Microsomal epoxide hydrolase

Last updated
Microsomal epoxide hydrolase
Microsomal Epoxide Hydrolase.png
A cartoon depiction of microsomal epoxide hydrolase from Aspergillus niger
Identifiers
EC no. 3.3.2.9
Databases
IntEnz IntEnz view
BRENDA BRENDA entry
ExPASy NiceZyme view
KEGG KEGG entry
MetaCyc metabolic pathway
PRIAM profile
PDB structures RCSB PDB PDBe PDBsum
Search
PMC articles
PubMed articles
NCBI proteins

In enzymology, a microsomal epoxide hydrolase (mEH) (EC 3.3.2.9) is an enzyme that catalyzes the hydrolysis reaction between an epoxide and water to form a diol.

Contents

Hydrolysis of an epoxide ring Epoxide Hydrolysis.png
Hydrolysis of an epoxide ring

This enzyme plays a role in the uptake of bile salts within the large intestine. It functions as a Na+ dependent transporter. This enzyme participates in metabolism of xenobiotics by cytochrome p450. mEH has been identified as playing a large role in the detoxification and bioactivation of a wide variety of substrates, such as polycyclic aromatic hydrocarbons (PAHs), which are known for their carcinogenic properties. [1]

The human homolog of microsomal epoxide hydrolase is EPHX1 and is located on chromosome 1. [2]

Nomenclature

This enzyme belongs to the family of hydrolases, specifically those acting on ether bonds (ether hydrolases). The systematic name of this enzyme class is cis-stilbene-oxide hydrolase. Other names in common use include epoxide hydratase (ambiguous), microsomal epoxide hydratase (ambiguous), epoxide hydrase, microsomal epoxide hydrase, arene-oxide hydratase (ambiguous), benzo[a]pyrene-4,5-oxide hydratase, benzo(a)pyrene-4,5-epoxide hydratase, aryl epoxide hydrase (ambiguous), cis-epoxide hydrolase, and mEH.

Structure

Microsomal epoxide hydrolase is a single polypeptide chain composed of 455 amino acids with a molecular weight of 52.96 kilodaltons. It is known that the N-terminal region of the enzyme is responsible for anchoring the protein to the cell membrane, [3] while the C-terminal region of the enzyme contains catalytic residues. [4] Microsomal epoxide hydrolase belongs to the superfamily α/β-hydrolase fold enzymes. [5] The center of all α/β-hydrolase fold enzymes is an alpha/beta-sheet consisted of 8 beta strands connected by 6 alpha helices. [6] [7] The three dimensional structure of mEH has been elucidated from Aspergillus niger . [4] Although no 3D modeling has been solved for the mammalian mEH enzyme (EPHX1), the overall homology between fungal and mammalian mEH is relatively high. [8] [9] [10] This high homology has allowed for the elucidation overall general structure and subsequent catalytic mechanism of EPHX1 in humans by comparisons to existing structures of fungal mEH.

Mechanism

α/β-hydrolase fold enzymes use a catalytic triad in their active site. The catalytic triad present in microsomal epoxide hydrolase is composed of glutamine, histidine and aspartic acid. [10] The substrate is positioned in an orientation poised for nucleophilic attack through hydrogen bonding stabilization from two nearby tyrosine residues [11] [12] The proposed mechanism for the mEH-catalyzed reaction first involves a nucleophilic attack on the oxirane ring of the substrate from the aspartic acid residue near the active site, which forms an ester intermediate. [13] The second step in this mechanism is hydrolysis of the ester that occurs by an activated water molecule. [14] The activation of water is facilitated by proton abstraction via the catalytic triad between a water molecule, glutamine, and histidine. [15] After hydrolysis, the substrate is then released from its bond to the aspartic acid residue, liberating the diol product from the enzyme active site. [16]

The mechanism of microsomal epoxide hydrolase MEH-Mechanism.pdf
The mechanism of microsomal epoxide hydrolase

The active site of this enzyme lies within a hydrophobic pocket in the enzyme, which in turn leads to the enzyme's preferential reactivity with molecules with hydrophobic side-chains. [17] [11] The mEH enzyme typically binds to small organic epoxides, such as styrene epoxide and cis-stillbene-oxide. mEH does not catalyze the hydrolysis of bulkier molecules, as their large side-chains may sterically disrupt the charge relay system responsible for water activation. [11]

Active site of mEH from Aspergillus niger bound to small molecule 2-propoylpentanamide. MEH Active Site.png
Active site of mEH from Aspergillus niger bound to small molecule 2-propoylpentanamide.

Function

In humans, mEH has been found in the ovary, lung, kidney, lymphocytes, epithelial cells, and liver. [18] Microsomal epoxide hydrolase serves as a protective enzyme against potentially harmful small molecules derived from the external environment. [19] This hydrolysis of genotoxic epoxides causes subsequent effects in several signal transduction pathways, rendering this enzyme important to metabolism. [20] [21]

Disease relevance

Microsomal epoxide hydrolase plays a large role in its effects on human health. Studies have shown that mutations EPHX1 in humans may be the cause of hypercholanemia, [22] preeclampsia, [23] [24] and may contribute to fetal hydantoin syndrome. [25] Research also suggests that maternal polymorphisms in EPHX1 in pregnant women were related to facial malformations of children born from women taking phenytoin during their first trimester of pregnancy. [26] While mEH participates in the protection of human health via detoxification of various environmental substances, it also has been found to facilitate the activation of carcinogens. [1]

mEH detoxifies reactive epoxides that are commonly caused from cigarette smoke, and as such it is hypothesized that mutations in EPHX1 in humans may have an effect on an individual's susceptibility to COPD, emphysema and lung cancer. Some sources have demonstrated that individuals affected by COPD have a higher rate of containing an under-active variant of the EPHX1 gene, yet also demonstrated that the overactive variant of the gene was also found in higher frequencies in individuals affected by disease as well. [27] [28] Other research has provided evidence supporting the idea that EPHX1 variants do not contribute to susceptibility of disease, but do contribute to disease severity. [1] The role that mEH plays in lung cancer and COPD is still not fully elucidated, as the data on the topic in the literature is not completely unanimous. [29]

There is some evidence that mEH variants may contribute to the occurrence of childhood asthma in combination with variants on the GSTP1 gene. [30]

Compared to soluble epoxide hydrolase, the contribution of mEH to metabolism of beneficial epoxy fatty acids such as Epoxyeicosatrienoic acid is considered minor since they are relatively poor mEH substrates in vitro. Yet, in vivo, it was found that mEH can play a considerable role in regulation of EET levels [31] [32] and hence inhibition of mEH or dual inhibition of mEH and sEH might have therapeutic potential. Amide, amine and urea based mEH inhibitors have been explored. [33] Based on the most potent inhibitors characterized, an amide with a bulky alpha-substituent and a phenyl ring with lipophilic groups at meta-positions appear to be key pharmacophore units. [34]

The overall effect that mEH has on human health is still debated, with some sources finding evidence that the overactive EPHX1 gene is the culprit for some diseases, while other evidence supports that the under active variant is the cause of others.

Related Research Articles

<span class="mw-page-title-main">CYP2E1</span> Protein-coding gene in the species Homo sapiens

Cytochrome P450 2E1 is a member of the cytochrome P450 mixed-function oxidase system, which is involved in the metabolism of xenobiotics in the body. This class of enzymes is divided up into a number of subcategories, including CYP1, CYP2, and CYP3, which as a group are largely responsible for the breakdown of foreign compounds in mammals.

<span class="mw-page-title-main">Epoxide hydrolase</span> Enzyme that metabolizes compounds containing epoxides

Epoxide hydrolases (EHs), also known as epoxide hydratases, are enzymes that metabolize compounds that contain an epoxide residue; they convert this residue to two hydroxyl residues through an epoxide hydrolysis reaction to form diol products. Several enzymes possess EH activity. Microsomal epoxide hydrolase, soluble epoxide hydrolase, and the more recently discovered but not as yet well defined functionally, epoxide hydrolase 3 (EH3) and epoxide hydrolase 4 (EH4) are structurally closely related isozymes. Other enzymes with epoxide hydrolase activity include leukotriene A4 hydrolase, Cholesterol-5,6-oxide hydrolase, MEST (gene) (Peg1/MEST), and Hepoxilin-epoxide hydrolase. The hydrolases are distinguished from each other by their substrate preferences and, directly related to this, their functions.

β-Glucuronidase Class of enzymes

β-Glucuronidases are members of the glycosidase family of enzymes that catalyze breakdown of complex carbohydrates. Human β-glucuronidase is a type of glucuronidase that catalyzes hydrolysis of β-D-glucuronic acid residues from the non-reducing end of mucopolysaccharides such as heparan sulfate. Human β-glucuronidase is located in the lysosome. In the gut, brush border β-glucuronidase converts conjugated bilirubin to the unconjugated form for reabsorption. β-Glucuronidase is also present in breast milk, which contributes to neonatal jaundice. The protein is encoded by the GUSB gene in humans and by the uidA gene in bacteria.

<span class="mw-page-title-main">Glucocerebrosidase</span> Mammalian protein found in humans

β-Glucocerebrosidase is an enzyme with glucosylceramidase activity that cleaves by hydrolysis the β-glycosidic linkage of the chemical glucocerebroside, an intermediate in glycolipid metabolism that is abundant in cell membranes. It is localized in the lysosome, where it remains associated with the lysosomal membrane. β-Glucocerebrosidase is 497 amino acids in length and has a molecular mass of 59,700 Da.

The epoxyeicosatrienoic acids or EETs are signaling molecules formed within various types of cells by the metabolism of arachidonic acid by a specific subset of Cytochrome P450 enzymes termed cytochrome P450 epoxygenases. These nonclassic eicosanoids are generally short-lived, being rapidly converted from epoxides to less active or inactive dihydroxy-eicosatrienoic acids (diHETrEs) by a widely distributed cellular enzyme, Soluble epoxide hydrolase (sEH), also termed Epoxide hydrolase 2. The EETs consequently function as transiently acting, short-range hormones; that is, they work locally to regulate the function of the cells that produce them or of nearby cells. The EETs have been most studied in animal models where they show the ability to lower blood pressure possibly by a) stimulating arterial vasorelaxation and b) inhibiting the kidney's retention of salts and water to decrease intravascular blood volume. In these models, EETs prevent arterial occlusive diseases such as heart attacks and brain strokes not only by their anti-hypertension action but possibly also by their anti-inflammatory effects on blood vessels, their inhibition of platelet activation and thereby blood clotting, and/or their promotion of pro-fibrinolytic removal of blood clots. With respect to their effects on the heart, the EETs are often termed cardio-protective. Beyond these cardiovascular actions that may prevent various cardiovascular diseases, studies have implicated the EETs in the pathological growth of certain types of cancer and in the physiological and possibly pathological perception of neuropathic pain. While studies to date imply that the EETs, EET-forming epoxygenases, and EET-inactivating sEH can be manipulated to control a wide range of human diseases, clinical studies have yet to prove this. Determination of the role of the EETS in human diseases is made particularly difficult because of the large number of EET-forming epoxygenases, large number of epoxygenase substrates other than arachidonic acid, and the large number of activities, some of which may be pathological or injurious, that the EETs possess.

Thomas Alwyn Jones (born 30 August 1947) is a Welsh biophysicist and a professor at the Uppsala University in Sweden.

In enzymology, a hepoxilin-epoxide hydrolase is an enzyme that catalyzes the conversion of the epoxyalcohol metabolites arachidonic acid, hepoxilin A3 and hepoxilin B3 to their tri-hydroxyl products, trioxolin A3 and trioxilin B3, respectively. These reactions in general inactivate the two biologically active hepoxilins.

<span class="mw-page-title-main">Limonene-1,2-epoxide hydrolase</span>

In enzymology, a limonene-1,2-epoxide hydrolase (EC 3.3.2.8) is an enzyme that catalyzes the chemical reaction

The enzyme carboxylesterase (or carboxylic-ester hydrolase, EC 3.1.1.1; systematic name carboxylic-ester hydrolase) catalyzes reactions of the following form:

The enzyme lipid-phosphate phosphatase (EC 3.1.3.76) catalyzes the reaction

<span class="mw-page-title-main">Carboxylesterase 1</span> Protein-coding gene in the species Homo sapiens

Liver carboxylesterase 1 also known as carboxylesterase 1 is an enzyme that in humans is encoded by the CES1 gene. The protein is also historically known as serine esterase 1 (SES1), monocyte esterase and cholesterol ester hydrolase (CEH). Three transcript variants encoding three different isoforms have been found for this gene. The various protein products from isoform a, b and c range in size from 568, 567 and 566 amino acids long, respectively.

<span class="mw-page-title-main">Microsomal glutathione S-transferase 1</span> Protein-coding gene in the species Homo sapiens

Microsomal glutathione S-transferase 1 is an enzyme that in humans is encoded by the MGST1 gene.

<span class="mw-page-title-main">CYP4F8</span> Protein-coding gene in the species Homo sapiens

Cytochrome P450 4F8 is a protein that in humans is encoded by the CYP4F8 gene.

<span class="mw-page-title-main">CYP4F12</span> Protein-coding gene in the species Homo sapiens

Cytochrome P450 4F12 is a protein that in humans is encoded by the CYP4F12 gene.

<span class="mw-page-title-main">EPHX1</span> Protein-coding gene in the species Homo sapiens

Epoxide hydrolase 1 is an enzyme encoded by the EPHX1 gene in humans.

Epoxygenases are a set of membrane-bound, heme-containing cytochrome P450 enzymes that metabolize polyunsaturated fatty acids to epoxide products that have a range of biological activities. The most thoroughly studied substrate of the CYP epoxylgenases is arachidonic acid. This polyunsaturated fatty acid is metabolized by cyclooxygenases to various prostaglandin, thromboxane, and prostacyclin metabolites in what has been termed the first pathway of eicosanoid production; it is also metabolized by various lipoxygenases to hydroxyeicosatetraenoic acids and leukotrienes in what has been termed the second pathway of eicosanoid production. The metabolism of arachidonic acid to epoxyeicosatrienoic acids by the CYP epoxygenases has been termed the third pathway of eicosanoid metabolism. Like the first two pathways of eicosanoid production, this third pathway acts as a signaling pathway wherein a set of enzymes metabolize arachidonic acid to a set of products that act as secondary signals to work in activating their parent or nearby cells and thereby orchestrate functional responses. However, none of these three pathways is limited to metabolizing arachidonic acid to eicosanoids. Rather, they also metabolize other polyunsaturated fatty acids to products that are structurally analogous to the eicosanoids but often have different bioactivity profiles. This is particularly true for the CYP epoxygenases which in general act on a broader range of polyunsaturated fatty acids to form a broader range of metabolites than the first and second pathways of eicosanoid production. Furthermore, the latter pathways form metabolites many of which act on cells by binding with and thereby activating specific and well-characterized receptor proteins; no such receptors have been fully characterized for the epoxide metabolites. Finally, there are relatively few metabolite-forming lipoxygenases and cyclooxygenases in the first and second pathways and these oxygenase enzymes share similarity between humans and other mammalian animal models. The third pathway consists of a large number of metabolite-forming CYP epoxygenases and the human epoxygenases have important differences from those of animal models. Partly because of these differences, it has been difficult to define clear roles for the epoxygenase-epoxide pathways in human physiology and pathology.

<span class="mw-page-title-main">Epoxide hydrolase 2</span> Protein-coding gene in the species Homo sapiens

Soluble epoxide hydrolase (sEH) is a bifunctional enzyme that in humans is encoded by the EPHX2 gene. sEH is a member of the epoxide hydrolase family. This enzyme, found in both the cytosol and peroxisomes, binds to specific epoxides and converts them to the corresponding diols. A different region of this protein also has lipid-phosphate phosphatase activity. Mutations in the EPHX2 gene have been associated with familial hypercholesterolemia.

Cholesterol-5,6-oxide hydrolase (EC 3.3.2.11, cholesterol-epoxide hydrolase, ChEH) is an enzyme with systematic name 5,6alpha-epoxy-5alpha-cholestan-3beta-ol hydrolase. This enzyme catalyses the following chemical reaction

<span class="mw-page-title-main">Coronaric acid</span> Chemical compound

Coronaric acid (isoleukotoxin) is a mono-unsaturated, epoxide derivative of the di-saturated fatty acid, linoleic acid (i.e. 9(Z),12(Z) octadecadienoic acid). It is a mixture of the two optically active isomers of 12(Z) 9,10-epoxy-octadecenoic acid. This mixture is also termed 9,10-epoxy-12Z-octadecenoic acid or 9(10)-EpOME and when formed by or studied in mammalians, isoleukotoxin.

<span class="mw-page-title-main">Epoxide hydrolase 3</span> Protein-coding gene in the species Homo sapiens

Epoxide hydrolase 3 is a protein that in humans is encoded by the EPHX3 gene. It is the third defined isozyme in a set of epoxide hydrolase isozymes, the epoxide hydrolases. This set includes the Microsomal epoxide hydrolase ; the epoxide hydrolase 2 ; and the far less well defined enzymatically, epoxide hydrolase 4. All four enzyme contain an Alpha/beta hydrolase fold suggesting that they have Hydrolysis activity. EH1, EH2, and EH3 have been shown to have such activity in that they add water to epoxides of unsaturated fatty acids to form vicinal cis products; the activity of EH4 has not been reported. The former three EH's differ in subcellular location, tissue expression patterns, substrate preferences, and thereby functions. These functions include limiting the biologically actions of certain fatty acid epoxides, increasing the toxicity of other fatty acid epoxides, and contributing to the metabolism of drugs and other xenobiotics.

References

  1. 1 2 3 Kiyohara C, Yoshimasu K, Takayama K, Nakanishi Y (January 2006). "EPHX1 polymorphisms and the risk of lung cancer: a HuGE review". Epidemiology. 17 (1): 89–99. doi: 10.1097/01.ede.0000187627.70026.23 . PMID   16357600.
  2. Jackson MR, Craft JA, Burchell B (September 1987). "Nucleotide and deduced amino acid sequence of human liver microsomal epoxide hydrolase". Nucleic Acids Research. 15 (17): 7188. doi:10.1093/nar/15.17.7188. PMC   306212 . PMID   3502697.
  3. Craft JA, Baird S, Lamont M, Burchell B (August 1990). "Membrane topology of epoxide hydrolase". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1046 (1): 32–9. doi:10.1016/0005-2760(90)90091-B. PMID   2397243.
  4. 1 2 3 PDB: 3G0I ; Zou J, Hallberg BM, Bergfors T, Oesch F, Arand M, Mowbray SL, Jones TA (February 2000). "Structure of Aspergillus niger epoxide hydrolase at 1.8 A resolution: implications for the structure and function of the mammalian microsomal class of epoxide hydrolases". Structure. 8 (2): 111–22. doi: 10.1016/S0969-2126(00)00087-3 . PMID   10673439.
  5. Ollis DL, Cheah E, Cygler M, Dijkstra B, Frolow F, Franken SM, Harel M, Remington SJ, Silman I, Schrag J, Sussman JL (April 1992). "The α/β hydrolase fold. Protein Engineering, Design and Selection" (PDF). Protein Eng. 5 (3): 197–211. doi:10.1093/protein/5.3.197. hdl: 11370/2d4c057d-1a67-437d-ad10-701f7a60f1e6 . PMID   1409539.
  6. Ollis DL, Cheah E, Cygler M, Dijkstra B, Frolow F, Franken SM, Harel M, Remington SJ, Silman I, Schrag J (April 1992). "The alpha/beta hydrolase fold" (PDF). Protein Engineering. 5 (3): 197–211. doi:10.1093/protein/5.3.197. hdl: 11370/2d4c057d-1a67-437d-ad10-701f7a60f1e6 . PMID   1409539.
  7. Carr PD, Ollis DL (2009). "Alpha/beta hydrolase fold: an update". Protein and Peptide Letters. 16 (10): 1137–48. doi:10.2174/092986609789071298. PMID   19508187.
  8. Arand M, Oesch F (2002-02-14). Mammalian Xenobiotic Epoxide Hydrolases. John Wiley & Sons, Ltd. pp. 459–483. doi:10.1002/0470846305.ch12. ISBN   9780470846308.{{cite book}}: |work= ignored (help)
  9. Arand M, Hemmer H, Dürk H, Baratti J, Archelas A, Furstoss R, Oesch F (November 1999). "Cloning and molecular characterization of a soluble epoxide hydrolase from Aspergillus niger that is related to mammalian microsomal epoxide hydrolase". The Biochemical Journal. 344 (1): 273–80. doi:10.1042/0264-6021:3440273. PMC   1220641 . PMID   10548561.
  10. 1 2 3 Arand M, Müller F, Mecky A, Hinz W, Urban P, Pompon D, Kellner R, Oesch F (January 1999). "Catalytic triad of microsomal epoxide hydrolase: replacement of Glu404 with Asp leads to a strongly increased turnover rate". The Biochemical Journal. 337 (1): 37–43. doi:10.1042/0264-6021:3370037. PMC   1219933 . PMID   9854022.
  11. 1 2 3 4 Lewis DF, Lake BG, Bird MG (June 2005). "Molecular modelling of human microsomal epoxide hydrolase (EH) by homology with a fungal (Aspergillus niger) EH crystal structure of 1.8 A resolution: structure-activity relationships in epoxides inhibiting EH activity". Toxicology in Vitro. 19 (4): 517–22. doi:10.1016/j.tiv.2004.07.001. PMID   15826809.
  12. 1 2 Saenz-Méndez P, Katz A, Pérez-Kempner ML, Ventura ON, Vázquez M (April 2017). "Structural insights into human microsomal epoxide hydrolase by combined homology modeling, molecular dynamics simulations, and molecular docking calculations". Proteins. 85 (4): 720–730. doi:10.1002/prot.25251. PMID   28120429. S2CID   9772104.
  13. 1 2 Lacourciere GM, Armstrong RN (November 1993). "The catalytic mechanism of microsomal epoxide hydrolase involves an ester intermediate". Journal of the American Chemical Society. 115 (22): 10466–10467. doi:10.1021/ja00075a115.
  14. McCall PM, Srivastava S, Perry SL, Kovar DR, Gardel ML, Tirrell MV (April 2018). "Partitioning and Enhanced Self-Assembly of Actin in Polypeptide Coacervates". Biophysical Journal. 114 (7): 1636–1645. Bibcode:2018BpJ...114.1636M. doi:10.1016/j.bpj.2018.02.020. PMC   5954293 . PMID   29642033.
  15. Oesch F, Herrero ME, Hengstler JG, Lohmann M, Arand M (May 2000). "Metabolic detoxification: implications for thresholds". Toxicologic Pathology. 28 (3): 382–7. doi: 10.1177/019262330002800305 . PMID   10862554.
  16. Reetz MT, Bocola M, Wang LW, Sanchis J, Cronin A, Arand M, Zou J, Archelas A, Bottalla AL, Naworyta A, Mowbray SL (June 2009). "Directed evolution of an enantioselective epoxide hydrolase: uncovering the source of enantioselectivity at each evolutionary stage". Journal of the American Chemical Society. 131 (21): 7334–43. doi:10.1021/ja809673d. PMID   19469578.
  17. Václavíková R, Hughes DJ, Souček P (October 2015). "Microsomal epoxide hydrolase 1 (EPHX1): Gene, structure, function, and role in human disease". Gene. 571 (1): 1–8. doi:10.1016/j.gene.2015.07.071. PMC   4544754 . PMID   26216302.
  18. Bachmann K (2009). "Chapter 8: Drug Metabolism". Pharmacology. Elsevier. pp. 131–173. doi:10.1016/b978-0-12-369521-5.00008-7. ISBN   978-0-12-369521-5.
  19. Oesch F (May 1973). "Mammalian epoxide hydrases: inducible enzymes catalysing the inactivation of carcinogenic and cytotoxic metabolites derived from aromatic and olefinic compounds". Xenobiotica; the Fate of Foreign Compounds in Biological Systems. 3 (5): 305–40. doi:10.3109/00498257309151525. PMID   4584115.
  20. Samuelsson B, Dahlén SE, Lindgren JA, Rouzer CA, Serhan CN (September 1987). "Leukotrienes and lipoxins: structures, biosynthesis, and biological effects". Science. 237 (4819): 1171–6. Bibcode:1987Sci...237.1171S. doi:10.1126/science.2820055. PMID   2820055.
  21. Moghaddam MF, Grant DF, Cheek JM, Greene JF, Williamson KC, Hammock BD (May 1997). "Bioactivation of leukotoxins to their toxic diols by epoxide hydrolase". Nature Medicine. 3 (5): 562–6. doi:10.1038/nm0597-562. PMC   7095900 . PMID   9142128.
  22. Zhu QS, Xing W, Qian B, von Dippe P, Shneider BL, Fox VL, Levy D (July 2003). "Inhibition of human m-epoxide hydrolase gene expression in a case of hypercholanemia". Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 1638 (3): 208–16. doi:10.1016/s0925-4439(03)00085-1. PMID   12878321.
  23. Zusterzeel PL, Rütten H, Roelofs HM, Peters WH, Steegers EA (February 2001). "Protein carbonyls in decidua and placenta of pre-eclamptic women as markers for oxidative stress". Placenta. 22 (2–3): 213–9. doi:10.1053/plac.2000.0606. PMID   11170826.
  24. Laasanen J, Romppanen EL, Hiltunen M, Helisalmi S, Mannermaa A, Punnonen K, Heinonen S (September 2002). "Two exonic single nucleotide polymorphisms in the microsomal epoxide hydrolase gene are jointly associated with preeclampsia". European Journal of Human Genetics. 10 (9): 569–73. doi: 10.1038/sj.ejhg.5200849 . PMID   12173035.
  25. Buehler BA, Delimont D, van Waes M, Finnell RH (May 1990). "Prenatal prediction of risk of the fetal hydantoin syndrome". The New England Journal of Medicine. 322 (22): 1567–72. doi: 10.1056/NEJM199005313222204 . PMID   2336087.
  26. Azzato EM, Chen RA, Wacholder S, Chanock SJ, Klebanoff MA, Caporaso NE (January 2010). "Maternal EPHX1 polymorphisms and risk of phenytoin-induced congenital malformations". Pharmacogenetics and Genomics. 20 (1): 58–63. doi:10.1097/fpc.0b013e328334b6a3. PMID   19952982. S2CID   29336596.
  27. Smith CA, Harrison DJ (August 1997). "Association between polymorphism in gene for microsomal epoxide hydrolase and susceptibility to emphysema". Lancet. 350 (9078): 630–3. doi:10.1016/s0140-6736(96)08061-0. PMID   9288046. S2CID   23974600.
  28. Brøgger J, Steen VM, Eiken HG, Gulsvik A, Bakke P (April 2006). "Genetic association between COPD and polymorphisms in TNF, ADRB2 and EPHX1". The European Respiratory Journal. 27 (4): 682–8. doi: 10.1183/09031936.06.00057005 . PMID   16585076.
  29. Postma DS, Silverman EK (2009). "Chapter 4 - Genetics of Asthma and COPD". Genetics of Asthma and COPD. Elsevier. pp. 37–51. doi:10.1016/b978-0-12-374001-4.00004-3. ISBN   9780123740014.
  30. Salam MT, Lin PC, Avol EL, Gauderman WJ, Gilliland FD (December 2007). "Microsomal epoxide hydrolase, glutathione S-transferase P1, traffic and childhood asthma". Thorax. 62 (12): 1050–7. doi:10.1136/thx.2007.080127. PMC   2094290 . PMID   17711870.
  31. Marowsky A, Burgener J, Falck JR, Fritschy JM, Arand M (June 2009). "Distribution of Soluble and Microsomal Epoxide Hydrolase in the Mouse Brain and Its Contribution to Cerebral Epoxyeicosatrienoic Acid Metabolism". Neuroscience. 163 (2): 646–661. doi:10.1016/j.neuroscience.2009.06.033. PMID   19540314. S2CID   25808698.
  32. Edin ML, Hamedani BG, Gruzdev A, Graves JP, Lih FB, Arbes SJ, Singh R, Leon AO, Bradbury JA, DeGraff LM, Hoopes SL, Arand M, Zeldin DC (January 2018). "Epoxide hydrolase 1 (EPHX1) hydrolyzes epoxyeicosanoids and impairs cardiac recovery after ischemia". The Journal of Biological Chemistry. 293 (9): 3281–3292. doi: 10.1074/jbc.RA117.000298 . PMC   5836130 . PMID   29298899.
  33. Morisseau C, Newman JW, Dowdy DL, Goodrow MH, Hammock BD (April 2001). "Inhibition of Microsomal Epoxide Hydrolases by Ureas, Amides, and Amines". Chemical Research in Toxicology. 14 (4): 409–415. doi:10.1021/tx0001732. PMID   11304129.
  34. Barnych B, Singh N, Negrel S, Zhang Y, Magis D, Roux C, Hua X, Ding Z, Morisseau C, Tantillo DJ, Siegel JB, Hammock BD (March 2020). "Development of potent inhibitors of the human microsomal epoxide hydrolase". European Journal of Medicinal Chemistry. 193: 112206. doi: 10.1016/j.ejmech.2020.112206 . PMC   7366823 . PMID   32203787.

Further reading