DTX31 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of Antibodies

Antibodies are versatile tools used in research, diagnostics, and therapeutics. They can be broadly categorized into polyclonal and monoclonal antibodies based on their production methods and specificity.

  • Polyclonal Antibodies: These are derived from multiple B cells and recognize multiple epitopes on an antigen, offering robust recognition but with variability between batches .

  • Monoclonal Antibodies: Produced from a single B cell clone, these antibodies are highly specific and consistent across batches. They are widely used in therapeutic applications .

Applications of Antibodies

Antibodies are used in various applications, including:

  • Immunohistochemistry (IHC): For detecting specific proteins in tissue samples. For example, anti-CD31 antibodies are used to detect CD31/PECAM1 in tissues .

  • Western Blotting: To analyze protein expression levels in cell lysates.

  • Therapeutics: Monoclonal antibodies like REGEN-COV are used to treat diseases by targeting specific proteins .

Recombinant Antibodies

Recombinant antibodies are produced in vitro using defined sequences, ensuring consistent specificity and bioactivity across different batches. They are engineered for improved performance and are suitable for large-scale experiments .

Research Findings and Data

Since specific data on "DTX31 Antibody" is not available, here is a general table summarizing the characteristics of antibodies:

Antibody TypeProduction MethodSpecificityApplications
PolyclonalDerived from multiple B cellsMultiple epitopesRobust recognition, variable batches
MonoclonalSingle B cell cloneHighly specificTherapeutics, diagnostics
RecombinantIn vitro productionConsistent specificityLarge-scale experiments, improved performance

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
DTX31 antibody; RHS2 antibody; At1g12950 antibody; F13K23.21Protein DETOXIFICATION 31 antibody; AtDTX31 antibody; Multidrug and toxic compound extrusion protein 31 antibody; MATE protein 31 antibody; Protein ROOT HAIR SPECIFIC 2 antibody
Target Names
DTX31
Uniprot No.

Target Background

Function
This antibody positively mediates root hair elongation.
Database Links

KEGG: ath:AT1G12950

STRING: 3702.AT1G12950.1

UniGene: At.42053

Protein Families
Multi antimicrobial extrusion (MATE) (TC 2.A.66.1) family
Subcellular Location
Membrane; Multi-pass membrane protein.

Q&A

What is DTX31 and what cellular functions does it regulate?

DTX31 (also known as RHS2, At1g12950, or F13K23.21Protein) belongs to the deltex family of proteins that play regulatory roles in cellular signaling pathways. While specific information about DTX31 is limited in the current literature, deltex family proteins typically function as E3 ubiquitin ligases involved in protein degradation pathways. These proteins often participate in Notch signaling regulation, which impacts various developmental processes and cellular differentiation mechanisms. When working with DTX31 antibodies, researchers should consider these potential functional roles when designing experiments to investigate its cellular localization, interaction partners, or expression patterns in different tissue types.

What are the key considerations when selecting a DTX31 antibody for research applications?

When selecting a DTX31 antibody for research, consider:

  • Antibody format: Determine whether polyclonal or monoclonal antibodies better suit your needs. Polyclonal antibodies recognize multiple epitopes on DTX31, providing robust detection across applications but potential batch-to-batch variability. Monoclonal antibodies offer higher specificity and consistency.

  • Validation status: Verify the antibody has been validated for your specific application (IHC, ICC-IF, WB) and species of interest. Request validation data from manufacturers to ensure compatibility with your experimental system.

  • Epitope location: Consider whether the antibody targets regions that may be masked or modified in your experimental conditions.

  • Host species: Select an antibody raised in a species compatible with your experimental design, particularly for multi-color immunostaining experiments.

  • Production method: Custom-made antibodies may require significant lead time (14-16 weeks for DTX31 antibodies) but can offer targeted specificity for specialized applications.

How do I optimize western blotting protocols for DTX31 detection?

For optimal DTX31 detection via western blotting, follow these methodological steps:

  • Sample preparation: Extract proteins using lysis buffers containing protease inhibitors to prevent degradation. For membrane-associated proteins like DTX31, consider specialized detergent-based extraction methods.

  • Protein loading: Load 20-50 μg of total protein per lane, with precise quantification using BCA or Bradford assays.

  • Blocking optimization: Test both BSA and non-fat milk blocking solutions (3-5%) to determine optimal background reduction.

  • Antibody dilution: Begin with manufacturer-recommended dilutions (typically 1:500 to 1:2000) and optimize through titration experiments.

  • Incubation conditions: For primary antibody, incubate overnight at 4°C to maximize specific binding. For secondary antibody, room temperature incubation for 1-2 hours is typically sufficient.

  • Detection system: Choose chemiluminescence for general detection or fluorescence-based systems for quantitative analysis.

  • Controls: Always include positive controls (tissues/cells known to express DTX31) and negative controls (antibody diluent only) to validate specificity.

How can I address cross-reactivity concerns when using DTX31 antibodies in multiplex immunoassays?

Cross-reactivity represents a significant challenge in multiplex immunoassays involving DTX31 antibodies, particularly due to sequence homology with other deltex family proteins. To address this:

  • Perform comprehensive cross-reactivity testing by pre-absorbing the DTX31 antibody with recombinant proteins of related family members (DTX1, DTX2, DTX3, DTX4) and analyzing changes in signal intensity.

  • Implement competitive binding assays using increasing concentrations of purified DTX31 protein to demonstrate specificity through signal displacement.

  • Validate antibody specificity using genetic approaches:

    • Analyze antibody reactivity in DTX31 knockout/knockdown systems

    • Overexpress tagged DTX31 and confirm co-localization with antibody signals

  • For multiplex assays, conduct spectral overlap analysis when using fluorescently-tagged secondary antibodies to prevent false positives from emission spectrum overlap.

  • Employ bioinformatic analysis of epitope regions to predict potential cross-reactive domains and select antibodies targeting unique regions of DTX31.

What are the optimal fixation and antigen retrieval methods for DTX31 immunohistochemistry in different tissue types?

Optimizing fixation and antigen retrieval for DTX31 immunohistochemistry requires systematic evaluation across tissue types:

Fixation Protocol Comparison for DTX31 Detection:

Fixation MethodDurationTemperatureAdvantagesLimitationsBest For
4% Paraformaldehyde24-48 hrs4°CPreserves morphologyMay mask epitopesFrozen sections
10% Neutral Buffered Formalin24 hrsRTStandard histologyCross-linking may reduce signalFFPE tissues
Methanol/Acetone (1:1)10 min-20°CExcellent for membrane proteinsPotential tissue distortionCell preparations
Zinc-based fixatives24 hrsRTReduced epitope maskingLimited tissue penetrationSurface epitopes

Antigen Retrieval Optimization:

  • Heat-induced epitope retrieval (HIER): Test both citrate buffer (pH 6.0) and EDTA buffer (pH 9.0) at 95-98°C for 15-20 minutes.

  • Enzymatic retrieval: For heavily fixed tissues, try proteinase K (10-20 μg/mL) for 10-15 minutes at 37°C.

  • Dual retrieval approach: For difficult tissues, implement sequential HIER followed by mild enzymatic treatment.

  • Tissue-specific considerations: Neural tissues may require reduced fixation times and gentler retrieval methods to preserve cellular morphology while enabling DTX31 detection.

  • Validation: Always include positive control tissues with known DTX31 expression patterns to confirm protocol efficacy.

How can conflicting data between DTX31 antibody-based detection methods be reconciled?

When facing conflicting results between different DTX31 antibody-based detection methods:

  • Examine epitope accessibility differences:

    • Western blotting detects denatured proteins, exposing all epitopes

    • IHC/ICC detects proteins in semi-native states with potential conformational masking

    • Flow cytometry typically detects surface-accessible epitopes only

  • Implement orthogonal validation approaches:

    • Confirm DTX31 expression using RNA-based methods (qPCR, RNA-seq)

    • Use multiple antibodies targeting different DTX31 epitopes

    • Employ tagged-DTX31 expression systems for direct visualization

  • Evaluate post-translational modifications:

    • Analyze whether phosphorylation, glycosylation, or ubiquitination alters epitope recognition

    • Implement dephosphorylation or deglycosylation treatments prior to antibody staining

  • Quantitative comparison framework:

    • Standardize quantification methods across techniques

    • Normalize to appropriate housekeeping proteins for each method

    • Establish detection thresholds based on signal-to-noise ratios

  • Consider biological variables that might explain discrepancies:

    • Cell cycle-dependent expression patterns

    • Subcellular localization changes under different conditions

    • Tissue-specific isoform expression

What controls are essential for validating DTX31 antibody specificity in immunofluorescence studies?

A comprehensive validation strategy for DTX31 antibody specificity in immunofluorescence requires multiple control approaches:

  • Genetic controls:

    • DTX31 knockout/knockdown cells or tissues (negative control)

    • DTX31 overexpression systems (positive control)

    • Dose-dependent transfection of DTX31 to demonstrate signal correlation with expression level

  • Antibody controls:

    • Secondary antibody-only control to assess non-specific binding

    • Isotype control (irrelevant primary antibody of same isotype/host species)

    • Antibody pre-absorption with recombinant DTX31 protein (should abolish specific signal)

    • Comparative analysis using multiple DTX31 antibodies targeting different epitopes

  • Technical controls:

    • Autofluorescence control (untreated sample without antibodies)

    • Systematic titration of primary antibody concentration

    • Inclusion of known positive and negative tissue/cell types

  • Signal validation:

    • Co-localization studies with established markers of expected subcellular compartments

    • Comparative analysis of DTX31 localization under different physiological conditions

    • Quantitative analysis of signal intensity across multiple independent experiments

How do I design experiments to investigate DTX31 protein interactions using co-immunoprecipitation with anti-DTX31 antibodies?

Effective co-immunoprecipitation (co-IP) experiments to investigate DTX31 protein interactions require careful design:

  • Lysis buffer optimization:

    • Test multiple lysis conditions (NP-40, RIPA, digitonin-based) to preserve protein complexes

    • Include appropriate protease and phosphatase inhibitors

    • Optimize salt concentration (150-300 mM) to balance complex preservation with non-specific binding reduction

  • Antibody selection and coupling:

    • Choose DTX31 antibodies validated for immunoprecipitation

    • Test both direct coupling to beads and indirect capture using Protein A/G

    • Determine optimal antibody-to-lysate ratio through titration experiments

  • Experimental design:

    • Include IgG control IP from same species as DTX31 antibody

    • Perform reciprocal IPs using antibodies against suspected interaction partners

    • Consider crosslinking approaches for transient interactions

    • Include both DTX31-overexpressing systems and endogenous expression models

  • Washing and elution optimization:

    • Test stringency gradient in wash buffers to maximize signal-to-noise ratio

    • Compare different elution methods (pH, competitive, denaturing) for efficiency

    • Validate elution conditions do not interfere with downstream applications

  • Detection methods:

    • Western blot for targeted detection of suspected partners

    • Mass spectrometry for unbiased identification of interaction partners

    • Proximity ligation assays to confirm interactions in intact cells

What approaches can be used to quantify DTX31 expression levels across different tissue samples?

Comprehensive quantification of DTX31 expression across tissues requires multi-modal approaches:

Protein-Level Quantification Methods:

MethodSensitivitySpatial InformationThroughputKey Considerations
Western BlottingModerateNoneLow-ModerateRequires optimization of extraction methods for different tissues
ELISAHighNoneHighHighly quantitative but requires validation across tissue types
IHC/IF with Digital PathologyModerateHighModerateEnables spatial analysis but requires consistent staining protocols
Mass SpectrometryHighLimitedModerateCan detect post-translational modifications but requires specialized equipment

Transcript-Level Quantification:

  • qRT-PCR: Design primers specific to DTX31 with careful validation against related family members.

  • RNA-seq: Provides comprehensive transcriptome data but requires bioinformatic expertise for analysis.

  • RNA in situ hybridization: Offers spatial resolution of transcript expression in tissue context.

Integrated Analysis Approach:

  • Establish tissue-specific standard curves using recombinant DTX31 protein spiked into negative control lysates.

  • Normalize protein expression to appropriate housekeeping controls selected for stability across the tissue types being compared.

  • Correlate protein-level measurements with transcript quantification to identify potential post-transcriptional regulation.

  • When comparing across tissues, account for differences in cellularity and protein extraction efficiency through normalization to total protein content.

  • Validate findings using at least two independent quantification methods.

What are common pitfalls in DTX31 antibody applications and how can they be addressed?

Researchers frequently encounter several challenges when working with DTX31 antibodies:

  • Non-specific binding:

    • Cause: Insufficient blocking or antibody cross-reactivity

    • Solution: Optimize blocking conditions (concentration, time), test different blocking agents (BSA, casein, commercial blockers), and increase washing stringency

  • Inconsistent staining patterns:

    • Cause: Batch-to-batch antibody variation, inconsistent fixation/permeabilization

    • Solution: Validate each new antibody lot, standardize sample preparation protocols, and include positive control samples in each experiment

  • Signal detection issues:

    • Cause: Suboptimal antibody concentration or incubation conditions

    • Solution: Perform systematic titration of primary and secondary antibodies, optimize incubation time/temperature

  • Epitope masking:

    • Cause: Fixation-induced cross-linking or protein-protein interactions

    • Solution: Test multiple fixation protocols, implement antigen retrieval methods, consider native versus denaturing conditions

  • Subcellular localization discrepancies:

    • Cause: Sample preparation artifacts or biological regulation

    • Solution: Validate with multiple antibodies targeting different epitopes, compare with tagged DTX31 constructs, use subcellular fractionation to confirm localization

  • Signal variability across tissues:

    • Cause: Tissue-specific expression levels or isoforms

    • Solution: Normalize to appropriate tissue-specific controls, validate with orthogonal techniques (qPCR, RNA-seq)

How can researchers distinguish between specific and non-specific signals when using DTX31 antibodies?

To differentiate specific DTX31 signals from background or non-specific binding:

  • Implement a multi-layered validation strategy:

    • Genetic validation: Compare staining patterns in DTX31 knockout/knockdown models versus wild-type

    • Antibody validation: Test multiple antibodies targeting different DTX31 epitopes

    • Signal validation: Confirm expected molecular weight, subcellular localization, and expression pattern

  • Perform systematic controls:

    • Peptide competition/pre-absorption: Pre-incubate antibody with recombinant DTX31 or immunizing peptide

    • Secondary antibody-only controls: Identify background from secondary antibody

    • Isotype controls: Use irrelevant primary antibody of same isotype and concentration

  • Optimize signal-to-noise ratio:

    • Titrate antibody concentration to determine optimal working dilution

    • Modify blocking conditions to reduce background

    • Adjust detection parameters (exposure time, gain settings) based on control samples

  • Employ complementary techniques:

    • Confirm protein expression using orthogonal methods (RNA-seq, qPCR)

    • Use tagged DTX31 constructs as positive controls

    • Implement proximity ligation assays for interaction studies

  • Quantitative approach:

    • Establish signal threshold based on negative controls

    • Calculate signal-to-background ratios across multiple experiments

    • Implement statistical methods to differentiate specific from non-specific signals

What emerging technologies might enhance DTX31 antibody-based research?

Several emerging technologies hold promise for advancing DTX31 antibody-based research:

  • Advanced imaging approaches:

    • Super-resolution microscopy (STORM, PALM, SIM) for nanoscale localization of DTX31

    • Expansion microscopy for improved spatial resolution in tissues

    • Light-sheet microscopy for rapid 3D imaging of DTX31 distribution in intact specimens

  • Single-cell analysis platforms:

    • Mass cytometry (CyTOF) for multi-parameter DTX31 analysis at single-cell level

    • Microfluidic-based single-cell Western blotting for quantitative protein measurement

    • Spatial transcriptomics combined with protein detection for correlative analysis

  • Antibody engineering advances:

    • Nanobodies against DTX31 for improved tissue penetration

    • Recombinant antibody fragments with site-specific conjugation

    • Intrabodies for live-cell tracking of DTX31 dynamics

  • High-throughput screening approaches:

    • Antibody arrays for systematic DTX31 interaction partner screening

    • CRISPR screening combined with DTX31 antibody-based readouts

    • Automated immunostaining platforms for consistent large-scale studies

  • Integrated multi-omic approaches:

    • Combined proteomics and transcriptomics for comprehensive DTX31 pathway analysis

    • Antibody-based proximity labeling (BioID, APEX) for DTX31 interaction networks

    • Chromatin immunoprecipitation followed by sequencing (ChIP-seq) to identify DTX31-associated genomic regions

How might systems biology approaches incorporate DTX31 antibody-based data for pathway analysis?

Systems biology frameworks can leverage DTX31 antibody-based data through:

  • Multi-scale integration strategies:

    • Correlate DTX31 protein levels with transcriptomic data across tissues/conditions

    • Map DTX31 interactions to known signaling networks

    • Model DTX31 regulation in context of cellular pathways

  • Network analysis approaches:

    • Construct protein-protein interaction networks centered on DTX31

    • Identify network motifs and regulatory hubs connected to DTX31

    • Perform perturbation studies to validate network connections

  • Temporal dynamics modeling:

    • Track DTX31 expression changes over developmental timepoints

    • Monitor DTX31 localization during cellular processes

    • Develop mathematical models of DTX31-regulated pathways

  • Computational prediction tools:

    • Use antibody-derived localization data to inform subcellular targeting predictions

    • Predict post-translational modifications based on antibody epitope mapping

    • Model structural interactions based on co-immunoprecipitation data

  • Data integration platforms:

    • Incorporate DTX31 antibody-based data into pathway databases

    • Develop visualization tools for multi-parameter DTX31 data

    • Create integrated models combining genetic, transcriptomic, and proteomic data

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.