DOF4.4 Antibody

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
14-16 week lead time (made-to-order)
Synonyms
DOF4.4 antibody; At4g21050 antibody; T13K14.210Dof zinc finger protein DOF4.4 antibody; AtDOF4.4 antibody
Target Names
DOF4.4
Uniprot No.

Target Background

Function
DOF4.4 is a transcription factor that exhibits specific binding affinity for the 5'-AA[AG]G-3' consensus core sequence.
Database Links

KEGG: ath:AT4G21050

STRING: 3702.AT4G21050.1

UniGene: At.54450

Subcellular Location
Nucleus.

Q&A

What is the DRD4 antibody and what is its role in vision research?

The dopamine receptor D4 (DRD4) antibody is an essential research tool used to detect and visualize the expression of DRD4 in tissue samples, particularly in vision research. DRD4 plays a crucial role in visual function, making reliable antibodies critical for investigating its expression patterns . These antibodies typically target specific peptide sequences of the DRD4 receptor and are designed to bind with high specificity.

For vision researchers, DRD4 antibodies enable the study of dopaminergic signaling in the retina through techniques like immunohistochemistry (IHC) and immunoblotting. When selecting a DRD4 antibody for vision research, it's essential to choose one validated specifically in retinal tissue. According to published studies, only certain antibodies demonstrate sufficient specificity and sensitivity for retinal DRD4 detection—for example, the N-20 antibody has shown effectiveness in both immunoblot analysis of DRD4-transfected cells and IHC of mouse retinal sections .

How can researchers validate antibodies for specific research applications?

Antibody validation requires a multi-step approach to ensure specificity and reliability for intended applications:

Validation Methodology Table for Research Antibodies:

Validation MethodDescriptionAdvantagesRecommended Controls
Transfection TestingExpress target protein in mammalian cellsConfirms binding to target in cellular contextEmpty vector transfection
Application-specific TestingTest antibody in intended applications (IHC, WB)Confirms utility in specific methodologiesPositive and negative tissue samples
Cross-reactivity AnalysisTest against similar proteinsIdentifies potential off-target bindingRelated protein panels
Multiple Antibody ComparisonTest several antibodies targeting different epitopesConfirms expression patternsComparison across antibodies

For DRD4 antibodies specifically, validation should include:

  • In vitro testing using transfected mammalian cells expressing DRD4

  • Application-specific testing in both immunoblot and IHC applications

  • Negative controls using non-transfected cells or DRD4-negative tissues

  • Comparative analysis between multiple antibodies targeting different DRD4 epitopes

As demonstrated in published research, even commercially available antibodies can show significant variation in specificity and application utility. For example, when six different antibodies raised against DRD4 peptides were tested, only a subset were successful in IHC of transfected cells, and only one (N-20) demonstrated effectiveness across multiple applications .

What critical factors affect antibody performance in immunohistochemistry?

Multiple technical factors can significantly impact antibody performance in immunohistochemical applications:

  • Fixation Method: The type and duration of fixation can preserve or mask epitopes. For DRD4 detection in retinal tissue, paraformaldehyde fixation protocols must be carefully optimized to maintain receptor structure while allowing antibody access.

  • Antigen Retrieval: Heat-induced or enzymatic antigen retrieval may be necessary to expose epitopes altered by fixation. Different DRD4 antibodies may require specific retrieval methods based on their epitope targets.

  • Antibody Concentration: Titration experiments are essential to determine optimal antibody dilutions that maximize specific signal while minimizing background. For DRD4 antibodies, dilutions typically range from 1:500 to 1:2000 for immunoblotting applications .

  • Incubation Conditions: Temperature, duration, and buffer composition during antibody incubation can affect binding kinetics and specificity.

  • Detection Systems: The choice between direct fluorescence, enzyme-based detection, or amplification systems impacts sensitivity.

  • Tissue Preparation: Section thickness, permeabilization protocols, and blocking methods significantly influence antibody penetration and non-specific binding.

Researchers should systematically optimize each of these parameters when establishing IHC protocols for DRD4 detection. Documentation of a validated antibody's optimal working conditions is essential for reproducible results.

How can researchers effectively characterize antibody specificity for receptors like DRD4?

Characterizing antibody specificity for G protein-coupled receptors (GPCRs) like DRD4 requires a comprehensive approach that addresses multiple aspects of antibody-target interactions:

Multi-dimensional Specificity Analysis:

  • Expression System Validation: Testing in multiple expression systems provides robust evidence of specificity. For DRD4 antibodies, this includes:

    • Transiently transfected HEK293 cells with controlled DRD4 expression

    • Stable cell lines expressing physiological levels of DRD4

    • Native tissue samples with known DRD4 expression patterns (e.g., retina)

  • Knockout/Knockdown Controls: Utilizing genetic approaches to eliminate or reduce target expression:

    • CRISPR/Cas9-mediated knockout cells for complete DRD4 elimination

    • siRNA knockdown for partial reduction of DRD4 expression

    • Tissue from DRD4 knockout animals as gold-standard negative controls

  • Cross-family Specificity Testing: Evaluating binding to related dopamine receptors:

    • Testing against cells expressing DRD1, DRD2, DRD3, and DRD5

    • Epitope sequence comparison across dopamine receptor family

    • Competitive binding assays with peptides from related receptors

  • Multiple Application Assessment: Evaluating performance across diverse methodologies:

    • Western blot under reducing and non-reducing conditions

    • Immunoprecipitation to verify native protein recognition

    • Flow cytometry for cell-surface DRD4 detection

    • Immunohistochemistry with multiple fixation protocols

Published research demonstrates that comprehensive validation is essential—among six DRD4 antibodies tested, three Santa Cruz antibodies (D-16, N-20, and R-20) successfully detected transfected DRD4 in IHC, but only N-20 showed effectiveness across multiple applications, including immunoblotting and IHC of mouse retinal sections .

What strategies can improve antibody validation in complex tissue samples?

Validating antibodies in complex tissues presents unique challenges compared to cell culture systems. Advanced strategies to enhance confidence in tissue-specific antibody validation include:

  • Orthogonal Validation: Confirming protein expression using antibody-independent methods:

    • mRNA analysis (RT-PCR, RNA-Seq, in situ hybridization)

    • Mass spectrometry-based proteomics

    • Reporter gene expression in transgenic animals

  • Spatial Resolution Analysis: Comparing subcellular localization patterns:

    • Super-resolution microscopy to confirm expected receptor distribution

    • Co-localization with known interaction partners

    • Fractionation studies to verify membrane association of DRD4

  • Functional Correlation: Linking antibody staining with functional assays:

    • Coupling immunostaining with electrophysiological recordings

    • Correlating staining intensity with receptor signaling activity

    • Verifying changes in staining patterns following pharmacological manipulation

  • Multiplex Validation: Using multiple antibodies against different epitopes:

    • Confirmation of staining patterns using independently-derived antibodies

    • Epitope mapping to understand which receptor domains are accessible in tissue

    • Competitive binding assays to verify epitope specificity

  • Species Cross-reactivity Assessment: Comparing detection across evolutionary related species:

    • Testing in tissues from multiple species with conserved DRD4 sequences

    • Aligning epitope sequences across species to predict cross-reactivity

For DRD4 specifically, researchers should consider the receptor's native conformation in neuronal and retinal tissues, where post-translational modifications and protein-protein interactions may affect epitope accessibility.

How can active learning approaches optimize antibody-antigen binding experiments?

Active learning strategies represent a cutting-edge approach to optimize experimental design and resource allocation in antibody research. These methods can significantly improve efficiency in binding assays:

Active Learning Implementation for Antibody Research:

  • Iterative Experimental Design: Active learning begins with a small subset of labeled data and strategically selects the most informative additional experiments:

    • Using initial binding data to identify patterns and areas of uncertainty

    • Selecting experimental conditions that maximize information gain

    • Iteratively refining predictions with each new experimental dataset

  • Uncertainty-based Sampling: Prioritizing experiments in regions of highest prediction uncertainty:

    • Testing antibody-antigen pairs where binding outcomes are least certain

    • Focusing resources on boundary cases between binding and non-binding

    • Reducing redundant testing of clearly positive or negative interactions

  • Model-guided Experimentation: Using machine learning predictions to direct laboratory work:

    • Developing predictive models from initial binding data

    • Updating models as new data becomes available

    • Directing experiments toward conditions that improve model performance

Recent research has demonstrated that active learning approaches can significantly reduce experimental costs in antibody-antigen binding studies. In a library-on-library setting, the best active learning algorithms reduced the number of required antigen mutant variants by up to 35% and accelerated the learning process by 28 steps compared to random selection approaches .

Comparative Performance of Active Learning Methods:

Active Learning StrategyEfficiency ImprovementKey AdvantagesBest Application Scenario
Uncertainty Sampling20-35% reduction in experimentsSimple implementationInitial screening phases
Diversity-based Selection15-25% reduction in experimentsBroader explorationNovel epitope discovery
Model-based Active Learning25-35% reduction in experimentsHighest performanceComplex binding landscapes

These approaches are particularly valuable for out-of-distribution prediction scenarios, where test antibodies and antigens differ from training data—a common challenge in antibody research .

How can researchers troubleshoot cross-reactivity issues with antibodies targeting specific receptor subtypes?

Cross-reactivity remains one of the most challenging aspects of antibody-based receptor subtype detection. A systematic troubleshooting approach includes:

  • Epitope Analysis: Examine the sequence homology between the target receptor (e.g., DRD4) and related subtypes:

    • Align amino acid sequences of dopamine receptor subtypes

    • Identify regions of high conservation that may lead to cross-reactivity

    • Select antibodies targeting unique regions or splice variants

  • Absorption Controls: Pre-incubate antibodies with peptide antigens:

    • Use the immunizing peptide to verify specific binding

    • Test absorption with peptides from related receptors

    • Quantify the degree of signal reduction following absorption

  • Titration Optimization: Establish the minimal effective concentration:

    • Perform systematic dilution series (e.g., 1:500-1:2000 for immunoblotting )

    • Identify concentrations that maximize specific:non-specific signal ratios

    • Document optimal conditions for each application

  • Detection Method Refinement: Modify secondary detection systems:

    • Test alternative detection antibodies with different host species

    • Compare enzyme-based versus fluorescence-based detection

    • Implement signal amplification for weak but specific signals

  • Alternative Antibody Formats: Consider using different antibody types:

    • Compare polyclonal versus monoclonal antibodies

    • Evaluate different antibody isotypes (IgG vs. IgM)

    • Test recombinant antibody fragments with engineered specificity

For DRD4 antibodies specifically, published research indicates significant variation in specificity—among six tested antibodies, several showed non-specific binding or below-detection performance in retinal tissues despite manufacturer claims of specificity .

What methodological approaches can improve antibody performance in challenging applications?

When working with difficult targets or applications, several methodological refinements can significantly improve antibody performance:

  • Sample Preparation Optimization:

    • For membrane proteins like DRD4, use gentle detergents that preserve native conformation

    • Implement non-denaturing conditions when possible for conformational epitopes

    • Consider native-PAGE for immunoblotting GPCRs like DRD4

    • Optimize fixation protocols (duration, temperature, fixative composition)

  • Signal Enhancement Strategies:

    • Implement tyramide signal amplification for low-abundance targets

    • Use biotin-streptavidin amplification systems

    • Consider proximity ligation assays for improved specificity and sensitivity

    • Optimize antigen retrieval methods specifically for the target epitope

  • Blocking Optimization:

    • Test different blocking agents (BSA, casein, normal sera)

    • Implement dual-blocking with protein and detergent combinations

    • Use antibody diluents with background-reducing components

    • Pre-absorb secondary antibodies with tissue homogenates

  • Protocol Modifications for Specific Applications:

    For Immunohistochemistry:

    • Extend primary antibody incubation time (overnight at 4°C)

    • Use thinner tissue sections for improved antibody penetration

    • Implement two-step fixation protocols for membrane proteins

    For Immunoblotting:

    • Test different membrane transfer conditions (wet vs. semi-dry)

    • Optimize protein loading concentrations

    • Consider native vs. reducing conditions for conformation-dependent epitopes

Published research demonstrates the importance of application-specific validation—for DRD4 antibodies, some performed well in immunohistochemistry but failed in immunoblotting, highlighting the need for comprehensive testing across all intended applications .

What are the current methodological approaches for engineering antibodies with improved specificity?

Antibody engineering has evolved significantly, offering researchers powerful tools to improve specificity for challenging targets like DRD4:

  • Directed Evolution Approaches:

    • Phage display selection under stringent conditions to isolate highly specific binders

    • Yeast display systems allowing for fluorescence-activated cell sorting (FACS)

    • Bacterial display platforms for rapid screening

    • Mammalian display systems for complex epitopes requiring post-translational modifications

  • Rational Design Strategies:

    • Structure-guided mutagenesis of complementarity-determining regions (CDRs)

    • Computational modeling of antibody-antigen interfaces

    • Humanization of antibody frameworks while preserving binding specificity

    • Framework modifications to improve biophysical properties

  • Affinity Maturation Techniques:

    • Error-prone PCR to generate CDR variants

    • Site-directed mutagenesis of key binding residues

    • CDR shuffling between related antibodies

    • Affinity selection under increasingly stringent conditions

  • Fragment-based Approaches:

    • Generation of single-domain antibodies with high specificity

    • Creation of bispecific antibodies targeting unique epitope combinations

    • Development of antibody fragments (Fab, scFv) with improved tissue penetration

    • Engineering of multivalent constructs for avidity enhancement

Current antibody engineering relies heavily on understanding structure-function relationships. The immunoglobulin fold, characterized by tightly packed anti-parallel β-sheets, provides the structural framework for engineering efforts . CDRs, particularly the hypervariable loops CDR-H3, offer the primary targets for specificity engineering .

What methodological considerations are important when using antibodies to study post-translational modifications?

Studying post-translational modifications (PTMs) of receptors like DRD4 requires specialized antibody approaches:

  • Modification-specific Antibody Generation:

    • Design immunogens containing the exact modified residue (phosphorylation, glycosylation, etc.)

    • Use carrier proteins that don't interfere with the modification epitope

    • Implement negative selection strategies to eliminate antibodies recognizing the unmodified sequence

    • Validate using synthetic peptides with and without modifications

  • Enrichment Strategies for Low-abundance Modified Proteins:

    • Use immunoprecipitation with modification-specific antibodies prior to detection

    • Implement affinity chromatography for enrichment

    • Consider chemical labeling approaches to enhance detection sensitivity

    • Use phosphatase or glycosidase treatments as negative controls

  • Validation Requirements for PTM-specific Antibodies:

    • Demonstrate absence of signal after enzymatic removal of the modification

    • Show correlation between signal intensity and biochemical quantification of the modification

    • Verify specificity using synthetic peptides containing the modification at different sites

    • Confirm biological relevance through pharmacological manipulation of the modification

  • Technical Considerations for Different Modifications:

    For Phosphorylation:

    • Include phosphatase inhibitors throughout sample preparation

    • Use phosphatase treatments as controls

    • Consider the impact of fixation on phospho-epitope preservation

    For Glycosylation:

    • Evaluate the impact of deglycosylating enzymes

    • Consider native vs. denatured conditions for complex glycan structures

    • Address potential steric hindrance from bulky glycan structures

  • Multiplex Detection Approaches:

    • Combine antibodies recognizing total protein and modified forms

    • Use different fluorophores to visualize distinct modifications simultaneously

    • Implement sequential probing strategies for multiple modifications

    • Consider proximity ligation assays for co-occurrence of modifications

These methodological considerations are particularly important for receptors like DRD4, where phosphorylation and glycosylation can significantly impact ligand binding, signaling, and subcellular localization.

How might advances in recombinant antibody technology impact DRD4 research?

The recombinant antibody field is rapidly evolving, offering new opportunities for DRD4 research:

  • Synthetic Antibody Libraries:

    • Generation of fully human antibodies without animal immunization

    • Creation of specialized libraries targeting GPCR-specific structural features

    • Development of conformation-specific antibodies for active vs. inactive DRD4 states

    • Engineering of antibodies recognizing specific receptor-ligand complexes

  • Single-domain Antibodies (Nanobodies):

    • Improved access to cryptic epitopes due to small size

    • Enhanced penetration of tight tissue junctions for in vivo imaging

    • Potential for intracellular expression to study DRD4 trafficking

    • Generation of conformation-specific nanobodies as research tools

  • Site-specific Conjugation Technologies:

    • Precise attachment of fluorophores or enzymes at defined positions

    • Development of homogeneous antibody-drug conjugates for targeted approaches

    • Creation of bifunctional reagents combining detection and modification capabilities

    • Orientation-controlled immobilization for biosensor applications

  • Engineered Fc Domains:

    • Silent Fc regions for applications requiring minimal effector function

    • Extended half-life variants for in vivo applications

    • pH-dependent binding for improved intracellular targeting

    • Engineered cross-species reactivity for translational research

These technologies promise to address current limitations in DRD4 research by providing more specific, consistent, and versatile reagents. The recombinant nature ensures batch-to-batch consistency and eliminates animal immunization variability, which has been a significant challenge in traditional antibody development.

What methodological innovations are improving reproducibility in antibody-based experiments?

Several methodological innovations are addressing the reproducibility crisis in antibody research:

  • Standardized Validation Criteria:

    • Implementation of application-specific validation requirements

    • Adoption of minimal reporting standards for antibody characteristics

    • Generation of validation packages with positive and negative controls

    • Development of standard reference materials for antibody benchmarking

  • Recombinant Antibody Advantages:

    • Sequence-defined reagents with eliminated batch-to-batch variation

    • Renewable source independent of animal immunization

    • Ability to engineer desired properties (affinity, specificity, stability)

    • Consistent performance across applications and over time

  • Open Science Initiatives:

    • Public antibody validation resources and databases

    • Community-based validation efforts sharing raw data

    • Pre-registration of antibody validation protocols

    • Transparent reporting of negative results and limitations

  • Advanced Analytical Approaches:

    • Quantitative image analysis replacing subjective interpretation

    • Statistical methods for distinguishing specific from non-specific signals

    • Multiplex detection systems for internal validation

    • Machine learning algorithms for pattern recognition in staining profiles

  • Reproducibility-focused Experimental Design:

    • Implementation of blinding procedures

    • Inclusion of appropriate positive and negative controls

    • Replicate experiments across different lots of antibodies

    • Multi-laboratory validation of critical findings

Published research highlights the importance of these approaches—among six antibodies raised against DRD4 peptides, significant variation in specificity and application performance was observed, underscoring the need for comprehensive validation .

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.