EPHX1 Human

Epoxide Hydrolase 1 Microsomal Human Recombinant
Shipped with Ice Packs
In Stock

Description

EPHX1 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 451 amino acids (21-455a.a) and having a molecular mass of 52.2kDa.
EPHX1 is fused to a 16 amino acid T7-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Epoxide Hydrolase 1 Microsomal (EPHX1), a crucial enzyme in biotransformation, converts epoxides generated from aromatic compound degradation into trans-dihydrodiols. These trans-dihydrodiols can then be conjugated and eliminated from the body. EPHX1 plays a dual role in both the activation and detoxification of epoxides. Mutations in the EPHX1 gene can lead to various conditions, including preeclampsia, epoxide hydrolase deficiency, or elevated epoxide hydrolase activity.
Description
Recombinant human EPHX1, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 451 amino acids (21-455a.a). With a molecular weight of 52.2kDa, it includes a 16 amino acid T7-tag fused at the N-terminus. Purification is achieved through proprietary chromatographic methods.
Physical Appearance
A sterile, filtered solution that is colorless.
Formulation
The EPHX1 solution (0.25mg/ml) is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4M Urea.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended periods, store frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. It's essential to avoid repeated freeze-thaw cycles.
Purity
The purity of the product is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Epoxide hydrolase 1, microsomal (xenobiotic), MEH, EPHX, EPOX, Epoxide hydratase, Microsomal epoxide hydrolase.
Source
Escherichia Coli.
Amino Acid Sequence
MASMTGGQQM GRGSHMRDKE ETLPLEDGWW GPGTRSAARE DDSIRPFKVE TSDEEIHDLH QRIDKFRFTP PLEDSCFHYG FNSNYLKKVI SYWRNEFDWK KQVEILNRYP HFKTKIEGLD IHFIHVKPPQ LPAGHTPKPL LMVHGWPGSF YEFYKIIPLL TDPKNHGLSD EHVFEVICPS IPGYGFSEAS SKKGFNSVAT ARIFYKLMLR LGFQEFYIQG GDWGSLICTN MAQLVPSHVK GLHLNMALVL SNFSTLTLLL GQRFGRFLGL TERDVELLYP VKEKVFYSLM RESGYMHIQC TKPDTVGSAL NDSPVGLAAY ILEKFSTWTN TEFRYLEDGG LERKFSLDDL LTNVMLYWTT GTIISSQRFY KENLGQGWMT QKHERMKVYV PTGFSAFPFE LLHTPEKWVR FKYPKLISYS YMVRGGHFAA FEEPELLAQD IRKFLSVLER Q.

Q&A

What is the genomic organization of human EPHX1?

Human EPHX1 is located on chromosome 1 (1q42.12), spans approximately 35 kb, and consists of nine exons . The gene encodes three transcription variants that differ in the 5'-untranslated region, while each translated protein product contains 455 amino acids . The complex regulation of EPHX1 expression involves alternative promoters - the proximal E1 promoter driving liver expression, and the alternative E1-b promoter controlling expression in other tissues . An additional novel human EPHX1 transcript (E1-b′) has been identified with highest expression in human ovary, suggesting a potential transcription-independent regulatory role .

How is EPHX1 expression regulated at the transcriptional level?

EPHX1 expression is regulated by a diverse array of transcription factors binding to specific regulatory sequences. Key transcriptional regulators include:

Transcription FactorRole in EPHX1 RegulationOMIM Number
GATA4Major activator600576
HNF3 (FOXA1, FOXB1)Co-repressor in HepG2 cells602294, 600288
CEBPAInteracts with DNA-bound NFY116897
NFY (A, B, C subunits)Regulator of EPHX1 expression189903, 189904, 605344
HNF4A, CAR (NR1I3), RXRBind to proximal EPHX1 promoter600281, 603881, 180245/180246

Additionally, hormonal regulation has been observed, with insulin positively and glucagon negatively regulating EPHX1 expression in rat hepatocytes, while progesterone regulates EPHX1 expression in the endometrium during the menstrual cycle .

What is the tissue distribution pattern of EPHX1 in humans?

EPHX1 shows tissue-specific, age-specific, and sex-specific expression patterns with high inter-individual variation among humans . The highest EPHX1 transcript and protein levels are found in human liver and skin . EPHX1 transcripts have been detected in human primary bronchial epithelial cells but not in alveolar macrophages . Strong to moderate immunohistochemical EPHX1 protein staining has been observed in synovial blood vessels and lining cells . The topology of EPHX1 on the cell surface varies considerably between different cell types, which may have important functional implications for substrate accessibility and metabolism .

What are the most common polymorphisms in human EPHX1?

The human EPHX1 protein exhibits polymorphism primarily at two amino acid positions: Y113H (tyrosine to histidine at position 113) and H139R (histidine to arginine at position 139) . These substitutions result in four possible allelic variants: Y113/H139 (reference protein), H113/H139, Y113/R139, and H113/R139 . While additional non-synonymous SNPs have been identified, the Y113H and H139R variants remain the most common in human populations . Currently, there are 142 EPHX1 gene variations listed in the National Cancer Institute dbSNP database, including 18 copy number variations and 4 SNPs with potential clinical significance .

How do EPHX1 polymorphisms affect enzyme activity?

The functional impact of EPHX1 polymorphisms has been investigated using both purified protein preparations and human liver microsomes:

EPHX1 VariantRelative Activity with cSO/BaPO (Purified Protein)Activity in Human Liver Microsomes
Y113/H139 (reference)~2-fold greater than other variantsNo major differences between genotypes
H113/H139Reduced compared to referenceNo major differences between genotypes
Y113/R139Reduced compared to referenceNo major differences between genotypes
H113/R139Reduced compared to referenceNot specifically measured

What methodological approaches can be used to analyze EPHX1 polymorphisms?

For investigating EPHX1 polymorphisms and their functional consequences, researchers can employ the following methodological approaches:

  • Genotyping methods:

    • PCR-RFLP (Polymerase Chain Reaction-Restriction Fragment Length Polymorphism)

    • Real-time PCR with allele-specific probes

    • Sanger sequencing or next-generation sequencing

  • Functional analysis methods:

    • Expression of recombinant EPHX1 variants in heterologous systems (e.g., baculovirus-infected Sf9 cells)

    • Purification by column chromatography for in vitro enzymatic assays

    • Analysis of human liver microsomes from individuals with known EPHX1 genotypes

    • Western immunoblot analysis to determine specific EPHX1 content in samples

  • Substrate specificity assessment:

    • Using model substrates like cis-stilbene oxide (cSO) and benzo[a]pyrene-4,5-oxide (BaPO)

    • Measuring formation of trans-dihydrodiol metabolites by HPLC or other analytical methods

How does EPHX1 contribute to carcinogenesis and cancer susceptibility?

EPHX1 plays a dual role in cancer development through its ability to both detoxify and bioactivate carcinogenic compounds:

  • Detoxification: EPHX1 typically converts epoxides to trans-dihydrodiols, which are generally less reactive and more water-soluble, facilitating excretion .

  • Bioactivation: In certain instances, the initial trans-dihydrodiol metabolites can be further activated by subsequent P450 catalysis to form highly electrophilic and reactive dihydrodiol-epoxides that can form covalent adducts with DNA in a stereoselective manner .

Epidemiological studies have investigated associations between EPHX1 polymorphisms and cancer risk with varying results:

  • Hepatocellular carcinoma (HCC): Initially associated with the EPHX1 H113 allele in a Chinese population, particularly relevant given EPHX1's role in aflatoxin B1 metabolism, but subsequent studies showed conflicting results .

  • Other cancers: EPHX1 polymorphisms have been variously associated with colorectal polyp formation, lung cancer, orolaryngeal cancer, and sensitivity to 1,3-butadiene .

These contradictory findings may result from differences in study populations, environmental exposures, or interactions with other genetic polymorphisms that collectively influence cancer risk.

What hereditary disorders are associated with EPHX1 mutations?

Mutations and polymorphisms in EPHX1 have been linked to several hereditary disorders:

DisorderOMIM NumberAssociation with EPHX1
Preeclampsia189800EPHX1 mutations may contribute to development
Hypercholanemia607748EPHX1 mutations may cause this disorder
Fetal hydantoin syndrome-EPHX1 is suspected to contribute
Craniofacial abnormalities-Two maternally transmitted EPHX1 SNPs (rs1051740 and rs2234922) associated with risk in children of women taking phenytoin during pregnancy

The mechanisms by which EPHX1 variants contribute to these disorders likely involve altered metabolism of endogenous or exogenous substrates that affect specific physiological processes .

What are the methodological challenges in studying EPHX1-disease associations?

Researchers investigating EPHX1-disease associations face several methodological challenges:

  • Expression variability: High inter-individual variation in EPHX1 expression may confound genotype-phenotype correlations .

  • Tissue specificity: EPHX1 shows tissue-specific, age-specific, and sex-specific expression patterns, necessitating careful study design and tissue selection .

  • Functional complexity: The dual role of EPHX1 in both detoxification and bioactivation makes it difficult to predict the net effect of polymorphisms on disease risk .

  • In vitro vs. in vivo correlation: Limited correlation between in vitro enzymatic activities and in vivo function, as demonstrated by differences between purified proteins and human liver microsomes .

  • Study design considerations:

    • Need for large, well-characterized study populations

    • Importance of controlling for environmental exposures

    • Consideration of gene-gene interactions with other biotransformation enzymes

How can researchers investigate the regulatory mechanisms of EPHX1 expression?

The complex regulation of EPHX1 gene expression can be studied using the following experimental approaches:

  • Promoter analysis:

    • Reporter gene assays with EPHX1 promoter constructs to identify regulatory elements

    • Deletion and mutation analysis to map specific transcription factor binding sites

    • Investigation of DNAseI hypersensitive sites (HS-1 and HS-2) in the intronic region between E-1b and E1

  • Transcription factor binding:

    • Chromatin immunoprecipitation (ChIP) to detect binding of transcription factors (GATA4, HNF3, CEBPA, NFY, HNF4A, CAR, RXR) to the EPHX1 promoter

    • Electrophoretic mobility shift assays (EMSA) to confirm protein-DNA interactions

    • Co-immunoprecipitation to study protein-protein interactions among transcription factors

  • Post-transcriptional regulation:

    • Analysis of upstream open reading frames (uORFs) and their impact on translation efficiency

    • Investigation of the novel E1-b′ transcript and its potential transcription-independent regulatory role

    • RNA stability assays to determine post-transcriptional control mechanisms

  • Tissue-specific expression:

    • Comparison of E1 and E1-b promoter activities across different tissues

    • Analysis of Sp1/Sp3 binding sites in the E1-b promoter region

    • Single-cell RNA sequencing to characterize cell-type-specific expression patterns

What are the optimal methods for assessing EPHX1 enzymatic activity?

For measuring EPHX1 enzymatic activity in research settings, the following methodological approaches are recommended:

  • Protein expression systems:

    • Baculovirus-infected Spodoptera frugiperda-9 (Sf9) insect cells for expression of recombinant EPHX1 variants

    • Purification by column chromatography to obtain homogeneous protein preparations

  • Enzyme sources:

    • Purified recombinant EPHX1 proteins

    • Human liver microsomes from donors with characterized EPHX1 genotypes

    • Cell lines with different levels of EPHX1 expression

  • Activity assays:

    • Using model substrates like cis-stilbene oxide (cSO) and benzo[a]pyrene-4,5-oxide (BaPO)

    • HPLC or LC-MS/MS analysis to measure formation of trans-dihydrodiol metabolites

    • Spectrophotometric assays for high-throughput screening

  • Normalization methods:

    • Western immunoblot analysis to determine specific EPHX1 content in samples

    • Calculation of specific activity (activity per unit of EPHX1 protein)

    • Internal standards for quantitative analysis

  • Experimental controls:

    • Inclusion of positive and negative controls

    • Use of specific EPHX1 inhibitors to confirm enzyme specificity

    • Comparative analysis of different EPHX1 variants under identical conditions

How can researchers study EPHX1 interactions with other biotransformation enzymes?

The functional role of EPHX1 in xenobiotic metabolism often involves interactions with other biotransformation enzymes, particularly cytochrome P450s. To study these interactions, researchers can employ:

  • Co-expression systems:

    • Co-expression of EPHX1 with relevant P450 enzymes in cellular models

    • Sequential metabolism studies using purified enzymes

    • Microsomes containing both EPHX1 and P450 enzymes

  • Metabolic pathway analysis:

    • Metabolite profiling using LC-MS/MS

    • Tracking formation of intermediates and final metabolites

    • Inhibitor studies to block specific steps in the biotransformation pathway

  • Genetic association studies:

    • Analysis of combined genotypes (EPHX1 plus other biotransformation enzymes)

    • Haplotype analysis across multiple genes

    • Gene-gene interaction modeling in epidemiological studies

  • Structural biology approaches:

    • Protein-protein interaction studies

    • Molecular modeling of enzyme complexes

    • Investigation of membrane co-localization of biotransformation enzymes

  • Systems biology approaches:

    • Pathway-based analysis of xenobiotic metabolism

    • Integration of transcriptomic, proteomic, and metabolomic data

    • Mathematical modeling of multi-enzyme metabolic networks

What are emerging areas of EPHX1 research beyond xenobiotic metabolism?

While EPHX1 is primarily known for its role in xenobiotic metabolism, several emerging research directions extend beyond this traditional focus:

  • Endogenous substrate metabolism:

    • Identification of endogenous epoxide-containing lipids as EPHX1 substrates

    • Role in signaling lipid metabolism and regulation

    • Potential impact on inflammatory pathways and immune regulation

  • Developmental biology:

    • Investigation of EPHX1's role in embryonic development

    • Mechanisms underlying the association with craniofacial abnormalities in fetal hydantoin syndrome

    • Potential interactions with developmental signaling pathways

  • Cell biology:

    • Significance of variable EPHX1 topology across different cell types

    • Role of cell-surface EPHX1 in cell-cell interactions

    • Impact of EPHX1 localization on its access to substrates

  • Precision medicine applications:

    • Development of EPHX1 genotyping panels for personalized drug therapy

    • Prediction of individual susceptibility to environmental toxicants

    • EPHX1-based biomarkers for disease risk assessment

How can contradictory findings in EPHX1 epidemiological studies be reconciled?

The literature contains numerous contradictory findings regarding associations between EPHX1 polymorphisms and disease risk. To reconcile these contradictions, researchers should consider:

  • Meta-analysis approaches:

    • Systematic reviews combining data from multiple studies

    • Stratification by ethnicity, exposure, and other relevant factors

    • Assessment of publication bias and study quality

  • Gene-environment interaction analysis:

    • Evaluation of specific environmental exposures that may modify the effect of EPHX1 variants

    • Investigation of dose-response relationships

    • Consideration of timing and duration of exposures

  • Advanced genetic approaches:

    • Whole gene sequencing rather than focusing solely on common polymorphisms

    • Haplotype analysis to capture combined effects of multiple variants

    • Epigenetic studies to assess EPHX1 regulation beyond genetic sequence

  • Functional validation:

    • Correlation of epidemiological findings with functional assays

    • Development of more physiologically relevant in vitro models

    • Use of humanized animal models expressing human EPHX1 variants

What technological advances might enhance EPHX1 research in the coming decade?

Several emerging technologies and methodological advances are likely to significantly impact EPHX1 research:

  • CRISPR-Cas9 genome editing:

    • Creation of precise EPHX1 variants in cell lines and animal models

    • Generation of conditional and tissue-specific EPHX1 knockout models

    • High-throughput screening of EPHX1 regulatory elements

  • Single-cell technologies:

    • Single-cell RNA sequencing to characterize cell-type-specific EPHX1 expression patterns

    • Single-cell proteomics to assess EPHX1 protein levels and modifications

    • Spatial transcriptomics to map EPHX1 expression within tissue architecture

  • Structural biology advances:

    • Cryo-electron microscopy for high-resolution EPHX1 structure determination

    • Molecular dynamics simulations to understand polymorphism effects on protein function

    • Structure-based drug design for EPHX1 modulators

  • Artificial intelligence and machine learning:

    • Prediction of EPHX1 substrate specificity and activity

    • Integration of multi-omics data to understand EPHX1 function in complex systems

    • Development of models to predict individual response to xenobiotics based on EPHX1 genotype

  • Organoid and microphysiological systems:

    • 3D organoid models to study EPHX1 function in tissue-specific contexts

    • Liver-on-a-chip and other organ models to assess EPHX1-mediated metabolism

    • Multi-tissue microphysiological systems to study systemic effects of EPHX1 activity

Product Science Overview

Introduction

Epoxide Hydrolase 1 (EH1), also known as microsomal epoxide hydrolase (mEH) or EPHX1, is an enzyme that plays a crucial role in the metabolism of both xenobiotic and endogenous epoxides. This enzyme is part of the hydrolase family and is involved in the detoxification processes within the body. The recombinant form of this enzyme, produced through genetic engineering techniques, is used extensively in research and therapeutic applications.

Structure and Function

mEH is a membrane-bound enzyme found in the endoplasmic reticulum of cells. It catalyzes the hydrolysis of epoxides to their corresponding diols, which are less reactive and more water-soluble, facilitating their excretion from the body. This reaction is essential for the detoxification of harmful compounds, including those derived from environmental pollutants, drugs, and endogenous metabolic processes .

Genetic Variants and Polymorphisms

The EPHX1 gene, located on chromosome 1q42.1, encodes the mEH enzyme. Several polymorphisms in this gene have been identified, which can affect the enzyme’s activity. For instance, the His139Arg polymorphism has been associated with variations in enzyme activity and susceptibility to certain diseases . These genetic variations can influence an individual’s response to environmental toxins and their risk of developing conditions such as cancer, preeclampsia, and neurological disorders .

Biological Roles and Clinical Significance

While mEH is primarily known for its role in xenobiotic metabolism, it also has significant endogenous functions. It metabolizes epoxides derived from fatty acids, such as epoxyeicosatrienoic acids (EETs), which are involved in various physiological processes, including inflammation and cardiovascular regulation . The enzyme’s activity can influence the levels of these bioactive lipids, thereby impacting cardiovascular health and disease .

Recombinant mEH in Research and Therapeutics

The recombinant form of mEH is produced using human cells or other expression systems to ensure it closely mimics the natural enzyme. This recombinant enzyme is used in various research applications, including the study of drug metabolism, toxicology, and the development of therapeutic interventions. For example, recombinant mEH is utilized in enzyme-linked immunosorbent assays (ELISAs) to quantify the enzyme’s levels in biological samples, aiding in the diagnosis and monitoring of diseases .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.