Gliadin Gamma 18.6kDa Wheat

Gliadin Gamma 18.6kD Wheat Recombinant
Shipped with Ice Packs
In Stock

Description

Key Features:

PropertyDetailReference
Molecular Weight (Theoretical)18.6 kDa (recombinant fragment) / 37,945.14 Da (full-length)
Observed SDS-PAGE Mobility~50 kDa (discrepancy due to high proline content and low pI)
Sequence HighlightsHigh proline content; His-tag fusion at C-terminus (6 His)
Deamidation EpitopesContains transglutaminase-mediated deamidated neo-epitopes (critical for celiac disease)

Amino Acid Composition:
The protein exhibits a repetitive proline-rich domain, with a sequence including motifs such as QPQQPQQPFLQPQQPFPQQPQQPFPQTQQPQQPFPQQPQQPFPQTQQPQQPFPQLQQPQQPFPQPQQQLPQPQQPQQSFPQQQRPFIQPSL .

Role in Celiac Disease and Immune Response

Gliadin Gamma 18.6kDa Wheat is a major immunogen in celiac disease, triggering autoimmune responses in genetically predisposed individuals. Its deamidated epitopes, formed by tissue transglutaminase, are recognized by IgG and IgA autoantibodies .

Mechanism of Action:

  1. Epitope Formation: Deamidation of glutamine residues at specific sites enhances immunogenicity.

  2. Antibody Binding: Recombinant Gliadin Gamma 18.6kDa is used to detect anti-gliadin antibodies in diagnostic assays .

Recombinant Production and Biochemical Properties

The protein is heterologously expressed in E. coli with a His-tag for purification, ensuring high yield and specificity.

Production Details:

ParameterSpecificationReference
Host OrganismEscherichia coli
Purification MethodNickel affinity chromatography (His-tag)
Purity>90% (SDS-PAGE confirmed)
Storage Conditions-20°C (long-term), 4°C (short-term); avoid freeze-thaw cycles
Formulation Buffer10 mM Tris-HCl (pH 7.2), 20% glycerol ; 20 mM HEPES (pH 8.0), 200 mM NaCl

Research Applications

Gliadin Gamma 18.6kDa Wheat is pivotal in diagnostic and immunological studies.

Key Applications:

ApplicationMethodPurposeReference
ELISAPlate coating (0.15–0.5 µg/ml)Quantification of anti-gliadin antibodies in patient sera
Immunodot AssaysMembrane-based detectionQualitative screening for celiac disease biomarkers
Epitope MappingCompetitive inhibition assaysIdentifying deamidated epitopes recognized by celiac patient sera

Product Specs

Introduction
Wheat gliadin and related gluten components from barley, rye, and potentially oats can trigger an abnormal immune reaction known as Celiac disease. This disease is a chronic gastrointestinal disorder characterized by the flattening of the jejunal mucosa and intestinal lesions of varying severity in genetically predisposed individuals. Although not a classic autoimmune disease, Celiac disease is linked to anti-tissue transglutaminase antibodies. Gliadin antibody tests are highly recommended for screening at-risk populations for Celiac disease and other gluten-sensitive enteropathies. Historically, serologic tests for gliadin antibodies lacked precision and were insufficient for accurate diagnosis due to the absence of deamidated epitopes in the authentic gliadin fraction used in conventional diagnostic kits. THE BioTek's deamidated Gliadin isoform addresses this limitation by matching the deamidated neo-epitopes, which are formed in the natural antigen through transglutaminase-mediated glutamine side chain deamidation.
Description

Gliadin, encoded by a cDNA, represents an epitope-carrying fragment of a wheat gamma-gliadin isoform. It possesses a molecular mass of 19 kDa (the discrepancy between the calculated molecular weight and the observed electrophoretic mobility of approximately 50 kDa on standard SDS-PAGE is likely attributed to its high proline content and low pI) and a pH of 4.6. The epitopes are designed to correspond to the deamidated neo-epitopes, which are generated in the natural antigen through transglutaminase-mediated deamidation of glutamine side chains. The gliadin protein is fused to a hexa-histidine purification tag.

Physical Appearance
A clear, sterile-filtered solution.
Formulation
Gliadin is provided in a buffer solution containing 20mM HEPES (pH 8.0), 200mM NaCl, and 20% glycerol.
Stability

For short-term storage (2-4 weeks), maintain at 4°C.
For long-term storage, freeze at -20°C.
Minimize repeated freeze-thaw cycles.

Immunological Functions

1. Binds to human IgG and IgA autoantibodies associated with celiac disease.

2. Suitable for standard ELISA tests (checkerboard analysis of positive and negative sera panels) and immunodot assays.

Applications
Suitable for Western blot analysis using a monoclonal anti-hexa-His-tag antibody.
Coating Concentration
The recommended coating concentration ranges from 0.15 to 0.5 µg/ml, depending on the specific ELISA plate type and coating buffer used.
Purity

Purity exceeds 95% as determined by SDS-PAGE analysis.

Source
Escherichia Coli.

Q&A

What is Gliadin Gamma 18.6kDa Wheat protein and how is it structurally distinct from other gliadins?

Gliadin Gamma 18.6kDa is a specific wheat protein component belonging to the gamma gliadin family, which constitutes one of the major fractions of gluten proteins. Unlike omega gliadins that serve as key components triggering immune responses in gluten-related disorders, gamma gliadins possess distinct structural characteristics and immunogenic properties . The recombinant version is typically produced using a cDNA sequence encoding the specific gamma gliadin protein, expressed in bacterial systems such as E. coli, and purified to greater than 94% homogeneity as determined by SDS-PAGE .

The molecular structure features characteristic cysteine residues that form disulfide bonds, distinguishing it from omega gliadins which typically lack cysteine. This structural difference contributes to its unique functional properties and potentially different immunogenic profiles in celiac disease research.

How do recombinant versus native forms of Gliadin Gamma 18.6kDa compare in research applications?

Recombinant Gliadin Gamma 18.6kDa protein offers several advantages over native extraction, including:

  • Consistent protein sequence without contamination from other wheat proteins

  • Precise control over post-translational modifications

  • Higher purity levels (>94%) achievable through affinity purification using the His-tag

  • Reduced batch-to-batch variation critical for reproducible immunological studies

What buffer conditions optimize Gliadin Gamma 18.6kDa stability for long-term storage and experimental use?

For optimal stability, Gliadin Gamma 18.6kDa is typically supplied in 20mM HEPES buffer at pH 8.0, containing 200mM NaCl and 20% glycerol . This buffer composition helps maintain protein solubility and prevents aggregation.

Storage recommendations:

  • Short-term use (2-4 weeks): Store at 4°C

  • Long-term storage: Maintain at -20°C

  • Avoid repeated freeze-thaw cycles which promote denaturation and aggregation

For extended storage periods beyond one year, researchers should consider maintaining aliquots at -70°C or colder to preserve full biological activity. When planning experimental timelines, note that the protein is typically guaranteed stable for two years when stored at -70°C or colder, and one year when aliquoted and stored at -20°C .

What are the optimal protocols for using Gliadin Gamma 18.6kDa in ELISA-based autoantibody detection?

When developing ELISA assays for detection of anti-gliadin autoantibodies using recombinant Gliadin Gamma 18.6kDa:

  • Plate coating:

    • Recommended concentration: 0.15-0.5 μg/ml depending on plate type and coating buffer

    • Optimal coating buffer: Typically carbonate-bicarbonate buffer (pH 9.6)

    • Coating time: 16-18 hours at 4°C for maximum protein adherence

  • Blocking:

    • Use 2-5% BSA or casein-based blockers to minimize background

    • Avoid milk-based blockers which may contain trace gluten proteins

  • Sample dilution:

    • For serum samples: Initial screening at 1:100 dilution

    • For cerebrospinal fluid: Initial testing at 1:10 dilution

  • Antibody detection:

    • The protein effectively binds both IgG and IgA-type human autoantibodies associated with celiac disease

    • Include isotype-specific secondary antibodies for differential diagnosis

  • Control inclusion:

    • Always run checkerboard analysis with validated positive/negative sera panels

    • Include cross-reactivity controls with other gluten proteins

This methodological approach maximizes sensitivity while maintaining specificity for celiac disease-associated antibodies.

How should researchers design immunodot assays using Gliadin Gamma 18.6kDa to differentiate celiac-specific responses?

When implementing immunodot assays with Gliadin Gamma 18.6kDa:

  • Membrane selection:

    • PVDF membranes typically provide better protein retention than nitrocellulose

    • Pre-activation with methanol improves hydrophobic protein binding

  • Protein application:

    • Apply 1-5 μg protein per dot in a grid pattern

    • Allow complete drying before blocking

  • Differential diagnosis design:

    • Create multi-antigen arrays including:

      • Gliadin Gamma 18.6kDa

      • Omega gliadins (for comparison with different immunogenic profiles)

      • Tissue transglutaminase (tTG)

      • Deamidated gliadin peptides

  • Sample processing:

    • Pre-absorb sera with E. coli lysates if using recombinant proteins to reduce background

    • Test multiple dilutions (1:50, 1:100, 1:200) to identify optimal signal-to-noise ratio

  • Signal development:

    • Chemiluminescent detection provides quantifiable results for comparison

    • Document results with densitometry for semi-quantitative analysis

This approach enables researchers to differentiate between various gluten protein-specific responses and identify clinically relevant autoantibody patterns.

What purification strategies yield the highest purity Gliadin Gamma 18.6kDa preparations for crystallization studies?

For researchers requiring ultra-pure Gliadin Gamma 18.6kDa preparations:

  • Initial purification:

    • For His-tagged recombinant protein, use IMAC (Immobilized Metal Affinity Chromatography)

    • Optimize imidazole concentration in elution buffer to minimize co-purification of contaminating proteins

  • Secondary purification:

    • Size exclusion chromatography to separate monomeric from aggregated forms

    • Ion exchange chromatography to remove charged contaminants

  • Tag removal:

    • If tag-free protein is required, implement specific protease cleavage (TEV or thrombin)

    • Perform reverse IMAC to separate cleaved protein from tag and uncleaved material

  • Final polishing:

    • Hydroxyapatite chromatography can separate proteins with similar characteristics

    • Achieve >99% purity required for crystallization

  • Quality assessment:

    • Verify by multiple methods:

      • SDS-PAGE with silver staining

      • Mass spectrometry

      • Dynamic light scattering for aggregation analysis

These sequential purification steps consistently yield preparations suitable for structural biology applications including crystallization and high-resolution structural studies.

How do epitope mapping approaches for Gliadin Gamma 18.6kDa differ from those used for omega gliadins?

Epitope mapping approaches for Gliadin Gamma 18.6kDa require specialized techniques distinct from those used for omega gliadins:

  • Peptide synthesis strategy:

    • For gamma gliadins: Focus on cysteine-rich repeats and unique C-terminal domains

    • For omega gliadins: Emphasize proline and glutamine-rich repetitive sequences

  • Overlapping peptide design:

    • Create 15-20mer peptides with 5-amino acid overlaps

    • Include native and deamidated peptide variants (Q→E substitutions) to mimic tissue transglutaminase modifications

  • Conformational epitope considerations:

    • Gamma gliadins require methods that preserve disulfide bonds

    • Use circular dichroism to confirm secondary structure retention in synthetic peptides

  • T-cell versus B-cell epitope mapping:

    • For T-cell epitopes: HLA-DQ2/DQ8 binding assays followed by T-cell proliferation tests

    • For B-cell epitopes: Competitive inhibition ELISA with patient sera

  • Cross-reactivity assessment:

    • Test mapped epitopes against other gliadin families to determine uniqueness

    • Evaluate homology with non-gluten proteins to identify potential cross-reactive epitopes

This comprehensive approach provides critical information about disease-specific immunogenic regions that may inform diagnostic test development and therapeutic interventions.

What are the most effective protocols for studying Gliadin Gamma 18.6kDa interactions with innate immune receptors?

For investigating interactions between Gliadin Gamma 18.6kDa and innate immune receptors:

  • Receptor binding assays:

    • Surface Plasmon Resonance (SPR) with immobilized receptors

    • ELISA-based binding with recombinant receptor ectodomains

    • Cell-based reporter assays using HEK293 cells expressing specific pattern recognition receptors

  • Cell culture systems:

    • Primary dendritic cells or macrophages for physiological relevance

    • Cell lines (THP-1, U937) for standardized responses

    • Intestinal epithelial cell models (Caco-2, T84) for barrier function studies

  • Signaling pathway analysis:

    • Phospho-flow cytometry for single-cell signaling responses

    • Western blotting for NF-κB, MAPK, and inflammasome pathway activation

    • RNA-seq for comprehensive transcriptional profiling

  • Comparison controls:

    • Include LPS as positive control for TLR4 activation

    • Use omega gliadins to differentiate gliadin subtype-specific responses

    • Test both native and recombinant proteins to identify processing-dependent effects

  • Validation in complex systems:

    • Ex vivo intestinal tissue culture from celiac and non-celiac individuals

    • Organoid models to assess epithelial-immune cell interactions

These methodological approaches enable researchers to characterize fundamental mechanisms by which different gliadin subtypes trigger innate immune activation and potentially contribute to celiac disease pathogenesis.

What experimental design best resolves conflicting data on Gliadin Gamma 18.6kDa deamidation by tissue transglutaminase?

When addressing conflicting findings regarding Gliadin Gamma 18.6kDa deamidation patterns:

  • Sequential deamidation analysis:

    • Time-course experiments with tissue transglutaminase (tTG)

    • Mass spectrometry to identify specific glutamine residues modified over time

    • Correlation of deamidation patterns with immunogenicity

  • Comparative approach:

    • Side-by-side testing of multiple gamma gliadin subtypes

    • Inclusion of omega gliadins as differential controls

    • Analysis of native versus recombinant proteins to identify production-dependent modifications

  • Functional validation:

    • HLA-DQ2/DQ8 binding assays of native versus deamidated peptides

    • T-cell proliferation assays using cells from celiac patients

    • Competitive peptide binding to quantify relative affinities

  • Structural biology integration:

    • Circular dichroism before and after deamidation

    • NMR analysis of structural changes induced by specific deamidation events

    • Computational modeling of deamidation effects on peptide-MHC interactions

  • Comprehensive data integration:

    • Multi-laboratory validation using standardized protocols

    • Meta-analysis of published deamidation data with consistent statistical methods

    • Development of predictive algorithms for preferred deamidation sites

This systematic approach resolves apparent contradictions by identifying experimental variables that influence deamidation patterns and establishing definitive structure-function relationships relevant to celiac disease pathogenesis.

How can researchers overcome solubility challenges when working with Gliadin Gamma 18.6kDa in physiological buffers?

Gliadin proteins, including Gamma 18.6kDa, present significant solubility challenges. Researchers can implement these solutions:

  • Buffer optimization strategy:

    • Start with HEPES buffer (20mM, pH 8.0) containing 200mM NaCl and 20% glycerol as baseline

    • Systematically test pH range (7.0-9.0) with 0.5 increments

    • Evaluate salt concentration effects (100-300mM NaCl)

  • Solubility enhancers:

    • Non-ionic detergents: Add 0.05-0.1% Tween-20 or Triton X-100

    • Zwitterionic additives: CHAPS (0.5-1%) preserves native conformation

    • Arginine (50-100mM) or proline (25-50mM) as chemical chaperones

  • Physical processing techniques:

    • Gentle sonication (10-second pulses, 30% amplitude)

    • Temperature cycling between 4°C and room temperature

    • Filtration through 0.22μm filters after solubilization

  • Concentration determination methods:

    • BCA assay with BSA standard curves for soluble fractions

    • Amino acid analysis for absolute quantification

    • UV spectroscopy with extinction coefficient correction

  • Long-term stability approach:

    • Store concentrated stock solutions (>1mg/ml)

    • Use small aliquots to avoid repeated freeze-thaw cycles

    • Add protease inhibitors to prevent degradation during storage

These approaches enable researchers to maintain Gliadin Gamma 18.6kDa in solution at physiologically relevant concentrations while preserving its native conformation and biological activity.

What strategies effectively distinguish between specific and non-specific antibody responses to Gliadin Gamma 18.6kDa in research samples?

To differentiate specific from non-specific antibody responses:

  • Competitive inhibition assays:

    • Pre-incubate samples with soluble Gliadin Gamma 18.6kDa

    • Compare binding before and after absorption

    • True specific responses show dose-dependent inhibition

  • Cross-reactivity assessment:

    • Test binding against multiple gliadin subtypes

    • Include non-gluten proteins as negative controls

    • Determine antibody recognition patterns across protein panels

  • Antibody isotype profiling:

    • Evaluate IgG subclasses (IgG1-4) and IgA responses

    • Compare profiles between patient and control samples

    • Identify disease-specific isotype distributions

  • Epitope-specific assays:

    • Use synthetic peptides representing specific epitopes

    • Compare whole protein versus peptide recognition

    • Map response to known immunogenic regions

  • Functional validation:

    • Assess ability of purified antibodies to activate complement

    • Measure effects on epithelial permeability in cell models

    • Correlate antibody binding with biological activity

This comprehensive approach provides multiple layers of validation to distinguish genuine antigen-specific responses from background or cross-reactive antibodies, essential for accurate interpretation of research findings.

What are the critical parameters for reproducible immunogenicity studies using Gliadin Gamma 18.6kDa in animal models?

To ensure reproducible immunogenicity studies using Gliadin Gamma 18.6kDa in animal models:

  • Animal model selection:

    • Consider genetic background (HLA-DQ2/DQ8 transgenic mice)

    • Control for microbiome variations through co-housing or fecal transplantation

    • Use age and sex-matched animals with documented genetic background

  • Protein preparation consistency:

    • Standardize recombinant protein expression and purification protocols

    • Verify batch-to-batch consistency via SDS-PAGE and mass spectrometry

    • Validate biological activity before each experimental series

  • Administration protocol:

    • Define precise dosing regimen (10-100μg per dose)

    • Select appropriate route (intraperitoneal, subcutaneous, or oral)

    • Include adjuvant controls to distinguish intrinsic from adjuvant-dependent immunogenicity

  • Comprehensive readouts:

    • Measure antibody responses (titer, isotype, epitope specificity)

    • Assess T-cell responses (proliferation, cytokine production)

    • Evaluate tissue pathology (intestinal morphology, inflammatory markers)

  • Environmental standardization:

    • Maintain consistent housing conditions (temperature, humidity, light cycles)

    • Control diet composition, especially avoiding inadvertent gluten exposure

    • Document health status including presence of infections or stress factors

This systematic approach minimizes variability and enables meaningful comparisons between different experimental groups, essential for translating findings to human disease mechanisms.

How might comparative proteomics between different wheat varieties inform selection of specific Gliadin Gamma 18.6kDa variants for research?

Comparative proteomics offers significant opportunities for advancing Gliadin Gamma 18.6kDa research:

  • Variant identification protocol:

    • LC-MS/MS analysis of gliadin extracts from diverse wheat varieties

    • De novo sequencing of gamma gliadin variants

    • Correlation of sequence variations with immunogenic potential

  • Functional comparison approach:

    • Express recombinant variants representing major haplotypes

    • Compare T-cell stimulatory capacity in celiac disease models

    • Identify naturally occurring hypoimmunogenic variants

  • Evolutionary analysis integration:

    • Phylogenetic mapping of gamma gliadin variants across Triticum species

    • Identification of ancestral versus derived sequence features

    • Correlation with domestication and breeding history

  • Bioinformatic prediction framework:

    • Develop algorithms to predict immunogenicity based on sequence

    • Create databases of variant-specific epitope maps

    • Design tools for rational selection of research-relevant variants

  • Translation to breeding applications:

    • Identify marker sequences for low-immunogenicity varieties

    • Develop screening tools for wheat breeding programs

    • Characterize environmental effects on gliadin expression profiles

This comprehensive approach enables researchers to select the most relevant Gliadin Gamma 18.6kDa variants for specific research questions, improving model relevance and translational potential.

What emerging technologies might revolutionize structure-function studies of Gliadin Gamma 18.6kDa in celiac disease research?

Several cutting-edge technologies show promise for transforming Gliadin Gamma 18.6kDa research:

  • Cryo-EM applications:

    • High-resolution structural determination without crystallization

    • Visualization of protein-antibody complexes

    • Mapping conformational epitopes at near-atomic resolution

  • Single-cell immunoprofiling:

    • Paired TCR-BCR sequencing of gliadin-reactive lymphocytes

    • Spatial transcriptomics of intestinal tissues during gliadin exposure

    • Clonal tracking of gliadin-specific T cells during disease progression

  • Organoid-on-chip systems:

    • Patient-derived intestinal organoids with immune components

    • Real-time monitoring of epithelial-immune interactions

    • High-throughput screening of immunomodulatory compounds

  • Computational biology integration:

    • Molecular dynamics simulations of gliadin-receptor interactions

    • Machine learning algorithms for epitope prediction

    • Systems biology modeling of immune network responses

  • CRISPR-based approaches:

    • Precise genome editing to create improved celiac disease models

    • Development of modified wheat lines with reduced immunogenicity

    • High-throughput screening of genetic factors influencing gliadin responses

These technologies collectively provide unprecedented resolution in understanding Gliadin Gamma 18.6kDa structure-function relationships and will likely accelerate development of diagnostic and therapeutic approaches for celiac disease.

Product Science Overview

Molecular Characteristics

Gliadin Gamma 18.6kD Wheat Recombinant is a cDNA coding for an epitope-carrying fragment of a wheat gamma-gliadin isoform. It has a molecular mass of approximately 18,616 Daltons . The high proline content and low isoelectric point (pI) of gliadins contribute to the observed discrepancy between the calculated molecular weight and the electrophoretic mobility observed on standard SDS-PAGE, which is around 47-50 kDa .

Expression and Purification

This recombinant protein is typically expressed in Escherichia coli (E. coli) and is purified using a hexa-histidine purification tag . The protein is supplied in a sterile filtered clear solution, formulated in 20mM HEPES buffer (pH 8.0), 200mM NaCl, and 20% glycerol . The purity of the protein is greater than 94-95% as determined by SDS-PAGE .

Immunological Significance

Gliadin Gamma 18.6kD Wheat Recombinant is particularly significant in the context of celiac disease. Celiac disease is a chronic gastrointestinal disorder characterized by an abnormal immune response to gluten. The disease leads to the flattening of the jejunal mucosa and intestinal lesions of varying severity in genetically predisposed individuals .

The recombinant gliadin protein contains deamidated neo-epitopes, which are formed by transglutaminase-mediated glutamine side chain deamidation in the natural antigen . These epitopes are crucial for the binding of IgG- and IgA-type human autoantibodies associated with celiac disease .

Applications

Gliadin Gamma 18.6kD Wheat Recombinant is used in various immunological assays, including:

  • Western Blotting: It is used with monoclonal anti-hexa-His-tag antibodies .
  • ELISA (Enzyme-Linked Immunosorbent Assay): It is used in standard ELISA tests for the analysis of positive/negative sera panels .
  • Immunodot Assays: It is used for the detection of autoantibodies in celiac disease .
Storage and Stability

The recombinant protein should be stored at 4°C if the entire vial will be used within 2-4 weeks. For longer storage periods, it should be frozen at -20°C. It is important to avoid multiple freeze-thaw cycles to maintain the protein’s stability .

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.