations are associated with either increased or de-
some in vitro models (Spector, 2009). EPA epoxides inhibit endothelial creased risk of ischemic stroke (Fornage et al., 2005). Three SNPs in
cell proliferation (Cui et al., 2011). In a number of in vivo rodent models the sEH gene are associated with ischemic stroke. Two SNPs were
the DHA epoxides seem to have similar biological effects as the EETs; located in introns, but the third was R287Q (Gschwendtner et al.,
however they appear to be more potent. These include reduction in en- 2008). Neuronal cells transfected with the R287Q mutant exhibit in-
hanced pain perception or nociception (Morisseau et al., 2010) and re- creased survival after oxygen–glucose deprivation, supporting a pos-
duction in angiotensin-II driven blood pressure (Ulu et al., in press). sible role for sEH in neuron survival (Koerner et al., 2007). The R287Q
T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74 65

SNP is associated with a lower risk of ischemic stroke in a Chinese disease. There is a critical need to follow up on these observations with
population, but only in the case of nonsmokers (Zhang et al., 2008a). experimental models in animals and clinical interventions in man.
R287Q was associated with insulin resistance in patients with type 2
diabetes (Ohtoshi et al., 2005). Finally, no association was identified be- 7. Mammalian gene expression
tween K55R and hypertension or coronary heart disease in patients that
received restenosis after percutaneous coronary intervention (PCI, also 7.1. Introduction
called coronary angioplasty) (Kullmann et al., 2009).
Interestingly SNPs in the non-coding regions of the sEH gene, as well sEH was first identified through its activity on substrates such as
as in the 5′ and 3′ UTRs, are associated with human diseases. SNPs locat- juvenile hormone, as well as lipid epoxides such as epoxystearate
ed in introns are associated with carotid and coronary artery calcified (Hammock et al., 1976). Through gel filtration and the assessment of dif-
plaques, as well as carotid artery intima-media thickness in African and ferential hydrolysis of epoxystearate, the epoxide hydrolase activity in
European Americans participating in the Diabetic Heart Study (Burdon soluble fractions was determined to be distinct from the microsomal frac-
et al., 2008). A SNP in the 3′ region of the gene is associated with de- tion obtained from the liver and kidneys of mice (Gill and Hammock,
creased semen quality and male infertility in a Chinese population (Qin 1979). Subsequent studies using a lipid epoxide as a substrate detected
et al., 2012). In rodents, variation in the sEH promoter region segregates this activity in the soluble fraction of multiple organs, though at a lower
with heart failure in the spontaneous hypertensive heart failure rat level than in liver and kidney (Gill and Hammock, 1980). The enzyme ac-
model (Monti et al., 2008). This variation results in increased gene ex- tivity was detected in rabbits, mice and rats, and the proposed enzyme
pression and epoxide hydrolase activity. The authors further show that was named soluble epoxide hydratase or hydrase. Establishment of sys-
sEH gene expression in humans with heart failure is downregulated rel- tematic nomenclature by the International Union of Pure and Applied
ative to control patients. Finally, a mutation in the 3′ UTR is associated Chemistry has lead to the acceptance of soluble epoxide hydrolase as
with differences in kidney function in patients that have undergone the name of the enzyme.
kidney transplant. Specifically, the absence of this SNP is predictive of There are sEH homologs in plants such as potato and cress, and an-
kidney failure (Lee et al., 2008). These “non-coding” SNPs may play a imals such as nematode, sea urchin, chicken, cat, and dog where it
role in sEH regulation through microRNA or other mechanisms. was cloned and expressed (Morisseau and Hammock, 2013). Al-
Taken together, these data show that the race and sex of patients, as though sEH activity is highest in the liver and kidney, it is implicated
well as patient history must be considered when assessing the effect of in important physiological processes in the brain, lung, and other or-
sequence variation in the sEH gene. Coronary heart disease is associated gans. sEH was first identified and localized to tissues through its ep-
with R287Q, a SNP that displays decreased sEH epoxide hydrolase activ- oxide hydrolase activity. To focus this review, we have confined our
ity. In addition, K55R, a SNP that displays decreased phosphatase activity, discussion to sEH mRNA and protein expression. A summary of the
is associated with ischemic stroke. Many of these studies need to be re- significant results discussed in this section, as well as additional re-
peated with higher numbers of patients and more refined criteria of sults not discussed, is contained in Table 1.

Table 1
Modulation of sEH expression in selected tissues.

Ref Organ/cell Localization Species Conditions Result Detection method

Zhang et al. (2012a) HUVECs Human Hcy treatment Upregulation Western qRT-PCR
Zhao et al. (2005) Vasculature Mesenteric artery Rat Lean vs. obese Zucker Upregulation Western qRT-PCR
Mark-Kappeler et al. (2011) Vasculature Mesenteric artery Rat Ethinyl estradiol, tamoxifen, and Upregulation cDNA microarray
raloxifene-treated ovariectomized
animals
Zhang et al. (2012a) Aorta Intima Mouse Hcy treatment Upregulation qRT-PCR
Maresh et al. (2005) Aorta Endothelium Mouse Exposure to cigarette smoke Upregulation qRT-PCR
Gao et al. (2011) Coronary artery Mouse Renal artery stenosis Upregulation Western
Aboutabl et al. (2009) Heart Rat 3-methylcholanthrene and Upregulation Western qRT-PCR
benzo(a)pyrene treatment
Xu et al. (2006) Heart Atrial and ventricular Mouse Thoracic aortic constriction No change Western qRT-PCR
myocytes
Zordoky et al. (2008) Heart Mouse Isoproterenol treatment Upregulation qRT-PCR
Ai et al. (2009) Heart Left ventricle Rat SHR vs. Wistar control Upregulation Western
Ai et al. (2009) Heart Left ventricle Rat Ang-II treatment Upregulation Western
Ai et al. (2009) Heart Left ventricle Rat Exercise training No change Western
Ai et al. (2009) Heart Left ventricle Rat Norepinephrine treatment No change Western
Yu et al. (2000) Kidney Cortex Rat Upregulation Western Northern
Seubert et al. (2005) Kidney Rat SHR vs. Wistar control Upregulation cDNA microarray Northern
Koeners et al. (2011) Kidney Rat SHR vs. Wistar control Upregulation qRT-PCR
Imig et al. (2002) Kidney Cortex Rat Ang-II infusion Upregulation Western
(Zhao et al., 2004) Kidney Cortex Rat Ang-II infusion Upregulation Western
Zhao et al. (2004) Kidney Microvessels Rat Ang-II infusion Upregulation Western
Walkowska et al. (2010) Kidney Cortex Rat Renal artery stenosis Upregulation Western
Jung et al. (2010) Kidney Mouse 5/6 nephrectomy Downregulation Western qRT-PCR
Oguro et al. (2009) Kidney Mouse Streptozotocin treatment Downregulation Western qRT-PCR
Oguro et al. (2009) Liver Mouse Streptozotocin treatment Downregulation Western
Fife et al. (2008) Liver Mouse LPS treatment Downregulation qRT-PCR
Sellers et al. (2005) Brain Brain stem Hypothalamus Rat SHR vs. Wistar control Upregulation Western qRT-PCR
Koerner et al. (2008) Brain Cortex Rat Estradiol treatment of Downregulation Western
ovariectomized animals
Revermann et al. (2009) Lung Rat Monocrotaline treatment Downregulation Western
Keseru et al. (2010) Lung Human Hypoxia Downregulation Western qRT-PCR
Morin et al. (2010) Lung Bronchi Human Asthma vs. control Upregulation Immunohistochem
Rodriguez and Clare-Salzler Macrophages Bone marrow derived Rat LPS treatment of non-obese Upregulation qRT-PCR
(2006) diabetic animals
66 T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74

7.2. Subcellular localization Receptor-dependent regulation of sEH was apparent since the en-
zyme was first identified in rodents. In human, exploration of the role
Originally identified in the soluble fraction of organ lysates, the of PPAR receptors in the regulation of sEH expression will provide a
difficulties of subcellular fractionation and/or the permeability of sub- connection between EpFA signaling and lipid homeostasis and pa-
cellular compartments lead to contradictory results. After the initial thologies such as metabolic syndrome. The involvement of sEH in
reports of sEH activity, sEH was detected in mitochondrial, microsom- the renin–angiotensin system (RAAS) has wide ranging implications
al and cytosolic fractions by activity or in the peroxisomes alone by for inflammatory diseases and hypertension.
immunohistochemistry (Hollinshead and Meijer, 1988; Kaur and
Gill, 1986; Waechter et al., 1983). Eventually it was determined that 7.4. Vasculature
the activity in the mitochondrial fractions was an artifact from perox-
isomal contamination, and the same enzyme was responsible for sEH is implicated in the hydrolysis of the vasoactive EETs. sEH SNPs
the activity in the cytosol and peroxisomes (Chang and Gill, 1991; are associated with cardiovascular disease. Taken together, these
Eriksson et al., 1991). The small amount of sEH either trapped in the data suggest a role for sEH in the vasculature. sEH is localized in the
microsomes or tightly associated with them can be differentiated rat mesenteric artery (Olearczyk et al., 2006; Zhao et al., 2005).
with selective substrates or inhibitors. A more recent comprehensive Vasodilatory effects of sEH inhibitors are not affected when the endo-
examination of multiple tissues reported that sEH peroxisomal local- thelium is stripped from isolated rat vessels, indicating that sEH in the
ization may be tissue specific (Enayetallah et al., 2006a). The peroxi- vascular smooth muscle may be responsible for the vasodilatory effects
somal localization is complex because targeting sequences in the (Olearczyk et al., 2006). (An aside for future in vitro experiments: at
mammalian sEH are perfect glyoxysome targeting signals in plants least some cultured VSMCs do not contain sEH or lose the enzyme
but imperfect in mammals. after repeated passages (Davis et al., 2006).) In the obese Zucker rat,
the mesenteric microvessel endothelium-dependent dilation is reduced
relative to lean Zucker rats (Zhao et al., 2005). Interestingly, sEH is
7.3. sEH, PPAR agonists, and angiotensin-II upregulated in the mesenteric arteries of these rats (Zhao et al., 2005).
Treatment with ethinyl estradiol, tamoxifen, and raloxifene in ovariec-
A challenge facing the investigation of sEH in isolated cells is that tomized rats upregulates sEH mRNA expression in mesenteric arteries
sEH is not uniformly expressed in immortalized cell lines. In addition, (Mark-Kappeler et al., 2011). sEH protein is expressed in red blood
most cell lines do not express a full complement of CYPs, major pro- cells (Jiang et al., 2008). sEH mRNA and protein expression is decreased
ducers of the EETs and other EpFAs that are substrates for sEH in the red blood cells (RBCs) in the spontaneous hypertensive rat, com-
(Langenbach et al., 1992; Tanaka et al., 2008). However, in the absence pared to the Wistar control (Jiang et al., 2011).
of good cell-based systems, a number of inducers were identified in These results support the involvement of sEH in paracrine and auto-
vivo. Most recently, treatment with arsenic-III increased sEH mRNA ex- crine signaling in the vasculature, through its metabolism of vasoactive
pression in the lung, liver and kidney of mice (Anwar-Mohamed et al., EpFA signals produced in the region of VSMCs and endothelial cells. The
2012). The protein levels increased in the lung, did not change in the pool of sEH in RBCs could exert its effects systemically, having enhanced
liver and decreased in the kidney. effects when blood cells are in close proximity to vessel walls. This
The first identified inducer of sEH activity was clofibrate, a peroxi- occurs in the micro-capillaries of peripheral tissue, for example.
some proliferator that increased sEH activity in rodent liver (Hammock
and Ota, 1983). Clofibrate induces sEH mRNA expression in intestine, 7.5. Heart
kidney and heart, as well as the liver of mice (Johansson et al., 1995). In-
terestingly, the human 5′ UTR does not contain PPARα response ele- sEH is expressed in the aorta and in both atrial and ventricular tis-
ments (Tanaka et al., 2008). sues of rodents (Chawengsub et al., 2008; Xu et al., 2006). Because the
PPARγ activation is linked to sEH expression in human endothelial EETs display vasoactive properties on isolated coronary artery rings,
cells (Liu et al., 2005). The laminar flow produced by a flow chamber the expression of sEH in coronary arteries is of special interest
apparatus simulates anti-inflammatory pathways in these cells and (Fang et al., 1999). In endothelial nitric oxide synthase (eNOS) knock-
activates PPARγ. In this model, sEH mRNA and protein expression is out animals fed a high salt (HS) diet, sEH protein expression in the
downregulated under conditions of increased laminar flow and the aortas is reduced in HS-fed eNOS−/− and eNOS+/+ mice relative
inhibition of sEH epoxide hydrolase activity further enhances the to mice fed a normal salt diet, with the most significant reduction in
PPARγ response. A mouse heterozygote for a PPARγ deletion has re- the eNOS+/+ mice (Nayeem et al., 2011). This link between eNOS
duced sEH mRNA expression in the heart (Pang et al., 2011). Addi- and sEH expression implies that there is interplay between different
tionally, in an angiotensin-II infused rat model, the increase in sEH endothelium-derived hyperpolarization factors in models that exhibit
protein levels can be reversed by treatment with the PPARγ agonist endothelium dysfunction. For example, exposure to tobacco smoke
rosiglitazone (Pang et al., 2011). Rosiglitazone increases mouse sEH upregulates sEH mRNA expression in the endothelial cells of aortas
mRNA expression in epididymal and subcutaneous fat pads, but not of mice (Maresh et al., 2005) and hypertension induced by renal ar-
in the liver and kidney (De Taeye et al., 2010). The link between tery stenosis increases sEH protein expression in the endothelial
PPARγ activation and sEH expression is an ongoing area of research cells of coronary arteries of mice (Gao et al., 2011). In summary,
(Imig et al., 2012; Nayeem et al., 2013; Xu et al., in press). sEH is implicated in proper functioning of the endothelium of coro-
Angiotensin-II (Ang-II) is a peptide hormone with vasoconstric- nary arteries and the aorta.
tive properties involved in the regulation of blood pressure and the The SNPs identified in the CARDIA and ARIC studies suggest that
pathology of hypertension. Ang-II treatment of HUVECs results in an variation in sEH epoxide hydrolase activity may play a role in cardio-
over two-fold increase in mRNA expression of sEH (Ai et al., 2007). vascular disease. sEH expression is modulated in the heart in several
The authors attribute this increase to the action of c-Jun binding to disease models, prominently in models of cardiac hypertrophy.
putative AP-1 binding sites located in the human sEH promoter re- sEH protein levels increase in the heart of a rat Ang-II induced
gion, since deletion constructs of the promoter lacking AP-1 sites model of cardiac hypertrophy (Ai et al., 2009). This increase in sEH
lose their response to Ang-II. ChIP assays confirmed that cJun binds was not detected in hypertrophy induced by norepinephrine or in
to two elements in the promoter. This result was replicated in rat so-called physiological hypertrophy induced by exercise training. In-
endothelial cells, and in two in vivo models, where sEH transcript creased expression of sEH mRNA and protein also occurs in the
expression is specifically upregulated in the rat aortic intima. heart in a rat isoproterenol-induced model of cardiac hypertrophy
T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74 67

(Althurwi et al., 2013; Zordoky et al., 2008). There are conflicting presumable through its ability to metabolize EpFA with osmoregulatory
reports of sEH expression in mouse pressure-induced models of hy- and vasoactive properties.
pertrophy, where no change or a reduction in sEH protein and mRNA In rodent models of diabetes, sEH expression varies from no
expression is detected in heart tissue (Morgan et al., in press; Xu et al., change to a decrease in expression in the kidney. Fructose-fed insulin
2006). Chronic treatment with doxorubicin increases hypertrophic, in- resistant hypertensive rats do not exhibit a change in sEH protein
flammatory and apoptotic markers as well as sEH mRNA expression in expression in whole kidney preparations (Mustafa et al., 2010). How-
the rat heart (Alsaad et al., 2012). Finally, arsenic-III treatment increases ever, sEH protein and mRNA expression is decreased in the kidneys of
mRNA expression of hypertrophic markers as well as protein and mRNA diabetic mice in a streptozotocin (STZ)-induced mouse model, where
levels of sEH in the mouse heart (Anwar-Mohamed et al., 2012). These sEH is immunolocalized prominently to the proximal tubules (Oguro
data imply a distinct role for sEH in the progression of pathological et al., 2009). This illustrates the difficulty in comparing immunohisto-
hypertrophy. chemistry to data from tissue homogenates in highly structured or-
sEH is not upregulated in norepinephrine-induced or pressure- gans such as the kidney. Because of the localization of sEH, whole
induced hypertrophy or so-called physiological hypertrophy, indicating tissue homogenates may mask important changes in expression. Rel-
that sEH is not merely a marker of hypertrophy, or even a general marker evant to these diabetic models, sEH is expressed in human pancreatic
of pathological hypertrophy, as is the fetal gene marker beta myosin beta islet cells (Enayetallah et al., 2004). Finally, modulation in sEH
heavy chain, for example. Its upregulation in the heart in Ang-II induced expression is linked to kidney damage. First, there is a decrease in
hypertrophy shows a further link between the renin–angiotensin– renal levels of sEH mRNA and protein in a mouse model of progres-
aldosterone system (RAAS) and sEH, and demonstrates that sEH is in- sive renal disease where one kidney is surgically impaired and dam-
volved in specific physiological effects of chronic dysregulation of the aged and the second kidney is removed (Jung et al., 2010). A second
RAAS. EpFAs appear so intimately involved in the RAAS that possibly it group has recently reported an increase in sEH protein expression
should be termed the ERAAS to indicate the importance of EpFA in the in the renal proximal tubules of human patients with glomerulone-
system. phritis (Wang et al., 2012). They also report an increase in sEH pro-
Use of sEH inhibitors and sEH genetic models has implicated the tein expression in the renal cortex of mice with adriamycin-induced
enzyme in cardiac ischemia/reperfusion injury. However, sEH protein nephritis. These results raise the general issue of EpFA and organ pro-
levels are not upregulated in the mouse left ventricle free wall in a tection. It is possible that EpFAs play a role in the protection of partic-
myocardial infarction model (Li et al., 2009). Similarly, sEH mRNA ex- ular organs from damage due to chronic disease states as well as
pression does not change in the left ventricle in a rat chronic heart acute injuries.
failure model (Merabet et al., 2012).
7.7. Liver

7.6. Kidney Epoxide hydrolase activity is present in the cytosolic fractions pre-
pared from the livers of monkeys and rodents. This activity is upregulated
sEH is expressed in the renal vasculature as well as the tubules to a by clofibrate and disease states and conditions such as STZ-induced diabe-
lesser degree, as determined by immunohistochemistry on human kid- tes and food restriction (Hammock and Ota, 1983; Moody et al., 1985;
neys (Yu et al., 2004). The weaker immunoreactivity in the glomeruli Pacifici et al., 1983; Thomas et al., 1989). Treatment with xenobiotics
and tubules is homogeneous, but in the arteries sEH is localized to the such as the drug doxorubicin or the environmental toxin acrylonitrile
smooth muscle layers of the arterial wall (Yu et al., 2004). In mice, does not alter sEH expression in the rat liver (Chanas et al., 2003;
sEH mRNA and protein expression is greater in the kidneys of male Zordoky et al., 2011). Mirroring the results from kidney, there is a differ-
mice, compared to female mice (Chanas et al., 2003; Sinal et al., 2000). ence in sEH expression in males and females; male rats have greater sEH
Female and male spontaneously hypertensive rats (SHRs) have protein expression than female rats in the liver (Chanas et al., 2003).
elevated levels of sEH mRNA and protein in the kidney cortex com- sEH expression levels have both increased and decreased in
pared to Wistar-Kyoto “control” rats (WKY) (Koeners et al., 2011; models that involve modulation of dietary fat. In a mouse high-fat
Yu et al., 2000). In a cDNA microarray experiment, sEH mRNA expres- (HF)-diet-induced fatty liver model, a 16 week diet increased the ex-
sion was increased in the SHR kidney compared to the WKY controls pression of sEH mRNA in the liver (Liu et al., 2012). This model used a
and sEH was the primary outlier at 3 and 9 weeks of age (Seubert 60% versus 10% fat diet of unspecified composition. When the fatty
et al., 2005). sEH protein expression is greater in male SHR kidneys acid composition but not the total amount of fat in the diet is manip-
compared to females, and ovariectomy increases the expression ulated, supplementation with fish oil decreases the mRNA and pro-
(Martin et al., 2012). This difference in the sexes mirrors the situation tein levels of sEH in the mouse liver compared to animals fed a diet
in the mouse. supplemented with a high-oleic acid sunflower-seed oil or DHA-rich
Another model of hypertension maintains Ang-II infusion via an im- oil (Mavrommatis et al., 2010). The authors conclude that EPA may
planted osmotic mini-pump. In general, Ang-II infusion in rats increases be responsible for the decrease in expression of hepatic sEH. This re-
sEH protein expression in the renal cortex and microvasculature (Imig sult indicates that researchers who use high fat diets in their disease
et al., 2002; Zhao et al., 2004). In Ang-II infused mice, sEH is highly models should carefully examine the composition of the added fat,
expressed in the distal tubuli and expressed at lower levels in the glo- since it appears that sEH expression in the liver may be altered by
meruli, as determined by immunohistochemistry (Jung et al., 2005). specific lipids. This is not surprising, since several different classes
Similarly, in the two-kidney-one-clip model of hypertension, sEH is of fatty acids can be metabolized by CYPs to form potential EpFA sig-
upregulated in the renal cortex (Kopkan et al., 2012; Walkowska et naling molecules and some of these are substrates for sEH, such as the
al., 2010). These results point to a role for sEH in the vasculature of epoxides of the omega three fatty acids DHA and EPA, the epoxides of
the kidney in the maintenance of hypertension. Interestingly, there is linoleic acid, as well as the EETs (Morisseau et al., 2010) (see Fig. 2).
no difference in sEH protein expression in the kidney cortex and medul-
la of transgenic mice with inducible expression of the renin gene when 7.8. Brain
the control and induced groups are compared (Honetschlagerova et al.,
2011). Similarly, sEH expression in the kidney did not change in a die- In the human brain, sEH is immunolocalized primarily in astro-
tary salt-loading model of hypertension (Sinal et al., 2000). The specific cytes, oligodendrocytes and neuronal cell bodies (Sura et al., 2008).
localization and high expression of sEH in the kidney is an indication sEH is also detected in the smooth muscle cells of arterioles in the
of the important role sEH plays in the control of blood pressure, brain vasculature (Sura et al., 2008). In rat cerebral parenchymal
68 T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74

microvessels, sEH is immunolocalized to the vascular smooth muscle This may explain the model and insult-dependent changes in sEH ex-
cells (VSMCs) of the microvasculature and the perivascular nerves pression. The variation in sEH expression is reminiscent of the brain,
that innervate the middle cerebral artery (Iliff et al., 2007). The au- where control of both temporal blood flow and long term blood
thors suggest that nerve-derived EETs may act on the cerebral vascu- flow to certain regions has physiological relevance.
lature (Iliff et al., 2009). In the mouse brain, sEH is immunolocalized
in astrocytes rather than in neurons in most regions except the cen- 7.10. Fat
tral amydgala, where it is primarily expressed in neurons and may
play a role in the release of neuropeptides (Marowsky et al., 2009). Fat is a highly structured organ that contains both adipocytes and
Thus, it is possible that sEH acts in both VSMCs and nerve cells to reg- non-fat stromal cells arranged in a defined architecture. Stromal cells
ulate blood flow in the brain, through the metabolism of lipid media- include endothelial cells that form part of the vasculature, as well as in-
tors with vasoactive properties or perhaps the ability to trigger the flammatory cells such as macrophages. In mice fed a high fat diet, sEH
release of neuronal signaling molecules. Elucidation of mechanism was immunolocalized in both endothelial and macrophages in adipose
will require further examination of regional localization. Supporting tissue, as would be expected from previous studies (De Taeye et al.,
a role for sEH outside the regulation of cerebral blood flow, mouse 2010). In the high fat diet, 42% rather than 12% of the total calories
sEH was immunolocalized in medium and large sized myelinated were derived from fat. Normalized concentrations of sEH mRNA or pro-
axons in dorsal root ganglia that are involved in fast nociception tein did not change in total fat, but there was variation between sEH ex-
and the development of allodynia (Brenneis et al., 2011). pressions in perirenal, epididymal, pericardial, and subcutaneous fat
sEH expression in the rat brain is modulated by genetic background pads, indicating specific sEH expression. After immunohistochemistry
and disease states. In rats subjected to middle cerebral artery occlusion, suggested that sEH was also expressed in adipocytes, the group isolated
treatment with salvianolic acid A reduced infarct volume and sEH pro- epididymal fat. sEH mRNA expression was four times higher in the
tein expression in the hippocampus, as determined by immunohisto- adipocyte-enriched fraction compared to the stromal fraction. Addi-
chemistry (Wang et al., 2013). In the SHR, sEH mRNA and protein tionally, in cultured 3T3-L1 preadipocytes, differentiation was accom-
expression is elevated in the brainstem and hypothalamus compared panied with an increase in sEH protein and mRNA expression. As
to the control animals (Sellers et al., 2005). Both these areas are adipose tissue expands in obesity, peripheral resistance increases with
known to be cardioregulatory, adding yet another mechanism by tissue mass and vascularization. The observation that sEH increases
which sEH, through its metabolism of EpFAs or other mediators, may with adipocyte maturation suggests that anti-inflammatory and anti-
be regulating blood pressure. However, Bianco et al. did not detect a hypertensive EpFAs will be reduced. Thus one can envision a positive
change in blood pressure after sEH was overexpressed in the brain feedback where increased adiposity in turn increases vascular resis-
(Bianco et al., 2009). Additionally, stroke-prone SHRs have lower sEH tance and Ang-II levels, leading to increased sEH levels and decreases
mRNA expression than stroke-resistant SHRs, due to sequence variation in EETs and other EpFAs. The presence of sEH in adipocytes suggests a
in the promoter region of the gene (Corenblum et al., 2008). The change role for sEH beyond the regulation of vascular tone in fat. It is especially
in sEH activity does not result in a significant alteration of plasma EET intriguing that sEH expression varies between different fat regions. Fu-
levels, but the stroke-prone SHRs have lower plasma DHET levels. This ture studies may discover a role for sEH in the physiological changes
calls attention to the current lack of knowledge of the biological activity that occur within fat in response to chronic disease states. Perhaps
of the DHETs. It also illustrates the differences in lipid metabolism that sEH expression in fat pads in proximity to the surface of organs such
may occur when sEH is permanently downregulated in an organism. as the kidney or heart will have some relevance to particular disease
As in the liver and kidney, male rodents have a higher sEH expres- states such as diabetes or hypertension.
sion in the brain than females, a result confirmed in primary cortical
neurons from both mice and rats, measuring both protein and mRNA 7.11. Inflammatory system
levels (Fairbanks et al., 2012; Zhang et al., 2009). Estradiol replacement
in ovariectomized (OVX) rats significantly reduces sEH expression in sEH is implicated in the regulation of inflammation in several
the cerebral cortex, implicating sex hormones in the regulation of sEH models. Rat alveolar and peritoneal macrophages and peritoneal neu-
protein expression in the brain (Koerner et al., 2008). trophils contain detectable amounts of sEH by Western blot (Draper
It appears that in addition to its well-established role in the regu- and Hammock, 1999). Macrophages isolated from the bone marrow
lation of blood flow through metabolism of the EETs, the enzyme may of non-obese diabetic mice and treated with LPS for 12 h display a
be involved in regulation of the release of neurotransmitters or even greater increase in sEH expression than cells isolated from control
memory. This new function may or may not involve the EETs, and mice (Rodriguez and Clare-Salzler, 2006). This indicates that the dis-
will require tools that allow for a finer distinction between the roles ease background has a sensitizing effect. sEH protein level was elevat-
of sEH in particular brain regions. Because of the potential complica- ed at the graft–vein junction of synthetic arteriovenous grafts in a
tions of these overlapping functions of the enzyme, and the possibility porcine model, and continued to increase up to 4 weeks post surgery,
that the second, phosphatase activity of the enzyme may be involved, judged by immunohistochemistry (Sanders et al., 2012). sEH expres-
genetic tools will be of particular help. sion appeared to spread beyond the media of the vessel to the adven-
titia by the first week. The authors then cultured human monocytes
7.9. Lung and pulmonary arteries and confirmed sEH expression by Western blot. Finally, in a variety
of inflammation-based assays inhibitors of the sEH are more potent
sEH is immunolocalized to smooth muscle tissue surrounding pul- at reducing inflammation in vivo than NSAIDs and COXIBs and
monary venules in mice, as well as the endothelium of the alveolar synergize with them (Morisseau and Hammock, 2013; Shen and
capillaries to a lesser degree (Zheng et al., 2001). In humans, sEH pro- Hammock, 2012). These results support the view that sEH and by im-
tein was identified in lysates prepared from the parenchymal, bron- plication EpFA are broadly involved in the regulation of inflammation.
chial, and pulmonary arteries (Senouvo et al., 2011). Patients with
asthma exhibit a higher expression of sEH in the bronchi, judged by 7.12. Reproductive system
immunohistochemistry (Morin et al., 2010). In contrast, in the rat
model of monocrotaline-induced pulmonary hypertension sEH pro- sEH is expressed in porcine granulosa and theca cells, where protein
tein levels were decreased in lung tissue (Revermann et al., 2009). levels are lower in follicles harvested after the luteinizing hormone
Blood flow is regulated in a complex manner in the lung with differ- surge (Newman et al., 2004). In the mouse, sEH transcript levels in
ent regions oxygenated at different stages of the respiratory cycle. the ovary increase upon stimulation of the estrous cycle, and peak in
T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74 69

the luteal phase, when the ovaries contain corpora lutea (Hennebold et ω-hydroxylase metabolite, 20-HETE (see Fig. 4), was enhanced
al., 2005). The mouse also expresses a splice variant of sEH during the 4-fold in kidney lysates. When the plasma lipids were analyzed, the
periovulatory phase of a stimulated estrous cycle that has not been EpFAs were increased and the corresponding diols were decreased and
identified in other mammals (Hennebold et al., 2005). In the primate there was no change in lipoxygenase- and cyclooxygenase-dependent
corpus luteum, the sEH protein and mRNA levels peak in the late luteal oxylipins. Other differences between the sEH null mice and wild-type an-
phase of the menstrual cycle (Irusta et al., 2007). The authors suggest imals include variation in plasma testosterone, sperm count and testicular
that sEH may play a role in either the initiation of corpus luteum forma- size in males and a decrease in plasma cholesterol levels (Luria et al.,
tion, and/or in the regression of the structure. In pig, sEH protein in- 2009). Adenosine-induced relaxation of coronary artery rings isolated
creases in the prepubertal cumulus–oocyte complexes (COCs) from knockout animals depends on the upregulation or downregulation
compared to cyclic females (Paczkowski and Krisher, 2010). Finally, of several receptors compared to the wild-type mice, including A2A AR,
sEH protein is expressed in the human myometrium, as well as in the A1 AR, PPARγ, and PPARα (Nayeem et al., 2013). This indicates that
placenta (Corriveau et al., 2009). Taken together, these results imply compensatory mechanisms in the null mice involve complex changes in
that sEH may play a role in fertility, not necessarily limited to the action signaling pathways relating to the maintenance of vascular tone. These
of the corpus luteum. knockout strains, particularly when coupled with chemical knockouts
In the male reproductive system, sEH protein is expressed in the using sEH inhibitors and EET antagonists have proven very valuable
rat epididymis, specifically the caput, corpus, and cauda (DuTeaux in elucidating the physiological role of sEH. Some important findings
et al., 2004). Interestingly, unlike the liver and kidney, sEH in the rat are summarized in Table 2. Hopefully, conditional, tissue-selective and
epididymis is not induced by clofibrate treatment. This observation other modifications will provide even greater insight in the future.
fits the pattern that sEH induction by PPARα and PPARγ ligands Transgenic mice that overexpress human sEH in the endothelium
varies among organs. were produced by inserting sEH cDNA under the control of a Tie2 pro-
The difference between sEH expressions between the sexes, as moter (Edin et al., 2011). Tie2-sEH Tr mice have a reduced plasma
well as modulation of sEH expression by estrogens shows that careful epoxide-to-diol ratios of 14,15 EET and 12,13-epoxyoctadecenoic
attention should be paid to the sex of the animals chosen for in vivo acid and do not exhibit decreased infarct size or improved heart func-
experiments. In the case of estrogens, this is a further example of tion after ischemia/reperfusion (Edin et al., 2011). Aortic endothelial
receptor-dependent regulation of sEH, and the enzyme's presence at cells isolated from Tie2-sEH Tr mice display reduced migration com-
specific moments during the fertility cycle indicates a specific, and pared to wild-type cells (Panigrahy et al., 2012).
possibly, novel role for sEH and EpFA in reproduction.
8.1. Acute inflammation and sepsis
7.13. Neoplasms and tumors
The injection of lipopolysaccharide (LPS), a fragment from the
A 2006 study examined the expression of human sEH in 15 differ- bacterial wall, is an established model of acute inflammation and
ent neoplastic tissues (Enayetallah et al., 2006b). The authors noticed septic shock. Recovery from acute inflammation induced by an intra-
a significant reduction in sEH protein expression compared to healthy peritoneal LPS injection of 10 mg/kg was enhanced in the null relative
tissue in renal and liver malignant neoplasms, but an increase in other to the wild-type, as assessed by a return of blood pressure to pre-
cancers, such as advanced ovarian cancer. This reduction in sEH ex- treatment levels (Luria et al., 2007). This was reflected in a higher sur-
pression was detected in the tissue surrounding the neoplasms. sEH vival rate: 100% of the sEH null mice survived the challenge while 18%
protein expression is reduced in a mouse Lewis lung carcinoma of the wild-type mice died within 24 h. Recovery of healthy blood
model, where sEH protein levels are reduced in tumor endothelial pressure in the null animals is enhanced by treatment with aspirin
cells versus normal endothelial cells, as well as in tumor lysates or the 5-lipoxygenase activation protein (FLAP) inhibitor MK886
from large versus small tumors (Panigrahy et al., 2012). sEH protein (Liu et al., 2010). Aspirin or MK886 treatment does not improve the
expression is also reduced in melanoma liver metastases versus recovery of wild-type animals, indicating that the sEH null back-
healthy liver (Panigrahy et al., 2012). These data indicate that sEH ex- ground increases the anti-inflammatory effect of these compounds.
pression is increased or decreased in specific cancers. Given the com- However, treatment with indomethacin does not produce an additive
plex process of tumor initiation, growth and metastasis, more work anti-inflammatory effect (Oni-Orisan et al., in press). sEH null mice
will be required to elucidate the role of sEH in cancer. Questions display a reduction in nuclear factor (NF)-κB signaling compared to
will include how sEH expression affects the vascularization of larger controls after 40 mg/kg LPS injection (Deng et al., 2011). They also
tumors, as well as the role the enzyme plays in preneoplastic lesions display reduced cellular adhesion molecule, chemokine and cytokine
and surrounding neoplasms that eventually develop into tumors. expression, as well as a reduction in neutrophil infiltration in lungs.
sEH null mice injected with a lower dose of LPS (1 mg/kg) did not dis-
8. sEH genetic models play a reduction in inflammatory gene expression in the liver com-
pared to controls (Fife et al., 2008). In general, these results support
The first sEH knockout mouse colony was created by disruption of the view that sEH null animals have reduced acute inflammatory
exon one by a Neo cassette (Sinal et al., 2000). Early generations of response and/or enhanced recovery from LPS challenge.
these sEH null mice displayed lower resting blood pressure than
wild-type, though females displayed no change in blood pressure. 8.2. Ischemia/reperfusion injury in the heart and brain
Later, this strain was backcrossed into C57Black/6 genetic back-
ground, and a new colony established by embryo transfer (Luria et When isolated hearts in a Langendorff apparatus are subjected to is-
al., 2007). There was no difference in resting blood pressure between chemia and reflow, hearts from sEH null mice display enhanced recovery
sEH null mice from the new colony and wild-type mice. An indepen- of left ventricular developed pressure compared to controls (Chaudhary
dent sEH null colony engineered by Boehringer Ingelheim also did not et al., in press; Seubert et al., 2006). In an in vivo model where the left cor-
display a change in blood pressure, raising the question of compensa- onary artery was surgically occluded and then released from its snare to
tory mechanisms in the knockout animals (Luria et al., 2007). The au- allow reperfusion, the infarction area was reduced in sEH null animals, in-
thors did not detect sEH protein expression in the liver, kidney, or dicating reduced injury (Motoki et al., 2008). However, in an in vivo
heart from animals of either the Boehringer or the backcrossed colony mouse model of cardiac arrest followed by cardiopulmonary resuscitation
and homogenates generated from liver and kidney displayed an in- (CPR), the knockouts displayed a longer time before restoration of spon-
crease in EpFA. However, the vasoconstrictive and hypertensive taneous circulation during CPR and a decreased survival rate (Hutchens
70 T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74

Table 2
sEH null mice and disease models.

Ref Disease Model Summary of physiological differences


compared to wild-type animals

Deng et al. (2011), Fife et al. (2008), Acute inflammation LPS-induced Increased survival
Liu et al. (2010), Luria et al. (2007) Reduced inflammatory cytokine and chemokine
expression
Enhanced return to basal blood pressure (BP)
from hypotensive state
Seubert et al. (2006) Cardiac ischemia/reperfusion injury Isolated heart in Langendorff apparatus Enhanced recovery of LVDP
Reduced infarct size
Motoki et al. (2008) Cardiac ischemia/reperfusion injury Surgical occlusion Reduced infarct size
Hutchens et al. (2008) Cardiac arrest Cardiac arrest and CPR Longer time to recover BP
Reduced survival
Zhang et al. (2008a,b) Cerebral ischemia/reperfusion injury Middle cerebral artery occlusion/reperfusion Smaller infarct
Increased cerebral blood flow
Jia et al. (2011) Brain trauma Increased blood volume at the lesion site one
month after injury
Increased rate of revascularization
Zhang et al. (2012a), Zhang et al. Inflammatory bowel disease sEH(−/−)/IL-10(−/−) fed piroxicam Reduction in inflammatory cytokines
(2012b) vs. IL-10(−/−) mice fed piroxicam Decreased ulcer formation
Decreased inflammatory cell infiltration in the
bowel
Reduced number and size of tumors
Luo et al. (2010) Diabetes Streptozotocin (STZ)-induced Improved glucose tolerance
Improved insulin secretion
Reduction in islet apoptosis
Elmarakby et al. (2011) Diabetes STZ-induced Reduced renal inflammation and damage
Chen et al. (2012) Diabetes STZ-induced Reduced renal tubular apoptosis
Luria et al. (2011) Diabetes Type II high fat dietary model Increased islet size and pancreatic vascularization
Improved insulin signaling
Keseru et al. (2010) Pulmonary Hypertension Hypoxia-induced Increased right heart hypertrophy
Enhanced pulmonary artery muscularization
Decreased exercise performance
Manhiani et al. (2009) Hypertension Deoxycorticosterone acetate treatment plus Reduction in mean arterial BP
high salt diet (DOCA–salt) Reduction in urinary MCP-1 excretion
Reduced macrophage infiltration
Reduced albuminuria
Revermann et al. (2010) Arteriosclerosis Femoral cuff in Apo(−/−) animals Reduction of neointima formation
Reduction in proinflammatory gene expression
in femoral arteries
Hercule et al. (2009) Hypertension L-NAME induced No difference in BP and EDHF responses
Inceoglu et al. (2012) Niacin flushing Reduction in flushing
Liu et al. (2012) Kidney damage Cisplatin-induced Reduction in serum urea nitrogen and creatinine
Reduction in renal tubular damage
Reduced neutrophil infiltration
Sanders et al. (2012) Wound healing Dermal wound on ear Accelerated wound closure

et al., 2008). This may reflect the altered blood pressure regulation in the test compared to wild-type mice before and after traumatic brain injury
knockout mouse reported by Luria et al. as well as the complex relation- induced by unilateral controlled cortical impact (Strauss et al., in press).
ship between sEH function in the vasculature and in heart muscle cells A promising tool for the further investigation of the role of sEH in the
(Luria et al., 2007). brain is a transgenic mouse model with neuronal over-expression of
The knockout animals display less tissue damage and enhanced re- sEH created by fusing the rat synapsin 1 promoter to a mouse sEH
covery in cerebral ischemia/reperfusion injury and acute brain trauma cDNA (Bianco et al., 2009).
models. sEH null animals subjected to a single round of middle cerebral
occlusion followed by reperfusion have a smaller infarct size (Zhang et 8.3. Inflammatory bowel disease
al., 2008b). Cerebral blood flow rates are also increased in the knockout
animals, mirroring the results in heart (Zhang et al., 2008b). Interesting- The NSAID piroxicam synergizes the development of inflammato-
ly, the authors report no difference in free 14,15-EET levels between ry bowel disease (IBD) in IL-10(−/−) mice (Zhang et al., in press). A
knockout and wild-type brains. This highlights the diverse roles of EET reduction in inflammatory cytokines was noted in IL-10(−/−)/
signaling in specific organs and disease states, and cautions against sEH(−/−) animals relative the IL-10(−/−) mice after piroxicam
making generalizations about the sEH knockout animals based on plas- treatment. There was also less ulcer formation and infiltration of
ma levels of EpFA. Compensatory mechanisms may operate differently inflammatory cells in the bowels of the IL-10(−/−)/sEH(−/−) ani-
in different tissues and produce localized effects not reflected systemi- mals. In this model, the eicosanoid profile, including several metabo-
cally. When optical micro-angiography was used to monitor revascular- lites from all three branches of the ARA cascade, was shifted away
ization in mice that had undergone a traumatic brain injury, the sEH from propagation of inflammation. Null mice displayed a lower
null mice exhibited accelerated revascularization and enhanced blood tumor multiplicity, tumor size, and a decrease in precancerous dys-
volume at the lesion site one month after the injury (Jia et al., 2011). plasia (Zhang et al., 2012b). The NSAID-induced gastrointestinal ul-
These data indicate that repair of the tissue is due to revascularization cers were also reduced in this model, an important observation,
after the injury, rather than a decrease in the damage or immediate re- particularly for individuals who must use high doses of NSAIDs for ex-
pair after injury. sEH null mice display improved motor coordination on tended periods. However, humans appear quite resistant to NSAID-
a beam walk task but an impaired performance in a Morris water maze induced gastrointestinal lesions compared to canine, equine, and feline
T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74 71

patients. An increase in the EET-to-diol ratio, including 14,15-EET, such as the lung and brain. Similarly, the localization of sEH in macro-
was noted in the plasma of the knockout and IL-10(−/−)/sEH(−/−) phages and other inflammatory cells, as well as the increase in sEH
animals compared to the IL-10(−/−) mice. expression in the region of grafts is consistent with its role in the reg-
ulation of inflammation. The interesting localization of sEH in specific
8.4. Diabetes brain regions or the tissues of the reproductive system or fat cells
may indicate as yet undiscovered functions for sEH that go beyond the
The sEH knockouts have improved function in a streptozotocin maintenance of blood pressure and regulation of inflammation. Such
(STZ)-induced model of diabetes (Luo et al., 2010). They do not suffer roles could involve pain, cognition and other neurological processes as
from hyperglycemia, and have improved insulin secretion and glu- well as reproductive processes ranging from ovulation to birth.
cose tolerance. The islet cells in the sEH knockout animals exhibit The sEH knockout mice are valuable models that have associated the
an increase in glucose-stimulated insulin secretion (GSIS) and intra- sEH protein with disease states and physiological function. The appar-
cellular calcium levels. There is also a decrease in islet cell apoptosis. ent adjustment to the genetic elimination of the sEH does complicate
This anti-apoptotic effect may contribute to the organ protection ob- interpretation of results, however. Additionally, sEH contains two active
served in brain, heart and kidney. Supporting enhanced organ protec- sites with two different catalytic activities. For this reason, comparison
tion in the nulls, knockout mice display a decrease in urinary markers of the knockouts to inhibition studies that use sEH epoxide hydrolase
of kidney inflammation and damage such as albumin and monocyte inhibitors must be made carefully. Also, the complete elimination of
chemoattractant protein-1 in the STZ model (Elmarakby et al., the sEH activity throughout the life of the mouse is expected to generate
2011). This protective effect occurs in kidney tubules, in addition to different biological effects than the transient and partial inhibition of
the pancreas. The sEH knockout animals exhibit decreased renal tu- the enzyme with pharmaceutical probes. Greater insight can be antici-
bular apoptosis when treated with STZ (Chen et al., 2012). These pated from tissue selective and temporally dependent genetic alter-
last two papers report an increase in urinary EET over DHET relative ations in sEH activity in the future. Combining these genetic tools with
to the wild-type controls. The organ protective effect observed in the pharmacological inhibitors of the sEH and mimics of EpFA that function
sEH knockout animals extends beyond the STZ models. In a model of as stable agonists and antagonists will provide still greater insight.
type-II diabetes where mice are fed a high fat diet, the knockout animals
display larger islets, increased pancreatic vasculature, and improved in-
sulin signaling (Luria et al., 2011). In the STZ and the type-II high fat diet Conflict of interest statement
models, the sEH knockout animals have improved islet function, as well
as an increased resistance to islet cell apoptosis, implicating EpFA in the The University of California, Davis has filed patents in the area of
soluble epoxide hydrolase inhibitors for the treatment of diseases.
protection of the kidney and pancreas.
All authors have read the journal's policy on disclosure of potential
conflicts of interest.
8.5. Pulmonary hypertension

sEH is implicated in the pathology of pulmonary hypertension Acknowledgments


(Keseru et al., 2010). When the sEH null mice are subjected to chronic
hypoxia, they develop a more pronounced right heart hypertrophy, This work was supported by NIEHS grant R37 ES02710. T.R.H. is
enhanced pulmonary artery muscularization, and decreased exercise also supported by NIH T32 training grant in basic and translational
performance compared to the wild-type animals. Interestingly, long cardiovascular science (T32 HL86350). B.D.H. is a George and Judy
term treatment with compounds that inhibit the epoxide hydrolase Marcus Senior Fellow of the American Asthma Society.
activity does not duplicate the phenotype observed in the knockout
mice, raising the possibility that the phosphatase activity of the en-
zyme is important to the development or progression of pulmonary References
hypertension. This is a reminder that the knockout sEH animals are Aboutabl, M.E., Zordoky, B.N., El-Kadi, A.O., 2009. 3-methylcholanthrene and benzo(a)
deficient in both epoxide hydrolase and phosphatase activities, and pyrene modulate cardiac cytochrome P450 gene expression and arachidonic acid
that examination of the difference in phenotypes obtained with in- metabolism in male Sprague Dawley rats. Br. J. Pharmacol. 158, 1808–1819.
Ai, D., et al., 2007. Angiotensin II up-regulates soluble epoxide hydrolase in vascular
hibitors and the knockout animals may give some insight into the
endothelium in vitro and in vivo. Proc. Natl. Acad. Sci. U. S. A. 104, 9018–9023.
function of the sEH phosphatase activity. There is a need for future Ai, D., et al., 2009. Soluble epoxide hydrolase plays an essential role in angiotensin
knockout and knock-in models where the sEH and phosphatase cata- II-induced cardiac hypertrophy. Proc. Natl. Acad. Sci. U. S. A. 106, 564–569.
lytic activities are dealt with separately and the possible structural Alsaad, A.M., Zordoky, B.N., El-Sherbeni, A.A., El-Kadi, A.O., 2012. Chronic doxorubicin
cardiotoxicity modulates cardiac cytochrome P450-mediated arachidonic acid
roles of the sEH protein are treated apart from the catalytic roles. metabolism in rats. Drug Metab. Dispos. 40, 2126–2135.
Althurwi, H.N., Tse, M.M., Abdelhamid, G., Zordoky, B.N., Hammock, B.D., El-Kadi, A.O.,
9. Conclusion 2013. Soluble epoxide hydrolase inhibitor, TUPS, protects against isoproterenol-
induced cardiac hypertrophy. Br. J. Pharmacol. 168, 1794–1807.
Anwar-Mohamed, A., El-Sherbeni, A.A., Kim, S.H., Althurwi, H.N., Zordoky, B.N., El-Kadi, A.O.,
This review has not fully addressed the use of substrates to distin- 2012. Acute arsenic toxicity alters cytochrome P450 and soluble epoxide hydrolase and
guish sEH from microsomal EH. Interested readers are referred to his- their associated arachidonic acid metabolism in C57Bl/6 mouse heart. Xenobiotica 42,
1235–1247.
torical reviews (Imig, 2012; Morisseau and Hammock, 2008). Also, Argiriadi, M.A., Morisseau, C., Hammock, B.D., Christianson, D.W., 1999. Detoxification
this review has not discussed sEH inhibition. sEH inhibitors are pow- of environmental mutagens and carcinogens: structure, mechanism, and evolution
erful tools for the elucidation of sEH function and are being pursued of liver epoxide hydrolase. Proc. Natl. Acad. Sci. U. S. A. 96, 10637–10642.
Beetham, J.K., Tian, T., Hammock, B.D., 1993. cDNA cloning and expression of a soluble
as possible therapies for diseases such as hypertension. Readers inter- epoxide hydrolase from human liver. Arch. Biochem. Biophys. 305, 197–201.
ested in the history of the development of sEH inhibitors are referred Beetham, J.K., et al., 1995. Gene evolution of epoxide hydrolases and recommended
to several recent reviews (Morisseau and Hammock, 2013; Shen, nomenclature. DNA Cell Biol. 14, 61–71.
Bianco, R.A., Agassandian, K., Cassell, M.D., Spector, A.A., Sigmund, C.D., 2009. Charac-
2010; Shen and Hammock, 2012) and included references.
terization of transgenic mice with neuron-specific expression of soluble epoxide
The localization of sEH in the VSMCs and endothelial cells in sev- hydrolase. Brain Res. 1291, 60–72.
eral tissues is consistent with its proposed role in the metabolism in Brenneis, C., et al., 2011. Soluble epoxide hydrolase limits mechanical hyperalgesia
the vasoactive EETs. By converting EETs to their corresponding diols, during inflammation. Mol. Pain 7, 78.
Burdon, K.P., et al., 2008. Genetic analysis of the soluble epoxide hydrolase gene,
sEH can exert a regulatory effect on vascular tone and systemic EPHX2, in subclinical cardiovascular disease in the Diabetes Heart Study. Diab.
blood pressure or create regional differences in blood flow in organs Vasc. Dis. Res. 5, 128–134.
72 T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74

Chanas, B., Wang, H., Ghanayem, B.I., 2003. Differential metabolism of acrylonitrile to Gill, S.S., Hammock, B.D., 1980. Distribution and properties of a mammalian soluble
cyanide is responsible for the greater sensitivity of male vs female mice: role of epoxide hydrase. Biochem. Pharmacol. 29, 389–395.
CYP2E1 and epoxide hydrolases. Toxicol. Appl. Pharmacol. 193, 293–302. Gomez, G.A., Morisseau, C., Hammock, B.D., Christianson, D.W., 2004. Structure of
Chang, C., Gill, S.S., 1991. Purification and characterization of an epoxide hydrolase from human epoxide hydrolase reveals mechanistic inferences on bifunctional catalysis
the peroxisomal fraction of mouse liver. Arch. Biochem. Biophys. 285, 276–284. in epoxide and phosphate ester hydrolysis. Biochemistry 43, 4716–4723.
Chaudhary, K.R., et al., 2013. Differential effects of soluble epoxide hydrolase inhibition Grant, D.F., Storms, D.H., Hammock, B.D., 1993. Molecular cloning and expression of
and CYP2J2 overexpression on postischemic cardiac function in aged mice. Prosta- murine liver soluble epoxide hydrolase. J. Biol. Chem. 268, 17628–17633.
glandins Other Lipid Mediat. (in press). Grant, D.F., et al., 1994. Chromosomal mapping and expression levels of a mouse solu-
Chawengsub, Y., et al., 2008. Identification of 15-hydroxy-11,12-epoxyeicosatrienoic ble epoxide hydrolase gene. Pharmacogenetics 4, 64–72.
acid as a vasoactive 15-lipoxygenase metabolite in rabbit aorta. Am. J. Physiol. Gschwendtner, A., et al., 2008. Genetic variation in soluble epoxide hydrolase (EPHX2)
Heart Circ. Physiol. 294, H1348–H1356. is associated with an increased risk of ischemic stroke in white Europeans. Stroke
Chen, G., et al., 2012. Genetic disruption of soluble epoxide hydrolase is protective 39, 1593–1596.
against streptozotocin-induced diabetic nephropathy. Am. J. Physiol. Endocrinol. Haeggstrom, J.Z., Funk, C.D., 2011. Lipoxygenase and leukotriene pathways: biochemis-
Metab. 303, E563–E575. try, biology, and roles in disease. Chem. Rev. 111, 5866–5898.
Corenblum, M.J., Wise, V.E., Georgi, K., Hammock, B.D., Doris, P.A., Fornage, M., 2008. Al- Hammock, B.D., Ota, K., 1983. Differential induction of cytosolic epoxide hydrolase, mi-
tered soluble epoxide hydrolase gene expression and function and vascular disease crosomal epoxide hydrolase, and glutathione S-transferase activities. Toxicol. Appl.
risk in the stroke-prone spontaneously hypertensive rat. Hypertension 51, 567–573. Pharmacol. 71, 254–265.
Corriveau, S., Berthiaume, M., Rousseau, E., Pasquier, J.C., 2009. Why eicosanoids could Hammock, B.D., Gill, S.S., Stamoudis, V., Gilbert, L.I., 1976. Soluble mammalian epoxide
represent a new class of tocolytics on uterine activity in pregnant women. Am. hydratase: action on juvenile hormone and other terpenoid epoxides. Comp.
J. Obstet. Gynecol. 201 (420), e1–e7. Biochem. Physiol. B 53, 263–265.
Cronin, A., et al., 2003. The N-terminal domain of mammalian soluble epoxide hydro- Harris, T.R., Aronov, P.A., Hammock, B.D., 2008a. Soluble epoxide hydrolase homologs
lase is a phosphatase. Proc. Natl. Acad. Sci. U. S. A. 100, 1552–1557. in Strongylocentrotus purpuratus suggest a gene duplication event and subsequent
Cui, P.H., Petrovic, N., Murray, M., 2011. The omega-3 epoxide of eicosapentaenoic acid in- divergence. DNA Cell Biol. 27, 467–477.
hibits endothelial cell proliferation by p38 MAP kinase activation and cyclin D1/CDK4 Harris, T.R., Aronov, P.A., Jones, P.D., Tanaka, H., Arand, M., Hammock, B.D., 2008b. Iden-
down-regulation. Br. J. Pharmacol. 162, 1143–1155. tification of two epoxide hydrolases in Caenorhabditis elegans that metabolize
Davis, B.B., Morisseau, C., Newman, J.W., Pedersen, T.L., Hammock, B.D., Weiss, R.H., 2006. mammalian lipid signaling molecules. Arch. Biochem. Biophys. 472, 139–149.
Attenuation of vascular smooth muscle cell proliferation by 1-cyclohexyl-3-dodecyl Hennebold, J.D., et al., 2005. Identification and characterization of an ovary-selective
urea is independent of soluble epoxide hydrolase inhibition. J. Pharmacol. Exp. isoform of epoxide hydrolase. Biol. Reprod. 72, 968–975.
Ther. 316, 815–821. Hercule, H.C., Schunck, W.H., Gross, V., Seringer, J., Leung, F.P., Weldon, S.M., da Costa
De Taeye, B.M., et al., 2010. Expression and regulation of soluble epoxide hydrolase in Goncalves, A., Huang, Y., Luft, F.C., Gollasch, M., 2009. Interaction between P450 eicosa-
adipose tissue. Obesity (Silver Spring) 18, 489–498. noids and nitric oxide in the control of arterial tone in mice. Arterioscler Thromb. Vasc.
Deng, Y., et al., 2011. Endothelial CYP epoxygenase overexpression and soluble epoxide Biol. 29, 54–60.
hydrolase disruption attenuate acute vascular inflammatory responses in mice. Hollinshead, M., Meijer, J., 1988. Immunocytochemical analysis of soluble epoxide
FASEB J. 25, 703–713. hydrolase and catalase in mouse and rat hepatocytes demonstrates a peroxisomal
Draper, A.J., Hammock, B.D., 1999. Soluble epoxide hydrolase in rat inflammatory cells is localization before and after clofibrate treatment. Eur. J. Cell Biol. 46, 394–402.
indistinguishable from soluble epoxide hydrolase in rat liver. Toxicol. Sci. 50, 30–35. Honetschlagerova, Z., et al., 2011. Renal mechanisms contributing to the antihyperten-
Dreisbach, A.W., et al., 2005. The prevalence of CYP2C8, 2C9, 2J2, and soluble epoxide sive action of soluble epoxide hydrolase inhibition in Ren-2 transgenic rats with in-
hydrolase polymorphisms in African Americans with hypertension. Am. J. Hypertens. ducible hypertension. J. Physiol. 589, 207–219.
18, 1276–1281. Hutchens, M.P., Nakano, T., Dunlap, J., Traystman, R.J., Hurn, P.D., Alkayed, N.J., 2008.
DuTeaux, S.B., et al., 2004. Epoxide hydrolases in the rat epididymis: possible roles in Soluble epoxide hydrolase gene deletion reduces survival after cardiac arrest and
xenobiotic and endogenous fatty acid metabolism. Toxicol. Sci. 78, 187–195. cardiopulmonary resuscitation. Resuscitation 76, 89–94.
Edin, M.L., et al., 2011. Endothelial expression of human cytochrome P450 epoxygenase Iliff, J.J., Close, L.N., Selden, N.R., Alkayed, N.J., 2007. A novel role for P450 eicosanoids in
CYP2C8 increases susceptibility to ischemia–reperfusion injury in isolated mouse the neurogenic control of cerebral blood flow in the rat. Exp. Physiol. 92, 653–658.
heart. FASEB J. 25, 3436–3447. Iliff, J.J., Wang, R., Zeldin, D.C., Alkayed, N.J., 2009. Epoxyeicosanoids as mediators of
Elmarakby, A.A., Faulkner, J., Al-Shabrawey, M., Wang, M.H., Maddipati, K.R., Imig, J.D., neurogenic vasodilation in cerebral vessels. Am. J. Physiol. Heart Circ. Physiol.
2011. Deletion of soluble epoxide hydrolase gene improves renal endothelial func- 296, H1352–H1363.
tion and reduces renal inflammation and injury in streptozotocin-induced type 1 Imig, J.D., 2012. Epoxides and soluble epoxide hydrolase in cardiovascular physiology.
diabetes. Am. J. Physiol. Regul. Integr. Comp. Physiol. 301, R1307–R1317. Physiol. Rev. 92, 101–130.
Enayetallah, A.E., French, R.A., Thibodeau, M.S., Grant, D.F., 2004. Distribution of soluble Imig, J.D., Hammock, B.D., 2009. Soluble epoxide hydrolase as a therapeutic target for
epoxide hydrolase and of cytochrome P450 2C8, 2C9, and 2J2 in human tissues. cardiovascular diseases. Nat. Rev. Drug Discov. 8, 794–805.
J. Histochem. Cytochem. 52, 447–454. Imig, J.D., Zhao, X., Capdevila, J.H., Morisseau, C., Hammock, B.D., 2002. Soluble epoxide
Enayetallah, A.E., French, R.A., Barber, M., Grant, D.F., 2006a. Cell-specific subcellular lo- hydrolase inhibition lowers arterial blood pressure in angiotensin II hypertension.
calization of soluble epoxide hydrolase in human tissues. J. Histochem. Cytochem. Hypertension 39, 690–694.
54, 329–335. Imig, J.D., et al., 2012. Soluble epoxide hydrolase inhibition and peroxisome proliferator
Enayetallah, A.E., French, R.A., Grant, D.F., 2006b. Distribution of soluble epoxide activated receptor gamma agonist improve vascular function and decrease renal
hydrolase, cytochrome P450 2C8, 2C9 and 2J2 in human malignant neoplasms. injury in hypertensive obese rats. Exp. Biol. Med. (Maywood) 237, 1402–1412.
J. Mol. Histol. 37, 133–141. Inceoglu, A.B., Clifton, H.L., Yang, J., Hegedus, C., Hammock, B.D., Schaefer, S., 2012. Inhibition
Eriksson, A.M., Zetterqvist, M.A., Lundgren, B., Andersson, K., Beije, B., DePierre, J.W., of soluble epoxide hydrolase limits niacin-induced vasodilation in mice. J Cardiovasc
1991. Studies on the intracellular distributions of soluble epoxide hydrolase and Pharmacol 60, 70–75.
of catalase by digitonin-permeabilization of hepatocytes isolated from control Irusta, G., Murphy, M.J., Perez, W.D., Hennebold, J.D., 2007. Dynamic expression of
and clofibrate-treated mice. Eur. J. Biochem. 198, 471–476. epoxyeicosatrienoic acid synthesizing and metabolizing enzymes in the primate
Fairbanks, S.L., Young, J.M., Nelson, J.W., Davis, C.M., Koerner, I.P., Alkayed, N.J., 2012. Mech- corpus luteum. Mol. Hum. Reprod. 13, 541–548.
anism of the sex difference in neuronal ischemic cell death. Neuroscience 219, 183–191. Jia, Y., Grafe, M.R., Gruber, A., Alkayed, N.J., Wang, R.K., 2011. In vivo optical imaging of
Fang, X., Weintraub, N.L., Stoll, L.L., Spector, A.A., 1999. Epoxyeicosatrienoic acids revascularization after brain trauma in mice. Microvasc. Res. 81, 73–80.
increase intracellular calcium concentration in vascular smooth muscle cells. Jiang, H., et al., 2008. Hydrolysis of cis- and trans-epoxyeicosatrienoic acids by rat red
Hypertension 34, 1242–1246. blood cells. J. Pharmacol. Exp. Ther. 326, 330–337.
Fava, C., et al., 2010. Homozygosity for the EPHX2 K55R polymorphism increases the Jiang, H., et al., 2011. Increases in plasma trans-EETs and blood pressure reduction in spon-
long-term risk of ischemic stroke in men: a study in Swedes. Pharmacogenet. taneously hypertensive rats. Am. J. Physiol. Heart Circ. Physiol. 300, H1990–H1996.
Genomics 20, 94–103. Johansson, C., Stark, A., Sandberg, M., Ek, B., Rask, L., Meijer, J., 1995. Tissue specific
Fife, K.L., et al., 2008. Inhibition of soluble epoxide hydrolase does not protect against basal expression of soluble murine epoxide hydrolase and effects of clofibrate on
endotoxin-mediated hepatic inflammation. J. Pharmacol. Exp. Ther. 327, 707–715. the mRNA levels in extrahepatic tissues and liver. Arch. Toxicol. 70, 61–63.
Fornage, M., et al., 2002. Polymorphism in soluble epoxide hydrolase and blood pres- Jung, O., et al., 2005. Soluble epoxide hydrolase is a main effector of angiotensin II-induced
sure in spontaneously hypertensive rats. Hypertension 40, 485–490. hypertension. Hypertension 45, 759–765.
Fornage, M., Boerwinkle, E., Doris, P.A., Jacobs, D., Liu, K., Wong, N.D., 2004. Polymor- Jung, O., et al., 2010. Inhibition of the soluble epoxide hydrolase promotes albuminuria
phism of the soluble epoxide hydrolase is associated with coronary artery calcifica- in mice with progressive renal disease. PLoS One 5, e11979.
tion in African-American subjects: the Coronary Artery Risk Development in Young Kaur, S., Gill, S.S., 1986. Distribution and nature of epoxide hydrolase activity in subcel-
Adults (CARDIA) study. Circulation 109, 335–339. lular organelles of mouse liver. Biochem. Pharmacol. 35, 1299–1308.
Fornage, M., et al., 2005. The soluble epoxide hydrolase gene harbors sequence varia- Keseru, B., et al., 2010. Hypoxia-induced pulmonary hypertension: comparison of
tion associated with susceptibility to and protection from incident ischemic stroke. soluble epoxide hydrolase deletion vs. inhibition. Cardiovasc. Res. 85,
Hum. Mol. Genet. 14, 2829–2837. 232–240.
Fromel, T., et al., 2012. Soluble epoxide hydrolase regulates hematopoietic progenitor cell Koeners, M.P., et al., 2011. Soluble epoxide hydrolase in the generation and mainte-
function via generation of fatty acid diols. Proc. Natl. Acad. Sci. U. S. A. 109, 9995–10000. nance of high blood pressure in spontaneously hypertensive rats. Am. J. Physiol.
Gao, J., et al., 2011. Soluble epoxide hydrolase inhibition prevents coronary endothelial Endocrinol. Metab. 300, E691–E698.
dysfunction in mice with renovascular hypertension. J. Hypertens. 29, 1128–1135. Koerner, I.P., et al., 2007. Polymorphisms in the human soluble epoxide hydrolase gene
Gill, S.S., Hammock, B.D., 1979. Hydration of cis- and trans-epoxymethyl stearates by the cy- EPHX2 linked to neuronal survival after ischemic injury. J. Neurosci. 27,
tosolic epoxide hydrase of mouse liver. Biochem. Biophys. Res. Commun. 89, 965–971. 4642–4649.
T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74 73

Koerner, I.P., Zhang, W., Cheng, J., Parker, S., Hurn, P.D., Alkayed, N.J., 2008. Soluble ep- inflammatory effects in the lung: role of soluble epoxide hydrolase. Am. J. Respir. Cell
oxide hydrolase: regulation by estrogen and role in the inflammatory response to Mol. Biol. 43, 564–575.
cerebral ischemia. Front. Biosci. 13, 2833–2841. Morisseau, C., Hammock, B.D., 2008. Gerry Brooks and epoxide hydrolases: four de-
Kopkan, L., et al., 2012. Soluble epoxide hydrolase inhibition exhibits antihypertensive cades to a pharmaceutical. Pest Manag. Sci. 64, 594–609.
actions independently of nitric oxide in mice with renovascular hypertension. Morisseau, C., Hammock, B.D., 2013. Impact of soluble epoxide hydrolase and
Kidney Blood Press. Res. 35, 595–607. epoxyeicosanoids on human health. Annu. Rev. Pharmacol. Toxicol. 53, 37–58.
Kullmann, S., et al., 2009. Variation in the human soluble epoxide hydrolase gene Morisseau, C., et al., 2010. Naturally occurring monoepoxides of eicosapentaenoic acid and
and risk of restenosis after percutaneous coronary intervention. BMC Cardiovasc. docosahexaenoic acid are bioactive antihyperalgesic lipids. J. Lipid Res. 51, 3481–3490.
Disord. 9, 48. Motoki, A., et al., 2008. Soluble epoxide hydrolase inhibition and gene deletion are pro-
Langenbach, R., Smith, P.B., Crespi, C., 1992. Recombinant DNA approaches for the tective against myocardial ischemia–reperfusion injury in vivo. Am. J. Physiol.
development of metabolic systems used in in vitro toxicology. Mutat. Res. 277, 251–275. Heart Circ. Physiol. 295, H2128–H2134.
Larsson, C., White, I., Johansson, C., Stark, A., Meijer, J., 1995. Localization of the human Mustafa, S., Vasudevan, H., Yuen, V.G., McNeill, J.H., 2010. Renal expression of arachi-
soluble epoxide hydrolase gene (EPHX2) to chromosomal region 8p21–p12. Hum. donic acid metabolizing enzymes and RhoA/Rho kinases in fructose insulin resis-
Genet. 95, 356–358. tant hypertensive rats. Mol. Cell. Biochem. 333, 203–209.
Lee, C.R., et al., 2006. Genetic variation in soluble epoxide hydrolase (EPHX2) and risk Nayeem, M.A., Zeldin, D.C., Boegehold, M.A., Falck, J.R., 2011. Salt modulates vascular
of coronary heart disease: the Atherosclerosis Risk in Communities (ARIC) study. response through adenosine A(2A) receptor in eNOS-null mice: role of CYP450
Hum. Mol. Genet. 15, 1640–1649. epoxygenase and soluble epoxide hydrolase. Mol. Cell. Biochem. 350, 101–111.
Lee, S.H., et al., 2008. Genetic variations in soluble epoxide hydrolase and graft function Nayeem, M.A., Pradhan, I., Mustafa, S.J., Morisseau, C., Falck, J.R., Zeldin, D.C., 2013. Adenosine
in kidney transplantation. Transplant. Proc. 40, 1353–1356. A2A receptor modulates vascular response in soluble epoxide hydrolase-null mice
Lee, J., Dahl, M., Grande, P., Tybjaerg-Hansen, A., Nordestgaard, B.G., 2010. Genetically through CYP-epoxygenases and PPARgamma. Am. J. Physiol. Regul. Integr. Comp.
reduced soluble epoxide hydrolase activity and risk of stroke and other cardiovas- Physiol. 304, R23–R32.
cular disease. Stroke 41, 27–33. Newman, J.W., et al., 2004. Cytochrome p450-dependent lipid metabolism in preovula-
Lee, C.R., et al., 2011. Genetic variation in soluble epoxide hydrolase (EPHX2) is associ- tory follicles. Endocrinology 145, 5097–5105.
ated with forearm vasodilator responses in humans. Hypertension 57, 116–122. Newman, J.W., Morisseau, C., Hammock, B.D., 2005. Epoxide hydrolases: their roles and
Li, N., et al., 2009. Beneficial effects of soluble epoxide hydrolase inhibitors in myocar- interactions with lipid metabolism. Prog. Lipid Res. 44, 1–51.
dial infarction model: insight gained using metabolomic approaches. J. Mol. Cell. Oguro, A., Fujita, N., Imaoka, S., 2009. Regulation of soluble epoxide hydrolase (sEH) in mice
Cardiol. 47, 835–845. with diabetes: high glucose suppresses sEH expression. Drug Metab. Pharmacokinet.
Liu, Y., et al., 2005. The antiinflammatory effect of laminar flow: the role of PPARgamma, 24, 438–445.
epoxyeicosatrienoic acids, and soluble epoxide hydrolase. Proc. Natl. Acad. Sci. U. S. A. Ohtoshi, K., et al., 2005. Association of soluble epoxide hydrolase gene polymorphism with
102, 16747–16752. insulin resistance in type 2 diabetic patients. Biochem. Biophys. Res. Commun. 331,
Liu, J.Y., et al., 2010. Inhibition of soluble epoxide hydrolase enhances the anti- 347–350.
inflammatory effects of aspirin and 5-lipoxygenase activation protein inhibitor in Olearczyk, J.J., Field, M.B., Kim, I.H., Morisseau, C., Hammock, B.D., Imig, J.D., 2006.
a murine model. Biochem. Pharmacol. 79, 880–887. Substituted adamantyl-urea inhibitors of the soluble epoxide hydrolase dilate
Liu, Y., Dang, H., Li, D., Pang, W., Hammock, B.D., Zhu, Y., 2012. Inhibition of soluble mesenteric resistance vessels. J. Pharmacol. Exp. Ther. 318, 1307–1314.
epoxide hydrolase attenuates high-fat-diet-induced hepatic steatosis by reduced Oni-Orisan, A., et al., 2013. Dual modulation of cyclooxygenase and CYP epoxygenase
systemic inflammatory status in mice. PLoS One 7, e39165. metabolism and acute vascular inflammation in mice. Prostaglandins Other Lipid
Luo, P., et al., 2010. Inhibition or deletion of soluble epoxide hydrolase prevents hyper- Mediat. (in press).
glycemia, promotes insulin secretion, and reduces islet apoptosis. J. Pharmacol. Pacifici, G.M., Lindberg, B., Glaumann, H., Rane, A., 1983. Styrene oxide metabolism in
Exp. Ther. 334, 430–438. rhesus monkey liver: enzyme activities in subcellular fractions and in isolated he-
Luria, A., et al., 2007. Compensatory mechanism for homeostatic blood pressure regu- patocytes. J. Pharmacol. Exp. Ther. 226, 869–875.
lation in Ephx2 gene-disrupted mice. J. Biol. Chem. 282, 2891–2898. Paczkowski, M., Krisher, R., 2010. Aberrant protein expression is associated with de-
Luria, A., et al., 2009. Alteration in plasma testosterone levels in male mice lacking creased developmental potential in porcine cumulus–oocyte complexes. Mol.
soluble epoxide hydrolase. Am. J. Physiol. Endocrinol. Metab. 297, E375–E383. Reprod. Dev. 77, 51–58.
Luria, A., et al., 2011. Soluble epoxide hydrolase deficiency alters pancreatic islet size Pang, W., Li, N., Ai, D., Niu, X.L., Guan, Y.F., Zhu, Y., 2011. Activation of peroxisome
and improves glucose homeostasis in a model of insulin resistance. Proc. Natl. proliferator-activated receptor-gamma downregulates soluble epoxide hydrolase
Acad. Sci. U. S. A. 108, 9038–9043. in cardiomyocytes. Clin. Exp. Pharmacol. Physiol. 38, 358–364.
Manhiani, M., Quigley, J.E., Knight, S.F., Tasoobshirazi, S., Moore, T., Brands, M.W., Panigrahy, D., et al., 2012. Epoxyeicosanoids stimulate multiorgan metastasis and
Hammock, B.D., Imig, J.D., 2009. Soluble epoxide hydrolase gene deletion attenuates tumor dormancy escape in mice. J. Clin. Invest. 122, 178–191.
renal injury and inflammation with DOCA-salt hypertension. Am. J. Physiol. Renal. Przybyla-Zawislak, B.D., et al., 2003. Polymorphisms in human soluble epoxide hydro-
Physiol. 297, F740–8. lase. Mol. Pharmacol. 64, 482–490.
Maresh, J.G., Xu, H., Jiang, N., Gairola, C.G., Shohet, R.V., 2005. Tobacco smoke dysregulates Qin, Y., Han, X., Peng, Y., Shen, R., Guo, X., Cao, L., Song, L., Sha, J., Xia, Y., Wang, X., 2012.
endothelial vasoregulatory transcripts in vivo. Physiol. Genomics 21, 308–313. Genetic variants in epoxide hydrolases modify the risk of oligozoospermia and
Markaverich, B.M., Crowley, J., Rodriquez, M., Shoulars, K., Thompson, T., 2007. asthenospermia in Han-Chinese population. Gene 510, 171–714.
Tetrahydrofurandiol stimulation of phospholipase A2, lipoxygenase, and cyclooxy- Revermann, M., et al., 2009. Inhibition of the soluble epoxide hydrolase attenuates
genase gene expression and MCF-7 human breast cancer cell proliferation. Envi- monocrotaline-induced pulmonary hypertension in rats. J. Hypertens. 27, 322–331.
ron. Health Perspect. 115, 1727–1731. Revermann, M., Schloss, M., Barbosa-Sicard, E., Mieth, A., Liebner, S., Morisseau, C.,
Mark-Kappeler, C.J., Martin, D.S., Eyster, K.M., 2011. Estrogens and selective estrogen Geisslinger, G., Schermuly, R.T., Fleming, I., Hammock, B.D., Brandes, R.P., 2010. Soluble
receptor modulators regulate gene and protein expression in the mesenteric arter- epoxide hydrolase deficiency attenuates neointima formation in the femoral cuff
ies. Vascul. Pharmacol. 55, 42–49. model of hyperlipidemic mice. Arterioscler Thromb. Vasc. Biol. 30, 909–914.
Marowsky, A., Burgener, J., Falck, J.R., Fritschy, J.M., Arand, M., 2009. Distribution of sol- Rodriguez, M., Clare-Salzler, M., 2006. Eicosanoid imbalance in the NOD mouse is relat-
uble and microsomal epoxide hydrolase in the mouse brain and its contribution to ed to a dysregulation in soluble epoxide hydrolase and 15-PGDH expression. Ann.
cerebral epoxyeicosatrienoic acid metabolism. Neuroscience 163, 646–661. N. Y. Acad. Sci. 1079, 130–134.
Martin, D.S., Klinkova, O., Eyster, K.M., 2012. Regional differences in sexually dimorphic Saito, S., Iida, A., Sekine, A., Eguchi, C., Miura, Y., Nakamura, Y., 2001. Seventy genetic
protein expression in the spontaneously hypertensive rat (SHR). Mol. Cell. Biochem. variations in human microsomal and soluble epoxide hydrolase genes (EPHX1
362, 103–114. and EPHX2) in the Japanese population. J. Hum. Genet. 46, 325–329.
Mavrommatis, Y., et al., 2010. Intervention with fish oil, but not with docosahexaenoic Sandberg, M., Meijer, J., 1996. Structural characterization of the human soluble epoxide
acid, results in lower levels of hepatic soluble epoxide hydrolase with time in apoE hydrolase gene (EPHX2). Biochem. Biophys. Res. Commun. 221, 333–339.
knockout mice. Br. J. Nutr. 103, 16–24. Sandberg, M., Hassett, C., Adman, E.T., Meijer, J., Omiecinski, C.J., 2000. Identification
Merabet, N., et al., 2012. Soluble epoxide hydrolase inhibition improves myocardial per- and functional characterization of human soluble epoxide hydrolase genetic poly-
fusion and function in experimental heart failure. J. Mol. Cell. Cardiol. 52, 660–666. morphisms. J. Biol. Chem. 275, 28873–28881.
Moghaddam, M., Motoba, K., Borhan, B., Pinot, F., Hammock, B.D., 1996. Novel metabolic Sanders, W.G., Morisseau, C., Hammock, B.D., Cheung, A.K., Terry, C.M., 2012. Soluble
pathways for linoleic and arachidonic acid metabolism. Biochim. Biophys. Acta 1290, epoxide hydrolase expression in a porcine model of arteriovenous graft stenosis
327–339. and anti-inflammatory effects of a soluble epoxide hydrolase inhibitor. Am. J. Phys-
Moghaddam, M.F., Grant, D.F., Cheek, J.M., Greene, J.F., Williamson, K.C., Hammock, iol. Cell Physiol. 303, C278–C290.
B.D., 1997. Bioactivation of leukotoxins to their toxic diols by epoxide hydrolase. Sellers, K.W., et al., 2005. Novel mechanism of brain soluble epoxide hydrolase-mediated
Nat. Med. 3, 562–566. blood pressure regulation in the spontaneously hypertensive rat. FASEB J. 19,
Monti, J., et al., 2008. Soluble epoxide hydrolase is a susceptibility factor for heart failure 626–628.
in a rat model of human disease. Nat. Genet. 40, 529–537. Senouvo, F.Y., Tabet, Y., Morin, C., Albadine, R., Sirois, C., Rousseau, E., 2011. Improved
Moody, D.E., Loury, D.N., Hammock, B.D., 1985. Epoxide metabolism in the liver of mice bioavailability of epoxyeicosatrienoic acids reduces TP-receptor agonist-induced
treated with clofibrate (ethyl-alpha-(p-chlorophenoxyisobutyrate)), a peroxisome tension in human bronchi. Am. J. Physiol. Lung Cell. Mol. Physiol. 301, L675–L682.
proliferator. Toxicol. Appl. Pharmacol. 78, 351–362. Seubert, J.M., et al., 2005. Differential renal gene expression in prehypertensive and hyper-
Morgan, L.A., et al., 2013. Soluble epoxide hydrolase inhibition does not prevent cardiac re- tensive spontaneously hypertensive rats. Am. J. Physiol. Renal Physiol. 289, F552–F561.
modeling and dysfunction following aortic constriction in rats and mice. J. Cardiovasc. Seubert, J.M., et al., 2006. Role of soluble epoxide hydrolase in postischemic recovery of
Pharmacol. (in press). heart contractile function. Circ. Res. 99, 442–450.
Morin, C., Sirois, M., Echave, V., Albadine, R., Rousseau, E., 2010. 17,18-Epoxyeicosatetraenoic Shen, H.C., 2010. Soluble epoxide hydrolase inhibitors: a patent review. Expert Opin.
acid targets PPARgamma and p38 mitogen-activated protein kinase to mediate its anti- Ther. Pat. 20, 941–956.
74 T.R. Harris, B.D. Hammock / Gene 526 (2013) 61–74

Shen, H.C., Hammock, B.D., 2012. Discovery of inhibitors of soluble epoxide hydro- Xu, D., et al., 2006. Prevention and reversal of cardiac hypertrophy by soluble epoxide
lase: a target with multiple potential therapeutic indications. J. Med. Chem. 55, hydrolase inhibitors. Proc. Natl. Acad. Sci. U. S. A. 103, 18733–18738.
1789–1808. Xu, D.Y., et al., 2013. A potent soluble epoxide hydrolase inhibitor, t-AUCB, acts through
Sinal, C.J., Miyata, M., Tohkin, M., Nagata, K., Bend, J.R., Gonzalez, F.J., 2000. Targeted PPARgamma to modulate the function of endothelial progenitor cells from patients
disruption of soluble epoxide hydrolase reveals a role in blood pressure regulation. with acute myocardial infarction. Int. J. Cardiol. (in press).
J. Biol. Chem. 275, 40504–40510. Yu, Z., et al., 2000. Soluble epoxide hydrolase regulates hydrolysis of vasoactive
Smith, W.L., Urade, Y., Jakobsson, P.J., 2011. Enzymes of the cyclooxygenase pathways epoxyeicosatrienoic acids. Circ. Res. 87, 992–998.
of prostanoid biosynthesis. Chem. Rev. 111, 5821–5865. Yu, Z., et al., 2004. Vascular localization of soluble epoxide hydrolase in the human
Spector, A.A., 2009. Arachidonic acid cytochrome P450 epoxygenase pathway. J. Lipid kidney. Am. J. Physiol. Renal Physiol. 286, F720–F726.
Res. 50 (Suppl.), S52–S56. Zhang, L., et al., 2008a. Genetic variation in cytochrome P450 2J2 and soluble epoxide
Spector, A.A., Norris, A.W., 2007. Action of epoxyeicosatrienoic acids on cellular func- hydrolase and risk of ischemic stroke in a Chinese population. Pharmacogenet.
tion. Am. J. Physiol. Cell Physiol. 292, C996–C1012. Genomics 18, 45–51.
Srivastava, P.K., Sharma, V.K., Kalonia, D.S., Grant, D.F., 2004. Polymorphisms in human Zhang, W., et al., 2008b. Soluble epoxide hydrolase gene deletion is protective against
soluble epoxide hydrolase: effects on enzyme activity, enzyme stability, and qua- experimental cerebral ischemia. Stroke 39, 2073–2078.
ternary structure. Arch. Biochem. Biophys. 427, 164–169. Zhang, W., Iliff, J.J., Campbell, C.J., Wang, R.K., Hurn, P.D., Alkayed, N.J., 2009. Role of
Strauss, K.I., Gruzdev, A., Zeldin, D.C., 2013. Altered behavioral phenotypes in soluble soluble epoxide hydrolase in the sex-specific vascular response to cerebral ischemia.
epoxide hydrolase knockout mice: effects of traumatic brain injury. Prostaglandins J. Cereb. Blood Flow Metab. 29, 1475–1481.
Other Lipid Mediat. (in press). Zhang, D., Ai, D., Tanaka, H., Hammock, B.D., Zhu, Y., 2010. DNA methylation of the pro-
Sura, P., Sura, R., Enayetallah, A.E., Grant, D.F., 2008. Distribution and expression of moter of soluble epoxide hydrolase silences its expression by an SP-1-dependent
soluble epoxide hydrolase in human brain. J. Histochem. Cytochem. 56, 551–559. mechanism. Biochim. Biophys. Acta 1799, 659–667.
Tanaka, H., et al., 2008. Transcriptional regulation of the human soluble epoxide hydro- Zhang, D., Xie, X., Chen, Y., Hammock, B.D., Kong, W., Zhu, Y., 2012a. Homocysteine
lase gene EPHX2. Biochim. Biophys. Acta 1779, 17–27. upregulates soluble epoxide hydrolase in vascular endothelium in vitro and in
Thomas, H., Schladt, L., Knehr, M., Oesch, F., 1989. Effect of diabetes and starvation on vivo. Circ. Res. 110, 808–817.
the activity of rat liver epoxide hydrolases, glutathione S-transferases and peroxi- Zhang, W., et al., 2012b. Soluble epoxide hydrolase gene deficiency or inhibition atten-
somal beta-oxidation. Biochem. Pharmacol. 38, 4291–4297. uates chronic active inflammatory bowel disease in IL-10(−/−) mice. Dig. Dis. Sci.
Ulu, A., et al., 2013. Anti-inflammatory effects of omega-3 polyunsaturated fatty acids 57, 2580–2591.
and soluble epoxide hydrolase inhibitors in angiotensin-II dependent hyperten- Zhang, W., et al., 2013. Reduction of inflammatory bowel disease-induced tumor devel-
sion. J. Cardiovasc. Pharmacol. (in press). opment in IL-10 knockout mice with soluble epoxide hydrolase gene deficiency.
Waechter, F., Bentley, P., Bieri, F., Staubli, W., Volkl, A., Fahimi, H.D., 1983. Epoxide hy- Mol. Carcinog. (in press).
drolase activity in isolated peroxisomes of mouse liver. FEBS Lett. 158, 225–228. Zhang, G.D., et al., 2013. Epoxy metabolites of docosahexaenoic acid (DHA) inhibit
Wagner, K., Inceoglu, B., Hammock, B.D., 2011. Soluble epoxide hydrolase inhibition, angiogenesis and tumorigenesis. Proc. Natl. Acad. Sci. U. S. A. 110, 6530–6535.
epoxygenated fatty acids and nociception. Prostaglandins Other Lipid Mediat. 96, Zhao, X., et al., 2004. Soluble epoxide hydrolase inhibition protects the kidney from
76–83. hypertension-induced damage. J. Am. Soc. Nephrol. 15, 1244–1253.
Walkowska, A., et al., 2010. Intrarenal cytochrome P-450 metabolites of arachidonic Zhao, X., et al., 2005. Decreased epoxygenase and increased epoxide hydrolase expres-
acid in the regulation of the nonclipped kidney function in two-kidney, one-clip sion in the mesenteric artery of obese Zucker rats. Am. J. Physiol. Regul. Integr.
Goldblatt hypertensive rats. J. Hypertens. 28, 582–593. Comp. Physiol. 288, R188–R196.
Wang, S.B., et al., 2012. Protection of salvianolic acid A on rat brain from ischemic damage Zheng, J., Plopper, C.G., Lakritz, J., Storms, D.H., Hammock, B.D., 2001. Leukotoxin-diol:
via soluble epoxide hydrolase inhibition. J. Asian Nat. Prod. Res. 14, 1084–1092. a putative toxic mediator involved in acute respiratory distress syndrome. Am.
Wang, Q., Pang, W., Cui, Z., Shi, J., Liu, Y., Liu, B., Zhou, Y., Guan, Y., Hammock, B.D., J. Respir. Cell Mol. Biol. 25, 434–438.
Wang, Y., Zhu, Y., 2013. Upregulation of soluble epoxide hydrolase in proximal Zordoky, B.N., Aboutabl, M.E., El-Kadi, A.O., 2008. Modulation of cytochrome P450 gene
tubular cells mediated proteinuria-induced renal damage. Am. J. Physiol. Renal. expression and arachidonic acid metabolism during isoproterenol-induced cardiac
Physiol. 304, F168–176. hypertrophy in rats. Drug Metab. Dispos. 36, 2277–2286.
Wei, Q., et al., 2007. Sequence variation in the soluble epoxide hydrolase gene and sub- Zordoky, B.N., Anwar-Mohamed, A., Aboutabl, M.E., El-Kadi, A.O., 2011. Acute doxorubicin
clinical coronary atherosclerosis: interaction with cigarette smoking. Atherosclerosis toxicity differentially alters cytochrome P450 expression and arachidonic acid
190, 26–34. metabolism in rat kidney and liver. Drug Metab. Dispos. 39, 1440–1450.

You might also like