NGEF Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timelines.
Synonyms
ARHGEF27 antibody; Eph receptor-interacting exchange protein antibody; Eph-interacting exchange protein antibody; EPHEXIN antibody; Ephexin-1 antibody; Guanine nucleotide exchange factor, neuronal antibody; Neuronal guanine nucleotide exchange factor antibody; Ngef antibody; NGEF_HUMAN antibody
Target Names
NGEF
Uniprot No.

Target Background

Function
NGEF functions as a guanine nucleotide exchange factor (GEF) that selectively activates the GTPases RHOA, RAC1, and CDC42. It plays a crucial role in axon guidance by regulating ephrin-induced growth cone collapse and dendritic spine morphogenesis. Upon activation by ephrin through EPHA4, the GEF activity shifts towards RHOA, leading to its activation. Activated RHOA promotes cone retraction at the expense of RAC1- and CDC42-stimulated growth cone extension.
Gene References Into Functions
  1. A single nucleotide polymorphism (SNP), rs11678490, within the NGEF gene was identified in Korean men and associated with visceral fat accumulation. PMID: 26340433
  2. FGFR-mediated phosphorylation of ephexin1 enhances the guanine nucleotide exchange activity towards RhoA without affecting the activity to Rac1 or Cdc42. PMID: 17702745
Database Links

HGNC: 7807

OMIM: 605991

KEGG: hsa:25791

STRING: 9606.ENSP00000264051

UniGene: Hs.97316

Subcellular Location
Cytoplasm. Membrane. Cell projection, growth cone.
Tissue Specificity
Highly expressed in brain specifically in caudate nucleus and to a lower extent in amygdala and hippocampus. Also detected in lung.

Q&A

What is the principle behind biotin conjugation of antibodies?

Biotin conjugation involves the covalent attachment of biotin molecules to antibodies through a chemical reaction. This technique leverages biotin's relatively small size (240 Da), flexible valeric side chain, and ease of conjugation, which make it exceptionally well-suited for protein labeling without altering the interaction of the antibody with its target ligand . The resulting biotin-labeled antibodies can then bind with extraordinary high affinity to streptavidin (Kd ~ 10–15 M), creating one of the strongest non-covalent bonds in biological systems. This interaction forms the foundation for numerous detection and purification strategies in immunoassays .

How does the biotin-streptavidin system enhance antibody-based detection methods?

The biotin-streptavidin system provides several distinct advantages in antibody-based detection:

  • Signal amplification: The system enables the use of highly diluted primary antibodies while maintaining strong signal output .

  • Exceptional stability: The interactions remain stable against proteolytic enzymes, pH variations, temperature changes, and various denaturing reagents .

  • Versatility: The system allows for indirect interaction between biomolecules while preserving their natural binding properties .

  • Improved sensitivity: In comparative studies, biotin-streptavidin based lateral flow test strips showed enhanced sensitivity (95.21%) and specificity (99.29%) compared to conventional antibody systems .

  • Flexibility in experimental design: Researchers can employ various streptavidin-conjugated reporter molecules (enzymes, fluorophores, etc.) with the same biotinylated antibody preparation .

What are the optimal storage conditions for maintaining biotin-conjugated antibody activity?

For biotin-conjugated antibodies, including those targeting NGEF, the following storage guidelines should be observed:

  • Short-term storage (frequent use): Store at 4°C .

  • Long-term storage: Keep at -20°C in a manual defrost freezer for up to two years without detectable loss of activity .

  • Avoid repeated freeze-thaw cycles as these can compromise antibody integrity .

  • The thermal stability can be quantified by the loss rate, which should be less than 5% within the expiration date under appropriate storage conditions .

  • Both stored and freshly prepared biotin-conjugated antibodies can show equivalent performance, as demonstrated in lateral flow test strip applications .

What factors should be optimized when biotinylating NGEF antibodies for experimental use?

Successful biotinylation of NGEF antibodies requires optimization of several critical parameters:

  • Antibody-to-biotin ratio: Test different concentrations of antibody (e.g., 5 µg, 10 µg, and 15 µg) with standardized amounts of biotinylation reagent to identify optimal conjugation ratios .

  • Buffer conditions: Typically, PBS at pH 7.4 provides an appropriate environment for the biotinylation reaction .

  • Reaction time: Standard protocols recommend approximately 20 minutes at room temperature in dark conditions .

  • Quenching: Proper termination of the reaction with quenching reagents prevents over-biotinylation .

  • Purification: Removal of excess unreacted biotin is essential to reduce background in subsequent applications.

  • Functional validation: Compare the reactivity of biotinylated antibodies with gold-conjugated streptavidin to verify retention of binding capacity .

In published protocols, 5 µg of antibodies (1mg/ml) has demonstrated superior performance when conjugated to gold-streptavidin compared to higher concentrations .

How can researchers verify the specificity of biotin-conjugated NGEF antibodies?

Verification of specificity requires a multi-faceted approach:

  • Comparative analysis: Test the antibody against multiple tissue types, including those known to express NGEF (such as lung adenocarcinoma tissues) and those without NGEF expression .

  • Western blot validation: Confirm the antibody detects bands of the expected molecular weight for NGEF .

  • Knockdown/knockout controls: Verify reduced or absent signal in samples where NGEF expression has been genetically suppressed.

  • Correlation with mRNA data: Compare antibody detection patterns with NGEF mRNA expression profiles from techniques like qPCR .

  • Multiple epitope targeting: Use antibodies against different NGEF epitopes to confirm consistent localization patterns.

  • Cross-reactivity assessment: Test against related proteins to ensure specificity for NGEF rather than similar proteins.

  • Immunoprecipitation followed by mass spectrometry: Confirm the identity of the pulled-down protein as NGEF.

What controls are essential when using biotin-conjugated NGEF antibodies in immunoassays?

A comprehensive set of controls should include:

  • Positive controls: Samples with confirmed NGEF expression, such as specific LUAD tissue samples or cell lines with known high NGEF expression .

  • Negative controls: Samples known to lack NGEF expression or normal adjacent tissue from cancer patients .

  • Isotype controls: Non-specific antibodies of the same isotype as the NGEF antibody to assess background binding.

  • Blocking controls: Pre-incubation with excess unconjugated NGEF antibody to demonstrate specific binding.

  • Endogenous biotin blocking: Particularly for tissue samples, to prevent streptavidin binding to endogenous biotin.

  • Streptavidin-only controls: To assess non-specific binding of the detection system without the primary antibody.

  • Gradient dilution controls: Serial dilutions of the antibody to establish dose-dependent signal patterns.

  • Biotinylation efficiency controls: Comparison of different biotinylation batches to ensure consistency.

How can the biotin-streptavidin system be adapted for proximity labeling to study NGEF protein interactions?

The Biotinylation by Antibody Recognition (BAR) method offers a sophisticated approach for studying NGEF interactions:

  • Sample preparation: Fix and permeabilize cells or tissue samples containing NGEF .

  • Primary targeting: Apply primary antibody specific to NGEF to localize the protein of interest .

  • Secondary conjugation: Add HRP-conjugated secondary antibody that binds to the primary antibody .

  • Proximity labeling: In the presence of hydrogen peroxide and phenol biotin, the HRP creates free radicals that result in biotinylation of proteins in close proximity to NGEF .

  • Harsh extraction: Use stringent conditions for reverse cross-linking and protein solubilization .

  • Purification: Employ streptavidin-coated beads to precipitate the biotinylated proteins .

  • Identification: Analyze precipitated proteins using tandem mass-spectrometry .

  • Quantification: When possible, use Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) to distinguish genuine signals from background .

This approach enables identification of protein-protein interactions directly from primary tissue and can identify interactors of insoluble proteins forming higher-order structures .

How can researchers quantitatively assess biotinylation efficiency for NGEF antibodies?

Quantitative assessment of biotinylation efficiency can be performed through multiple methods:

  • HABA assay (4'-hydroxyazobenzene-2-carboxylic acid): Measures the displacement of HABA from avidin by biotin, allowing calculation of biotin-to-protein ratio.

  • Mass spectrometry: Provides precise determination of the number and positions of biotin molecules per antibody.

  • Functional titration: Compare activity of biotinylated antibody against a standard curve of antibodies with known biotin incorporation.

  • Colorimetric assays: Use reagents that react specifically with either biotin or the protein component to determine relative concentrations.

  • Fluorescence-based methods: Employ fluorescently-labeled streptavidin to quantify accessible biotin molecules.

  • Direct comparison: Test different biotinylation protocols and antibody concentrations (e.g., 5 µg, 10 µg, and 15 µg) on standardized detection platforms to determine optimal functional performance .

For optimal detection without steric hindrance, the ideal range is typically 3-8 biotin molecules per antibody molecule.

What are the molecular mechanisms underlying the high-affinity interaction between biotin and streptavidin?

The exceptional binding between biotin and streptavidin involves several molecular features:

  • Structural components: Streptavidin is a basic glycoprotein composed of four identical subunits, each containing a biotin-binding pocket .

  • Binding affinity: Each subunit binds one biotin molecule with extraordinary affinity (Kd ~ 10–15 M), making it one of the strongest non-covalent interactions in biology .

  • Bond formation: The interaction involves multiple hydrogen bonds and van der Waals forces between biotin and the binding pocket residues.

  • Stability factors: The complex exhibits remarkable resistance to extreme conditions including:

    • Proteolytic enzymes

    • pH variations

    • Temperature fluctuations

    • Denaturing reagents

  • Cooperative binding: The tetrameric structure of streptavidin enables multivalent interactions with biotinylated molecules.

  • Accessibility: Biotin's flexible valeric side chain allows it to extend from the antibody surface and readily access the streptavidin binding pocket .

This combination of features creates an interaction that remains stable under conditions that would disrupt most protein-ligand complexes.

What are the critical differences between using biotin-conjugated NGEF antibodies for tissue versus cell culture applications?

The application of biotin-conjugated NGEF antibodies across different sample types requires consideration of several key differences:

ParameterTissue SamplesCell Culture Samples
Fixation requirementsMore extensive fixation needed; formalin commonly usedMilder fixation sufficient; paraformaldehyde often adequate
PermeabilizationMore stringent permeabilization required for tissue penetrationGentler permeabilization sufficient
Endogenous biotinHigher levels present, especially in biotin-rich tissues (liver, kidney)Generally lower levels, but varies by cell type
Biotin blockingEssential pre-treatment stepMay be necessary but less critical
Antibody concentrationHigher concentrations typically required (5-30 µg/mL) Lower concentrations often sufficient (5-20 µg/mL)
Incubation timesLonger incubations for adequate penetrationShorter incubations usually effective
Background issuesMore complex background due to tissue heterogeneityMore uniform background
AutofluorescenceHigher, requiring specific quenching strategiesLower, easier to manage
ControlsAdjacent normal tissue idealIsogenic cell lines with NGEF knockdown/knockout

For NGEF specifically, researchers should be aware that its expression patterns vary significantly across different tissue types and are particularly elevated in lung adenocarcinoma compared to normal lung tissue .

How can the sensitivity of biotin-streptavidin systems be maximized for detecting low-abundance NGEF?

To enhance sensitivity for low-abundance NGEF detection:

  • Signal amplification strategies:

    • Employ multilayered detection systems using biotinylated tyramine for signal enhancement

    • Utilize poly-HRP streptavidin conjugates instead of standard streptavidin-HRP

    • Implement gold-streptavidin nanoparticles for increased sensitivity

  • Sample preparation optimization:

    • Use antigen retrieval methods appropriate for NGEF epitopes

    • Optimize fixation to preserve antigen while maintaining tissue morphology

    • Consider using specialized permeabilization buffers to improve antibody access

  • Detection method refinement:

    • Employ dual gold conjugation of complementary binding proteins (e.g., ACE-2 and Nanobodies for viral proteins)

    • Utilize chemiluminescent substrates for enhanced signal-to-noise ratio

    • Consider biotin-based surrogate detection methods for improved sensitivity

  • Technical enhancements:

    • Use biotin-modified lateral flow test strips, which have demonstrated improved sensitivity (95.21%) compared to conventional methods

    • Implement overnight incubations at 4°C to increase binding efficiency

    • Consider specialized buffers containing signal enhancers

Studies have shown that modified biotin-streptavidin approaches can significantly improve both sensitivity and specificity compared to traditional methods .

How can researchers address false positive signals when using biotin-conjugated NGEF antibodies?

False positives with biotin-conjugated antibodies can arise from several sources:

  • Endogenous biotin interference:

    • Implement avidin/biotin blocking kits prior to primary antibody application

    • Use specialized blocking reagents specifically designed for biotin-rich tissues

  • Non-specific binding:

    • Increase blocking stringency with combinations of BSA, normal serum, and commercial blocking reagents

    • Test multiple wash buffer formulations with varying salt and detergent concentrations

    • Include competitive binding controls with excess unconjugated antibody

  • Cross-reactivity issues:

    • Validate antibody specificity against knockout/knockdown samples

    • Test multiple anti-NGEF antibodies targeting different epitopes

    • Perform peptide competition assays to confirm binding specificity

  • Technical considerations:

    • Optimize antibody dilution through careful titration experiments

    • Ensure proper storage of biotin-conjugated antibodies to prevent aggregation

    • Compare fresh versus stored antibody preparations to evaluate stability effects

    • Implement stringent washing protocols with increased frequency and duration

  • Data interpretation:

    • Establish clear thresholds for positive signals based on appropriate controls

    • Use quantitative image analysis to distinguish specific from non-specific signals

    • Consider dual labeling approaches to confirm specificity

What approaches resolve inconsistent results between different detection methods using biotin-conjugated NGEF antibodies?

When facing inconsistent results across different detection platforms:

  • Methodological analysis:

    • Examine fundamental differences in detection principles between methods

    • Compare sample preparation protocols for potential incompatibilities

    • Assess epitope accessibility under different experimental conditions

  • Technical evaluation:

    • Standardize antibody concentrations across methods based on optimization for each platform

    • Control incubation times and temperatures precisely

    • Ensure consistent biotinylation level across experiments

  • Sample-specific considerations:

    • Assess matrix effects from different sample types

    • Evaluate whether NGEF undergoes post-translational modifications that affect detection

    • Consider whether different isoforms are preferentially detected by different methods

  • Validation strategies:

    • Implement orthogonal detection methods not relying on antibodies

    • Correlate protein detection with NGEF mRNA levels by qPCR

    • Perform spike-in experiments with recombinant NGEF at known concentrations

  • Reference standards:

    • Develop internal reference standards for cross-platform normalization

    • Use calibrated positive controls across all methods

    • Consider advanced statistical methods for integrating data from multiple platforms

Studies examining NGEF as a prognostic biomarker have successfully employed multiple validation techniques including western blot, qPCR, and immunohistochemistry to confirm expression patterns .

What factors affect the quantitative relationship between NGEF expression level and detected signal using biotin-streptavidin systems?

The quantitative relationship between expression and signal is influenced by:

  • Analytical variables:

    • Degree of antibody biotinylation (optimal range typically 3-8 biotin molecules per antibody)

    • Binding affinity of the primary antibody for NGEF

    • Efficiency of streptavidin-reporter conjugate

    • Signal amplification cascade characteristics

  • Biological variables:

    • Epitope accessibility in different cellular compartments

    • Post-translational modifications affecting antibody recognition

    • Protein-protein interactions potentially masking epitopes

    • Heterogeneity of NGEF expression within samples

  • Technical considerations:

    • Linear dynamic range of the detection system

    • Signal saturation at high expression levels

    • Background contribution to total signal

    • Detector sensitivity limits at low expression levels

  • Standardization approaches:

    • Use of calibration curves with recombinant NGEF

    • Internal reference standards for normalization

    • Multi-point standard curves to define linear response range

    • Statistical methods to correct for non-linear response regions

For accurate quantitation, researchers should determine whether their biotin-streptavidin detection system exhibits linear, logarithmic, or sigmoidal response characteristics within their expected NGEF concentration range.

How can the specificity of proximity labeling be verified when studying NGEF protein interactions?

To confirm the specificity of proximity-labeled proteins as genuine NGEF interactors:

  • Distance-dependent controls:

    • Compare labeling patterns using primary antibodies targeting different regions of NGEF

    • Implement antibodies against proteins known to be proximal or distant from NGEF

    • Vary biotin phenol reaction time to create distance-dependent labeling profiles

  • Validation experiments:

    • Perform reciprocal proximity labeling using antibodies against potential interactors

    • Confirm interactions through traditional co-immunoprecipitation

    • Validate key interactions using techniques like FRET or PLA

  • Quantitative approaches:

    • Use SILAC to distinguish specific from non-specific labeling

    • Implement statistical thresholding based on enrichment ratios

    • Analyze replicate samples to identify consistently labeled proteins

  • Biological verification:

    • Correlate identified interactions with known biological pathways

    • Test functional relationships through genetic perturbation

    • Examine co-expression patterns in relevant tissues

  • Computational validation:

    • Compare identified interactions with protein interaction databases

    • Analyze structural compatibility of proposed interactions

    • Implement machine learning approaches to classify high-confidence interactions

The BAR (Biotinylation by Antibody Recognition) method has been validated for detecting proximal proteins in intact tissues and can quantify changes in interaction patterns under different conditions or in the presence of disease mutations .

How can biotin-conjugated NGEF antibodies advance understanding of its role in cancer progression?

Biotin-conjugated NGEF antibodies enable several approaches to understanding cancer biology:

These approaches collectively build a comprehensive understanding of NGEF's multifaceted roles in cancer biology and potential clinical applications.

What methodological considerations are important when analyzing NGEF expression in patient-derived samples?

Working with clinical specimens requires attention to several methodological aspects:

  • Sample acquisition and processing:

    • Standardized protocols for tissue collection and preservation

    • Consistent fixation methods to maintain epitope integrity

    • Proper biobanking procedures with detailed clinical annotation

  • Technical considerations:

    • Rigorous blocking of endogenous biotin, particularly in biotin-rich tissues

    • Standardized antibody concentrations and incubation protocols

    • Inclusion of appropriate positive and negative control tissues

  • Data normalization:

    • Adjustment for tissue heterogeneity and tumor content

    • Standardization across multiple batches of patient samples

    • Consideration of variations in tissue quality and preservation

  • Clinical correlation:

    • Detailed patient stratification by tumor stage, grade, and molecular subtypes

    • Comprehensive collection of treatment history and response data

    • Long-term follow-up for accurate survival correlation

  • Ethical and regulatory compliance:

    • Proper informed consent procedures

    • Institutional review board approval

    • Compliance with biospecimen research regulations

Studies have demonstrated that high NGEF expression is an independent prognostic factor for lung adenocarcinoma, highlighting the importance of rigorous methodological approaches when evaluating its clinical significance .

How might biotin-conjugated NGEF antibodies be integrated into diagnostic or prognostic assays for clinical use?

Development of clinically viable NGEF-based assays could include:

  • Immunohistochemical applications:

    • Standardized scoring systems for NGEF expression in tumor biopsies

    • Integration into existing pathology workflows

    • Development of companion diagnostic assays for therapies targeting NGEF-related pathways

  • Liquid biopsy approaches:

    • Detection of circulating NGEF or NGEF-expressing cells

    • Biotin-streptavidin systems for capturing NGEF-positive circulating tumor cells

    • Development of sensitive ELISA or similar assays for NGEF in patient serum

  • Prognostic panel development:

    • Integration of NGEF with other prognostic biomarkers

    • Creation of risk stratification algorithms incorporating NGEF expression

    • Development of biotin-based lateral flow or microarray platforms for multi-biomarker assessment

  • Predictive applications:

    • Correlation of NGEF expression with response to specific therapies

    • NGEF as a component of treatment selection algorithms

    • Monitoring changes in NGEF expression during treatment as a response indicator

  • Implementation considerations:

    • Analytical validation across multiple laboratories

    • Clinical validation in prospective patient cohorts

    • Standardization of reagents and protocols for reliable clinical deployment

What emerging technologies might enhance the utility of biotin-conjugated NGEF antibodies in cancer research?

Several innovative approaches show promise for expanding NGEF research capabilities:

  • Advanced imaging technologies:

    • Super-resolution microscopy for nanoscale localization of NGEF

    • Expansion microscopy combined with biotin-streptavidin detection for enhanced spatial resolution

    • Real-time in vivo imaging using biotin-conjugated antibodies with streptavidin-reporter systems

  • Next-generation proximity labeling:

    • TurboID and miniTurbo systems for more efficient proximity labeling

    • Split-biotin ligase approaches for detecting specific protein-protein interactions

    • Spatially-restricted enzymatic tagging for subcellular interaction mapping

  • Single-cell applications:

    • Integration with single-cell RNA sequencing for correlating NGEF protein and mRNA at cellular resolution

    • Mass cytometry (CyTOF) with biotinylated antibodies for high-dimensional protein profiling

    • Microfluidic approaches for analyzing NGEF expression in rare cell populations

  • Computational enhancements:

    • Machine learning algorithms for automated quantification of NGEF expression patterns

    • Network analysis tools for interpreting NGEF interaction data

    • Integrative multi-omics approaches incorporating NGEF protein data with genomic and transcriptomic profiles

  • Therapeutic applications:

    • Development of antibody-drug conjugates targeting NGEF

    • NGEF-directed CAR-T cell approaches

    • Small molecule discovery pipelines targeting NGEF or its key interaction partners

The biotin-streptavidin system's exceptional sensitivity and specificity make it particularly well-suited for integration with these emerging technologies .

How can researchers address the current limitations of biotin-conjugated antibodies for NGEF detection and analysis?

Current limitations and potential solutions include:

  • Endogenous biotin interference:

    • Development of alternative high-affinity pairs beyond biotin-streptavidin

    • Advanced blocking strategies specifically targeting endogenous biotin

    • Engineering modified streptavidin variants with higher specificity for exogenous biotin

  • Quantification challenges:

    • Creation of calibrated reference standards for absolute quantification

    • Development of digital counting approaches for precise NGEF quantification

    • Implementation of internal normalization controls for cross-sample comparison

  • Multiplexing limitations:

    • Exploration of orthogonal labeling strategies compatible with biotin-streptavidin

    • Development of sequential elution and re-probing methods

    • Integration with mass spectrometry imaging for highly multiplexed analysis

  • Tissue penetration issues:

    • Engineering smaller biotin-binding fragments for improved tissue access

    • Development of clearing methods compatible with biotin-streptavidin detection

    • Optimization of amplification strategies that maintain spatial precision

  • Reproducibility concerns:

    • Establishment of standardized protocols across laboratories

    • Development of automated systems for antibody production and validation

    • Creation of reference materials and proficiency testing programs

Newer approaches like the modified biotin-streptavidin based lateral flow test strips have already demonstrated improvements in sensitivity and specificity compared to conventional systems , suggesting continued innovation in this space.

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.