BD5 Human

Beta Defensin-5 Human Recombinant
Shipped with Ice Packs
In Stock

Description

BD5 Human Recombinant produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 51 amino acids and having a molecular mass of 5.8 kDa. The Beta Defensin-5 is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Defensins, encompassing both alpha and beta types, are positively charged peptides that exhibit a broad spectrum of antimicrobial activity, representing a crucial component of the innate immune system. Humans possess six beta-defensins: BD-1, BD-2, BD-3, BD-4, BD-5, and BD-6. These defensins are found on certain white blood cells and epithelial surfaces. In addition to their direct antimicrobial properties, beta-defensins can function as chemoattractants, guiding immature dendritic cells and memory T cells. Structurally, beta-defensins possess a distinct motif of six cysteine residues that form three intramolecular disulfide bonds.
Description
Recombinant human BD5, produced in E. coli, is a single, non-glycosylated polypeptide chain composed of 51 amino acids, resulting in a molecular weight of 5.8 kDa. The purification of Beta Defensin-5 is achieved through proprietary chromatographic methods.
Physical Appearance
Sterile, white powder obtained through lyophilization (freeze-drying).
Formulation
Lyophilized from a 0.2 µm filtered concentrated solution in phosphate-buffered saline (PBS) at a pH of 7.4.
Solubility
To reconstitute the lyophilized Beta Defensin-5, it is advised to dissolve it in sterile 18 MΩ·cm H₂O at a concentration of at least 100 µg/ml. Subsequently, it can be further diluted in other aqueous solutions.
Stability
Lyophilized Beta Defensin-5 demonstrates stability at room temperature for a duration of 3 weeks. However, for optimal storage, it is recommended to keep it desiccated at a temperature below -18°C. Following reconstitution, BD5 should be stored at 4°C for a period of 2 to 7 days. For extended storage, it is advisable to store it below -18°C. To ensure long-term stability during storage, the addition of a carrier protein (0.1% HSA or BSA) is recommended. It is crucial to avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 95.0% through the following methods: (a) Analysis by reverse-phase high-performance liquid chromatography (RP-HPLC). (b) Analysis by sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE).
Synonyms
Beta-defensin 105, eta-defensin 5, BD-5, DEFB-5, Defensin, beta 105, DEFB105A, BD5, DEFB105, DEFB5, DEFB105B.
Source
Escherichia Coli.
Amino Acid Sequence
GLDFSQPFPS GEFAVCESCK LGRGKCRKEC LENEKPDGNC RLNFLCCRQR I.

Q&A

What is Bromodomain Factor 5 (BDF5) and how does it function in Leishmania research?

BDF5 is an epigenetic reader protein in Leishmania that recognizes and binds to acetylated lysine residues in histone proteins. This protein plays a crucial role in the parasite's gene expression regulation, making it an attractive target for antileishmanial drug discovery. Leishmaniasis is a neglected tropical disease endemic in approximately 100 countries with an estimated prevalence of over four million cases in 2019 .

BDF5 contains two bromodomains, designated as BD5.1 and BD5.2, which function as readers of acetylated histones H2B and H4. Research has shown that BDF5 associates with extended transcriptional start regions in Leishmania and promotes gene expression . This mechanism suggests that acetylated H2B and H4 at transcriptional start regions can recruit BDF5, consistent with its known ability to associate with the extended transcriptional start regions of L. mexicana .

Researchers working with BDF5 typically use recombinant protein expression systems to produce the individual bromodomains for structural and functional studies. These recombinant domains can then be used in various biophysical assays to characterize their interactions with acetylated histone peptides and small molecule inhibitors .

How should researchers design experiments to study BD5 interactions with human bromodomain inhibitors?

When designing experiments to investigate BD5 interactions with human bromodomain inhibitors, researchers should follow a systematic approach that incorporates multiple orthogonal biophysical assays. Begin by establishing a clear research question, identifying your independent variable (e.g., inhibitor concentration or type) and your dependent variable (e.g., binding affinity or cellular response) .

A comprehensive experimental design for studying BD5-inhibitor interactions should include:

  • Thermal Shift Assays (TSA): This technique provides an initial screening method to identify potential inhibitors by measuring changes in protein thermal stability upon ligand binding .

  • Fluorescence Polarization: This approach allows for quantitative determination of binding affinities between BD5 domains and fluorescently labeled acetylated peptides or competitive inhibitors .

  • Nuclear Magnetic Resonance (NMR): NMR provides atomic-level details of the interactions and can confirm binding observed in other assays .

  • X-ray Crystallography: This method determines the three-dimensional structure of BD5-inhibitor complexes, revealing the precise binding mode and interactions .

  • Cellular Viability Assays: To assess the biological relevance of inhibitor binding, testing compounds against Leishmania promastigotes can establish their antiparasitic potential .

What methodological approaches are most effective for evaluating contradictions in BD5 inhibitor binding data?

When evaluating contradictions in BD5 inhibitor binding data, researchers should implement a structured approach that rigorously analyzes discrepancies across multiple experimental techniques. Contradiction detection in scientific data requires systematic evaluation of results obtained through different methodological approaches .

The DECODE (DialoguE COntradiction DEtection) approach, though developed for dialogue analysis, offers a valuable framework that can be adapted for analyzing scientific data contradictions . For BD5 inhibitor binding, this would involve:

  • Structured Data Comparison: Compare binding data across different assay platforms (TSA, NMR, crystallography) in a structured manner, evaluating each data point against corresponding results from other methods .

  • Identifying Systematic Biases: Determine whether contradictions arise from methodological differences or represent true biological phenomena. For example, some inhibitors may show different binding properties in solution-based assays versus crystallography .

  • Statistical Analysis of Outliers: Apply statistical methods to identify significant deviations that may represent true contradictions rather than experimental noise.

  • Validation Through Orthogonal Methods: When contradictions are identified, employ additional methodologies to resolve discrepancies. For instance, if TSA and NMR data conflict regarding an inhibitor's binding, isothermal titration calorimetry could provide a third perspective .

This methodological approach is more robust and transferable than unstructured contradiction analysis, particularly when evaluating out-of-distribution data or unexpected results .

How do structural differences between BD5.1 and BD5.2 affect their interactions with human bromodomain inhibitors?

The structural differences between BD5.1 and BD5.2 significantly influence their interactions with human bromodomain inhibitors, presenting important considerations for structure-based drug design. Crystal structure analysis of apo BD5.2 compared with the BD5.2-bromosporine complex reveals specific binding pocket characteristics that determine inhibitor selectivity and affinity .

BD5.1 and BD5.2 exhibit distinct binding preferences for human bromodomain inhibitors. While both domains can interact with compounds like bromosporine, SGC-CBP30, and I-BRD9, the binding affinities and thermodynamic profiles often differ substantially . These differences stem from variations in the composition of the acetyl-lysine binding pocket, including differences in hydrophobic residues and hydrogen bonding networks.

Researchers should approach BD5.1 and BD5.2 as distinct targets requiring separate optimization strategies. When designing inhibitor screening campaigns, both domains should be evaluated independently using orthogonal biophysical assays such as thermal shift, fluorescence polarization, and NMR spectroscopy .

To systematically address these structural differences in drug discovery efforts, researchers should:

  • Perform comprehensive structure-activity relationship (SAR) studies with chemical series showing activity against either domain

  • Utilize molecular dynamics simulations to understand domain-specific binding pocket flexibility

  • Consider dual-targeting compounds that can effectively inhibit both domains simultaneously

The differential binding of SGC-CBP30 to BD5.1, which correlates with antileishmanial activity in cell viability assays, demonstrates how these structural distinctions can be exploited for therapeutic development .

What experimental design considerations are critical when testing BD5 inhibitors against Leishmania in cellular models?

When testing BD5 inhibitors against Leishmania in cellular models, researchers must implement a rigorous experimental design that accounts for parasite biology, compound properties, and appropriate controls. The experimental approach should carefully define independent variables (inhibitor concentration, exposure time) and dependent variables (parasite viability, infectivity) .

Critical experimental design considerations include:

  • Cell Model Selection: Choose appropriate Leishmania life cycle stages (promastigotes vs. amastigotes) and strain variations. Amastigotes within macrophages represent the clinically relevant stage but are more challenging to work with than axenic promastigotes .

  • Control Implementation: Include positive controls (established antileishmanial drugs like amphotericin B), negative controls (vehicle only), and cytotoxicity controls (uninfected host cells) to distinguish antiparasitic effects from host cell toxicity .

  • Treatment Parameters Optimization: Determine optimal inhibitor exposure time and concentration ranges based on preliminary dose-finding experiments. Consider the compound's stability in culture conditions and potential metabolism by host cells .

  • Assay Selection for Outcome Measurement: Choose appropriate methods to quantify parasite burden, such as:

    • Luciferase-expressing parasites for bioluminescence readouts

    • Fluorescence microscopy with automated image analysis

    • qPCR-based parasite DNA quantification

  • Statistical Design: Implement randomized block designs or factorial designs to efficiently test multiple variables (inhibitor combinations, concentrations) while minimizing experimental bias .

The experimental design should also include measures to control extraneous variables that might confound results, such as passage number of parasites, host cell activation state, and environmental conditions .

How can researchers effectively distinguish between BD5-mediated effects and off-target activities of human bromodomain inhibitors in Leishmania?

Distinguishing between BD5-mediated effects and off-target activities of human bromodomain inhibitors in Leishmania requires a multi-faceted approach combining genetic, biochemical, and pharmacological methods. This methodological challenge is central to validating BD5 as a therapeutic target and developing selective inhibitors .

To effectively differentiate on-target from off-target effects, researchers should implement the following methodological strategies:

  • Genetic Validation:

    • Generate BD5 knockout or knockdown Leishmania strains through CRISPR-Cas9 or RNAi technologies

    • Create point mutations in the acetyl-lysine binding pocket of BD5.1 and BD5.2 that prevent inhibitor binding

    • Compare inhibitor efficacy between wild-type and genetically modified parasites; reduced efficacy in mutants suggests BD5-specific activity

  • Structure-Activity Relationship Analysis:

    • Synthesize close structural analogs with varying BD5 binding affinities but similar physicochemical properties

    • Correlate BD5 binding affinity (measured by biophysical assays) with antiparasitic activity

    • Develop negative control compounds that maintain similar physicochemical properties but cannot bind BD5

  • Cellular Target Engagement Assays:

    • Implement cellular thermal shift assays (CETSA) to confirm BD5 engagement in intact parasites

    • Use competitive pulldown assays with biotinylated inhibitors to identify all proteins bound by the inhibitor

  • Transcriptomic Profiling:

    • Compare gene expression changes induced by different inhibitors

    • Identify signature patterns consistent with BD5 inhibition versus other mechanisms

    • Cross-reference with expression changes in BD5 knockdown parasites

By systematically applying these methods, researchers can build a comprehensive evidence base to distinguish on-target BD5 inhibition from off-target effects. This approach is particularly important when repurposing human bromodomain inhibitors for antiparasitic applications, where selectivity profiles may differ substantially from human targets .

What methodologies should be employed to analyze contradictions in BD5 inhibitor efficacy between in vitro binding and cellular activity?

Analyzing contradictions between in vitro BD5 inhibitor binding and observed cellular activity requires sophisticated methodological approaches that bridge biochemical and biological systems. These contradictions can provide valuable insights into inhibitor mechanism of action, but demand systematic analysis .

The following methodological framework should be employed:

  • Quantitative Structure-Activity Relationship (QSAR) Analysis:

    • Plot biochemical binding data (Kd, IC50) against cellular efficacy metrics

    • Identify outlier compounds that exhibit strong binding but weak cellular activity (or vice versa)

    • Cluster compounds based on their deviation from expected correlation patterns

  • Compound Property Assessment:

    • Evaluate physicochemical properties that might explain discrepancies:

      • Cell permeability measurements using artificial membrane systems

      • Protein binding assays to assess compound availability

      • Metabolic stability studies to identify rapid degradation

  • Target Engagement Confirmation:

    • Implement thermal shift assays in cell lysates to verify inhibitor-BD5 binding occurs in cellular environment

    • Use photoaffinity labeling to identify all proteins interacting with the inhibitor

    • Develop proximal ligation assays to visualize inhibitor-target interaction locations

  • Structured Contradiction Resolution:

    • Apply the structured utterance-based approach from contradiction detection frameworks

    • Systematically document and categorize contradictions between assays

    • Develop decision trees for resolving conflicting data based on assay reliability hierarchies

  • Pharmacokinetic/Pharmacodynamic (PK/PD) Modeling:

    • Develop mathematical models relating inhibitor concentration to BD5 occupancy and parasite inhibition

    • Identify threshold effects or non-linear relationships that might explain discrepancies

This methodological approach recognizes that contradictions between biochemical and cellular data often provide the most valuable insights into biology and drug action. When properly analyzed, these discrepancies can reveal new aspects of BD5 biology, inhibitor mechanisms, or Leishmania physiology that would otherwise remain hidden .

How can researchers design comprehensive experiments to evaluate the role of BD5 in histone acetylation recognition?

Designing comprehensive experiments to evaluate BD5's role in histone acetylation recognition requires an integrated approach that combines biochemical, structural, and cellular methods. Researchers should structure their experimental design around specific, testable hypotheses that address different aspects of BD5-histone interactions .

A thorough experimental design should include:

  • Acetylation Site Profiling:

    • Perform systematic peptide arrays containing all possible acetylation sites on histones H2B and H4

    • Quantify binding affinity of BD5.1 and BD5.2 for each acetylated peptide using fluorescence polarization

    • Create a comprehensive binding profile that ranks acetylation sites by affinity

  • Structural Characterization:

    • Determine crystal structures of BD5.1 and BD5.2 in complex with high-affinity acetylated peptides

    • Identify key residues in the binding pocket that confer specificity

    • Use mutagenesis to validate the importance of these residues

  • Chromatin Association Studies:

    • Implement chromatin immunoprecipitation sequencing (ChIP-seq) to map BD5 genome-wide localization

    • Correlate BD5 binding with histone acetylation patterns using sequential ChIP

    • Compare wild-type BD5 binding with binding-deficient mutants

  • Functional Validation:

    • Create BD5 variants with mutations in key acetyl-lysine binding residues

    • Express these variants in BD5-depleted parasites

    • Assess the impact on gene expression, parasite viability, and life cycle progression

The experimental design should incorporate appropriate controls, randomization, and blinding where possible to minimize experimental bias . This comprehensive approach allows researchers to connect biochemical mechanisms to biological functions, providing a complete picture of BD5's role in acetylation recognition.

Research has demonstrated that BD5 bromodomains interact with acetylated sequences in Leishmania histones H2B and H4, suggesting that acetylated H2B and H4 at transcriptional start regions can recruit BD5 . Further adaptation of proximity biotinylation workflows could be used to identify BD5-proximal acetylation sites in vivo .

What methodological approaches should be used to develop selective BD5 inhibitors based on human bromodomain inhibitor scaffolds?

Developing selective BD5 inhibitors based on human bromodomain inhibitor scaffolds requires a methodical, structure-guided approach that maximizes selectivity while maintaining potency. Given that human bromodomain inhibitors like SGC-CBP30, bromosporine, and I-BRD9 have demonstrated binding to Leishmania BD5 domains and antiparasitic activity, they provide valuable starting points for selective inhibitor development .

The following methodological approach is recommended:

  • Comparative Structural Analysis:

    • Analyze crystal structures of human bromodomain-inhibitor complexes alongside BD5 structures

    • Identify regions of similarity and difference in binding pockets

    • Create structural alignments to guide rational design of selective inhibitors

  • Fragment-Based Optimization:

    • Screen fragment libraries against BD5 domains using thermal shift assays

    • Identify fragments with binding preference for BD5 over human bromodomains

    • Develop hybrid molecules incorporating selective fragments into existing scaffolds

  • Structure-Activity Relationship (SAR) Development:

    • Systematically modify existing inhibitors like SGC-CBP30 through:

      • Addition of parasite-specific binding elements

      • Removal of human bromodomain-specific interactions

      • Optimization of physicochemical properties for parasite penetration

    • Test each analog in parallel against BD5 and human bromodomains

    • Track selectivity indices throughout optimization

  • Computational Design and Screening:

    • Generate homology models for BD5 domains

    • Perform virtual screening against these models

    • Use molecular dynamics simulations to account for protein flexibility

    • Implement docking studies to predict binding modes of modified inhibitors

Researchers should consider implementing this iterative process in the context of the observed binding of human bromodomain inhibitors to BD5.1 and BD5.2 . For example, SGC-CBP30 has demonstrated both binding to BD5.1 and inhibitory effects against Leishmania promastigotes in cell viability assays, making it a promising starting point for selective inhibitor development .

Human Bromodomain InhibitorBD5.1 BindingBD5.2 BindingAntiparasitic Activity
SGC-CBP30StrongModerateDemonstrated
BromosporineModerateStrongNot reported
I-BRD9ModerateModerateNot reported

This table summarizes the binding characteristics of human bromodomain inhibitors to BD5 domains based on data from orthogonal biophysical assays .

Product Science Overview

Introduction

Beta Defensin-5 (hBD-5) is a member of the beta-defensin family, which are small cationic peptides known for their broad-spectrum antimicrobial properties. These peptides play a crucial role in the innate immune system by providing the first line of defense against pathogens. The recombinant form of Beta Defensin-5 is produced using advanced biotechnological methods, making it a valuable tool for research and potential therapeutic applications.

Characteristics

Beta Defensin-5 is a peptide consisting of 51 amino acids with a molecular mass of approximately 5.8 kDa . It is characterized by a six-cysteine motif that forms three intramolecular disulfide bonds, which are essential for its stability and function . The peptide is typically produced in Escherichia coli (E. coli) and purified using proprietary chromatographic techniques to achieve a purity greater than 95% .

Mode of Action

Beta Defensin-5 exhibits antimicrobial activity by disrupting the membrane integrity of bacteria, fungi, and viruses. It achieves this through its cationic nature, which allows it to interact with the negatively charged microbial membranes, leading to membrane permeabilization and cell death . Additionally, Beta Defensin-5 can act as a chemoattractant for immature dendritic cells and memory T cells, thereby enhancing the immune response .

Production and Stability

The recombinant form of Beta Defensin-5 is produced in E. coli and is available as a lyophilized (freeze-dried) powder . It is recommended to reconstitute the lyophilized peptide in sterile water to a concentration of at least 100 µg/ml, which can then be further diluted as needed . The lyophilized form is stable at room temperature for up to three weeks, but for long-term storage, it should be kept desiccated below -18°C . Once reconstituted, the peptide should be stored at 4°C for short-term use and below -18°C for long-term use, with the addition of a carrier protein to prevent freeze-thaw cycles .

Applications

Beta Defensin-5 has promising therapeutic applications due to its antimicrobial properties. It is being studied for its potential use in treating drug-resistant infections, enhancing wound healing, and as an adjuvant in vaccines . Its ability to modulate the immune response also makes it a candidate for research in autoimmune diseases and cancer .

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.