Recombinant Rat UDP-glucuronosyltransferase 2B7 (Ugt2b7)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. If you require a specific format, please specify this in your order notes; we will fulfill your request to the best of our ability.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping is available upon request with an additional charge. Please contact us in advance to arrange this.
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 collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and may serve as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type will be determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Ugt2b7; Ugt2b8; UDP-glucuronosyltransferase 2B7; UDPGT 2B7; UGT2B7; UGT2B-RH4
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
18-530
Protein Length
Full Length of Mature Protein
Species
Rattus norvegicus (Rat)
Target Names
Ugt2b7
Target Protein Sequence
FRSGNCGKVLVWPLEYSHWMNLKIILDELVQRGHEVTVLRPSSSVSLDPKKASGLVYETS PTTSNNDEVEKSFYPVGDMWTYDVPKYTCLRYYPSLNKMFGQFSDLWLQLCREVVSNKEL IAKLKESQFDVVLSDAVGPCGELIAEILQLPFVYSLRFATAPGIEKYSAGQPFPPSYVPI ILSGFSGQMTFMERVENMLCLLYFDSWFESFPAKDWDPFFSEILGRPTTMVDTMKKAEIW LIRSYWDLEFPRPSLPNIEFVGGLHCQPAKPLPKEMEDFAQSSGEHGVWVFSLGSMIRNI TQERANTIASALAQIPQKVFWRFEGKKPDTLGPNTRVFKWIPQNDLLGHPKTKAFVTHGG ANGIYESIHYGIPPMVGIPLFAEQRDNVAHMVAKGAAVSIDFHTMSSSDLLNALKAVINN PSYKKKVMWLSAIHHDQPLKPLDRAVFWIEFVMRHKGAKHLRPLAHNLALVSVHSLDVIG FLLACVLAIVLLAVKCCLFLYRFFVKVAKNKRD
Uniprot No.

Target Background

Function

Recombinant Rat UDP-glucuronosyltransferase 2B7 (UGT2B7) is a phase II biotransformation enzyme that conjugates lipophilic substrates with glucuronic acid, enhancing water solubility and facilitating urinary or biliary excretion. This enzyme plays a crucial role in the detoxification and elimination of drugs, xenobiotics, and endogenous compounds. UGT2B7 catalyzes the glucuronidation of steroid hormones (androgens and estrogens), retinoic acid (a vitamin A metabolite involved in apoptosis, cell growth, and differentiation), and bile acids. It also participates in the metabolism of several pharmaceuticals, including angiotensin receptor antagonists (losartan, candesartan, and olmesartan) and the immunosuppressant mycophenolate.

Gene References Into Functions
  1. Acute liver failure significantly increases oral plasma exposure of zidovudine in rats, partly due to impaired hepatic UGT2B7 function and intestinal P-gp expression. PMID: 28770824
  2. UGT2B7 is the primary enzyme responsible for haloperidol O-glucuronidation in both rat and human liver microsomes. PMID: 22028316
Database Links

KEGG: rno:286989

UniGene: Rn.11131

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

Q&A

What is the primary function of rat UGT2B7 in drug metabolism?

Rat UGT2B7, similar to its human ortholog, plays a major role in phase II metabolism by catalyzing the glucuronidation reaction, which involves the transfer of glucuronic acid from UDP-glucuronic acid to various substrates. This process increases the water solubility of compounds, facilitating their elimination through urine or bile. The enzyme is particularly important for the detoxification of various endogenous compounds and xenobiotics . Unlike some other UGT isoforms, UGT2B7 demonstrates relatively broad substrate specificity, making it a crucial enzyme in pharmaceutical research and toxicology studies.

How does the tissue distribution of UGT2B7 in rats compare to other UGT enzymes?

UGT2B7 is predominantly expressed in rat liver, with lower expression levels observed in kidney and intestine. In a comprehensive transcriptome analysis of UGTs across different tissues, quantitative PCR data revealed significant variations in tissue-specific expression patterns . The expression of UGT2B7 in rat liver microsomes correlates with higher glucuronidation activity in this tissue compared to intestinal microsomes. This tissue-specific expression pattern is important to consider when designing experiments to assess drug metabolism and potential drug-drug interactions in rat models .

What are the main substrates of rat UGT2B7?

Rat UGT2B7 shows activity toward a wide range of substrates including:

  • Opioids (such as morphine)

  • Non-steroidal anti-inflammatory drugs (like diclofenac)

  • Steroid hormones

  • Bile acids such as hyodeoxycholic acid (HDCA)

  • Catechol estrogens like 4-hydroxyestrone

  • Zidovudine (AZT)

  • Naftopidil enantiomers

Particularly, rat UGT2B7 shows significant activity toward zidovudine (ZDV) glucuronidation, which is commonly used as a probe substrate to assess UGT2B7 activity in experimental settings . For diclofenac, rat UGT2B1 (related to human UGT2B7) demonstrated a glucuronidation rate of approximately 250 pmol/min/mg protein with a low apparent Km value similar to human UGT2B7 .

What expression systems are most effective for producing recombinant rat UGT2B7?

Multiple expression systems have been successfully employed for recombinant rat UGT2B7 production:

  • E. coli expression systems: Commonly used for producing His-tagged recombinant rat UGT2B7, typically expressing the protein from Phe18 to Val495, which excludes the transmembrane domain to improve solubility .

  • Mammalian cell expression systems: These provide proper post-translational modifications and more native-like protein folding. HEK293 cells have been particularly effective for producing functional rat UGT2B7 with various tags (His, Fc, Avi) .

  • Bac-to-Bac systems: Used for double expression studies investigating protein-protein interactions between UGT2B7 and other enzymes .

For optimal activity, researchers should consider the inclusion of proper tags (His, GST, etc.) and careful design of the expression construct to exclude the hydrophobic transmembrane domain while maintaining catalytic function .

How can rat UGT2B7 enzyme activity be reliably measured in liver microsomes?

For reliable measurement of rat UGT2B7 activity in liver microsomes, the following methodological considerations are critical:

  • Latency removal: UGT2B7 activity in rat liver microsomes (RLM) requires removal of enzyme latency using detergents such as Brij58 at optimized concentrations (0.1 mg/mg protein) .

  • BSA addition: Including 1% (w/v) bovine serum albumin (BSA) in the incubation mixture increases ZDVG formation by approximately 2.5-fold in RLM compared to reactions without BSA .

  • Linearity conditions: For accurate kinetic measurements, ensure that:

    • Metabolite formation is linear with protein concentration (up to 6 mg/mL for RLM)

    • Incubation time linearity extends up to 180 min at RLM concentration of 2 mg/mL

    • Metabolite formation doesn't exceed 30% of substrate conversion

  • Detection methods: HPLC with appropriate detection systems (typically UV or MS detection) provides reliable quantification of glucuronide metabolites .

The intrinsic clearance (Vmax/Km) of ZDV in rat liver microsomes is approximately 0.16 μL/mg/min, which is about ten times lower than in human liver microsomes (1.65 μL/mg/min) .

What analytical techniques are recommended for detection and quantification of rat UGT2B7-mediated glucuronidation?

For optimal detection and quantification of rat UGT2B7-mediated glucuronidation:

  • HPLC-UV/HPLC-MS/MS: These are the most commonly employed methods for analyzing glucuronide metabolites. LC-MS/MS offers higher sensitivity and specificity, particularly valuable for complex biological matrices .

  • Enzyme immunoassays: ELISA-based detection using UGT2B7-specific antibodies can be employed for protein quantification rather than activity measurement .

  • RT-PCR: For mRNA expression analysis of UGT2B7, real-time quantitative PCR with gene-specific primers provides reliable quantification relative to housekeeping genes like β-actin .

  • Appropriate internal standards: When using chromatographic methods, suitable internal standards should be employed to ensure accurate quantification.

For microplate-based assays, the enzyme-substrate reaction can be terminated by adding sulphuric acid solution, with color change measured spectrophotometrically at 450nm ± 10nm. The concentration of UGT2B7 in samples can then be determined by comparing the O.D. against a standard curve .

What are the key functional differences between rat and human UGT2B7?

Several important functional differences exist between rat and human UGT2B7 that researchers should consider:

These differences highlight the importance of caution when extrapolating data from rat models to predict human drug metabolism and interactions.

How reliable are rat models for predicting human UGT2B7-mediated drug interactions?

The reliability of rat models for predicting human UGT2B7-mediated drug interactions is limited by several factors:

Researchers should exercise caution when extrapolating inhibitory metabolic data from rats to humans and ideally conduct parallel studies in both species when possible. When using rat models, researchers should acknowledge these limitations and consider complementary approaches, such as in vitro studies with human microsomes or hepatocytes, to strengthen the translational relevance of their findings.

How do protein-protein interactions affect rat UGT2B7 activity?

Protein-protein interactions significantly impact rat UGT2B7 activity through several mechanisms:

  • Homo-oligomerization: UGT2B7 can form dimers and higher-order oligomers with itself, which can alter substrate binding and catalytic efficiency. Studies using fluorescence resonance energy transfer (FRET) and co-immunoprecipitation (Co-IP) techniques have revealed stable homo-dimerization of UGT enzymes .

  • Hetero-dimerization with other UGTs: UGT2B7 can form heterodimers with other UGT isoforms, which can significantly affect its catalytic activity. For instance, in one study, morphine 6-glucuronide formation was 4.5-fold higher in cells co-transfected with two UGT isoforms compared to cells transfected with a single UGT, demonstrating the modulatory effect of UGT-UGT interactions on enzyme activity .

  • Interactions with CYP enzymes: UGT2B7 can interact with cytochrome P450 enzymes, forming metabolosomes (functional units of metabolism). These interactions can modulate the activity of both enzyme systems. For example, co-expression of UGT2B7 with CYP3A4 decreased the Vmax value of CYP3A4 activity without affecting its Km value .

  • Other protein interactions: Immunoprecipitation studies followed by LC-MS/MS analysis have identified interactions between UGT2B7 and other drug-metabolizing enzymes including epoxide hydrolase 1, carboxylesterase 1, alcohol dehydrogenases, and glutathione S-transferases .

These protein-protein interactions can explain some of the variability observed in UGT2B7 activity across different experimental systems and highlight the importance of considering the cellular context when studying this enzyme.

What role does rat UGT2B7 play in the metabolism of endogenous compounds?

Rat UGT2B7 plays crucial roles in the metabolism of several endogenous compounds:

  • Bile acids: UGT2B7 participates in the detoxification of bile acids, particularly hyodeoxycholic acid (HDCA). The enzyme catalyzes the glucuronidation of these compounds, facilitating their elimination .

  • Steroid hormones: UGT2B7 metabolizes various steroid hormones, including 17β-estradiol. The enzyme shows specific activity toward 3,4-catechol estrogens and estriol, suggesting its importance in regulating the levels and activities of these potent estrogen metabolites .

  • Fatty acid derivatives: UGT2B7 metabolizes arachidonic acid metabolites such as 20-hydroxyeicosatetraenoic acid (20-HETE). Genetic polymorphisms in UGT2B7 can reduce 20-HETE glucuronidation, potentially affecting vascular function since 20-HETE is a potent vasoconstrictor .

  • Hydroxylated retinoids: Studies have shown that UGT2B7 is involved in the glucuronidation of retinoids and their oxidized derivatives. Interestingly, biologically active forms of retinoic acid can suppress the expression of UGT2B7 in intestinal cells, indicating a potential feedback mechanism .

Understanding the role of UGT2B7 in endogenous compound metabolism is important for interpreting how genetic variations or drug-induced changes in UGT2B7 activity might affect physiological processes beyond drug metabolism.

What techniques can be used to study the structure-function relationship of rat UGT2B7?

Several advanced techniques can be employed to investigate the structure-function relationship of rat UGT2B7:

  • Homology modeling: Due to the difficulty in crystallizing membrane-bound proteins like UGTs, homology modeling based on crystallized soluble forms of plant and bacterial UGTs has been used to predict the three-dimensional structure of UGT2B7. These models can provide information about substrate binding sites and protein-protein interaction interfaces .

  • Site-directed mutagenesis: This approach has been critical in identifying key amino acid residues that determine substrate specificity. For example, studies have shown that phenylalanine 33 in UGT2B4 (corresponding to tyrosine 33 in UGT2B7) is critical for substrate recognition and catalytic activity. Mutation of this aromatic residue significantly affected the ability to glucuronidate various substrates .

  • Chimeric proteins: Creating chimeric proteins between different UGT isoforms can help identify domains responsible for specific functions. This approach has been particularly useful for studying the N-terminal domain's role in substrate binding versus the C-terminal domain's role in UDP-glucuronic acid binding.

  • Fluorescence resonance energy transfer (FRET): This technique has been effectively used to study protein-protein interactions between UGT isoforms. FRET efficiency measurements can provide information about the spatial arrangement of UGT proteins in hetero-dimers and oligomers .

  • Co-immunoprecipitation (Co-IP): Co-IP followed by mass spectrometry analysis can identify protein interaction partners of UGT2B7, providing insights into its involvement in metabolic complexes .

When studying structure-function relationships, it's important to note that genetic polymorphisms can result in altered affinities to target proteins and differential effects on catalytic activities .

What compounds are known to inhibit rat UGT2B7 and how does this compare to human UGT2B7?

Various compounds have been identified as inhibitors of rat UGT2B7, with notable differences compared to human UGT2B7:

Inhibitors of rat UGT2B7:

  • Mitragynine: Inhibits rat UGT2B7 with an IC50 value of 51.20 ± 5.95 μM

  • Zerumbone: Potently inhibits rat UGT2B7 with an IC50 value of 8.14 ± 2.12 μM

  • Fluconazole: Used as an inhibitor of both UGT2B4 and UGT2B7 in experimental settings

Comparative inhibition between rat and human UGT2B7:

CompoundHuman UGT2B7 IC50 (μM)Rat UGT2B7 IC50 (μM)
Andrographolide6.18 ± 1.27No significant inhibition
Kaempferol-3-rutinoside18.56 ± 8.62No significant inhibition
Mitragynine8.11 ± 4.4851.20 ± 5.95
Zerumbone4.57 ± 0.238.14 ± 2.12

These differences highlight the species-specific nature of UGT2B7 inhibition and underscore the challenges in extrapolating inhibition data from rat to human models. When designing drug interaction studies using rat models, researchers should consider these species differences and ideally conduct parallel studies with human enzymes .

How is rat UGT2B7 expression regulated at the transcriptional level?

Rat UGT2B7 expression is regulated at the transcriptional level through several mechanisms:

  • Retinoid signaling: Biologically active retinoids such as all-trans retinoic acid (atRA) and 9-cis retinoic acid can suppress UGT2B7 mRNA expression. This effect appears to be specific to UGT2B7 and doesn't affect other UGT isoforms like UGT2B15 or UGT1A6. The suppression occurs in a tissue-specific manner, affecting intestinal cells but not hepatic cells .

  • Nuclear receptors: Evidence suggests that nuclear receptors play a role in regulating UGT2B7 expression. The RAR agonist TTNPB is a potent suppressor of UGT2B7 mRNA expression, suggesting involvement of retinoic acid receptors in UGT2B7 regulation .

  • Tissue-specific regulation: Transcriptional regulation of UGT2B7 varies across tissues. For example, regulatory mechanisms in liver differ from those in intestine, which explains the different expression patterns and responses to inducers/suppressors .

  • Oxidized retinoid derivatives: Interestingly, oxidized derivatives of retinoic acid (4-OH-atRA, 4-oxo-atRA, and 5,6-epoxy-atRA) do not suppress UGT2B7 expression, indicating that only biologically active forms of retinoic acid have this regulatory effect .

This complex regulation contributes to tissue-specific expression patterns of UGT2B7 and may provide mechanisms for enzyme induction or suppression in response to physiological stimuli or xenobiotic exposure.

How do polymorphic variants of rat UGT2B7 affect enzyme function and drug metabolism?

While less extensively studied than human UGT2B7 polymorphisms, variants of rat UGT2B7 can significantly impact enzyme function and drug metabolism:

  • Catalytic activity: Polymorphic variants can exhibit altered catalytic efficiency toward various substrates. Similar to human UGT2B7, where the UGT2B7*2 polymorphism reduces 20-hydroxyeicosatetraenoic acid (20-HETE) glucuronidation, rat UGT2B7 variants may show substrate-specific changes in activity .

  • Protein-protein interactions: Genetic polymorphisms can result in altered affinities for protein-protein interactions. FRET studies with polymorphic variants of UGT enzymes have shown variable FRET efficiencies and donor-acceptor distances, suggesting that genetic polymorphisms affect the spatial arrangement and interaction potential of these enzymes .

  • Substrate specificity: Mutations in key amino acid residues can dramatically alter substrate specificity. As demonstrated with UGT2B4 (related to UGT2B7), substitution of phenylalanine 33 by leucine suppressed activity toward hyodeoxycholic acid and impaired glucuronidation of several other substrates including 4-hydroxyestrone and 17-epiestriol .

  • Metabolic clearance: Variants may exhibit different intrinsic clearance values for specific substrates, potentially leading to altered pharmacokinetics of drugs metabolized by UGT2B7.

When studying rat UGT2B7, researchers should consider characterizing the specific variant used in their studies and acknowledge potential strain differences that might impact extrapolation of results.

What are the optimal conditions for assessing rat UGT2B7 activity in vitro?

For optimal assessment of rat UGT2B7 activity in vitro, researchers should consider these methodological factors:

  • Microsomal preparation:

    • Use fresh or properly stored (-80°C) rat liver microsomes

    • Typical protein concentration: 2 mg/mL for kinetic studies

    • Ensure removal of enzyme latency with detergents

  • Buffer and cofactor conditions:

    • Buffer: Usually 50-100 mM phosphate or Tris buffer, pH 7.4-7.5

    • Include MgCl2 (usually 5-10 mM)

    • Add 1% (w/v) bovine serum albumin to enhance activity

    • UDPGA (uridine diphosphoglucuronic acid) concentration: 2-5 mM

  • Detergent treatment:

    • Use Brij58 at 0.1 mg/mg protein to remove latency

    • Add detergent before adding substrate and cofactor

  • Incubation parameters:

    • Temperature: 37°C

    • Incubation time: Ensure linearity (up to 180 minutes for rat liver microsomes)

    • Ensure substrate consumption does not exceed 30% to maintain initial velocity conditions

    • Perform reactions in triplicate

  • Substrate considerations:

    • For general UGT2B7 activity: Zidovudine (AZT) is a commonly used probe substrate

    • Substrate concentration: Around Km value for kinetic studies, or a range of concentrations for determining Km and Vmax

    • Include appropriate vehicle controls (usually ≤1% organic solvent)

  • Reaction termination and analysis:

    • Terminate reactions with organic solvent (methanol, acetonitrile) or perchloric acid

    • Centrifuge to remove precipitated protein before analysis

    • Analyze by HPLC or LC-MS/MS with appropriate internal standards

These optimized conditions ensure reliable and reproducible measurement of rat UGT2B7 activity in vitro.

What controls and validation steps are essential when working with recombinant rat UGT2B7?

When working with recombinant rat UGT2B7, several critical controls and validation steps should be implemented:

  • Expression validation:

    • Western blot confirmation using UGT2B7-specific antibodies

    • Mass spectrometry validation of expressed protein sequence

    • Quantification of recombinant protein expression level

  • Activity validation:

    • Confirmation of enzymatic activity using well-established UGT2B7 substrates (e.g., zidovudine)

    • Determination of kinetic parameters (Km, Vmax) and comparison with literature values

    • Assessment of substrate specificity profile to confirm isoform identity

  • Essential controls:

    • Negative control: Expression system without UGT2B7 gene

    • Positive control: Well-characterized batch of recombinant UGT2B7 or rat liver microsomes

    • Vehicle control: To account for potential solvent effects

    • No-UDPGA control: To confirm dependence on the co-substrate

    • Heat-inactivated enzyme control: To distinguish enzymatic from non-enzymatic reactions

  • Batch consistency checks:

    • Regular activity checks with standard substrates

    • Protein concentration determination

    • Storage stability assessment at different time points

  • Protein quality assessment:

    • Size-exclusion chromatography to assess aggregation state

    • Thermal stability analysis

    • Assessment of glycosylation status if expressed in eukaryotic systems

  • Cross-reactivity controls:

    • When using antibody-based detection, validate lack of cross-reactivity with other UGT isoforms

    • For activity assays, confirm specificity using known selective inhibitors

These controls and validation steps ensure the reliability and reproducibility of data generated using recombinant rat UGT2B7.

How can researchers account for the impact of protein-protein interactions when studying rat UGT2B7 in different experimental systems?

To account for protein-protein interactions when studying rat UGT2B7 across different experimental systems, researchers should consider these methodological approaches:

  • Selection of appropriate expression systems:

    • Single expression systems: Useful for studying intrinsic UGT2B7 properties

    • Co-expression systems: Necessary for studying effects of protein-protein interactions

    • Comparison between reconstituted systems and native microsomes to identify interaction effects

  • Analytical techniques for detecting interactions:

    • FRET analysis: To detect and quantify protein-protein interactions in living cells

    • Co-immunoprecipitation: To identify interaction partners

    • Crosslinking studies: To capture transient interactions

    • Bioluminescence resonance energy transfer (BRET): Alternative to FRET for live-cell interaction studies

  • Functional assessment approaches:

    • Compare activity in various systems (e.g., reconstituted lipid vesicles vs. microsomes vs. whole cells)

    • Systematically introduce potential interaction partners to identify modulatory effects

    • Use selective modulators of protein-protein interactions

  • Data interpretation considerations:

    • Acknowledge system-dependent differences in kinetic parameters

    • Consider the potential impact of differential expression levels of interaction partners

    • Account for membrane environment differences between systems

    • Recognize that kinetic parameters obtained in isolated recombinant systems may not reflect in vivo behavior

  • Complementary approaches:

    • Molecular modeling of protein-protein interfaces

    • Mutagenesis of putative interaction domains

    • Peptide competition studies to disrupt specific interactions

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.