HGD Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Introduction

The "HGD Antibody, Biotin conjugated" is a specialized immunochemical reagent designed for detecting the homogentisate 1,2-dioxygenase (HGD) enzyme in research and diagnostic applications. This antibody is chemically linked to biotin, enabling high-sensitivity detection through streptavidin-biotin interaction systems . HGD plays a critical role in tyrosine metabolism, and its dysfunction is linked to alkaptonuria, a rare genetic disorder .

Applications in Research

This biotinylated antibody is validated for multiple techniques, including:

ApplicationDetailsSource
Western Blot (WB)Detects HGD in HepG2 cells, HeLa cells, and liver tissues (human, mouse, rat) .
Immunohistochemistry (IHC)Used in paraffin-embedded human liver and prostatic cancer tissues (antigen retrieval: citrate buffer pH 6.0) .
Flow CytometryIntracellular staining of HEPG2 cells with 90% specificity under optimized conditions .
ELISACompatible with biotin-streptavidin detection systems .

Conjugation Methodology

The biotin conjugation process involves:

  1. Chemical Crosslinking: NHS-PEG4-biotin is used to label the antibody, ensuring minimal interference with antigen-binding sites .

  2. Purification: Post-conjugation, the product is purified via size-exclusion chromatography (e.g., PD-10 columns) .

  3. Quality Control: Binding capability is validated using competition assays with dually labeled IgG references .

Key Research Findings

  • Cancer Research: HGD expression is detectable in liver and prostatic cancer tissues, suggesting a role in tumor metabolism .

  • Enzyme Function: HGD’s catalytic activity in homogentisate degradation is critical for studying metabolic disorders like alkaptonuria .

  • Conjugation Efficiency: Site-specific biotinylation methods (e.g., affinity-based techniques) improve assay sensitivity compared to random conjugation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
Synonyms
Homogentisate 1,2-dioxygenase (EC 1.13.11.5) (Homogentisate oxygenase) (Homogentisic acid oxidase) (Homogentisicase), HGD, HGO
Target Names
HGD
Uniprot No.

Target Background

Gene References Into Functions
  1. Twelve novel HGD gene variants have been identified in 99 alkaptonuria patients affecting the bones. PMID: 25804398
  2. A novel alkaptonuria mutation, c.87 + 1G > A, exhibits a significant founder effect and high prevalence within a nomadic Indian population. PMID: 24575791
  3. A study described two novel HGD mutations in a Chinese alkaptonuria family, the splicing mutation of IVS7 1G>C, a donor splice site of exon 7, and a missense mutation of F329C in exon 12. PMID: 23353776
  4. This report details mutations of the HGD gene in Jordanian alkaptonuria patients. PMID: 21437689
  5. The observed increase of HGD expression in Alkaptonuria cells is likely due to a compensatory mechanism to overcome the nearly absent catalytic activity of the deficient enzyme. PMID: 22105303
  6. An update on the molecular genetics of Alkaptonuria (AKU) is provided. PMID: 21720873
  7. Familial ochronotic arthropathy is attributed to a gene mutation of the homogentisic acid 1,2-dioxygenase (HGD) gene, traced back three hundred years in a Hungarian family. PMID: 20462779
  8. A comprehensive mutation analysis of 93 patients enrolled in this study, along with an extensive update of all previously published HGD mutations associated with alkaptonuria, is reported. PMID: 19862842
  9. A Turkish mutation shares an homogentisate 1,2-dioxygenase haplotype with the mutation found in Finland, Slovakia, and India, suggesting that R58fs is an ancient alkaptonuria mutation that likely originated in central Asia. PMID: 12872836
  10. Four distinct mutations of the HGD gene were identified in alkaptonuria and ochronotic arthropathy diagnoses. PMID: 16085442
  11. A single nucleotide deletion located in exon 3 resulted in a frameshift mutation in the HGD gene within a family with alkaptonuria. PMID: 18945288

Show More

Hide All

Database Links

HGNC: 4892

OMIM: 203500

KEGG: hsa:3081

STRING: 9606.ENSP00000283871

UniGene: Hs.368254

Involvement In Disease
Alkaptonuria (AKU)
Protein Families
Homogentisate dioxygenase family
Tissue Specificity
Highest expression in the prostate, small intestine, colon, kidney and liver.

Q&A

What is HGD Antibody and why is it used with biotin conjugation?

HGD Antibody recognizes Homogentisate-1,2-Dioxygenase, an enzyme involved in tyrosine catabolism pathway. It is commonly used in laboratory settings for protein detection and localization studies. Biotin conjugation provides significant advantages by enhancing signal strength through the strong affinity between biotin and streptavidin/avidin systems . This conjugation enables sensitive detection in multiple applications including Western blotting, immunohistochemistry, and immunocytochemistry . The biotin-streptavidin interaction is among the strongest non-covalent biological interactions known, providing stable complexes for detection systems.

What are the primary applications for biotin-conjugated HGD antibodies?

Biotin-conjugated HGD antibodies are utilized across multiple research applications:

ApplicationRecommended ConcentrationDilution RangePurpose
Western Blotting0.2-2 μg/mL1:250-2500Protein detection in cellular lysates
Immunohistochemistry5-20 μg/mL1:25-100Tissue localization studies
Immunocytochemistry5-20 μg/mL1:25-100Cellular localization studies

These applications are versatile for studying HGD expression patterns in cells and tissues, with optimal working dilutions determined by end-user requirements . The biotin conjugation significantly enhances detection sensitivity compared to unconjugated antibodies, particularly in complex tissue samples where endogenous signal might be low.

How should biotin-conjugated HGD antibodies be stored to maintain optimal activity?

For optimal preservation of biotin-conjugated antibody activity, storage conditions are critical. Store at 4°C for frequent use (short-term storage) . For long-term preservation, store at -20°C in a manual defrost freezer, where stability can be maintained for up to two years without detectable loss of activity . The antibodies are typically supplied in PBS buffer (pH 7.4) containing 0.02% sodium azide and 50% glycerol for stability . It is imperative to avoid repeated freeze-thaw cycles as these can lead to protein denaturation and subsequent loss of antibody activity. Aliquoting before freezing is recommended for antibodies that will be used periodically over extended periods.

What conjugation methods are most effective for creating biotin-linked HGD antibodies?

The most common approach for biotin conjugation to antibodies involves derivatization of amino groups on the antibody with biotin derivatives . Several methodologies exist:

  • Amine-reactive NHS-biotin conjugation: This method targets primary amines (lysine residues) on the antibody. While simple to perform, it creates randomly biotinylated antibodies that may have variable binding efficiency .

  • Site-specific conjugation methods: These more advanced techniques include:

    • Fc-specific conjugation using Protein A/G domains

    • Introduction of unique conjugation sites through genetic engineering

    • Enzymatic approaches for controlled biotinylation

Site-specific conjugation maintains antibody orientation and binding capacity, offering approximately twice the sensitivity in assays compared to randomly biotinylated antibodies . The selection of conjugation method significantly impacts experimental outcomes, with site-specific approaches generally preferred for quantitative research applications.

How can researchers validate the efficiency of biotin conjugation to HGD antibodies?

Validation of biotin conjugation efficiency is crucial for experimental reproducibility. Multiple approaches can be employed:

  • Spectrophotometric analysis: Measuring the biotin:protein ratio using absorbance at 280 nm (protein) and by using HABA (4'-hydroxyazobenzene-2-carboxylic acid) assay for biotin quantification.

  • Functional validation: Comparing the activity of conjugated versus unconjugated antibody in standard applications (Western blot, ELISA).

  • Mass spectrometry: For precise determination of conjugation sites and ratio.

  • SDS-PAGE mobility shift: Biotin conjugation increases molecular weight, resulting in slightly reduced mobility in gel electrophoresis.

A successful conjugation typically results in a biotin:antibody molar ratio of 3-8 biotin molecules per antibody molecule, though optimal ratios depend on specific applications . Over-conjugation can impair antigen binding, while under-conjugation may result in insufficient signal.

What troubleshooting approaches are recommended when biotin-conjugated HGD antibodies show low sensitivity?

When encountering low sensitivity with biotin-conjugated HGD antibodies, systematic troubleshooting is essential:

  • Antibody concentration optimization: Titrate antibody concentration using recommended ranges:

    • For Western blotting: 0.2-2 μg/mL (1:250-2500 dilution)

    • For IHC/ICC: 5-20 μg/mL (1:25-100 dilution)

  • Detection system evaluation: Compare different avidin/streptavidin conjugates (HRP, AP, fluorophores) to identify optimal signal-to-noise ratios.

  • Blocking optimization: Test different blocking reagents to reduce non-specific binding; BSA and casein are commonly effective.

  • Conjugation quality assessment: If possible, evaluate the degree of biotinylation—excessive conjugation may sterically hinder antigen recognition.

  • Sample preparation refinement: Ensure proper antigen exposure through optimized fixation, permeabilization, and epitope retrieval methods.

Creating a systematic optimization matrix across these variables can efficiently identify the source of sensitivity issues. Documentation of all optimization steps is critical for experimental reproducibility.

How do site-specific biotin conjugation methods compare to traditional random conjugation for HGD antibody performance?

Site-specific conjugation offers significant advantages over random conjugation approaches:

ParameterRandom NHS-Biotin ConjugationSite-Specific Conjugation
Binding EfficiencyVariable; potentially compromisedPreserved; consistent
Signal-to-Noise RatioLowerApproximately 2x higher
Batch-to-Batch ReproducibilityVariableHigh consistency
Requirement for MaterialStandard protocolsMay require genetic engineering
Orientation ControlNonePrecise control of biotin positioning

Research demonstrates that site-specifically conjugated antibodies provide approximately twice the sensitivity in assays such as sandwich ELISA compared to randomly biotinylated counterparts . This improvement stems from maintaining proper antibody orientation with exposed antigen-binding sites. For quantitative research applications requiring maximum sensitivity and reproducibility, site-specific approaches represent the superior methodology despite increased technical complexity.

What are the implications of different biotin linker types for HGD antibody performance in various applications?

The biotin linker structure significantly impacts antibody performance:

  • Short linkers (direct conjugation): Provide minimal spatial separation between biotin and antibody, which may reduce streptavidin binding efficiency due to steric hindrance.

  • Medium-length spacers (e.g., LC-biotin): Incorporate a 6-atom spacer that improves streptavidin accessibility while maintaining close proximity to the antibody.

  • Long spacers (e.g., PEG-based linkers): Offer maximal spatial separation, reducing steric hindrance and improving solution properties.

Studies demonstrate that longer linkers generally improve detection sensitivity in solution-based assays, while shorter linkers may be preferable for certain solid-phase applications . The chemical nature of the linker (hydrophobic vs. hydrophilic) also impacts antibody solubility and non-specific binding characteristics. Selection should be based on application requirements, with PEG-based linkers showing advantage in reducing non-specific interactions in complex biological samples.

How does the biotin-streptavidin detection system compare with direct enzyme conjugation for HGD antibodies in terms of sensitivity and specificity?

The biotin-streptavidin system offers distinct advantages and limitations compared to direct enzyme conjugation:

ParameterBiotin-Streptavidin SystemDirect Enzyme Conjugation
Signal AmplificationHigh (multiple binding sites)Limited (1:1 ratio)
SensitivitySuperior (3-5x higher)Lower
Background SignalCan be higher due to endogenous biotinTypically lower
FlexibilityCompatible with multiple detection systemsLimited to specific enzyme
StabilityExcellent long-term stabilityVariable enzyme stability
Protocol ComplexityMulti-step processSimpler, fewer steps

What recent advances in biotin conjugation technology are applicable to HGD antibody research?

Several cutting-edge approaches have recently enhanced biotin conjugation technology:

  • Click chemistry conjugation: Utilizes highly selective azide-alkyne cycloaddition reactions for site-specific biotin attachment, offering superior control over traditional methods .

  • Enzymatic biotinylation: Employs biotin ligase (BirA) for site-specific attachment at engineered recognition sequences, providing homogeneous conjugation products.

  • THIOMAB technology: Introduces unique cysteine residues for site-specific conjugation, allowing precise control of biotin positioning without affecting antibody structure .

  • Sortase-mediated conjugation: Utilizes bacterial transpeptidases for site-specific attachment of biotin to antibody C-terminus.

These advanced methodologies enable the creation of homogeneously biotinylated antibodies with preserved antigen-binding capacity . The THIOMAB approach, in particular, has demonstrated conjugation efficiencies exceeding 92% with conversions over 98%, making it increasingly amenable to scaled production for research applications . Implementation of these techniques requires specialized expertise but delivers superior conjugates with enhanced performance characteristics.

What are the critical differences between polyclonal and monoclonal biotin-conjugated HGD antibodies for research applications?

Polyclonal and monoclonal biotin-conjugated HGD antibodies present distinct profiles for research applications:

CharacteristicPolyclonal HGD AntibodiesMonoclonal HGD Antibodies
Epitope RecognitionMultiple epitopesSingle epitope
Signal StrengthTypically strongerMay require amplification
Batch-to-Batch VariationHigherMinimal
SpecificityModerate; potential cross-reactivityHigh; epitope-specific
CostGenerally lowerHigher
Application VersatilityBroad applicationsMay have application limitations
Production ScalabilityLimited by host animalHighly scalable

Polyclonal antibodies, like the available biotin-linked HGD antibody, recognize multiple epitopes on the target protein . This enhances signal strength but may increase background. Conversely, monoclonal antibodies provide consistent recognition of a single epitope with higher specificity. For applications like Western blotting where signal strength is paramount, polyclonal antibodies often excel. For highly specific detection or quantitative analyses, monoclonal antibodies typically offer superior performance despite potentially lower signal intensity.

How should researchers optimize protocols when transitioning from unconjugated to biotin-conjugated HGD antibodies?

When transitioning from unconjugated to biotin-conjugated HGD antibodies, several protocol adjustments are necessary:

  • Concentration adjustments: Biotin-conjugated antibodies generally require different working concentrations:

    • For Western blotting: Start with 0.2-2 μg/mL (1:250-2500 dilution)

    • For IHC/ICC: Begin with 5-20 μg/mL (1:25-100 dilution)

  • Incubation modifications:

    • Primary antibody incubation: May require shorter incubation (1-2 hours at room temperature or overnight at 4°C)

    • Secondary detection: Replace standard secondary antibody with streptavidin conjugate

  • Blocking considerations:

    • Include avidin/biotin blocking steps in tissues with high endogenous biotin

    • Consider specialized blocking reagents to reduce background

  • Detection system optimization:

    • Test different streptavidin conjugates (HRP, AP, fluorophores)

    • Evaluate signal amplification systems if needed

Systematically testing these variables will establish optimal conditions for biotin-conjugated antibodies. Begin with manufacturer recommendations and adjust as needed through controlled comparisons. Document all optimization steps for future protocol standardization.

What detection strategies offer the highest sensitivity when working with biotin-conjugated HGD antibodies in low-abundance target scenarios?

For detecting low-abundance targets with biotin-conjugated HGD antibodies, several advanced detection strategies can significantly enhance sensitivity:

  • Tyramide Signal Amplification (TSA): Combines biotin-streptavidin with catalyzed reporter deposition, offering 10-100x signal enhancement over conventional methods.

  • Poly-HRP streptavidin systems: Utilize streptavidin conjugated to multiple HRP molecules, increasing enzymatic signal output.

  • Rolling Circle Amplification (RCA): Couples biotinylated antibody detection with DNA circle amplification for exponential signal enhancement.

  • Quantum dot streptavidin conjugates: Provide brighter, more photostable fluorescence compared to conventional fluorophores.

  • Proximity Ligation Assay (PLA): Combines antibody recognition with DNA amplification for single-molecule sensitivity.

For low-abundance targets, TSA and poly-HRP systems consistently deliver substantial sensitivity improvements, often enabling detection of proteins below the threshold of conventional methods . These advanced techniques require additional optimization steps but can reveal protein expression patterns undetectable with standard approaches. Quantitative comparisons have demonstrated up to 100-fold enhancement in detection limits using TSA compared to conventional biotin-streptavidin systems.

What experimental controls are essential when using biotin-conjugated HGD antibodies for quantitative analysis?

Rigorous experimental controls are crucial for quantitative applications with biotin-conjugated HGD antibodies:

  • Primary antibody controls:

    • Isotype control: Biotin-conjugated immunoglobulin of the same isotype (IgG) and host species (rabbit)

    • Concentration-matched unconjugated HGD antibody (comparative control)

    • Antibody pre-absorption with recombinant HGD antigen (specificity control)

  • Detection system controls:

    • Streptavidin-only control (no primary antibody)

    • Endogenous biotin blocking validation

    • Known positive and negative control samples

  • Quantification controls:

    • Standard curve using recombinant HGD protein

    • Internal reference protein for normalization

    • Technical and biological replicates

  • System validation:

    • Alternative detection method confirmation (e.g., mass spectrometry)

    • siRNA knockdown or CRISPR knockout validation

These controls collectively ensure signal specificity, minimize background interference, and validate quantitative measurements. For publication-quality research, documentation of all validation steps is essential to demonstrate antibody specificity and assay reliability.

How can researchers assess potential background issues from endogenous biotin when using biotin-conjugated HGD antibodies?

Endogenous biotin presents a significant challenge when using biotin-conjugated antibodies, particularly in biotin-rich tissues like liver, kidney, and brain. Several approaches help assess and mitigate this interference:

  • Tissue-specific evaluation: Perform streptavidin-only controls on target tissues to quantify endogenous biotin levels.

  • Blocking strategies:

    • Pre-block tissues with unconjugated avidin/streptavidin

    • Use commercial avidin/biotin blocking kits

    • Validate blocking efficiency with streptavidin-only controls

  • Alternative fixation assessment: Compare different fixation methods as some preservatives can reduce accessible endogenous biotin.

  • Comparative analysis: Test serial tissue sections with:

    • Biotin-conjugated HGD antibody

    • Directly labeled (non-biotin) HGD antibody

    • No primary antibody (detection reagent only)

  • Signal quantification: Calculate signal-to-background ratios under different blocking conditions to determine optimal protocol.

For tissues with consistently high endogenous biotin, consider alternative detection strategies such as directly labeled primary antibodies or non-biotin amplification systems. The comparative approach provides clear evidence of signal specificity versus biotin-related background.

What analytical approaches best quantify the degree of biotinylation of HGD antibodies and its impact on functional performance?

Accurate quantification of biotinylation degree is essential for consistent antibody performance. Several analytical methods provide complementary information:

  • HABA Assay (4'-hydroxyazobenzene-2-carboxylic acid):

    • Spectrophotometric measurement of biotin displacement from HABA-avidin complex

    • Provides molar ratio of biotin:antibody

    • Optimal range: 3-8 biotin molecules per antibody

  • Mass Spectrometry:

    • MALDI-TOF or ESI-MS for precise molecular weight determination

    • Peptide mapping for identification of specific biotinylation sites

    • Differentiates population heterogeneity in conjugation

  • Functional Correlation Analysis:

    • Systematic testing of conjugates with varying biotinylation degrees

    • Generation of correlation curves between biotinylation ratio and:

      • Signal intensity

      • Background levels

      • Antigen binding affinity (SPR or BLI measurement)

A combined analytical approach provides comprehensive characterization. Research indicates that over-biotinylation (>8 biotin molecules per antibody) can significantly impair antigen recognition, particularly for antibodies with epitopes containing lysine residues . Conversely, under-biotinylation (<3 per antibody) may provide insufficient signal amplification. Documentation of biotinylation degree is critical for experimental reproducibility and accurate interpretation of results.

How might emerging protein engineering approaches enhance biotin-conjugated HGD antibody performance?

Emerging protein engineering approaches offer significant potential for enhancing biotin-conjugated HGD antibody performance:

  • Site-directed mutagenesis: Introduction of unique conjugation sites (cysteine or non-canonical amino acids) at optimal positions away from antigen-binding regions, allowing controlled biotinylation without compromising function .

  • Domain fusion strategies: Engineering biotin acceptor peptide (BAP) sequences directly into antibody structure for enzymatic biotinylation via BirA ligase, ensuring homogeneous conjugation products.

  • Antibody fragment engineering: Development of Fab, scFv, or nanobody formats with optimized biotin conjugation sites, potentially improving tissue penetration and signal-to-background ratios.

  • Computational design approaches: In silico analysis to identify optimal conjugation sites based on antibody structure, maximizing solvent accessibility while minimizing immunogenicity and impact on binding .

Studies utilizing THIOMAB technology have achieved conjugation products with >92% efficiency and excellent homogeneity . These engineered antibodies exhibit unique pharmacokinetic profiles with minimal changes to critical binding properties, suggesting significant advantages for quantitative research applications. As these technologies mature, researchers can anticipate antibody conjugates with unprecedented consistency and performance.

What alternative affinity-based detection systems might complement or replace biotin-streptavidin for HGD antibody applications?

Several innovative affinity-based detection systems show promise as complements or alternatives to biotin-streptavidin:

  • Click chemistry pairs: Strain-promoted azide-alkyne cycloaddition (SPAAC) and tetrazine-trans-cyclooctene reactions provide bioorthogonal coupling with rapid kinetics and high specificity .

  • Peptide tag systems:

    • SNAP/CLIP-tag technology

    • HaloTag ligand pairs

    • Sortase A recognition sequences
      These systems enable covalent labeling with diverse reporter molecules.

  • DNA-based amplification strategies:

    • Proximity ligation assay (PLA)

    • Hybridization chain reaction (HCR)

    • DNA-PAINT super-resolution approaches

  • Nanobody-based detection: Direct labeling of small (15 kDa) camelid-derived antibody fragments for improved tissue penetration and reduced background.

These alternatives offer distinct advantages in specific applications. Click chemistry approaches provide coupling efficiency approaching 100% with minimal non-specific interactions . DNA-based systems enable amplification factors exceeding 1000-fold with single-molecule sensitivity. For multicolor applications, orthogonal peptide tag systems permit simultaneous visualization of multiple targets without cross-reactivity. Selection of optimal system depends on specific research requirements including sensitivity needs, multiplexing requirements, and sample characteristics.

How might artificial intelligence and machine learning approaches optimize biotin-conjugated antibody experimental design and analysis?

Artificial intelligence and machine learning approaches are transforming biotin-conjugated antibody research across multiple dimensions:

  • Conjugation optimization:

    • Predictive modeling of optimal conjugation sites based on antibody structure

    • Identification of ideal biotin:antibody ratios for specific applications

    • Simulation of linker dynamics and accessibility

  • Image analysis enhancement:

    • Automated quantification of signal intensity and distribution

    • Background subtraction algorithms for complex tissues

    • Deep learning for pattern recognition in subcellular localization

  • Experimental design optimization:

    • Bayesian optimization approaches for protocol parameters

    • Design of experiments (DOE) for efficient multivariable optimization

    • Transfer learning from similar antibody classes to predict performance

  • Quality control automation:

    • Automated detection of batch-to-batch variations

    • Prediction of stability and shelf-life

    • Identification of potential cross-reactivity based on epitope analysis

These computational approaches significantly reduce experimental iterations required for optimization. Machine learning analysis of antibody structure can predict optimal conjugation positions with >80% accuracy, substantially improving conjugate performance without extensive experimental screening . For image analysis, convolutional neural networks demonstrate >95% concordance with expert manual scoring while reducing analysis time by orders of magnitude. As these technologies mature, researchers can expect increasingly automated optimization workflows that deliver superior conjugates with minimal empirical testing.

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.