DTX34 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

DTX34 (clone DTX.34) is a mouse-derived monoclonal antibody conjugated to fluorescein isothiocyanate (FITC) for flow cytometry applications . It targets TNF-α, a proinflammatory cytokine involved in systemic inflammation and immune cell regulation.

Flow Cytometry in Immune Studies

DTX34 has been utilized in multi-color flow cytometry panels to analyze cytokine production in human immune cells, including:

  • CD3+ lymphocytes (T cells)

  • CD3−CD8+ lymphocytes

  • Monocytes

In a study comparing serum and cell-specific cytokines in HIV-positive and HIV-negative individuals, DTX34 enabled precise detection of TNF-α expression across these cell types .

Key Findings

  • HIV-Positive Individuals: Elevated TNF-α levels correlated with immune activation in CD3+ T cells and monocytes .

  • Neutralization Mechanism: While DTX34 itself is not a neutralizing antibody, analogous TNF-α-neutralizing antibodies (e.g., NeutraKine™) show ND50 values of 10–40 ng/mL in blocking cytokine activity .

Table 1: Flow Cytometry Panel Featuring DTX34 Antibody

TargetFluorophoreCloneCell Type Analyzed
TNF-αFITCDTX.34CD3+ lymphocytes, monocytes
CD3PerCPLymphocytes
CD8APCLymphocytes

Table 2: Comparative Neutralization Data for TNF-α Antibodies

Antibody TargetCloneND50 RangeAssay Context
TNF-α69002-1-Ig10–40 ng/mLL-929 cell cytotoxicity

Experimental Validation

  • Staining Protocol: DTX34 was used in four-color cytometry (FACSCalibur) with CellQuest software, analyzing 50,000–80,000 cellular events per sample .

  • Clinical Relevance: TNF-α detection via DTX34 provided insights into cytokine dysregulation during HIV progression .

Limitations and Alternatives

  • Specificity: DTX34 is optimized for flow cytometry, not neutralization. For functional blockade, antibodies like NeutraKine™ are preferred .

  • Species Reactivity: Validated in human samples; cross-reactivity with other species remains untested .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
DTX34 antibody; At4g00350 antibody; A_IG005I10.20 antibody; F5I10.20Protein DETOXIFICATION 34 antibody; AtDTX34 antibody; Multidrug and toxic compound extrusion protein 34 antibody; MATE protein 34 antibody
Target Names
DTX34
Uniprot No.

Target Background

Database Links

KEGG: ath:AT4G00350

STRING: 3702.AT4G00350.1

UniGene: At.34572

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

Q&A

What is the peptide-based approach for antibody generation, and when is it most appropriate?

Peptide-based antibody generation is particularly valuable when either the complete protein is not available in sufficient quantities to carry out an adequate immunization protocol or when antibodies that recognize only specific regions of a polypeptide chain are desired. This approach involves careful analysis of the amino acid sequence of the target protein, selecting peptide regions based on local hydrophilicity (such as using the Hopp and Woods method), and conjugating these peptides to carrier proteins like keyhole limpet hemocyanin (KLH) for immunization or bovine serum albumin (BSA) for testing specificity. The selected peptides should ideally be 12-15 amino acids in length and come from the extracellular or exposed portions of the target protein to ensure accessibility for antibody binding .

How should researchers validate the specificity of newly developed antibodies?

Antibody validation requires a multi-method approach to confirm specificity. Begin with ELISA testing against both the immunizing peptide and unrelated control peptides to establish basic reactivity. Then proceed to Western blotting analysis using both positive control samples known to express the target protein and negative control samples that don't express it - observing a single band of the expected molecular weight indicates specificity. Further validation should include testing against multiple cell lines or tissue types with varying expression levels of the target. Assess cross-reactivity with structurally similar proteins, particularly within the same protein family. For more rigorous validation, use knockout or knockdown models where the target protein is absent or reduced, respectively. Finally, compare results with commercially available antibodies against the same target to benchmark performance .

What are the key differences between monoclonal and polyclonal antibodies in research applications?

Monoclonal antibodies (mAbs) are produced from a single B-cell clone, resulting in antibodies that recognize a single epitope with high specificity but potentially limited sensitivity. They offer excellent reproducibility between batches and are ideal for applications requiring high specificity such as therapeutic use or detecting specific protein modifications (like phosphorylation states). Production involves hybridoma technology, where B-cells from immunized animals are fused with myeloma cells and cloned by limiting dilution to isolate single clones. In contrast, polyclonal antibodies recognize multiple epitopes on the target antigen, providing higher sensitivity but potentially more cross-reactivity. They are generated by immunizing animals and collecting serum containing diverse antibodies. Polyclonal antibodies are better for detecting proteins in denatured states and are less affected by minor conformational changes in the target protein .

What factors determine the choice of carrier protein for peptide immunization?

The choice of carrier protein significantly impacts immunogenicity and antibody production efficacy. Keyhole limpet hemocyanin (KLH) is frequently preferred due to its large molecular weight (4-8 MDa) and highly immunogenic nature, which strongly stimulates the immune response. KLH contains numerous epitopes that activate T-helper cells, enhancing the immune response against the conjugated peptide. For testing purposes, bovine serum albumin (BSA) is commonly used since it has a defined molecular weight (66 kDa) that allows for easier monitoring of conjugation efficiency through techniques like SDS-PAGE. The choice should account for the experimental animals to be used, as some carrier proteins may provoke varying immune responses in different species. Additionally, consider using different carriers for immunization versus screening to avoid detecting antibodies against the carrier itself .

How should researchers optimize hybridoma selection following cell fusion in monoclonal antibody production?

After cell fusion, implement a strategic multi-stage selection process beginning with HAT (hypoxanthine, aminopterine, and thymidine) medium culture to eliminate unfused myeloma cells while allowing hybridomas to survive. Plate the fused cells with peritoneal macrophages as feeder layers to provide growth factors supporting hybridoma survival. During initial screening, test supernatants via ELISA against both the target antigen and unrelated controls to identify specific binders. For clones showing promise, perform limiting dilution at densities of 1-0.5 cells per well for at least three rounds to ensure monoclonality. Between each round, rescreen supernatants to confirm stable antibody production. For final clone selection, prioritize hybridomas demonstrating both high absorbance in ELISA and stable growth patterns. Further confirm monoclonality through microscopic examination of wells during early growth to ensure they originated from single cells .

What are the critical steps in isotype determination and antibody purification for research applications?

Isotype determination begins with ELISA-based methods using isotype-specific secondary antibodies (anti-IgG1, IgG2a, IgG2b, IgG3, IgM, and IgA) to identify both the heavy and light chain classes. This information is crucial as different isotypes have varying properties that affect purification strategies and downstream applications. For purification, select the appropriate affinity chromatography method based on isotype - Protein A works well for most IgG subclasses but with varying affinities, while Protein G is preferred for IgG3 or mouse IgG1. For IgM antibodies, consider mannose-binding protein columns. During purification, maintain appropriate buffer conditions to preserve antibody structure and function. Confirm purification success through SDS-PAGE under non-reducing conditions, expecting a single band at approximately 150 kDa for intact IgG. Finally, dialyze against a physiological buffer and determine protein concentration through absorbance at 280 nm, considering the specific extinction coefficient of the antibody isotype .

How can researchers effectively design experiments to evaluate antibody cross-reactivity with similar epitopes?

Design a comprehensive cross-reactivity testing protocol starting with ELISA against the target peptide alongside a panel of structurally similar peptides with single or multiple amino acid substitutions. Follow with Western blotting against lysates from multiple cell types expressing the target protein and related family members at varying levels. Implement competitive binding assays where unlabeled potential cross-reactants are pre-incubated with the antibody before adding the primary target to assess displacement effects. For advanced characterization, employ surface plasmon resonance (SPR) to quantitatively measure binding kinetics and affinity constants for both the target and potential cross-reactants. Additionally, use immunoprecipitation followed by mass spectrometry to identify all proteins captured by the antibody under native conditions. This multi-method approach allows for detailed mapping of antibody specificity and cross-reactivity profiles, essential for applications requiring high specificity discrimination between similar epitopes .

What methodological approaches can optimize antibody performance in Western blotting?

To optimize Western blotting with antibodies, begin with sample preparation considerations: determine if denaturing conditions affect epitope recognition and adjust protocols accordingly for native versus reduced conditions. Test multiple blocking agents (5% milk, BSA, or commercial blockers) as certain antibodies perform better with specific blockers - particularly for phospho-specific antibodies, which often require BSA instead of milk proteins that contain phosphatases. Establish an antibody titration curve testing concentrations from 0.1-10 μg/ml to determine the optimal signal-to-noise ratio. For incubation conditions, compare various times (1-16 hours) and temperatures (4°C, room temperature) to identify optimal parameters for specific antibody-epitope interactions. For signal development, evaluate both chemiluminescent and fluorescent detection methods, considering sensitivity requirements and quantification needs. Finally, implement appropriate positive and negative controls, including cell lines with known expression levels of the target protein, to validate results and troubleshoot unexpected outcomes .

How can biophysical modeling enhance the design of antibodies with tailored specificity profiles?

Biophysical modeling approaches can significantly enhance antibody specificity design through computational methods that analyze binding modes associated with particular ligands. The process begins with high-throughput sequencing data from phage display experiments against multiple related ligands, which provides input for constructing a computational model of binding preferences. The model disentangles different binding modes, even when they involve chemically similar epitopes that cannot be experimentally separated during selection. This computational framework enables researchers to predict mutations that would enhance binding to specific targets while reducing cross-reactivity with similar epitopes. The approach can be used to design antibodies with both highly specific binding to single targets and controlled cross-specificity across multiple desired targets. Importantly, this method overcomes limitations of traditional selection approaches by allowing in silico exploration of sequence space beyond what was experimentally tested, effectively expanding the practical library size and enabling more precise control over specificity profiles .

What strategies are effective for generating antibodies against challenging epitopes in transmembrane proteins?

Generating antibodies against transmembrane proteins presents unique challenges due to their complex topology and hydrophobicity. A strategic approach begins with careful epitope selection, targeting extracellular domains that are naturally exposed and accessible. For CD34 and similar transmembrane proteins, analyze the amino acid sequence using predictive algorithms to identify regions with high hydrophilicity, surface probability, and antigenic index. Select 12-15 amino acid peptides from these regions, avoiding transmembrane segments. Conjugate these peptides to carrier proteins like KLH using heterobifunctional crosslinkers that preserve critical epitope structures. Implement a multi-step immunization strategy with gradual antigen increases and appropriate adjuvants to overcome potential tolerance issues. For selection and screening, use multiple formats including ELISA against peptide-conjugates and cell-based assays with native protein expression. Additionally, consider phage display technologies with customized selection strategies that alternate between peptide targets and cells expressing the native protein to ensure recognition of the correctly folded epitope .

How do post-translational modifications affect antibody epitope recognition, and how can researchers address this in experimental design?

Post-translational modifications (PTMs) significantly impact antibody epitope recognition through structural and chemical alterations of the target protein. Phosphorylation, as seen with the MK2 (phospho T334) antibody, creates a negatively charged modification that can either create new epitopes or mask existing ones. When designing experiments involving PTM-sensitive antibodies, researchers should implement parallel detection strategies using antibodies that recognize both modified and unmodified versions of the protein to obtain a complete understanding of protein dynamics. Treatment of samples with specific enzymes (phosphatases, glycosidases, etc.) before antibody application can reveal modification-dependent binding. When working with phospho-specific antibodies like anti-MK2 (phospho T334), include appropriate positive controls such as lysates from cells treated with activators of the relevant signaling pathway, and negative controls using phosphatase treatment. Additionally, validate specificity through peptide competition assays using both phosphorylated and non-phosphorylated peptides to confirm PTM-specific recognition .

What approaches can integrate antibodies into multi-omics research frameworks?

Integrating antibodies into multi-omics frameworks requires strategic experimental design that connects protein-level data with other molecular datasets. Begin by selecting antibodies with thoroughly validated specificity profiles, preferably ones that recognize conserved epitopes to enable cross-species comparisons. For proteogenomic integration, design experiments where samples undergo parallel analysis by antibody-based methods (immunohistochemistry, Western blotting) and transcriptomic approaches (RNA-seq), enabling direct correlation between protein presence and gene expression. To connect with metabolomic data, consider using antibodies against key metabolic enzymes or transporters while simultaneously measuring metabolite levels. For spatial multi-omics, implement multiplex immunofluorescence with carefully selected antibody panels that don't interfere with each other, combined with in situ transcriptomics methods. Analysis frameworks should incorporate computational methods that can integrate these diverse data types, such as network-based approaches that map protein interactions detected through antibody-based methods onto transcriptional regulatory networks .

How should researchers address conflicting results between antibody-based detection and genetic expression data?

When confronting discrepancies between antibody-based protein detection and genetic expression data, implement a systematic troubleshooting approach. First, verify antibody specificity through additional validation methods beyond those initially used, including testing on knockout/knockdown models and peptide competition assays. Assess post-transcriptional regulation mechanisms that might explain the discrepancy, such as microRNA-mediated repression or altered protein stability, by measuring mRNA half-life and performing pulse-chase experiments to determine protein turnover rates. Consider technical factors including differences in detection sensitivity between antibody-based methods and nucleic acid techniques, as well as potential epitope masking due to protein-protein interactions or conformational changes. If using phospho-specific antibodies like anti-MK2 (phospho T334), remember that phosphorylation status may not correlate with total protein expression. Finally, examine temporal dynamics by time-course experiments, as mRNA and protein levels may peak at different times following cellular stimulation .

What statistical approaches are recommended for analyzing immunohistochemistry data generated with research antibodies?

Analyzing immunohistochemistry data requires rigorous statistical approaches tailored to the nature of the data and research question. Begin with appropriate scoring systems - either automated image analysis for quantitative assessment or standardized manual scoring by multiple blinded observers using defined criteria (H-score, Allred score, or percentage positive cells). For comparing expression between experimental groups, select statistical tests based on data distribution: parametric tests (t-test, ANOVA) for normally distributed data or non-parametric alternatives (Mann-Whitney U, Kruskal-Wallis) for non-normal distributions. When evaluating multiple markers or tissue regions, apply appropriate corrections for multiple testing (Bonferroni, Benjamini-Hochberg FDR). For correlative studies with clinical outcomes, utilize survival analysis methods including Kaplan-Meier curves with log-rank tests and Cox proportional hazards models with hazard ratios. Additionally, consider more advanced analytical approaches such as tissue microarray analysis for high-throughput screening or machine learning algorithms for pattern recognition in complex staining profiles .

How can researchers effectively interpret and troubleshoot unexpected Western blot results with phospho-specific antibodies?

When encountering unexpected Western blot results with phospho-specific antibodies like anti-MK2 (phospho T334), implement a systematic troubleshooting approach. First, verify sample handling protocols, as phosphorylation states are highly labile - samples should be collected with phosphatase inhibitors and processed rapidly. Examine positive control samples from cells treated with known activators of the target pathway alongside negative controls treated with pathway inhibitors or phosphatases. For multiple or missing bands, consider potential degradation products, splice variants, or cross-reactivity with related phosphorylation sites. Validate the activation state of upstream kinases in your samples, as unexpected results may reflect genuine biological variation in signaling pathway activity. Test different lysis buffers, as some may better preserve phosphorylated epitopes. If signals appear weak, optimize transfer conditions specifically for the molecular weight of your phosphorylated target, as high molecular weight phospho-proteins may require longer transfer times. Finally, consider alternative detection methods such as Phos-tag SDS-PAGE, which can enhance separation of phosphorylated from non-phosphorylated protein forms .

What are the best practices for quantifying and normalizing antibody-based assay results for publication?

For publication-quality quantification of antibody-based assays, implement rigorous methodological standards beginning with appropriate replicate structure: minimum three biological replicates with 2-3 technical replicates each to capture both biological variation and technical reproducibility. For Western blots, use fluorescent secondary antibodies when possible, as they provide wider linear dynamic range than chemiluminescence. Quantify band intensities using software that integrates peak area rather than maximum intensity, and subtract local background values individually for each lane. Normalize target protein signals to validated loading controls appropriate for your experimental conditions (avoiding housekeeping proteins affected by your treatment). For immunohistochemistry quantification, define precise scoring parameters before analysis and ensure multiple trained observers score independently using standardized criteria. When reporting results, include representative original images alongside quantification, clearly state normalization methods, and provide appropriate statistical tests with exact p-values rather than thresholds. Include validation data demonstrating antibody specificity relevant to the experimental context, particularly for phospho-specific antibodies like anti-MK2 (phospho T334) .

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.