BPHL Human

Biphenyl Hydrolase-Like Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to Biphenyl Hydrolase-Like Protein (BPHL)

BPHL (biphenyl hydrolase-like protein), also termed valacyclovir hydrolase, is a serine hydrolase encoded by the BPHL gene located on chromosome 6p25 . Initially cloned from human breast carcinoma cells, BPHL belongs to the α/β-hydrolase fold family and is highly expressed in the liver, kidney, and intestine . Its primary characterized roles include:

  • Hydrolytic activation of antiviral prodrugs (e.g., valacyclovir and valganciclovir) .

  • Detoxification of homocysteine thiolactone (HCTL), a toxic metabolite linked to cardiovascular and neurological diseases .

Gene and Protein Structure

  • Gene ID: 670 (NCBI Gene) .

  • Protein: 274 amino acids, molecular weight ~32.5 kDa .

  • Catalytic Triad: Ser122-Asp227-His255, with a conserved GSXSG motif .

  • Substrate Specificity: Preferentially hydrolyzes amino acid ester prodrugs and HCTL due to a hydrophobic acyl pocket and open leaving-group groove .

Key Functions

  1. Prodrug Activation
    BPHL converts valacyclovir to acyclovir (anti-herpes drug) and valganciclovir to ganciclovir (anti-CMV drug) with high specificity .

  2. HCTL Detoxification
    BPHL hydrolyzes HCTL with a catalytic efficiency (kcat/Kmk_{cat}/K_m) of 7.7×104M1s17.7 \times 10^4 \, \text{M}^{-1}\text{s}^{-1}, significantly surpassing other enzymes like paraoxonase-1 (PON1) and bleomycin hydrolase (Blmh) .

EnzymeCatalytic Efficiency (kcat/Kmk_{cat}/K_m)Substrate Saturation
BPHL7.7×104M1s17.7 \times 10^4 \, \text{M}^{-1}\text{s}^{-1}Saturated at 10 mM HCTL
Blmh<1×103M1s1< 1 \times 10^3 \, \text{M}^{-1}\text{s}^{-1}Not saturated at 20 mM
PON1<1×102M1s1< 1 \times 10^2 \, \text{M}^{-1}\text{s}^{-1}Low activity

Data sourced from .

Hyperhomocysteinemia

BPHL mitigates HCTL toxicity, a risk factor for protein misfolding in cardiovascular and neurodegenerative diseases . Knockout studies in mice show increased HCTL sensitivity and memory deficits .

Cancer Biology

  • Lung Adenocarcinoma (LUAD): BPHL is upregulated in tumor tissues (fold change = 2.253, P=8.87×1027P = 8.87 \times 10^{-27}) .

  • Functional Impact:

    • In vitro: BPHL knockdown reduces proliferation, colony formation, and metastasis in A549 and NCI-H1299 cells .

    • In vivo: Tumors from BPHL-silenced cells grow slower and exhibit increased apoptosis .

ParameterBPHL Knockdown vs. Control
Proliferation↓ 40–60% (MTT assay)
Caspase 3/7 Activity↑ 3–4 fold
Metastasis↓ 50–70% (Transwell assay)

Data from .

Therapeutic Implications

  • Prodrug Design: BPHL’s substrate specificity informs the development of ester-based antiviral and anticancer prodrugs .

  • Cancer Therapy: Targeting BPHL may inhibit tumor growth in LUAD, though redundancy in prodrug activation (e.g., carboxylesterases) limits its exclusivity .

Research Limitations and Future Directions

  • Redundant Enzymes: Valacyclovir hydrolysis persists in Bphl knockout mice, suggesting alternative pathways .

  • Unclear Physiological Role: While HCTL detoxification is established, broader metabolic functions remain underexplored .

Product Specs

Introduction
BPHL, a member of the AB hydrolase superfamily, is a serine hydrolase that catalyzes the hydrolytic activation of amino acid ester prodrugs of nucleoside analogs. It is highly expressed in the liver and kidneys, with lower levels found in the heart, intestines, and skeletal muscle. BPHL plays a role in detoxification processes and exhibits specific alpha-amino acid ester hydrolase activity, preferring small, hydrophobic, and aromatic side chains. While not strictly required, a primary alcohol is the preferred leaving group.
Description
Produced in E. coli, our BPHL is a single, non-glycosylated polypeptide chain comprising 275 amino acids (residues 38-291). It has a molecular weight of 31.1 kDa. For purification, a 21 amino acid His-tag is fused to the N-terminus, and the protein is purified using proprietary chromatographic methods.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The BPHL protein solution (0.5 mg/mL) is formulated in 20 mM Tris-HCl buffer (pH 8.0), 1 mM DTT, 50 mM NaCl, and 10% glycerol.
Purity
Purity exceeds 95% as determined by SDS-PAGE analysis.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Avoid repeated freeze-thaw cycles.
Synonyms

Biphenyl Hydrolase-Like (serine hydrolase), Bph-rp, Breast Epithelial Mucin-Associated Antigen, MCNAA, VACVASE, MGC41865, MGC125930.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSVTSAKVAV NGVQLHYQQT GEGDHAVLLL PGMLGSGETD FGPQLKNLNK KLFTVVAWDP RGYGHSRPPD RDFPADFFER DAKDAVDLMK ALKFKKVSLL GWSDGGITAL IAAAKYPSYI HKMVIWGANA YVTDEDSMIY EGIRDVSKWS ERTRKPLEAL YGYDYFARTC EKWVDGIRQF KHLPDGNICR HLLPRVQCPA LIVHGEKDPL VPRFHADFIH KHVKGSRLHL MPEGKHNLHL RFADEFNKLA EDFLQ

Q&A

What is Biphenyl Hydrolase-Like (BPHL) protein?

BPHL is a novel human serine hydrolase originally cloned from a breast carcinoma cDNA library . It functions as a valacyclovirase (VACVase) that catalyzes the hydrolytic activation of valine ester prodrugs of antiviral drugs such as acyclovir and ganciclovir . The enzyme has been extensively characterized as a 27-kDa basic protein with specific enzymatic activities toward various amino acid ester prodrugs of therapeutic nucleoside analogues. BPHL has gained significant attention in biochemical research due to its dual role as both a valacyclovir hydrolase and an efficient homocysteine thiolactonase (HCTLase) . Its physiological importance stems from its involvement in drug activation pathways and potential protective effects against homocysteine-related pathologies.

What is the catalytic efficiency of BPHL compared to other enzymes with similar functions?

BPHL demonstrates remarkably higher catalytic efficiency for homocysteine thiolactone (HCTL) hydrolysis compared to other enzymes. The catalytic efficiency (kcat/Km) of recombinant BPHL for HCTL hydrolysis is 7.7 × 10^4 M^-1s^-1, which is orders of magnitude higher than that of other known HCTL-hydrolyzing enzymes such as paraoxonase-1 (PON1) and bleomycin hydrolase (Blmh) . For comparison, the catalytic efficiency of PON1 for HCTL hydrolysis is approximately 100-fold lower than that of Blmh, which itself is not saturated even at 20 mM HCTL. This significant difference in efficiency suggests BPHL likely plays a more crucial physiological role in detoxifying HCTL, a reactive homocysteine metabolite associated with cardiovascular, autoimmune, and neurological diseases.

How does BPHL function in prodrug activation?

BPHL functions as a critical enzyme in the activation pathway of several important prodrugs, particularly valacyclovir. The activation mechanism involves hydrolytic cleavage of the ester bond between the parent drug and its promoiety. For valacyclovir (the 5'-valyl ester prodrug of acyclovir), BPHL efficiently hydrolyzes the ester bond to release the active drug acyclovir . This activation typically occurs after intestinal absorption of the prodrug via the hPEPT1 peptide transporter. BPHL's enzymatic action contributes significantly to the increased bioavailability of acyclovir (three to five times higher) when administered orally as valacyclovir compared to direct administration of acyclovir . The enzyme exhibits significant specificity constants (kcat/Km) of 420 mm^-1·s^-1 for valacyclovir and 53.2 mm^-1·s^-1 for valganciclovir, demonstrating its efficiency in prodrug activation .

How does substrate specificity affect BPHL's hydrolytic activity?

BPHL demonstrates a clear preference for certain substrates based on their chemical structures and properties. The enzyme exhibits high specificity for amino acid ester prodrugs, particularly those with hydrophobic amino acyl promoieties such as the valine ester in valacyclovir . This specificity is directly related to the hydrophobic acyl-binding site formed by residues I158, G161, I162, and L229. BPHL shows remarkable efficiency in hydrolyzing valacyclovir and other valine ester prodrugs, while maintaining broad specificity for various nucleoside analogues as parent drugs . This dual characteristic of specific recognition of certain promoieties while accommodating diverse parent drug structures makes BPHL particularly valuable for prodrug activation. The enzyme's relatively less constrained nucleoside-binding site enables it to process various nucleoside analogues while still maintaining selectivity for the acyl portion of the molecule. This balance between specificity and adaptability is crucial for BPHL's physiological role and its potential applications in prodrug design .

What methods are most effective for measuring BPHL enzymatic activity?

For accurate measurement of BPHL enzymatic activity, researchers typically employ high-performance liquid chromatography (HPLC) methods to quantify the conversion of substrates to products. When studying valacyclovir hydrolysis, monitoring the formation of acyclovir provides a reliable measure of enzyme activity . For homocysteine thiolactonase activity, researchers measure the disappearance of homocysteine thiolactone (HCTL) or the formation of homocysteine using spectrophotometric or fluorometric assays . Recombinant expression of BPHL in Escherichia coli followed by purification offers a consistent source of enzyme for activity measurements. Mass spectrometry (MS) verification of both the recombinant protein sequence and the hydrolytic products ensures accuracy of the enzymatic characterization . Kinetic parameters such as Km, kcat, and catalytic efficiency (kcat/Km) are determined through standard enzyme kinetics approaches, typically using substrate concentration ranges that span below and above the Km value. For comprehensive characterization, researchers often employ multiple substrates to establish specificity profiles and compare catalytic efficiencies across different potential physiological substrates.

What role does BPHL play in homocysteine-related pathologies?

BPHL functions as a highly efficient homocysteine thiolactonase (HCTLase), hydrolyzing homocysteine thiolactone (HCTL) with remarkable catalytic efficiency . This activity has significant implications for homocysteine-related pathologies, as HCTL is a reactive metabolite that can cause homocysteinylation of lysine residues in proteins, leading to protein aggregation and malfunction. Such protein modifications are established risk factors for cardiovascular, autoimmune, and neurological diseases . The catalytic efficiency of BPHL for HCTL hydrolysis (7.7 × 10^4 M^-1s^-1) far exceeds that of other known HCTLases such as paraoxonase-1 (PON1) and bleomycin hydrolase (Blmh), suggesting BPHL may play a predominant physiological role in detoxifying HCTL . This protective function potentially mitigates the damage caused by protein homocysteinylation, thereby reducing risk for associated diseases. The discovery of BPHL's HCTLase activity provides a mechanistic link between this enzyme and various pathological conditions associated with elevated homocysteine levels, offering new perspectives for therapeutic interventions targeting homocysteine metabolism.

How can BPHL be targeted for prodrug design and improved drug delivery?

BPHL represents an important molecular target for rational prodrug design due to its efficient hydrolytic activity and broad specificity for nucleoside analogues . Researchers can exploit BPHL's substrate preferences to design prodrugs with improved pharmacokinetic properties. The enzyme shows high specificity for amino acid ester prodrugs, particularly those with hydrophobic amino acyl promoieties such as valine . This preference can be leveraged to design prodrugs that are efficiently absorbed via peptide transporters in the intestine and subsequently activated by BPHL. The structural model of BPHL provides valuable insights for prodrug design, highlighting the importance of the hydrophobic acyl-binding site and the relatively unrestricted nucleoside-binding pocket . When designing BPHL-activated prodrugs, researchers should consider: (1) incorporating valine or similar hydrophobic amino acids as promoieties, (2) ensuring the ester linkage is accessible to the catalytic triad, and (3) optimizing the parent drug structure to fit within the nucleoside-binding site. This approach has already proven successful with valacyclovir and valganciclovir, which show significantly improved bioavailability compared to their parent compounds .

What are the implications of BPHL genetic variations on drug metabolism?

Genetic variations in the BPHL gene may significantly impact drug metabolism, particularly for prodrugs that rely on BPHL for activation. While comprehensive population studies specifically on BPHL polymorphisms are still emerging, the potential clinical implications are substantial. Individuals with genetic variants resulting in reduced BPHL activity might experience diminished efficacy of prodrugs such as valacyclovir and valganciclovir due to incomplete conversion to the active drug form . Conversely, variants causing enhanced BPHL activity could potentially lead to more rapid prodrug activation, altering the pharmacokinetic profile and possibly affecting the therapeutic window. BPHL gene disruption studies can provide insights into how complete absence of the enzyme affects prodrug metabolism . The investigation of BPHL genetic variations could help explain inter-individual variability in response to certain medications and guide personalized medicine approaches. Researchers studying these variations typically employ gene sequencing, expression analysis, and functional enzyme assays to characterize the impact of specific polymorphisms on enzyme activity. Understanding the relationship between BPHL genotype and drug metabolism phenotype represents an important frontier in pharmacogenomics research with direct clinical implications.

What experimental approaches are optimal for investigating BPHL activity in tissue-specific contexts?

To effectively investigate tissue-specific BPHL activity, researchers should employ a multi-faceted approach combining molecular, cellular, and physiological techniques. Immunohistochemistry and in situ hybridization can precisely localize BPHL expression across different tissues, while quantitative PCR and western blotting provide quantitative measures of expression levels . For functional studies, tissue-specific enzyme activity assays using validated substrates such as valacyclovir or homocysteine thiolactone can determine regional variations in BPHL catalytic efficiency. Cell type-specific promoter analysis helps elucidate the regulatory mechanisms controlling tissue-specific expression patterns. Advanced approaches include generating conditional knockout mouse models with tissue-specific BPHL deletion to evaluate functional consequences, and developing fluorescent or radioactive tracers to monitor BPHL-dependent prodrug activation in vivo using imaging techniques. Single-cell RNA sequencing can reveal cell-specific expression patterns within heterogeneous tissues, while organoid models derived from different tissues provide systems for studying BPHL function in physiologically relevant three-dimensional contexts. These complementary approaches together provide comprehensive insights into how BPHL function varies across different tissues and cell types, critical for understanding its role in drug metabolism and disease processes.

How can computational modeling enhance our understanding of BPHL substrate interactions?

Computational modeling represents a powerful approach for elucidating BPHL-substrate interactions and guiding experimental design. Homology modeling based on related serine hydrolases, as demonstrated with the model developed using the crystal structure of 2-hydroxy-6-oxo-7-methylocta-2,4-dienoate hydrolase, provides valuable structural insights . Researchers can employ molecular docking studies to predict binding modes and affinities of various substrates, helping to explain experimental observations regarding substrate specificity. Molecular dynamics simulations offer deeper insights by capturing the dynamic nature of enzyme-substrate interactions, revealing transient binding events and conformational changes that may be critical for catalysis. Quantum mechanics/molecular mechanics (QM/MM) approaches can model the actual chemical reaction, providing mechanistic insights into the catalytic process. Virtual screening of compound libraries against the BPHL binding site can identify potential new substrates or inhibitors. Structure-based pharmacophore modeling helps identify the essential features required for substrate recognition. For investigating genetic variants, computational mutagenesis and stability analysis can predict the functional consequences of specific mutations. Integration of these computational approaches with experimental validation creates a powerful iterative process for advancing our understanding of BPHL function and leveraging this knowledge for applications in drug design and therapeutic development.

What are the current contradictions or knowledge gaps in BPHL research that require resolution?

Despite significant advances in understanding BPHL biology, several important knowledge gaps and contradictions persist in the field. A fundamental question remains regarding the relative physiological importance of BPHL's dual functions as both a valacyclovir hydrolase and a homocysteine thiolactonase . While BPHL demonstrates remarkably high catalytic efficiency for both functions in vitro, the predominant role in vivo under various physiological and pathological conditions remains unclear. The complete tissue distribution profile of BPHL activity across all human tissues has not been fully characterized, limiting our understanding of its potential site-specific functions. The comprehensive substrate profile of BPHL beyond the currently identified substrates remains to be determined, raising questions about other potential endogenous or xenobiotic compounds that might be processed by this enzyme. The regulation of BPHL expression under different physiological states, disease conditions, and in response to various stimuli is poorly understood. Limited information exists regarding potential protein-protein interactions that might modulate BPHL activity in cellular contexts. The clinical significance of genetic polymorphisms affecting BPHL function has not been systematically investigated across diverse populations. These knowledge gaps represent important opportunities for future research that could significantly advance our understanding of BPHL biology and its implications for human health and disease.

Product Science Overview

Introduction

Biphenyl Hydrolase-Like (BPHL) is a serine hydrolase enzyme that plays a significant role in the hydrolytic activation of amino acid ester prodrugs of nucleoside analogs, such as valacyclovir and valganciclovir. This enzyme is also known for its role in detoxification processes and has been identified as a highly efficient homocysteine thiolactonase (HCTLase).

Gene and Protein Structure

The BPHL gene is located on chromosome 6 and encodes a protein that belongs to the serine protease family of hydrolytic enzymes. These enzymes are characterized by the presence of a serine residue in their active site, which is crucial for their catalytic activity. The BPHL protein has several aliases, including Valacyclovir Hydrolase, Biphenyl Hydrolase-Related Protein, and Valacyclovirase .

Function and Mechanism

BPHL catalyzes the hydrolytic activation of amino acid ester prodrugs by cleaving the ester bond, thereby converting the prodrug into its active form. For instance, BPHL activates valacyclovir to acyclovir, an antiviral drug used to treat herpes infections. The enzyme prefers substrates with small, hydrophobic, and aromatic side chains and does not have a stringent requirement for the leaving group, other than preferring a primary alcohol .

In addition to its role in drug activation, BPHL has been identified as a highly efficient homocysteine thiolactonase. Homocysteine thiolactone (HCTL) is a reactive metabolite of homocysteine that can cause protein aggregation and malfunction, leading to cardiovascular, autoimmune, and neurological diseases. BPHL hydrolyzes HCTL, thereby detoxifying it and preventing its harmful effects .

Recombinant Expression

Recombinant BPHL (rBPHL) is produced by expressing the BPHL gene in a host organism, such as Escherichia coli. The recombinant protein is then purified and characterized to study its structure and function. The catalytic efficiency of rBPHL for HCTL hydrolysis is significantly higher than that of other known HCTL detoxifying enzymes, such as paraoxonase-1 (PON1) and bleomycin hydrolase (Blmh) .

Clinical and Research Implications

The ability of BPHL to activate antiviral prodrugs and detoxify harmful metabolites makes it a valuable target for clinical and pharmaceutical research. Understanding the structure and function of BPHL can lead to the development of more effective prodrugs and therapeutic strategies for diseases associated with homocysteine metabolism.

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.