BLVRB Mouse

Biliverdin Reductase B Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Role in Stress Hematopoiesis

Studies using Blvrb-deficient mice (>12 months old) revealed:

  • Defective Erythropoiesis: Impaired recovery of red blood cells post-5-fluorouracil (5-FU) stress .

  • Megakaryocyte Bias: 5-fold expansion of bone marrow megakaryocytes and elevated platelet counts during stress recovery .

  • Oxidant Mishandling: Increased lipid peroxidation in erythroid progenitors .

Cytoprotective Mechanisms

BLVRB mitigates oxidative stress by reducing biliverdin to bilirubin and flavins (e.g., FAD, FMN) via NADPH . Unlike its isoform BLVRA, BLVRB is specific to the biliverdin IXβ isomer .

Stress Hematopoiesis in Blvrb⁻/⁻ Mice

ParameterBlvrb⁻/⁻ MiceWild-Type Mice
Platelet Count (Day 14)>11 × 10⁶/μL ~1.8 × 10⁶/μL
Bone Marrow Megakaryocytes5-fold increase Baseline levels
Splenic Erythroid RecoveryImpaired Normal

Clinical Relevance

  • Biomarker for Atherosclerosis: Plasma BLVRB levels correlate with intraplaque hemorrhage (IPH) in carotid stenosis patients (737.32 vs. 520.94 MFI in IPH+ vs. IPH– patients) and predict recurrent stroke risk (HR = 1.6) .

  • Therapeutic Target: Anti-VEGFR-2 therapy reduced plasma BLVRB in murine models, suggesting utility in IPH prevention .

Inhibitor Development

BLVRB inhibitors like erythrosin extra bluish (IC₅₀ ~0.7 μM) and phloxine B (IC₅₀ ~1.8 μM) have been explored, though their drug-like properties remain suboptimal . Structural studies highlight Ser111 as critical for catalytic activity, guiding future inhibitor design .

Product Specs

Introduction

Biliverdin reductase B (BLVRB), also known as Enzyme Commission number (EC) 1.3.1.24, facilitates the transfer of electrons from reduced pyridine nucleotides to various molecules including flavins, methylene blue, pyrroloquinoline quinone, riboflavin, and methemoglobin. This enzyme plays a crucial role in protecting cells from oxidative stress and regulating iron metabolism. BLVRB catalyzes the reduction of biliverdin to bilirubin in the liver by converting a double bond between the second and third pyrrole ring into a single bond. It plays a vital role in the heme catabolic pathway of human erythrocytes and most mammalian species. BLVRB is highly expressed in organs such as the kidney, spleen, liver, and brain, while it is found at lower levels in the thymus and minimally detected in the testis.

Description

Recombinant Mouse BLVRB, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 229 amino acids, with 206 amino acids (1-206 a.a) forming the BLVRB protein and a 23 amino acid His-tag fused at the N-terminus. The molecular weight of the recombinant protein is 24.6 kDa. Purification is achieved using proprietary chromatographic techniques.

Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation

The BLVRB protein is supplied in a solution at a concentration of 0.5 mg/ml. The buffer consists of 20mM Tris-HCl at a pH of 8.0 and contains 10% glycerol.

Stability
For short-term storage (up to 2-4 weeks), the protein solution should be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To ensure stability during long-term storage, adding a carrier protein like HSA or BSA at a concentration of 0.1% is advisable. Repeated freezing and thawing of the protein solution should be avoided.
Purity
The purity of the BLVRB protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms

Flavin reductase (NADPH), FR, Biliverdin reductase B, BVR-B, Biliverdin-IX beta-reductase, NADPH-dependent diaphorase, NADPH-flavin reductase, FLR.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMTVKKIA IFGATGRTGL TTLAQAVQAG YEVTVLVRDS SRLPSEGPQP AHVVVGDVRQ AADVDKTVAG QEAVIVLLGT GNDLSPTTVM SEGTRNIVTA MKAHGVDKVV ACTSAFLLWD PTKVPPRLQD VTDDHIRMHK ILQESGLKYV AVMPPHIGDQ PLTGAYTVTL DGRGPSRVIS KHDLGHFMLR CLTTNEYDGH TTYPSHQYD.

Q&A

What is the primary function of BLVRB in mouse hematopoiesis?

BLVRB (flavin reductase NADPH) functions within the heme degradation pathway, specifically catalyzing the reduction of biliverdin to bilirubin. In mice, BLVRB exhibits isomer-restricted redox activity as part of the cytoprotective mechanisms involving heme oxygenases. While dispensable for steady-state hematopoiesis, BLVRB plays a crucial non-redundant cytoprotective role during stress hematopoiesis with significant impact on erythroid/megakaryocyte (E/Meg) fate determination . Research using computational models based on murine hematopoietic single-cell transcriptomic data has identified Blvrb as a biological driver connected to tumor necrosis factor stress pathways, establishing it as a key source of variation in defining hematopoietic cell heterogeneity .

How does BLVRB structure differ between mouse and human models?

Mouse BLVRB is a 206-amino acid protein with a molecular weight of approximately 24.6 kDa . The mouse protein shares significant homology with human BLVRB, enabling cross-species research applications. The mouse protein contains key functional domains that maintain its enzymatic activity, particularly its NAD(P)H-dependent reductase capacity. Structurally, the mouse protein contains the conserved catalytic domain responsible for biliverdin reduction, which appears functionally significant as mutations within this domain enhance megakaryocyte reactive oxygen species (ROS) production in hematopoietic cells . Commercial antibodies specific to BLVRB have been developed with cross-reactivity across human, mouse, and rat species , indicating substantial conservation of epitope regions among these mammals.

What are the standard mouse models used to study BLVRB function?

The primary mouse model for studying BLVRB function is the Blvrb-knockout (Blvrb−/−) mouse on a C57/BL6 background. These models are typically generated using targeting constructs designed to delete Blvrb exons 2-4 . Validation of successful knockout can be performed through immunoblot analysis and densitometric quantification of organs from Blvrb−/− mice, confirming the absence of the protein. F1 (heterozygote) crosses reveal genotypes of the expected Mendelian frequency (1:2:1 Blvrb+/+: Blvrb+/−: Blvrb−/−), confirming the viability of Blvrb−/− mice with no gender imbalance . For stress hematopoiesis studies, researchers commonly use 5-fluorouracil (5-FU) treatment to induce hematopoietic stress in both wild-type and Blvrb-deficient mice, allowing observation of differential recovery patterns in various blood cell lineages .

What are the recommended methods for validating BLVRB knockout in mouse models?

Validation of BLVRB knockout requires a multi-faceted approach:

  • Genotyping: PCR-based confirmation of deleted exons (typically exons 2-4) .

  • Protein Expression Analysis: Immunoblot analysis with densitometric quantification of tissues from Blvrb−/− mice compared to wild-type controls. This method can confirm the absence of the protein and assess any compensatory changes in related proteins (e.g., changes in Hmox1 and Blvra expression have been observed in Blvrb−/− spleens) .

  • Functional Validation: Assessment of biliverdin reductase activity using enzymatic assays with specific biliverdin isomers as substrates.

  • Phenotypic Characterization: Complete blood counts under both steady-state and stress conditions (e.g., post-5-FU treatment) to detect hematological abnormalities in knockout mice compared to wild-type controls .

When performing these validations, it's essential to include age-matched controls, as phenotypic changes may become more pronounced in aged Blvrb-deficient mice, particularly under stress conditions .

What techniques are most effective for studying BLVRB's role in stress hematopoiesis?

To effectively study BLVRB's role in stress hematopoiesis, researchers should implement a comprehensive experimental approach:

  • Stress Induction Protocol: Administration of 5-fluorouracil (5-FU) at 150 mg/kg via intraperitoneal injection is the established method to induce hematopoietic stress in both wild-type and Blvrb−/− mice .

  • Hematological Assessment: Serial complete blood counts (CBC) at defined intervals (typically days 7, 14, and 28 post-stress) to monitor recovery of erythrocytes, leukocytes, and platelets. Include reticulocyte and reticulated platelet counts to assess new cell production .

  • Flow Cytometry: Analysis of bone marrow and splenic cells for lineage markers, with particular attention to CD41+ (megakaryocyte) and erythroid populations. Analysis of ROS levels using ROS-sensitive dyes in conjunction with lineage markers (e.g., ROS+/CD41+ populations) .

  • Histological Examination: Bone marrow biopsies with immunohistochemical staining for von Willebrand factor (vWF) and other megakaryocyte markers. Spleen histology to assess changes in red pulp and white pulp constituents .

  • Molecular Analysis: Assessment of lipid peroxidation in erythroid cells as an indicator of oxidative stress and potentially disrupted cytoprotection in Blvrb−/− mice .

How can researchers effectively use recombinant BLVRB proteins in experimental designs?

Recombinant BLVRB proteins serve as valuable tools for mechanistic studies and can be incorporated into experimental designs through several approaches:

  • In vitro Enzymatic Assays: Using His-tagged recombinant mouse BLVRB (such as the commercially available protein expressed in E. coli) , researchers can perform kinetic studies to assess the enzyme's activity with various substrates and under different conditions. Standard assays monitor the NAD(P)H-dependent reduction of biliverdin isomers spectrophotometrically.

  • Structure-Function Analyses: Site-directed mutagenesis of recombinant BLVRB expression constructs allows investigation of specific amino acid residues critical for substrate binding or catalytic activity. The altered proteins can be expressed, purified, and characterized biochemically .

  • Protein-Protein Interaction Studies: Recombinant BLVRB can be used in pull-down assays, co-immunoprecipitation experiments, or surface plasmon resonance to identify and characterize interactions with other proteins in the heme degradation pathway or stress response networks.

  • Rescue Experiments: Transfection of BLVRB expression constructs into Blvrb-deficient cells can determine if wild-type or mutant versions of the protein can restore normal function, particularly in stress response contexts .

  • Antibody Validation: Recombinant proteins serve as positive controls for validating the specificity and sensitivity of anti-BLVRB antibodies used in Western blotting, immunohistochemistry, or flow cytometry .

When using recombinant proteins, researchers should consider the presence of tags (e.g., His-tag) that might affect protein folding or function, and validate that the recombinant protein displays enzymatic activity comparable to the native form.

How does BLVRB deficiency affect erythroid and megakaryocyte lineages during stress?

BLVRB deficiency creates a striking dichotomy in erythroid and megakaryocyte lineages during stress hematopoiesis:

Erythroid Effects:

  • Defective stress erythropoiesis in Blvrb−/− mice, particularly evident in aged animals

  • Impaired red blood cell recovery following 5-FU treatment

  • Disordered lipid peroxidation in erythroid cells, suggesting compromised cytoprotection against oxidative stress

  • Splenic atrophy with selective loss of red pulp constituents (erythrocytes/macrophages) while preserving white pulp

Megakaryocyte Effects:

  • Megakaryocyte-biased hematopoietic recovery following stress

  • ~5-fold expansion of megakaryocytes in Blvrb−/− bone marrow compared to wild-type controls

  • Formation of megakaryocyte clusters expressing von Willebrand factor (vWF)

  • Approximately 3-fold greater ROS+/CD41+ (megakaryocyte) subset in Blvrb−/− bone marrow post-5-FU treatment

  • Exaggerated rebound thrombocytosis that is not due to enhanced proplatelet formation

These observations suggest that BLVRB plays a role in erythrocyte/megakaryocyte partitioning during stress progenitor fate decisions, with its absence promoting megakaryopoiesis while impairing erythropoiesis . The link between BLVRB deficiency and increased ROS in megakaryocytes is particularly noteworthy, as ROS production is known to be required for megakaryocyte differentiation .

What molecular mechanisms underlie BLVRB's cytoprotective functions?

BLVRB's cytoprotective functions are mediated through several interconnected molecular mechanisms:

  • Antioxidant Pathway Participation: BLVRB functions within the heme degradation pathway that generates the antioxidant bilirubin. It specifically catalyzes the NAD(P)H-dependent reduction of biliverdin isomers to bilirubin, contributing to cellular antioxidant capacity .

  • Isomer-Specific Activity: BLVRB displays biliverdin isomer-restricted redox activity, suggesting specialized functions in processing specific metabolites generated during stress conditions .

  • Oxidative Stress Management: Deficiency in BLVRB leads to disordered lipid peroxidation, particularly in erythroid cells, indicating its role in protecting cellular membranes from oxidative damage .

  • Complementary Relationship with HMOX/BLVRA Pathway: BLVRB functions are distinct from and non-redundant with the predominant Hmox/Blvra pathway, establishing multiple layers of cytoprotection . Notably, Blvrb−/− mice exhibit approximately 50% reduction of Hmox1 and Blvra in the spleen, suggesting pathway crosstalk .

  • Stress Pathway Connection: Computational modeling has identified Blvrb as a biological driver linked to the tumor necrosis factor stress pathway, positioning it as a key factor in cellular responses to inflammatory stimuli .

The phenotypic similarity between stress-induced Blvrb−/− mice and age-dependent hematopoietic abnormalities (anemia with or without thrombocytosis) previously described in human and Hmox1−/− mice further supports BLVRB's role in an essential cytoprotective network .

How do aging and stress interact in BLVRB-deficient mouse models?

The interaction between aging and stress in BLVRB-deficient mouse models reveals important temporal aspects of BLVRB function:

  • Age-Dependent Phenotype Expression: While young Blvrb−/− mice show normal steady-state hematopoiesis, aged Blvrb−/− mice demonstrate more pronounced hematological abnormalities, particularly under stress conditions .

  • Stress Amplification: Hematopoietic stress (e.g., 5-FU treatment) magnifies the underlying defects in aged Blvrb−/− mice, resulting in:

    • More severe defects in erythroid recovery

    • More pronounced megakaryocyte expansion

    • Greater imbalance in erythroid/megakaryocyte lineage specification

  • Tissue-Specific Effects: Aging and stress in Blvrb−/− mice primarily affect the hematopoietic compartment, with detailed necropsy and histological studies confirming that post-stress phenotypes are restricted to hematopoietic tissues .

  • Temporal Recovery Patterns: While complete hematopoietic recovery occurs in Blvrb−/− mice by day 28 post-stress, the intermediate recovery phase (days 7-14) reveals significant differences in lineage-specific responses, suggesting BLVRB's importance during the acute stress response and recovery period rather than long-term homeostasis .

  • Similarity to Human Aging Conditions: The age-dependent and stress-responsive phenotype of Blvrb−/− mice parallels certain age-related hematological disorders in humans, such as anemia with or without thrombocytosis .

This age-stress interaction suggests that BLVRB functions as part of a compensatory mechanism that becomes increasingly important as organisms age and encounter more frequent or severe stressors, particularly in maintaining proper lineage balance during hematopoietic recovery .

How might BLVRB research inform therapeutic approaches for stress-induced hematological disorders?

BLVRB research offers several promising avenues for therapeutic development in stress-induced hematological disorders:

  • Targeted Cytoprotection: Understanding BLVRB's role in stress hematopoiesis could lead to targeted interventions that enhance cytoprotection specifically in vulnerable cell populations. For example, pharmacological enhancement of BLVRB activity might protect erythroid precursors during chemotherapy, radiation therapy, or other stress conditions .

  • Lineage Balance Modulation: The observed megakaryocyte-biased recovery in Blvrb−/− mice suggests that modulating BLVRB activity might allow controlled adjustment of erythroid/megakaryocyte lineage specification. This could be valuable in treating thrombocytopenia (low platelets) or polycythemia (excess erythrocytes) .

  • Oxidative Stress Management: BLVRB's connection to cellular redox status and lipid peroxidation indicates potential applications in conditions characterized by excessive oxidative stress. Enhancing BLVRB function could provide additional antioxidant protection in disorders where ROS contribute to pathology .

  • Age-Related Hematological Disorders: The phenotypic similarities between aged Blvrb−/− mice under stress and certain human age-related hematological disorders suggest BLVRB as a potential therapeutic target for addressing anemia and thrombocytosis in elderly populations .

  • Chemotherapy Adjuvants: Understanding the molecular basis of differential recovery in hematopoietic lineages following 5-FU treatment in Blvrb−/− mice could inform the development of adjuvant therapies that mitigate the hematological side effects of chemotherapy while preserving its anti-cancer effects .

Future therapeutic development will require deeper characterization of BLVRB's tissue-specific functions and the consequences of its modulation in diverse physiological and pathological contexts.

What are the recommended experimental controls when analyzing ROS levels in BLVRB-deficient hematopoietic cells?

When analyzing reactive oxygen species (ROS) levels in BLVRB-deficient hematopoietic cells, researchers should implement comprehensive controls to ensure accurate and interpretable results:

  • Genotype Controls:

    • Wild-type (Blvrb+/+) matched for age and background strain

    • Heterozygous (Blvrb+/−) mice to assess gene dosage effects

    • Positive controls such as Hmox1−/− cells, which are known to exhibit elevated ROS

  • Methodological Controls:

    • Unstained cells for autofluorescence baseline determination

    • ROS-inducing agent (e.g., hydrogen peroxide) treated cells as positive control

    • ROS scavenger (e.g., N-acetylcysteine) treated cells as negative control

    • Multiple ROS-sensitive dyes with different specificities to detect various ROS types

  • Lineage-Specific Controls:

    • Isolated lineage populations (CD41+ megakaryocytes, erythroid precursors, etc.)

    • Analysis of ROS in relation to cellular differentiation markers to distinguish effects at different maturation stages

    • Comparison of ROS+/CD41+ megakaryocyte subsets between genotypes and under various conditions

  • Temporal Controls:

    • Baseline measurements (pre-stress)

    • Multiple time points post-stress induction

    • Age-matched comparisons for both young and aged mice

  • Functional Validation:

    • Correlation of ROS levels with functional outcomes (cell viability, proliferation, differentiation)

    • Assessment of lipid peroxidation as a consequence of elevated ROS

    • Measurement of antioxidant enzyme activities (catalase, superoxide dismutase, etc.)

Given the observed ~3-fold greater ROS+/CD41+ subset in Blvrb−/− bone marrow post 5-FU treatment and the known requirement for ROS in megakaryocyte differentiation, these controls are essential for determining whether ROS changes are cause or consequence of altered lineage specification .

How can researchers differentiate between direct BLVRB effects and compensatory mechanisms in knockout models?

Differentiating between direct BLVRB effects and compensatory mechanisms in knockout models requires sophisticated experimental approaches:

  • Temporal Gene Manipulation:

    • Inducible knockout systems (e.g., Cre-ERT2) allowing BLVRB deletion at specific time points

    • Acute knockdown using siRNA or shRNA compared to constitutive knockout

    • Early vs. late analysis after BLVRB deletion to distinguish primary from secondary effects

  • Pathway Component Analysis:

    • Comprehensive analysis of related pathway components (e.g., Hmox1, Blvra) at protein and activity levels

    • Documented changes such as ~50% reduction of Hmox1 and Blvra in Blvrb−/− spleens may represent compensatory adjustments

    • Sequential inhibition studies (e.g., inhibiting potential compensatory pathways in Blvrb−/− cells)

  • Rescue Experiments:

    • Re-expression of BLVRB in knockout cells to determine which phenotypes are reversible

    • Expression of enzymatically inactive BLVRB mutants to distinguish catalytic from structural roles

    • Timing-controlled rescue to determine critical windows for BLVRB function

  • Cross-Species Validation:

    • Comparison of Blvrb−/− mouse phenotypes with BLVRB deficiency models in other species

    • Parallel analysis of mutations affecting the BLVRB catalytic domain across species

  • Multi-omics Approaches:

    • Transcriptomic analysis of Blvrb−/− cells before and after stress to identify adaptive responses

    • Metabolomic profiling to detect accumulated substrates or alternative pathway products

    • Proteomic studies to identify altered protein-protein interactions or post-translational modifications

  • Tissue-Specific Analysis:

    • Comparison of effects across different tissues (e.g., bone marrow vs. spleen)

    • Analysis of cell-autonomous effects using chimeric models or isolated cell populations

    • Examination of non-cell-autonomous factors through conditioned media experiments

The observation that CD163 transcript levels remain comparable between Blvrb−/− and Blvrb+/+ spleens despite changes in splenic architecture suggests selective maintenance of certain cell populations, highlighting the complexity of distinguishing direct effects from compensatory mechanisms .

What factors should researchers consider when selecting BLVRB antibodies for different applications?

When selecting BLVRB antibodies for research applications, several critical factors should be considered:

  • Species Cross-Reactivity: Choose antibodies validated for appropriate species reactivity. Some commercially available antibodies demonstrate cross-reactivity across human, mouse, and rat BLVRB , which can be advantageous for comparative studies but may lack specificity for species-specific epitopes.

  • Application-Specific Validation: Ensure the antibody has been validated for your specific application:

    • Western blotting: Confirm the antibody detects the expected ~24.6 kDa band in mouse tissues

    • Immunohistochemistry: Verify staining patterns in relevant tissues (particularly liver, spleen, and bone marrow)

    • Flow cytometry: Validate antibody performance in cell suspensions under fixation conditions

    • Immunoprecipitation: Confirm the antibody effectively captures native BLVRB protein

  • Epitope Considerations: Understand the target epitope location relative to functional domains of BLVRB. Antibodies targeting the catalytic domain might interfere with activity assays, while those targeting N- or C-terminal regions might be more suitable for detecting truncated variants.

  • Controls Availability: Ensure appropriate controls are available:

    • Positive controls: Wild-type mouse tissues known to express BLVRB

    • Negative controls: Blvrb−/− mouse tissues or cells

    • Recombinant protein standards: Purified mouse BLVRB proteins for standardization

  • Detection Method Compatibility: Select antibodies compatible with your detection system (fluorescent, enzymatic, etc.) and consider secondary antibody requirements for optimal signal-to-noise ratio.

  • Batch Consistency: For longitudinal studies, confirm lot-to-lot consistency or purchase sufficient quantity from a single lot to complete the study.

Proper antibody selection and validation are essential for accurate interpretation of BLVRB expression, localization, and function in experimental systems.

How should researchers optimize experimental conditions for studying BLVRB enzymatic activity?

Optimizing experimental conditions for studying BLVRB enzymatic activity requires careful consideration of multiple parameters:

  • Substrate Preparation and Handling:

    • Biliverdin isomers are light-sensitive and prone to oxidation; store under nitrogen in amber vials at -80°C

    • Prepare fresh substrate solutions immediately before use

    • Consider solubility limitations; biliverdin may require DMSO or other co-solvents for dissolution

  • Reaction Buffer Optimization:

    • pH: Test range 7.0-8.5 to determine optimal pH for mouse BLVRB

    • Salt concentration: Optimize NaCl or KCl concentration (typically 50-150 mM)

    • Stabilizing agents: Consider including glycerol (5-10%) or BSA (0.1-1 mg/mL)

    • Reducing environment: Include DTT or β-mercaptoethanol to maintain reduced state

  • Cofactor Considerations:

    • NADPH vs. NADH: Test both cofactors, as BLVRB can utilize either with potential differences in efficiency

    • Cofactor concentration: Typically 100-500 μM, but optimize based on Km determination

    • Regenerating system: Consider including glucose-6-phosphate/glucose-6-phosphate dehydrogenase to regenerate NADPH during longer reactions

  • Enzyme Source and Preparation:

    • Recombinant protein: Use His-tagged recombinant mouse BLVRB protein with confirmed purity (>95%)

    • Tissue extracts: Prepare under conditions that preserve enzymatic activity (4°C, protease inhibitors)

    • Enzyme concentration: Determine linear range for activity measurements

  • Detection Methods:

    • Spectrophotometric: Monitor NADPH oxidation at 340 nm or bilirubin formation at 450 nm

    • HPLC: For detailed analysis of substrate consumption and product formation

    • Fluorometric: May offer increased sensitivity for low enzyme concentrations

  • Controls and Validation:

    • No-enzyme controls to account for non-enzymatic reactions

    • Known inhibitors as positive controls for inhibition studies

    • Heat-inactivated enzyme as negative control

    • Parallel assays with human BLVRB to assess species differences

  • Data Analysis:

    • Determine kinetic parameters (Km, Vmax) under optimized conditions

    • Assess substrate specificity across different biliverdin isomers

    • Evaluate effects of potential modulators or inhibitors

Optimization should be performed systematically, changing one parameter at a time while maintaining others constant to identify truly optimal conditions for mouse BLVRB activity measurements.

What considerations are important when working with recombinant BLVRB proteins in functional studies?

When working with recombinant BLVRB proteins in functional studies, researchers should consider several critical factors to ensure reliable and interpretable results:

  • Expression System Effects:

    • The E. coli expression system commonly used for mouse BLVRB production lacks post-translational modifications present in mammalian cells

    • Compare activity of E. coli-expressed protein with that from mammalian expression systems if post-translational modifications are suspected to affect function

    • Consider potential effects of bacterial endotoxin contamination on cellular assays

  • Protein Tag Influences:

    • His-tagged recombinant BLVRB (commonly available) may exhibit altered properties compared to the native protein

    • When possible, compare tagged and untagged versions of the protein

    • Consider tag removal using specific proteases if the tag interferes with activity or interactions

  • Storage and Stability:

    • Optimal storage conditions include 20mM Tris-HCl buffer (pH 8.0) containing 10% glycerol

    • Short-term storage at 2-8°C (up to 1 week) versus long-term storage at -20°C to -80°C

    • Avoid repeated freeze-thaw cycles that can compromise protein integrity

    • Assess activity retention over time under various storage conditions

  • Protein Quality Assessment:

    • Verify purity (>95%) by SDS-PAGE

    • Confirm molecular weight (approximately 24.6 kDa for mouse BLVRB) by MALDI-TOF

    • Assess oligomeric state by size exclusion chromatography or native PAGE

    • Verify proper folding through circular dichroism or fluorescence spectroscopy

  • Functional Validation:

    • Determine specific activity under standardized conditions before complex experiments

    • Compare activity with literature values or endogenous BLVRB from tissues

    • Establish dose-response relationships for cellular studies

    • Confirm substrate specificity matches that of the native enzyme

  • Experimental Controls:

    • Include enzymatically inactive mutants as negative controls

    • Use proteins from related family members to assess specificity

    • Consider species-matching (using mouse BLVRB for mouse cell studies) to avoid cross-species complications

  • Delivery Methods for Cellular Studies:

    • For cell uptake studies, consider protein transduction domains or nanoparticle encapsulation

    • Monitor cellular localization of delivered protein using fluorescent tags or immunofluorescence

    • Assess functional complementation in Blvrb-deficient cells

These considerations help ensure that observations made with recombinant BLVRB accurately reflect the physiological functions of the native protein and provide a solid foundation for mechanistic studies and therapeutic development.

Product Science Overview

Introduction

Biliverdin Reductase B (BLVRB) is an enzyme that plays a crucial role in the heme metabolism pathway. It is responsible for the reduction of biliverdin to bilirubin, a process that is essential for the clearance of heme in mammals . This enzyme is part of the biliverdin reductase family and is encoded by the BLVRB gene.

Classification

Biliverdin Reductase B belongs to the short-chain dehydrogenase/reductase (SDR) family. It is also known by other names such as flavin reductase (NADPH) and NAD(P)H dehydrogenase (quinone) 1 . The enzyme is highly conserved across different species, including humans, mice, and rats .

Biological Properties

BLVRB is a cytosolic enzyme that exhibits dual cofactor specificity, meaning it can utilize both NADH and NADPH as electron donors . This property is significant for its role in various cellular processes, including oxidative stress response and cellular redox homeostasis.

Functions

The primary function of BLVRB is to catalyze the conversion of biliverdin to bilirubin. This reaction is crucial for the detoxification of heme, which is released during the breakdown of hemoglobin in senescent or damaged red blood cells . Bilirubin, the end product of this reaction, is a potent antioxidant that protects cells from oxidative damage .

Modes of Action

BLVRB operates by binding to biliverdin and reducing it to bilirubin using NAD(P)H as a cofactor. The enzyme’s active site contains a conserved tyrosine residue that is essential for its catalytic activity . The reduction process involves the transfer of electrons from NAD(P)H to biliverdin, resulting in the formation of bilirubin .

Regulatory Mechanisms

The expression and activity of BLVRB are regulated at multiple levels, including transcriptional, post-transcriptional, and post-translational modifications. Various factors such as oxidative stress, hypoxia, and inflammatory signals can influence the expression of BLVRB . Additionally, the enzyme’s activity can be modulated by phosphorylation and other post-translational modifications .

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.