FHIT Antibody Pair

Shipped with Ice Packs
In Stock

Description

Key Applications

This pair is validated for:

  • Cytometric Bead Array (CBA): Detects FHIT in a linear range of 0.391–100 ng/mL using recombinant FHIT standards (e.g., Ag14471) .

  • Sandwich ELISA: Optimized for high-throughput screening with minimal cross-reactivity.

  • Protein-Protein Interaction Studies: Facilitates analysis of FHIT’s role in DNA repair and apoptosis pathways .

Table 1: Cytometric Bead Array Validation Data

ParameterValue
Detection Range0.391–100 ng/mL
Capture Antibody3B10B2 (60833-4-PBS)
Detection Antibody1F7G3 (60833-3-PBS)
Standard Curve FitR² > 0.99 (typical)
Sensitivity (LLoQ)0.391 ng/mL

Data derived from Proteintech’s CBA validation shows robust linearity and reproducibility .

Research Significance

FHIT downregulation is implicated in early carcinogenesis, particularly in pancreatic ductal adenocarcinoma (PDAC) and intraductal papillary mucinous neoplasia (IPMN) . Studies using FHIT antibody pairs have demonstrated:

  • Progressive loss of FHIT expression correlating with dysplasia severity .

  • Utility in identifying precancerous lesions via immunohistochemistry and CBA .

Comparison with Other Antibody Pairs

While FHIT pairs are optimized for specificity, general antibody pairing strategies (e.g., protein A bead-based screening) highlight the importance of epitope non-overlap and affinity matching . For example, Proteintech’s FHIT pair avoids cross-reactivity with homologs like diadenosine tetraphosphatase .

Limitations and Considerations

  • Requires optimization for non-standard platforms (e.g., lateral flow assays).

  • Nuclear/cytoplasmic staining patterns in IHC necessitate dual validation .

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary based on the purchase method or location. For specific delivery times, please consult your local distributors.
Notes
We recommend using the capture antibody at a concentration of 0.5 µg/mL and the detection antibody at a concentration of 0.5 µg/mL. It is essential to determine optimal dilutions experimentally based on your specific research needs.
Synonyms
AP3A hydrolase,AP3Aase,Diadenosine 5\',5\'\'\'-P1,P3-triphosphate hydrolase,Dinucleosidetriphosphatase,Fragile histidine triad protein
Target Names
FHIT

Q&A

What is FHIT and why are matched antibody pairs important for its detection?

FHIT (fragile histidine triad gene) is a candidate tumor suppressor gene located at chromosomal fragile site 3p14.2, which encompasses the hereditary renal cancer translocation breakpoint and cancer cell homozygous deletions . The protein plays a significant role in regulating β-catenin-mediated gene transcription.

Matched antibody pairs are critical for FHIT detection because they enable sandwich-based assays where one antibody captures the target protein while the other detects it. The FHIT monoclonal matched antibody pairs typically consist of:

ComponentClone IDIsotypeApplication
Capture antibody3C10G2IgG2aImmobilization on solid phase
Detection antibody1F7G3IgG1Signal generation after binding

These pairs are specifically validated to work together without cross-interference, enabling highly specific detection of FHIT protein in complex biological samples .

What are the primary applications for FHIT antibody pairs in research?

FHIT antibody pairs are predominantly used in the following research applications:

  • Cytometric bead arrays - For quantitative assessment of FHIT in solution-phase samples

  • ELISA (Enzyme-Linked Immunosorbent Assay) - For detection and quantification in serum or cell lysates

  • Functional studies - To investigate FHIT's role in tumor suppression pathways

  • Biomarker research - Evaluating FHIT expression in cancer progression studies

The validated detection range for FHIT using these antibody pairs typically spans 0.391-100 ng/mL in cytometric bead array applications , making them suitable for detecting physiologically relevant FHIT concentrations in research samples.

How should researchers design sandwich ELISA experiments using FHIT antibody pairs?

When designing sandwich ELISA experiments with FHIT antibody pairs, researchers should follow this methodological approach:

  • Coating step: Dilute capture antibody (3C10G2) to 1-5 μg/mL in coating buffer (typically PBS) and add 100 μL to each well of a 96-well plate. Incubate overnight at 4°C.

  • Blocking step: Block non-specific binding sites with 200 μL of 1-5% BSA in PBS for 1-2 hours at room temperature.

  • Sample addition: Prepare samples and standards in a range covering 0.391-100 ng/mL. Add 100 μL per well and incubate at 37°C for 30-60 minutes.

  • Detection step: After washing, add 100 μL of detection antibody (1F7G3) conjugated to HRP (typically at 0.5-2 μg/mL). Incubate at 37°C for 30 minutes.

  • Substrate reaction: Add 100 μL of TMB substrate and incubate at room temperature for 10-15 minutes, then stop the reaction with 50 μL of 2N H₂SO₄.

  • Data acquisition: Measure absorbance at 450 nm with 630 nm reference wavelength .

To optimize assay performance, it's crucial to titrate both antibodies to determine their optimal concentrations. The EC50 values should be calculated from binding curves using appropriate statistical software to accurately determine assay sensitivity .

What controls should be included when validating a new FHIT antibody pair application?

A comprehensive validation of FHIT antibody pairs should include the following controls:

Control TypePurposeImplementation
Positive controlConfirms assay functionalityRecombinant FHIT protein at known concentration
Negative controlAssesses non-specific bindingBuffer only (no analyte)
Isotype controlEvaluates antibody specificityIrrelevant antibodies of matching isotypes
Cross-reactivity controlTests for interferenceStructurally similar proteins
Spike-and-recoveryVerifies matrix effectsFHIT protein added to actual sample matrix

Additionally, when implementing new applications beyond manufacturer specifications, researchers should verify antibody performance in their specific experimental system by testing:

  • Antibody binding kinetics using methods like biolayer interferometry

  • Detection limits in the specific sample type

  • Reproducibility across multiple experiments

The significance of proper controls cannot be overstated, as they help distinguish true positive signals from artifacts, particularly when working with antibodies in complex biological samples.

How can researchers optimize the blocking conditions to minimize background when using FHIT antibody pairs?

Optimizing blocking conditions is critical for maximizing signal-to-noise ratio when using FHIT antibody pairs. A methodological approach includes:

  • Systematic testing of blocking agents:

    • 1-5% BSA in PBS

    • 1-5% non-fat dry milk in PBS

    • Commercial blocking buffers with proprietary formulations

    • Combination of different proteins (e.g., 1% BSA + 0.1% casein)

  • Optimization of blocking parameters:

    • Duration: Test 1, 2, and 4-hour blocking periods

    • Temperature: Compare room temperature vs. 37°C

    • Buffer additives: Addition of 0.05% Tween-20 or 0.1% normal serum from the species unrelated to the antibody source

  • Background reduction strategies:

    • Pre-adsorb detection antibody with proteins from the sample species

    • Include 0.05-0.1% irrelevant IgG from the same species as samples in the diluent

    • Use specialized buffers designed to minimize heterophilic antibody interference

Researchers should systematically record signal-to-noise ratios for each condition to identify optimal parameters. For FHIT detection specifically, our data suggests that 2% BSA in PBS with 0.05% Tween-20 for 2 hours at room temperature typically provides the best balance between effective blocking and maintained antibody sensitivity.

What are the best methods for determining the binding kinetics of FHIT antibody pairs?

Biolayer interferometry (BLI) offers an excellent approach for characterizing the binding kinetics of FHIT antibody pairs. The methodology involves:

  • Instrument setup: Using platforms like Octet or similar BLI systems with appropriate biosensors (typically streptavidin or protein A/G).

  • Experimental design:

    • Loading step: Immobilize FHIT capture antibody (3C10G2) on sensors (300 seconds)

    • Baseline establishment: 60 seconds in buffer

    • Association phase: 180 seconds with varying antigen concentrations

    • Dissociation phase: 60 seconds in buffer

  • Data analysis:

    • Fit binding curves to a 1:1 binding model

    • Extract key parameters: ka (association rate), kd (dissociation rate), and KD (equilibrium dissociation constant)

For FHIT antibodies, optimal antigen concentrations typically range from 12.5-100 nM for kinetic determinations, with loading levels adjusted to achieve 0.7-1.5 nm wavelength shift .

ParameterTypical Range for High-Quality FHIT Antibody Pairs
ka (association rate)10⁵-10⁶ M⁻¹s⁻¹
kd (dissociation rate)10⁻⁴-10⁻³ s⁻¹
KD (equilibrium constant)0.1-10 nM

This methodology not only provides binding affinity data but also reveals important information about binding stability, which directly impacts assay performance in various experimental conditions .

How should researchers address potential rheumatoid factor (RF) interference when using FHIT antibody pairs in clinical samples?

Rheumatoid factor (RF) and heterophilic antibodies in clinical samples can cross-link assay antibodies, leading to false-positive signals that compromise data interpretation . To mitigate RF interference when using FHIT antibody pairs:

  • Sample pre-treatment options:

    • Implement PEG 6000 precipitation for antibody removal

    • Apply immunoglobulin blockade using commercial blocking reagents

    • Utilize heterophilic blocking tubes (HBT) for sample preparation

  • Assay modifications:

    • Consider using specialized ELISA PathRF format which introduces a recombinant backbone to detection antibodies

    • Implement a heterophilic antibody detection step in parallel with the primary assay

  • Validation approach:

    • Test samples with and without RF removal/blocking

    • Include RF-positive control samples of known concentration

    • Compare results with alternative detection methods

The risk of interference is particularly significant when analyzing samples from patients with autoimmune conditions, cancer, chronic infections, and certain other diseases that may present with elevated RF levels . Implementing these mitigation strategies is essential for ensuring accurate FHIT quantification in clinical research contexts.

What statistical approaches are most appropriate for analyzing data from FHIT antibody pair assays in large sample cohorts?

When analyzing data from FHIT antibody pair assays across large sample cohorts, researchers should implement a systematic statistical framework:

  • Data preprocessing:

    • Assess normality using Shapiro-Wilk or Kolmogorov-Smirnov tests

    • Apply appropriate transformations (log, square root) for non-normal distributions

    • Identify and address outliers using Grubbs' test or box-plot methods

  • Statistical analysis strategies:

    • For comparing groups: ANOVA with post-hoc tests for multiple groups or t-tests/Mann-Whitney for two groups

    • For correlations with clinical parameters: Pearson/Spearman correlation coefficients

    • For survival analysis: Kaplan-Meier curves with log-rank tests and Cox regression

  • Advanced analytical approaches:

    • Machine learning algorithms to identify patterns in complex datasets

    • Multivariate analysis to control for confounding variables

    • Receiver operating characteristic (ROC) curve analysis to evaluate diagnostic potential

  • Multiple testing corrections:

    • Apply Bonferroni, Benjamini-Hochberg, or other FDR methods to control false positive rates

    • Use q-value approach for large-scale analyses

When analyzing autoantibody data specifically, researchers should be aware that the number of autoantibodies tends to increase with age until adolescence before plateauing , which may require age-stratified analysis of FHIT antibody data in heterogeneous cohorts.

How can computational modeling enhance the development and application of FHIT antibody pairs?

Computational approaches are transforming antibody research, with specific applications for FHIT antibody pairs:

  • Antibody structure prediction and optimization:

    • The Antibody Mutagenesis-Augmented Processing (AbMAP) framework adapts protein language models to antibody-specific tasks, focusing on hypervariable regions through contrastive augmentation and multitask learning

    • RFdiffusion models enable de novo design of antibodies targeting specific epitopes with novel CDR loops

    • tFold-Ag provides end-to-end 3D atomic-level structure predictions of antibody-antigen complexes

  • Epitope mapping and optimization:

    • Computational identification of FHIT epitopes can guide the selection of optimal antibody pairs

    • Structural analysis can predict whether epitopes will be accessible in native FHIT

    • Binding simulations can evaluate potential steric hindrances between antibody pairs

  • Applied methodology for FHIT antibody research:

    • Implement sequence-based predictions to identify optimal antibody pair combinations

    • Use structural models to predict binding interfaces and optimize binding conditions

    • Apply machine learning to predict cross-reactivity profiles

These computational approaches have demonstrated remarkable success rates, with AbMAP achieving 82% hit rates in antibody design efforts , suggesting similar approaches could be valuable for optimizing FHIT antibody pairs.

What are the most promising high-throughput approaches for discovering novel FHIT antibody pairs with improved specificity?

Several cutting-edge high-throughput technologies can be leveraged to discover novel FHIT antibody pairs:

  • Microfluidics-enabled single-cell encapsulation:

    • Encapsulate antibody-secreting cells (ASCs) into antibody capture hydrogels at rates of 10⁷ cells per hour

    • Use flow cytometry to identify antigen-specific ASCs

    • Sequence isolated cells to enable recombinant antibody expression

  • In vitro display technologies:

    • Phage display libraries for screening billions of antibody variants

    • Yeast or mammalian surface display for maintaining mammalian post-translational modifications

    • Ribosome display for entirely cell-free antibody selection

  • Bispecific antibody screening platforms:

    • High-throughput combinatorial phenotypic screening approach using bispecific formats

    • Fab-based fusion proteins allowing non-covalent heterodimeric association for screening

  • Immune repertoire sequencing combined with antigen-specific sorting:

    • Deep sequencing of B cell repertoires from immunized animals or human donors

    • Computational pairing of heavy and light chains

    • AbMAP analysis revealing structure-function convergence across repertoires

These approaches offer significant advantages over traditional hybridoma methods, potentially generating diverse FHIT-specific antibodies in 2-4 weeks instead of months, with higher probabilities of discovering pairs with optimal epitope coverage and binding properties .

What are the potential causes and solutions for cross-reactivity issues with FHIT antibody pairs?

Cross-reactivity represents a significant challenge when working with FHIT antibody pairs. Here are the major causes and their methodological solutions:

Cause of Cross-ReactivityDetection MethodRecommended Solution
Epitope similarity with related proteinsWestern blot with multiple cell linesUse knockout/knockdown controls to confirm specificity
Post-translational modificationsMass spectrometry of immunoprecipitated proteinsSelect antibody pairs targeting unmodified regions
Non-specific Fc interactionsFlow cytometry with Fc blockingInclude appropriate Fc receptor blocking reagents
Conformational changes in FHITNative vs. denatured protein binding assaysChoose antibody pairs recognizing different structural elements
Molecular mimicry with viral proteinsPeptide array screeningSelect epitopes unique to FHIT with no viral homology

When developing new FHIT detection assays, researchers should validate specificity through:

  • Comprehensive cross-reactivity testing against proteins with similar domains

  • Epitope binning experiments to confirm the paired antibodies recognize distinct, non-overlapping epitopes

  • Immunodepletion studies to verify signal reduction upon FHIT removal

  • Testing in various sample matrices to identify potential matrix-specific interfering factors

For FHIT specifically, paying attention to potential cross-reactivity with other proteins containing histidine triad domains is essential for ensuring accurate and specific detection.

How can researchers address discrepancies between results obtained using different FHIT antibody-based methodologies?

When facing discrepancies between different FHIT antibody-based methods (e.g., ELISA vs. Western blot), researchers should implement a systematic investigation approach:

  • Epitope accessibility analysis:

    • Different methods expose different protein conformations

    • Some epitopes may be masked in native conditions but exposed in denatured states

    • Solution: Map the epitopes recognized by each antibody and determine their accessibility in different experimental conditions

  • Methodological validation:

    • Compare dynamic ranges of each technique (ELISA typically 0.391-100 ng/mL for FHIT )

    • Assess potential matrix effects in each methodology

    • Implement spike-and-recovery experiments across platforms

  • Antibody performance characterization:

    • Evaluate binding kinetics using biolayer interferometry

    • Assess antibody stability under each assay's conditions

    • Test for potential interfering factors specific to each method

  • Reference standard harmonization:

    • Use consistent recombinant FHIT standards across all methods

    • Implement calibration curves specific to each methodology

    • Consider absolute quantification approaches where appropriate

One important insight from the literature on antibody-based methods is that clinical samples from patients with autoimmune conditions or cancer may contain heterophilic antibodies that can cross-link assay antibodies, creating discrepancies between methods with different susceptibilities to such interference . In such cases, specialized formats like ELISA PathRF should be considered for validation.

How might structural insights into FHIT and its antibody interactions lead to next-generation detection technologies?

Structural studies of FHIT and its antibody interactions could revolutionize detection methodologies in several ways:

  • Structure-guided antibody engineering:

    • By understanding the binding interfaces between FHIT and its antibodies, researchers can engineer higher-affinity variants

    • Computational approaches like AbMAP can improve prediction accuracy for various antibody properties by focusing on hypervariable regions

    • De novo design methods could generate antibodies with precise epitope targeting and minimal cross-reactivity

  • Novel detection format development:

    • Structural insights could enable the design of allosteric sensors where FHIT binding causes conformational changes in engineered antibody constructs

    • Split-antibody complementation systems based on structural understanding of binding interfaces

    • Structure-based rational design of bispecific antibodies for enhanced sensitivity

  • Point-of-care diagnostics evolution:

    • Knowledge of binding kinetics and thermal stability can guide development of robust lateral flow assays

    • Understanding conformational epitopes could lead to aptamer-based alternatives with superior stability

    • Integration with emerging technologies like CRISPR-based detection systems

Recent research has demonstrated that antibodies containing specific structural motifs in their HCDR3 regions, such as RX₁₋₂R/KX₁₋₂R/H (RKH) and YYYYY (Y₅), can dramatically influence binding properties , suggesting similar structural motifs could be exploited in next-generation FHIT detection systems.

What role might FHIT antibody pairs play in understanding the broader landscape of autoimmunity and cancer immunology?

FHIT antibody pairs could serve as valuable tools in exploring the complex relationship between autoimmunity and cancer immunology:

  • Autoantibody profiling studies:

    • FHIT antibodies can help characterize autoantibody responses in various conditions

    • Research has shown that healthy individuals share common autoantibody profiles, with the number increasing with age until adolescence

    • FHIT antibody pairs could help distinguish pathological from physiological autoantibody responses

  • Cancer immunotherapy monitoring:

    • As a tumor suppressor gene, FHIT expression changes may correlate with response to immunotherapy

    • Monitoring FHIT protein levels in liquid biopsies during treatment

    • Investigating the role of anti-FHIT autoantibodies in cancer patients

  • Cross-disease immune repertoire analysis:

    • FHIT antibody tools can help investigate molecular mimicry between FHIT and viral proteins

    • Studies suggest viral proteins with sequences similar to human proteins may initiate cross-reactive antibodies

    • FHIT antibody pairs could help elucidate these mechanisms in the context of both autoimmunity and cancer

  • Convergent repertoire investigations:

    • AbMAP analysis has revealed extensive structure-function convergence across antibody repertoires beyond sequence alignment

    • FHIT antibody pairs could help characterize this convergence in disease-specific contexts

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.