EPHX1 Human, Sf9

Epoxide Hydrolase 1 Microsomal Human Recombinant, sf9
Shipped with Ice Packs
In Stock

Description

Introduction to EPHX1 Human, Sf9

EPHX1 Human, Sf9 refers to recombinant human microsomal epoxide hydrolase (EC 3.3.2.9) expressed in Spodoptera frugiperda (Sf9) insect cells. This enzyme catalyzes the hydrolysis of epoxides to trans-dihydrodiols, playing dual roles in detoxifying xenobiotics and bioactivating procarcinogens . The Sf9 system is preferred for producing functional EPHX1 due to its ability to perform post-translational modifications and high-yield expression .

Production and Purification in Sf9 Cells

EPHX1 is expressed in Sf9 cells via baculovirus-mediated transfection. Key steps include:

  1. Infection Optimization: Viral titer, multiplicity of infection, and expression duration (5–6 days) are calibrated to maximize protein yield .

  2. Purification: Proteins are purified using chromatography (e.g., ion exchange) with >85% purity confirmed via SDS-PAGE .

  3. Formulation: The final product is stored in 20 mM Tris-HCl buffer (pH 8.0) with 50% glycerol and 1 mM DTT to maintain stability .

Table 1: Production Parameters of EPHX1 Human, Sf9

ParameterDetailSource
Host SystemSpodoptera frugiperda (Sf9) insect cells
Expression VectorBaculovirus
Purification MethodColumn chromatography, ion exchange
Purity>85%
Molecular Mass~51.5 kDa (including tags)

Enzyme Activity and Polymorphism Studies

Table 2: Functional Impact of EPHX1 Polymorphisms

Variant (rsID)Substrate Activity (cSO/BaPO)Human Liver MicrosomesClinical RelevanceSource
Y113H (rs1051740)↓ (in vitro)No differenceLung cancer risk
H139R (rs2234922)↓ (in vitro)No differenceCOPD susceptibility
p.Thr333ProLoss of activity (aggregation)Not studiedLipoatrophic diabetes
  • Key Findings:

    • Sf9-expressed EPHX1 variants show reduced hydrolysis rates in vitro but no significant differences in human liver microsomes, highlighting species-specific regulatory mechanisms .

    • Missense mutations (e.g., p.Thr333Pro, p.Gly430Arg) cause ER aggregation and loss of function, linked to metabolic disorders .

Clinical and Research Applications

EPHX1 Human, Sf9 is critical for:

  1. Cancer Research: Studying bioactivation of procarcinogens (e.g., polycyclic aromatic hydrocarbons) and genetic susceptibility .

  2. Neurological Disorders: Investigating roles in Alzheimer’s disease and drug dependency via EET metabolism .

  3. Diagnostic Tools: Development of nanobody-based ELISAs for quantifying EPHX1 in biological samples .

Product Specs

Introduction
Epoxide Hydrolase 1 Microsomal (EPHX1), also known as microsomal epoxide hydrolase (mEH), is an essential enzyme involved in the detoxification of reactive epoxides derived from the metabolism of various compounds, including drugs, environmental toxins, and endogenous metabolites. EPHX1 plays a crucial role in converting these epoxides into less reactive and more readily excretable diols, thereby protecting cells from oxidative stress and potential damage.
Description
EPHX1, expressed in Sf9 insect cells, is a single, glycosylated polypeptide chain with a molecular weight of 51.5 kDa. The recombinant protein consists of 442 amino acids, spanning residues 21 to 455, and includes a 6-amino acid His tag at the C-terminus to facilitate purification. The protein has been purified using proprietary chromatographic techniques to ensure high purity.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The EPHX1 protein solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 50% glycerol, 1 mM DTT, and 0.1 M NaCl.
Stability
For short-term storage (up to 2-4 weeks), the EPHX1 protein solution can be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. To further enhance stability during long-term storage, the addition of a carrier protein such as HSA or BSA (0.1%) is recommended. Avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of EPHX1 is determined to be greater than 85% as analyzed by SDS-PAGE.
Synonyms

Epoxide hydrolase 1, Epoxide hydratase, Microsomal epoxide hydrolase, Meh, EPHX1, EPHX, EPOX, Epoxide Hydrolase 1 Microsomal, Microsomal Epoxide Hydrolase,
EC 3.3.2.9, HYL1

Source

Sf9, Insect cells.

Amino Acid Sequence

MRDKEETLPL EDGWWGPGTR SAAREDDSIR PFKVETSDEE IHDLHQRIDK FRFTPPLEDS CFHYGFNSNY LKKVISYWRN EFDWKKQVEI LNRYPHFKTK IEGLDIHFIH VKPPQLPAGH TPKPLLMVHG WPGSFYEFYK IIPLLTDPKN HGLSDEHVFE VICPSIPGYG FSEASSKKGF NSVATARIFY KLMLRLGFQE FYIQGGDWGS LICTNMAQLV PSHVKGLHLN MALVLSNFST LTLLLGQRFG RFLGLTERDV ELLYPVKEKV FYSLMRESGY MHIQCTKPDT VGSALNDSPV GLAAYILEKF STWTNTEFRY LEDGGLERKF SLDDLLTNVM LYWTTGTIIS SQRFYKENLG QGWMTQKHER MKVYVPTGFS AFPFELLHTP EKWVRFKYPK LISYSYMVRG GHFAAFEEPE LLAQDIRKFL SVLERQHHHH HH.

Q&A

What is EPHX1 and what is its biological significance?

EPHX1 (Microsomal epoxide hydrolase 1) is an enzyme encoded by the EPHX1 gene in humans that plays a crucial role in the metabolism of potentially carcinogenic or genotoxic epoxides, including those derived from polyaromatic hydrocarbons. EPHX1 typically catalyzes the hydrolysis of epoxides to trans-dihydrodiols, which is important for both detoxification of harmful compounds and, in some cases, bioactivation of certain compounds to more reactive metabolites . EPHX1 protein is predominantly found in the membrane fraction of the endoplasmic reticulum, with highest expression in the liver, followed by adrenal gland, lung, kidney, and intestine .

Why are Sf9 cells commonly used for expressing human EPHX1?

Sf9 (Spodoptera frugiperda-9) insect cells provide an excellent system for expressing human EPHX1 because they allow for high-level expression of functional eukaryotic proteins with proper folding and post-translational modifications. The baculovirus expression system used with Sf9 cells enables researchers to produce sufficient quantities of protein for purification and enzymatic studies . This expression system is particularly valuable for studying membrane-associated proteins like EPHX1, and it allows for the expression of multiple protein variants under identical conditions, facilitating direct functional comparisons between polymorphic variants .

What are the major EPHX1 polymorphisms and their functional significance?

The most studied EPHX1 polymorphisms are Y113H (rs1051740) and H139R (rs2234922), which result in amino acid substitutions at positions 113 (tyrosine to histidine) and 139 (histidine to arginine), respectively . These polymorphisms have significant functional consequences:

  • Y113H polymorphism reduces EPHX1 activity by approximately 39%

  • H139R polymorphism increases activity by about 25% in vitro

The distribution of these polymorphisms varies across populations:

  • Y113H: Most common in East Asian (MAF ~48%) and European populations (MAF ~30%), less common in African populations (MAF ~14%)

  • H139R: Most common in African populations (MAF ~35%), less common in European (MAF ~16%) and East Asian populations (MAF ~12%)

These genetic variations have been studied for associations with multiple disease outcomes, including various cancers and preeclampsia .

What is the complete workflow for expressing human EPHX1 variants in Sf9 cells?

The expression of human EPHX1 variants in Sf9 cells follows a systematic workflow:

  • Cloning: Amplify the coding region of EPHX1 alleles using PCR with primers containing appropriate restriction sites (e.g., NotI, PstI)

  • Vector construction: Clone the amplified fragments into a baculovirus transfer vector (e.g., pFastBac)

  • Bacmid generation: Transform competent DH10Bac cells with the recombinant plasmids to generate recombinant bacmid DNA through site-specific transposition

  • Transfection: Transfect Sf9 cells with the bacmid DNA to produce recombinant baculovirus

  • Viral stock production: Harvest and amplify the initial viral stock for subsequent infections

  • Optimization: Determine optimal viral titer, multiplicity of infection, and expression time

  • Scale-up: Grow infected cells in suspension cultures (typically in Grace's Insect Medium with 10% fetal calf serum) for 5-6 days

  • Harvest: Collect cells, wash with PBS, and freeze cell pellets until purification

  • Purification: Extract and purify EPHX1 protein using appropriate buffers (e.g., 10 mM potassium phosphate buffer, pH 7.4, containing 1% Genapol C-100) and column chromatography

  • Quality control: Verify purified protein by Western blot analysis and enzymatic activity assays

This systematic approach ensures the production of functional EPHX1 protein variants for comparative enzymatic studies.

How should I optimize baculovirus infection conditions for maximum EPHX1 expression?

Optimization of baculovirus infection for EPHX1 expression requires systematic evaluation of several parameters:

  • Viral titer: Accurately quantify your viral stock using plaque assay or other methods

  • Multiplicity of infection (MOI): Test a range of MOI values (typically 1-10) to determine optimal viral load

  • Cell density at infection: Usually optimal at mid-log phase (1.5-2.0 × 10^6 cells/mL)

  • Expression time: Monitor EPHX1 expression at 24-hour intervals from 48 to 120 hours post-infection; optimal expression is typically achieved after 5-6 days for EPHX1

  • Temperature: Maintain cells at 27°C for optimal protein folding

  • Media composition: Use Grace's Insect Medium supplemented with 10% fetal calf serum

  • Monitoring expression: Use Western blot with EPHX1-specific antibodies and enzymatic activity assays with cis-stilbene oxide (cSO) to track expression levels

Each batch of recombinant baculovirus may have slightly different infection characteristics, so optimization should be performed when switching to new viral stocks. The goal is to identify conditions that provide the best balance between protein yield and enzymatic activity.

What purification strategy is most effective for EPHX1 expressed in Sf9 cells?

The most effective purification strategy for EPHX1 from Sf9 cells involves:

  • Cell lysis: Resuspend cell pellets in 10 mM potassium phosphate buffer (pH 7.4) containing 1% Genapol C-100 to solubilize membrane-bound EPHX1

  • Centrifugation: Remove cell debris by centrifugation

  • Column chromatography sequence: Following the method of Lacourciere et al. with modifications, using potassium phosphate buffer instead of Tris-HCl

    • Ion exchange chromatography

    • Hydrophobic interaction chromatography

    • Additional purification steps as needed

  • Quality control: Monitor protein fractions during purification for:

    • EPHX1 content by Western immunoblot analysis

    • Enzymatic activity by cSO hydrolysis assay

  • Storage: Store purified protein in 10 mM potassium phosphate buffer (pH 7.4) at −80°C

This purification approach has been successfully used to prepare functionally active EPHX1 variants for comparative enzymatic studies, yielding protein preparations of sufficient purity for detailed kinetic analysis .

What experimental controls are essential when studying EPHX1 polymorphisms in Sf9 cells?

When studying EPHX1 polymorphisms using Sf9 cells, implement these critical controls:

  • Reference allele: Include the wild-type reference (typically Y113/H139) in every experiment as a baseline for activity comparisons

  • Complete variant panel: Express all four allelic variants (Y113/H139, H113/H139, Y113/R139, H113/R139) in parallel under identical conditions

  • Negative controls: Include empty vector-transfected Sf9 cells to confirm lack of endogenous epoxide hydrolase activity

  • Purification controls: Process all variant proteins through identical purification protocols to eliminate methodology-based variations

  • Protein normalization: Determine protein concentration using reliable methods (e.g., BCA assay) to ensure accurate specific activity calculations

  • Expression verification: Confirm expression levels by Western blot to ensure that activity differences are not due to varying protein amounts

  • Multiple substrates: Test enzymatic activity with multiple substrates (e.g., cSO and BaPO) to detect substrate-specific effects of polymorphisms

  • Human tissue correlations: Compare results with human liver microsomes of known EPHX1 genotypes

These controls help ensure that observed differences in enzymatic activity are genuinely attributable to the polymorphic variations rather than experimental artifacts.

How do EPHX1 polymorphisms affect enzyme activity in purified preparations versus human liver microsomes?

A significant discrepancy exists between the enzymatic properties of EPHX1 variants in purified systems versus human tissue:

  • Purified protein from Sf9 cells:

    • The reference protein (Y113/H139) shows approximately 2-fold greater rates of cSO and BaPO hydrolysis compared to the other EPHX1 allelic variants

    • Clear differences in catalytic activity are observable between variants

  • Human liver microsomes:

    • Despite genotypic differences, human liver microsomal preparations show no major differences in EPHX1-specific reaction rates

    • Activities are more consistent across different genotypes than would be predicted from purified protein studies

This discrepancy suggests that:

  • The cellular environment significantly influences EPHX1 activity

  • Additional factors in human liver may compensate for or modulate the effects of polymorphisms

  • The structural differences encoded by Y113H and H139R variants may exert only modest impacts on EPHX1-specific enzymatic activities in vivo

This finding has important implications for interpreting epidemiological studies associating EPHX1 polymorphisms with disease risk, as the functional impact of these polymorphisms may be less pronounced in vivo than in vitro studies would suggest.

What are the epidemiological implications of EPHX1 polymorphisms in disease risk?

EPHX1 polymorphisms have been investigated as potential risk factors for several diseases:

The inconsistent results across epidemiological studies highlight the complexity of gene-environment interactions and the potential importance of considering:

  • Population-specific allele frequencies

  • Environmental exposures that vary between populations

  • The modest functional impact of these polymorphisms in vivo

  • Interactions with other genetic variants in related pathways

What substrates are most appropriate for measuring EPHX1 activity in purified preparations?

For functional analysis of EPHX1 variants expressed in Sf9 cells, several substrates are particularly appropriate:

  • cis-Stilbene oxide (cSO):

    • Standard substrate for measuring EPHX1 activity

    • Used extensively in published studies of EPHX1 variants

    • Provides reliable activity measurements for comparative analyses

  • Benzo[a]pyrene-4,5-oxide (BaPO):

    • Environmentally relevant substrate representing polycyclic aromatic hydrocarbon metabolism

    • Important for understanding EPHX1's role in carcinogen metabolism

    • Used successfully in comparative studies of EPHX1 variants

  • Additional relevant substrates:

    • Epoxy eicosatrienoic acids (EETs): Physiologically relevant endogenous substrates

    • Styrene oxide: Occupationally relevant substrate

    • Various environmental pollutants with epoxide metabolites

When selecting substrates, consider both the research question and practical aspects:

  • Basic activity comparisons: cSO provides reliable data for variant comparison

  • Carcinogen metabolism: BaPO offers insights into EPHX1's role in cancer risk

  • Physiological relevance: EETs help understand endogenous functions

The choice of substrates should align with the specific aspect of EPHX1 function being investigated.

How can I accurately measure kinetic parameters of different EPHX1 variants?

Accurate measurement of EPHX1 variant kinetic parameters requires:

  • Protein quality control:

    • Ensure equal amounts of purified protein using BCA or Bradford assays

    • Verify purity by SDS-PAGE and protein staining

    • Confirm identity by Western blotting with specific antibodies

  • Assay optimization:

    • Determine linear range for reaction time and enzyme concentration

    • Optimize buffer conditions (pH, ionic strength) for maximum activity

    • Ensure substrate solubility across the concentration range tested

  • Kinetic analysis methodology:

    • Use a range of substrate concentrations spanning 0.1-10× estimated Km

    • Include sufficient replicates for statistical validity

    • Measure initial reaction rates to avoid substrate depletion effects

  • Data analysis:

    • Use non-linear regression to fit Michaelis-Menten equations

    • Calculate catalytic efficiency (kcat/Km) for comprehensive comparison

    • Perform appropriate statistical analysis to determine significance of differences

  • Controls and validation:

    • Run all variants in parallel under identical conditions

    • Include reference standards

    • Test multiple substrates to identify substrate-specific effects

This methodical approach provides reliable kinetic parameters for meaningful comparisons of EPHX1 variants, revealing how polymorphisms affect substrate binding and catalytic efficiency.

How do the enzymatic properties of EPHX1 variants compare across different substrates?

The enzymatic properties of EPHX1 variants show substrate-dependent patterns:

  • Y113/H139 (Reference variant):

    • Demonstrates approximately 2-fold greater rates of hydrolysis for both cSO and BaPO compared to other variants when using purified preparations from Sf9 cells

    • Considered the "wild-type" with reference activity levels

  • H113/H139:

    • The Y113H substitution generally decreases enzymatic activity

    • Shows reduced activity with both cSO and BaPO substrates in purified preparations

  • Y113/R139:

    • Despite the H139R substitution typically being associated with increased activity in some studies, purified preparations of this variant from Sf9 cells show lower activity than the reference allele with both cSO and BaPO

  • H113/R139:

    • Combined variant shows reduced activity compared to the reference allele in purified preparations

Interestingly, while these differences are clearly observable with purified proteins from Sf9 cells, they become much less pronounced when examining human liver microsomes from individuals with different EPHX1 genotypes . This suggests that the cellular context significantly influences how polymorphisms affect enzyme function, with the structural differences encoded by the Y113H and H139R variants potentially exerting only modest impacts on EPHX1-specific enzymatic activities in vivo .

What methods are recommended for isolating human liver microsomes for EPHX1 studies?

For isolating functionally active human liver microsomes for comparative EPHX1 studies:

  • Tissue procurement and handling:

    • Use fresh human liver specimens or properly preserved tissues

    • Document donor characteristics, including EPHX1 genotype

    • Maintain cold chain throughout processing

  • Homogenization and fractionation:

    • Prepare microsomes using established differential centrifugation protocols

    • Carefully isolate the microsomal fraction containing endoplasmic reticulum membranes

    • Store microsomes appropriately to maintain enzyme activity

  • Quality assessment:

    • Determine protein concentration using standardized methods (e.g., BCA Assay)

    • Verify EPHX1 content by Western immunoblot analysis

    • Assess enzymatic activity using standard substrates like cSO

  • Genotyping:

    • Determine EPHX1 genotypes using reliable methods (PCR-RFLP, TaqMan assays, or sequencing)

    • Focus on the key polymorphisms Y113H and H139R

    • Group samples according to homozygous genotypes for clearest comparisons

Human liver microsomes provide an essential reference for validating findings from recombinant expression systems, offering insights into how EPHX1 polymorphisms function in their native context . The research indicates that human liver microsomes from donors with different EPHX1 genotypes show less pronounced differences in enzymatic activities than purified recombinant proteins, suggesting important contextual influences on enzyme function .

Why might EPHX1 activity differ between purified Sf9-expressed protein and human liver microsomes?

Several factors may explain the discrepancy between EPHX1 activity in purified Sf9-expressed protein versus human liver microsomes:

  • Cellular context differences:

    • Membrane composition and lipid environment differ between insect cells and human liver

    • Orientation and integration of EPHX1 in membranes may affect substrate access

    • Post-translational modifications may vary between expression systems

  • Regulatory mechanisms:

    • Human liver likely contains mechanisms that regulate EPHX1 activity in vivo

    • Protein-protein interactions present in native tissue may modulate activity

    • Compensatory mechanisms may normalize activity differences in human tissue

  • Experimental considerations:

    • Purification procedures may affect protein conformations differently

    • Detergent effects on purified protein versus native membrane environment

    • Different assay conditions between systems

  • Biological implications:

    • The modest impact of polymorphisms in vivo suggests evolutionary pressure to maintain consistent EPHX1 function

    • The cellular environment may buffer against genetic variations in enzyme structure

    • This finding explains why epidemiological associations between EPHX1 genotype and disease risk have been inconsistent

Understanding these differences is crucial for interpreting how laboratory findings translate to physiological relevance and for designing meaningful studies of EPHX1 polymorphisms in disease risk.

How should researchers correlate in vitro EPHX1 findings with clinical or epidemiological data?

To effectively correlate in vitro EPHX1 findings with clinical or epidemiological data:

  • Establish functional relevance:

    • Determine if in vitro activity differences are substrate-specific

    • Quantify the magnitude of effect for each polymorphism

    • Consider whether these differences persist in more physiologically relevant systems (e.g., human liver microsomes)

  • Contextual interpretation:

    • Recognize that purified protein studies may overestimate the functional impact of polymorphisms compared to whole-tissue studies

    • Consider that modest enzymatic differences may still be significant in specific contexts or exposures

    • Evaluate the biological plausibility of associations based on substrate specificity

  • Population considerations:

    • Account for population-specific allele frequencies

    • Consider gene-environment interactions specific to study populations

    • Evaluate haplotypes rather than individual polymorphisms

  • Study design recommendations:

    • Include functional validation in epidemiological studies

    • Consider measuring metabolite ratios as biomarkers of in vivo EPHX1 activity

    • Account for potential confounding factors (other polymorphisms in metabolic pathways)

  • Integrated approach:

    • Combine laboratory findings with human tissue studies and epidemiological data

    • Develop models that incorporate both genetic and environmental factors

    • Consider the collective impact of multiple polymorphisms in related pathways

This approach recognizes the complexity of translating in vitro findings to human disease risk and helps explain the sometimes contradictory results in epidemiological studies of EPHX1 polymorphisms .

How can EPHX1 variants expressed in Sf9 cells be used to study drug metabolism?

EPHX1 variants expressed in Sf9 cells provide a valuable system for studying drug metabolism through:

  • Comprehensive metabolite profiling:

    • Express all four common EPHX1 variants in Sf9 cells

    • Test metabolism of drugs known to form epoxide intermediates

    • Identify and quantify all metabolites using LC-MS/MS

    • Determine if polymorphisms affect metabolite profiles qualitatively or quantitatively

  • Drug safety assessment:

    • Evaluate whether polymorphisms affect the balance between detoxification and bioactivation

    • Assess metabolite toxicity differences between variants

    • Identify drugs that might pose greater risks for individuals with specific EPHX1 genotypes

  • Pharmacokinetic predictions:

    • Determine kinetic parameters for each variant with relevant drug substrates

    • Use data to develop physiologically-based pharmacokinetic models

    • Predict how polymorphisms might affect drug exposure in patients

  • Translational applications:

    • Compare recombinant enzyme findings with human liver microsomes of known genotypes

    • Correlate in vitro findings with clinical pharmacokinetic data

    • Develop recommendations for genotype-specific dosing adjustments

This approach provides mechanistic insights into how EPHX1 polymorphisms might influence individual responses to medications, with potential applications in precision medicine and drug development.

What role does EPHX1 play in endogenous signaling pathways?

EPHX1 contributes to endogenous signaling pathways through several mechanisms:

  • Epoxyeicosatrienoic acid (EET) metabolism:

    • EPHX1 converts EETs to dihydroxyeicosatrienoic acids (DHETs)

    • This affects vasodilation, inflammation, and angiogenesis signaling

    • EPHX1 polymorphisms may influence cardiovascular and inflammatory processes through altered EET metabolism

  • Endocannabinoid system:

    • EPHX1 metabolizes the endocannabinoid 2-arachidonoylglycerol (2-AG) to arachidonic acid

    • May play an important role in endocannabinoid signaling pathways

    • Could influence processes regulated by endocannabinoids (appetite, pain, mood, memory)

  • Bile acid transport:

    • EPHX1 mediates sodium-dependent transport of bile acids into hepatocytes

    • May impact bile acid homeostasis and related signaling pathways

  • Steroid metabolism:

    • Metabolizes endogenous substrates including androstene oxide and epoxyestratrienol

    • May influence steroid hormone signaling

  • Preeclampsia association:

    • EPHX1 polymorphisms affecting enzyme activity are associated with preeclampsia risk

    • This condition involves hypertension and proteinuria during pregnancy

    • Other genes involved in epoxy fatty acid metabolism have also been linked to preeclampsia

The involvement of EPHX1 in these diverse signaling pathways suggests that its polymorphisms may have wide-ranging physiological consequences beyond xenobiotic metabolism, potentially explaining some of the disease associations observed in epidemiological studies .

What emerging approaches can advance our understanding of EPHX1 function and polymorphisms?

Several emerging approaches can significantly advance EPHX1 research:

  • Structural biology techniques:

    • Cryo-electron microscopy to visualize EPHX1 variants in membrane environments

    • X-ray crystallography of purified variants to identify structural differences

    • Molecular dynamics simulations to understand how polymorphisms affect protein motion and substrate binding

  • Advanced analytical methods:

    • Untargeted metabolomics to identify novel endogenous substrates

    • Proteomics to identify protein-protein interactions affected by polymorphisms

    • High-resolution mass spectrometry for comprehensive metabolite profiling

  • Systems biology approaches:

    • Integrate EPHX1 polymorphism data with other metabolic pathway variations

    • Network analyses to understand EPHX1's position in broader metabolic and signaling networks

    • Computational modeling of metabolism incorporating genetic variation data

  • Translational methodologies:

    • Development of EPHX1 genotype-specific biomarkers for personalized medicine

    • Creation of genotype-activity correlation models for clinical applications

    • Integration of EPHX1 data into pharmacogenomic databases

  • Advanced cell and tissue models:

    • CRISPR-engineered cells with specific EPHX1 genotypes

    • Organ-on-a-chip systems incorporating EPHX1 variants

    • Patient-derived organoids to study EPHX1 function in disease-relevant contexts

These approaches collectively provide a more comprehensive understanding of how EPHX1 polymorphisms influence enzyme function in various biological contexts, potentially leading to clinical applications in precision medicine and toxicology .

What are the key considerations when designing EPHX1 research using Sf9 expression systems?

When designing EPHX1 research using Sf9 expression systems, researchers should consider:

  • Expression system optimization:

    • Carefully optimize viral titer, MOI, and expression time for each EPHX1 variant

    • Implement consistent purification protocols across all variants

    • Include appropriate controls to ensure comparable expression levels

  • Functional analysis design:

    • Test multiple substrates to capture the substrate-dependent effects of polymorphisms

    • Include both environmentally relevant and endogenous substrates

    • Normalize activity measurements appropriately to protein concentration

  • Translational validity:

    • Recognize that purified enzyme studies may overestimate polymorphism effects compared to in vivo function

    • Validate findings using human liver microsomes from individuals with known genotypes

    • Consider the cellular context when interpreting results

  • Comprehensive variant analysis:

    • Study all four major haplotypes (Y113/H139, H113/H139, Y113/R139, H113/R139)

    • Consider population-specific frequencies when designing studies

    • Look beyond the common polymorphisms when appropriate

  • Interdisciplinary approach:

    • Integrate structural, functional, and epidemiological data

    • Consider both xenobiotic metabolism and endogenous signaling roles

    • Collaborate across disciplines for comprehensive understanding

These considerations ensure that research findings accurately reflect the biological significance of EPHX1 polymorphisms and provide meaningful insights into their role in health and disease .

What future directions should EPHX1 research pursue?

Future EPHX1 research should focus on several promising directions:

  • Comprehensive substrate profiling:

    • Identify the complete range of xenobiotic and endogenous substrates affected by EPHX1 polymorphisms

    • Develop substrate-specific activity profiles for each variant

    • Determine if certain exposures pose greater risks for specific genotypes

  • Mechanistic disease associations:

    • Clarify the molecular mechanisms behind EPHX1 associations with diseases like preeclampsia

    • Investigate how modest enzymatic differences translate to disease risk

    • Explore interactions with other genetic and environmental factors

  • Endogenous signaling roles:

    • Further characterize EPHX1's role in endocannabinoid metabolism

    • Investigate its impact on epoxy fatty acid signaling in cardiovascular function

    • Explore the physiological consequences of variant-specific differences in these pathways

  • Clinical applications:

    • Develop genotype-specific dosing guidelines for medications metabolized by EPHX1

    • Create diagnostic tools based on EPHX1 genotype for personalized risk assessment

    • Establish EPHX1-targeted interventions for disease prevention

  • Integrated multi-omics approach:

    • Combine genomics, proteomics, metabolomics, and clinical data

    • Develop computational models predicting phenotypic outcomes

    • Explore gene-gene and gene-environment interactions

These future directions will deepen our understanding of how EPHX1 polymorphisms influence human health and disease, potentially leading to novel preventive and therapeutic strategies based on individual genetic profiles .

Product Science Overview

Introduction

Epoxide Hydrolase 1 (EH1), also known as microsomal epoxide hydrolase (mEH), is an enzyme encoded by the EPHX1 gene located on chromosome 1 (1q42.12) in humans . This enzyme plays a crucial role in the metabolism of both xenobiotic and endogenous compounds by converting epoxides to diols, which are more hydrophilic and can be easily excreted from the body .

Structure and Function

The human microsomal epoxide hydrolase is a membrane-bound enzyme consisting of 455 amino acids . It is primarily found in the endoplasmic reticulum of cells and is involved in the detoxification of reactive epoxides formed during the metabolism of various compounds, including drugs, environmental pollutants, and endogenous lipids .

Role in Metabolism

EH1 is a vital biotransformation enzyme that catalyzes the hydrolysis of epoxides to trans-dihydrodiols . This reaction is essential for the detoxification of potentially harmful epoxides, which can form adducts with DNA and proteins, leading to cellular damage and mutagenesis. By converting these reactive epoxides to less harmful diols, EH1 helps protect cells from oxidative stress and chemical-induced toxicity .

Clinical Significance

The activity of EH1 has been linked to various diseases and conditions. For instance, genetic polymorphisms in the EPHX1 gene can result in altered enzyme activity, which has been associated with an increased risk of developing certain cancers, preeclampsia, and neurological disorders . Additionally, EH1 has been studied as a potential biomarker for diseases such as metastatic cancer and viral hepatitis .

Recombinant Expression in sf9 Cells

The recombinant form of human microsomal epoxide hydrolase (EH1) is often produced using the baculovirus expression system in sf9 insect cells. This system allows for the high-level expression of functional EH1, which can be used for various research and therapeutic applications . The recombinant enzyme retains its catalytic activity and can be used to study the enzyme’s structure, function, and role in disease processes.

Applications

Recombinant EH1 is used in various applications, including:

  • Drug Metabolism Studies: Understanding how drugs are metabolized and identifying potential drug-drug interactions.
  • Toxicology Research: Investigating the detoxification pathways of environmental pollutants and other xenobiotics.
  • Biomarker Development: Developing assays to measure EH1 activity as a potential biomarker for disease diagnosis and prognosis .

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 2025 TheBiotek. All Rights Reserved.