Recombinant Rat UDP-glucuronosyltransferase 1-3 (Ugt1)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Rat UDP-glucuronosyltransferase 1-3 (Ugt1)

Recombinant Rat UDP-glucuronosyltransferase 1-3 (Ugt1) refers to a genetically engineered form of the UDP-glucuronosyltransferase enzyme from rats, specifically focusing on the Ugt1 subfamily. UDP-glucuronosyltransferases (UGTs) are crucial phase II drug-metabolizing enzymes responsible for the glucuronidation of various compounds, including drugs, hormones, and bilirubin. This process involves the transfer of glucuronic acid from UDP-glucuronic acid to lipophilic substrates, enhancing their solubility and facilitating their excretion from the body.

Function and Role of UGTs

UGTs are classified into two main subfamilies: UGT1 and UGT2. The UGT1 subfamily includes several isoforms, such as UGT1A1, UGT1A3, UGT1A4, etc., which exhibit distinct substrate specificities and tissue distributions. In rats, the Ugt1 subfamily plays a vital role in the metabolism of endogenous and exogenous compounds. Recombinant forms of these enzymes are used in research to study their enzymatic activities and interactions with various substrates.

Table 1: Glucuronidation Rates of Diclofenac by Different UGT Isoforms

UGT IsoformGlucuronidation Rate (pmol/min/mg protein)
Human UGT1A9166
Human UGT1A6<20
Human UGT2B7>500
Rat UGT2B1250

Inhibition Studies

Fatty acids like oleic acid, linoleic acid, and DHA have been shown to inhibit human UGT1A1 activity, with DHA being the most potent inhibitor . While similar studies on rat UGTs are less common, understanding the inhibitory effects of such compounds can provide insights into potential interactions with Recombinant Rat UGT1-3.

Table 2: Inhibition Constants (Ki) of Fatty Acids on Human UGT1A1

Fatty AcidInhibition Constant (Ki, μM)
Oleic Acid29.3
Linoleic Acid24.0
DHA4.3

Genetic Variants and Functionality

Genetic variants in UGT genes can significantly affect the metabolism of drugs and endogenous compounds, leading to variations in drug responses and susceptibility to diseases . While specific data on genetic variants of Recombinant Rat UGT1-3 is limited, understanding these variations is crucial for predicting metabolic outcomes.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to settle the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, serving as a guideline for your use.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
If you require a specific tag type, please inform us, and we will prioritize its implementation.
Synonyms
Ugt1a3; Ugt1; UDP-glucuronosyltransferase 1A3; UGT1A3; B3; UDP-glucuronosyltransferase 1-3; UDPGT 1-3; UGT1*3; UGT1-03; UGT1.3
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
26-531
Protein Length
Full Length of Mature Protein
Species
Rattus norvegicus (Rat)
Target Names
Ugt1a3
Target Protein Sequence
GKVLVFPMEGSHWLSIRDVVRELHARGHQAVVLAPEVTVHIKEEDFFTLQTYPVPYTKEE YRHHLLGHLQNFFETEFSLKLVLQTMAAVNNVSTFYVRSCRGLLHNTALIQSLNSSSFDV VLTDPFFPCGAVLAMYLRVPAVFFLQSMLCELEFEATNSPNPSSYIPRLLTLNSDHMSFL DRVKNMLYPVPWMYLCHVNYGSLARLASDLLQREVSVVEILRHASVWLLRKDFVFHYPRP FMPNMVFIGGINCANRKPLSQEFEAYVNASGEHGIVVFSLGSMVSEIPEKKAMEIAEALG RIPQTLLWRYTGTRPSNLAKNTILVKWLPQNDLLGHPKARAFITHSGSHGIYEGICNGVP MVMMPLFGDQMDNAKRMETRGAGVTLNVLEMTADDLENALKTVINNKSYKENIMRLSSLH KDRPIEPLDLAVFWVEYVMRHKGAPHLRPAAHDLTWYQYHSLDVIGFLLAIVLTVVFIVY KSCAYGCRKCFGGKGRVKKSHKSKTH
Uniprot No.

Target Background

Function

Recombinant Rat UDP-glucuronosyltransferase 1-3 (UGT1A1) is a phase II biotransformation enzyme that conjugates lipophilic substrates with glucuronic acid, enhancing water solubility and facilitating urinary or biliary excretion. Its role is crucial for the elimination and detoxification of drugs, xenobiotics, and endogenous compounds. Key functions include:

  • Glucuronidation of endogenous estrogens (estradiol and estrone).
  • Contribution to bile acid detoxification through glucuronidation.
  • Glucuronidation of calcidiol (the primary circulating form of vitamin D3), vital for calcium and phosphate homeostasis.
  • Glucuronidation of angiotensin receptor antagonists (losartan, candesartan, and zolarsartan), impacting angiotensin II effects.
Database Links

UniGene: Rn.26489

Protein Families
UDP-glycosyltransferase family
Subcellular Location
Endoplasmic reticulum membrane; Single-pass membrane protein.

Q&A

What is the basic function of UDP-glucuronosyltransferase 1-3 in rats and how does it compare to human UGT isoforms?

UDP-glucuronosyltransferases (UGTs) are phase II drug-metabolizing enzymes that catalyze glucuronidation reactions, forming covalent bonds between endogenous polar glucuronic acid and lipophilic compounds. In rats, as in humans, UGT1 family enzymes play critical roles in the metabolism of both endogenous compounds (bilirubin, bile acids, steroid hormones) and xenobiotics (drugs) .

While humans have 22 identified UGT isoforms, specific comparisons between rat UGT1-3 and human homologs show both similarities and differences in substrate specificity and catalytic efficiency. For example, rat UGT2B1 and human UGT2B7 display similar kinetic properties when metabolizing certain compounds like diclofenac, with both showing low apparent Km values (< 15 μM) .

How should I design an appropriate expression system for recombinant rat UGT1-3?

Methodological approach:

  • Select an appropriate expression vector containing a strong promoter suitable for mammalian protein expression

  • Engineer the construct to include:

    • Full rat Ugt1-3 cDNA sequence with optimized codons

    • N-terminal modifications to enhance expression (signal peptide optimization)

    • C-terminal purification tag (His6 or FLAG) that won't interfere with enzyme activity

  • Choose between expression systems:

    • Insect cell systems (Sf9, High Five) for higher protein yields

    • Mammalian cell lines (HEK293, CHO) for proper post-translational modifications

  • Confirm expression using Western blot analysis with UGT1-specific antibodies

  • Validate enzyme activity using known substrates (e.g., diclofenac for UGT2B1)

Remember that expression conditions dramatically affect enzymatic activity - temperature, induction time, and media composition should be optimized through factorial design experiments.

What cofactors and reaction conditions are essential for optimal rat UGT1-3 activity in vitro?

For optimal activity of recombinant rat UGT1-3, the following methodological parameters are critical:

  • Essential cofactor: UDP-glucuronic acid (UDPGA) at 0.5-5 mM concentration

  • Buffer conditions: 100 mM phosphate buffer at pH 7.4 provides optimal activity

  • Membrane activation: Include alamethicin (12.5 μg/mL) when working with microsomal preparations

  • Divalent cations: Mg²⁺ (5-10 mM) enhances activity

  • Temperature: 37°C is optimal for kinetic studies

  • Reaction time: Linearity should be established (typically 15-60 minutes)

  • Protein concentration: Determine optimal range to ensure linearity of reaction velocity

When designing an in vitro glucuronidation assay, always include negative controls: (i) without substrate, (ii) without cofactor UDPGA, and (iii) without enzyme source, as these are essential for validating the specificity of the reaction .

How do I determine substrate specificity profiles for recombinant rat UGT1-3?

Methodological approach:

  • Substrate screening panel setup:

    • Prepare a diverse panel of potential substrates (phenols, carboxylic acids, alcohols, amines)

    • Include known UGT substrates as positive controls (e.g., diclofenac, morphine)

    • Use fluorescent compounds like 7-hydroxycoumarin derivatives for ease of detection

  • Initial screening assay:

    • Conduct incubations with standardized conditions (pH 7.4, 37°C)

    • Use a fixed substrate concentration (10-50 μM) and enzyme amount

    • Measure substrate disappearance or glucuronide formation

  • Kinetic characterization for active substrates:

    • Determine Km and Vmax using 6-8 substrate concentrations

    • Analyze data using appropriate enzyme kinetic models (Michaelis-Menten, allosteric, substrate inhibition)

    • Compare kinetic parameters with human orthologs to identify species differences

  • Confirmation of glucuronidation:

    • Verify glucuronide formation using mass spectrometry

    • Conduct β-glucuronidase hydrolysis to confirm the nature of the metabolite

For optimal results, adjust protein concentration based on substrate turnover rate, using lower protein concentrations (6-7 mg/L) for rapidly metabolized substrates and higher concentrations (12-14 mg/L) for slower reactions .

What are the established kinetic differences between rat UGT1-3 and human UGT1A isoforms?

Comparative kinetic analysis between rat and human UGT isoforms reveals significant species differences that researchers should consider when extrapolating data:

ParameterRat UGT2B1Human UGT2B7Notes
Diclofenac Km<15 μM<15 μMSimilar affinity
Diclofenac Vmax0.3 nmol/min/mg2.8 nmol/min/mgHigher capacity in human
Glucuronidation Rate250 pmol/min/mg>500 pmol/min/mgHuman enzyme more efficient
Substrate SpecificityNarrowerBroaderHuman enzyme metabolizes more diverse compounds
Morphine GlucuronidationMajor pathwayMajor pathwayBoth enzymes conjugate morphine

The key methodological consideration is that while certain rat UGT isoforms may show similar substrate affinity (Km) to human orthologs, the catalytic efficiency (Vmax) often differs substantially. This explains why human liver microsomes typically display higher glucuronidation capacity (4.3 nmol/min/mg) than rat liver microsomes (0.9 nmol/min/mg) for substrates like diclofenac .

How can I develop selective fluorescent substrates to distinguish between rat UGT1-3 activity and other UGT isoforms?

Methodological approach for developing selective fluorescent substrates:

  • Rational design strategy:

    • Construct homology models of target UGT enzymes

    • Identify unique structural features in the substrate binding pocket

    • Design derivatives of fluorescent scaffolds (e.g., 7-hydroxycoumarin) with substituents targeting these unique features

  • Synthesis approach:

    • Start with inexpensive core structures like 7-hydroxycoumarin

    • Introduce C3-substitutions with varying properties:

      • Hydrophobic groups (e.g., 4-methylphenyl)

      • Electron-donating groups (e.g., 4-methoxyphenyl)

      • Electron-withdrawing groups (e.g., 4-fluorophenyl)

      • Polar groups (e.g., triazole)

  • Validation methodology:

    • Screen compounds against a panel of recombinant UGT enzymes

    • Compare glucuronidation rates between target and non-target UGTs

    • Measure fluorescence quenching upon glucuronidation (excitation at 390-405 nm, emission at 460 nm)

    • Confirm selectivity using tissue microsomes (e.g., liver vs. intestine)

This approach has successfully identified selective substrates for human UGT1A10, where compounds with 4-(dimethylamino)phenyl and triazole C3-substitutions showed high selectivity . Similar strategies can be employed to develop selective probes for rat UGT1-3.

How do genetic polymorphisms in rat UGT1-3 affect enzyme function and what methods can detect these variations?

While fewer genetic polymorphisms have been characterized in rat UGT1 compared to human UGTs, methodological approaches to study these variations include:

  • Identification methodology:

    • Whole genome sequencing of different rat strains

    • Targeted exome sequencing of UGT1 gene cluster

    • SNP genotyping in laboratory and wild rat populations

  • Functional characterization:

    • Site-directed mutagenesis to introduce identified polymorphisms

    • Expression of variant UGT1-3 in cellular systems

    • Comparative enzyme kinetics with multiple substrates

    • Protein stability and expression level assessment

  • Physiological impact assessment:

    • Develop rat models with specific UGT genetic variants

    • Compare pharmacokinetics of UGT substrates across rat strains

    • Correlate genotype with glucuronidation capacity in primary hepatocytes

The UGT1A gene family in humans contains numerous clinically significant polymorphisms that affect drug responses, such as UGT1A128 (associated with hyperbilirubinemia after atazanavir treatment) and UGT1A16 (associated with irinotecan-induced neutropenia) . Similar structure-function relationships likely exist in rat UGT1-3, particularly affecting metabolism of drugs like carvedilol and morphine.

What approaches can be used to determine structure-function relationships in recombinant rat UGT1-3?

Methodological approach for elucidating structure-function relationships:

  • Computational methods:

    • Homology modeling based on existing UGT crystal structures

    • Molecular docking of substrates to identify key binding residues

    • Molecular dynamics simulations to understand protein flexibility

  • Experimental validation:

    • Site-directed mutagenesis of predicted critical residues

    • Creation of chimeric enzymes between different UGT isoforms

    • Alanine scanning of substrate binding regions

    • Analysis of conserved motifs, particularly the 29 amino acids involved in UDP-glucuronic acid binding

  • Kinetic analysis of mutants:

    • Compare Km, Vmax, and catalytic efficiency (Vmax/Km) between wild-type and mutant enzymes

    • Analyze substrate specificity shifts resulting from mutations

    • Determine the effect of mutations on cofactor binding

For example, creating a H210M mutant in human UGT1A10 affected glucuronidation kinetics variably depending on the substrate . Similar approaches with rat UGT1-3 can reveal critical residues for substrate selectivity and catalytic efficiency, particularly when comparing the differences between rat UGT2B1 and human UGT2B7 in diclofenac metabolism .

What are the most effective analytical methods for quantifying glucuronide metabolites from rat UGT1-3 reactions?

Comprehensive analytical approach for glucuronide metabolite quantification:

  • Chromatographic separation techniques:

    • HPLC with optimized column selection (C18 for most glucuronides)

    • UPLC for higher resolution and faster analysis

    • Specialized column chemistries for hydrophilic glucuronides (HILIC)

  • Detection methods (by increasing specificity and sensitivity):

    • UV detection (appropriate for high concentrations, simple matrices)

    • Fluorescence detection (for substrates with fluorophores, e.g., 7-hydroxycoumarins)

    • Mass spectrometry:

      • Single quadrupole MS for molecular weight confirmation

      • Triple quadrupole MS/MS for quantitative analysis

      • High-resolution MS for structural characterization

  • Methodology for glucuronide confirmation:

    • Neutral loss scanning (m/z 176 for glucuronides)

    • Pre-column and post-column β-glucuronidase hydrolysis

    • NMR for positional isomer identification

  • Quantification strategy:

    • Use authentic glucuronide standards when available

    • Employ relative response factors when standards unavailable

    • Consider stable isotope labeled internal standards

Kinetic studies should employ analytical methods with sufficient sensitivity to determine initial reaction rates accurately. For fluorescent substrates like C3-substituted 7-hydroxycoumarins, fluorescence decrease measurements (excitation 390-405 nm, emission 460 nm) in 96-well plate format enable high-throughput analysis .

How can I design experiments to investigate the regulation of rat UGT1-3 expression in different experimental models?

Methodological approach for UGT regulation studies:

  • Transcriptional regulation analysis:

    • Promoter analysis using luciferase reporter assays

    • ChIP-seq to identify transcription factor binding sites

    • Investigation of nuclear receptors known to regulate UGTs:

      • PXR and CAR (xenobiotic receptors)

      • FXR (bile acid receptor)

      • LXR (oxysterol receptor)

      • PPARα (fatty acid receptor)

      • AhR (aromatic hydrocarbon receptor)

  • Expression model systems:

    • Primary rat hepatocytes (maintains physiological context)

    • Rat hepatoma cell lines (H4IIE, MH1C1)

    • Precision-cut liver slices (maintains 3D architecture)

    • Transgenic rat models with reporter constructs

  • Intervention methods:

    • Treatment with prototypical inducers:

      • Phenobarbital (CAR activator)

      • Rifampicin (PXR activator)

      • TCDD (AhR activator)

    • Specific nuclear receptor agonists and antagonists

    • NSAIDs (shown to regulate UGT2B1 in organ-specific manner)

    • Steroid hormones (regulate UGT expression in breast and prostate)

  • Quantification techniques:

    • RT-qPCR for mRNA expression

    • Western blotting with isoform-specific antibodies

    • Activity assays with selective substrates

    • Proteomics for global protein expression changes

Understanding the tissue-specific regulation of rat UGT1-3 is particularly important as NSAIDs have been shown to downregulate UGT2B1 mRNA in liver and kidneys while upregulating it in the heart , demonstrating complex regulatory mechanisms that must be considered in drug metabolism studies.

What approaches can resolve contradictory data when comparing rat UGT1-3 activity in different experimental systems?

Methodological approach for resolving contradictory UGT activity data:

  • Systematic experimental system comparison:

    • Recombinant enzymes vs. microsomes vs. hepatocytes vs. in vivo models

    • Standardize experimental conditions across systems:

      • Same buffer composition, pH, and temperature

      • Equivalent protein/enzyme concentrations

      • Identical analytical methods

  • Cofactor availability assessment:

    • Measure endogenous UDPGA levels in cellular systems

    • Supplement UDPGA in microsomes (0.5 mM is standard)

    • Consider the role of UDP-glucose dehydrogenase in UDPGA synthesis

  • Membrane environment effects analysis:

    • Add alamethicin (12.5 μg/mL) to disrupt microsomal membranes

    • Compare detergent-solubilized and membrane-bound UGTs

    • Consider lipid composition effects on enzyme activity

  • Expression system artifacts investigation:

    • Verify full-length protein expression (not truncated variants)

    • Assess post-translational modifications

    • Confirm absence of inhibitory contaminants

    • Examine potential enzyme inhibition by high substrate concentrations

When encountering contradictory data between systems, researchers should establish scaling factors. For example, when comparing diclofenac glucuronidation, recombinant rat UGT2B1 showed a rate of 250 pmol/min/mg while rat liver microsomes showed a Vmax of 0.9 nmol/min/mg , indicating the need for appropriate scaling factors when extrapolating from recombinant systems to tissue preparations.

How should I design experiments to compare rat and human UGT activities for translational research?

Methodological approach for interspecies UGT activity comparison:

  • Enzyme source standardization:

    • Recombinant enzymes expressed in identical systems

    • Species-matched microsomes (rat vs. human)

    • Hepatocytes from both species prepared with identical protocols

    • Matched tissue fractions (S9, cytosol, microsomes)

  • Experimental design considerations:

    • Use identical assay conditions for both species

    • Select substrate concentrations spanning below Km to above saturating levels

    • Include positive controls known to be metabolized by both species

    • Determine protein concentration ranges ensuring linear reaction rates

  • Substrate panel selection:

    • Include clinically relevant drugs (e.g., diclofenac, morphine)

    • Add selective substrates for specific UGT isoforms

    • Consider species-specific probe substrates

    • Use fluorescent substrates for high-throughput screening

  • Data analysis approach:

    • Calculate intrinsic clearance (Vmax/Km) for each substrate

    • Determine species differences in substrate selectivity

    • Apply appropriate scaling factors for in vitro to in vivo extrapolation

    • Create correlation plots between species for multiple substrates

For proper translational research, it's critical to recognize the differences in catalytic efficiency between species. For example, human UGT2B7 showed approximately 9 times higher Vmax for diclofenac glucuronidation (2.8 nmol/min/mg) compared to rat UGT2B1 (0.3 nmol/min/mg), despite similar Km values .

What methodologies can accurately predict human UGT-mediated drug metabolism from rat UGT1-3 data?

Methodological approach for interspecies extrapolation:

  • Quantitative structure-activity relationship (QSAR) models:

    • Develop models based on physicochemical properties

    • Include molecular descriptors relevant to UGT binding

    • Train models on compounds tested in both species

    • Validate with external test sets

  • Physiologically-based pharmacokinetic (PBPK) modeling:

    • Incorporate species-specific physiological parameters

    • Include protein binding differences between species

    • Account for differences in UGT expression levels

    • Apply scaling factors derived from probe substrates

  • Relative activity factor (RAF) approach:

    • Determine RAF values using isoform-selective substrates

    • Apply RAF correction to metabolism data

  • Integrated testing strategy:

    • In vitro metabolism studies with recombinant enzymes from both species

    • Verification in liver microsomes from both species

    • Confirmation in hepatocytes for species-specific differences

    • Validation in humanized rat models expressing human UGTs

When predicting human clearance from rat data, consider that while rat UGT2B1 and human UGT2B7 share similar substrate affinity for compounds like diclofenac, the catalytic efficiency can differ substantially . Additionally, genetic polymorphisms in human UGT1A genes can significantly impact drug metabolism and should be incorporated into prediction models .

How can advanced mutagenesis techniques be applied to create humanized rat UGT1-3 models?

Methodological approach for creating humanized rat UGT models:

  • Targeted mutagenesis strategy:

    • Identify critical amino acid differences between rat and human orthologs

    • Focus on substrate binding regions and catalytic sites

    • Design mutations based on homology models and alignment analysis

    • Apply site-directed mutagenesis to create single and multiple mutations

  • Advanced genetic engineering techniques:

    • CRISPR/Cas9 gene editing of rat UGT genes in cell lines

    • Creation of chimeric UGT enzymes (rat-human hybrids)

    • Domain swapping between species to identify functional regions

    • Whole gene replacement in transgenic rat models

  • Functional validation methodology:

    • Compare enzyme kinetics between wild-type, mutant, and human enzymes

    • Test substrate panels spanning diverse chemical classes

    • Analyze shifts in substrate selectivity and catalytic efficiency

    • Validate with probe substrates specific for human UGTs

  • Application to translational research:

    • Create cell lines expressing humanized rat UGTs

    • Develop transgenic rats with humanized UGT genes

    • Use humanized models to predict human-specific drug metabolism

    • Apply to drug-drug interaction studies

This approach has been demonstrated with UGT1A10, where a targeted H210M mutation based on molecular modeling affected substrate metabolism differently depending on the compound . Similar strategic mutations in rat UGT1-3 could create better models for human drug metabolism prediction.

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.