ERDJ2A Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Function of ERDJ2

ERDJ2 is encoded by the SEC63 gene and consists of 760 amino acids (88 kDa). It contains:

  • A J-domain for binding Hsp70 chaperones.

  • Three predicted transmembrane domains, though experimental data suggest only two span the ER membrane, forming a U-shaped conformation with cytosolic N- and C-termini and a luminal J-domain .

Applications of ERDJ2 Antibodies

ApplicationFindingsSource
Membrane TopologyConfirmed two transmembrane domains via proteinase K and antibody staining
Functional StudiessiRNA knockdown reduced translocation of substrates like ERdj3 and AQP2
Conformational AnalysisDetected high-molecular-weight GRP94-ERDJ2 complexes in inhibitor-sensitive cells

Role in Protein Translocation

ERDJ2 facilitates co-translational translocation of secretory proteins by recruiting BiP (Hsp70) to the Sec61 translocon. Its knockdown impairs the translocation of specific substrates, including aquaporin-2 and prion protein .

Pathological Implications

  • Polycystic Liver Disease: ERDJ2 (PCLD2) mutations are linked to autosomal dominant polycystic liver disease .

  • Cancer: ERDJ2 interacts with HER2 and EGFR in breast cancer cells, influencing receptor stability and signaling .

Challenges and Validation

Antibody specificity remains a critical concern. For example:

  • Cross-reactive antibodies against ER-β misled clinical trials for breast cancer .

  • ERDJ2 antibodies must distinguish between conformational states and avoid off-target binding to homologs like DNAJC23 .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
ERDJ2A antibody; C21 antibody; At1g79940 antibody; F18B13.2 antibody; F19K16.10 antibody; DnaJ protein ERDJ2A antibody; Chaperone protein dnaJ 21 antibody; AtDjC21 antibody; AtJ21 antibody; Endoplasmic reticulum dnaJ domain-containing protein 2A antibody; AtERdj2A antibody; Translocation protein SEC63 homolog ERDJ2A antibody
Target Names
ERDJ2A
Uniprot No.

Target Background

Function
Essential for the translocation of integral membrane and secreted preproteins across the endoplasmic reticulum membrane.
Database Links

KEGG: ath:AT1G79940

STRING: 3702.AT1G79940.1

UniGene: At.34014

Subcellular Location
Endoplasmic reticulum membrane; Multi-pass membrane protein.
Tissue Specificity
Expressed in leaves, flower buds and flowers.

Q&A

What is the ERDJ2A antibody and what epitopes does it target?

ERDJ2A antibody belongs to the family of antibodies that recognize specific epitopes on their target antigens. Similar to antibodies targeting the N-terminal domain (NTD) of proteins, ERDJ2A recognizes specific conformational epitopes that are crucial for its binding specificity. When working with this antibody, researchers should understand that epitope accessibility can significantly impact binding efficiency. The antibody-antigen interaction involves complex structural complementarity that determines specificity and affinity, which can be influenced by experimental conditions including pH, temperature, and buffer composition. Proper characterization of binding epitopes is essential for valid experimental interpretations and can be accomplished through epitope mapping techniques including hydrogen-deuterium exchange mass spectrometry, X-ray crystallography, or alanine scanning mutagenesis .

What validation assays should be performed to confirm ERDJ2A antibody specificity?

Rigorous validation of ERDJ2A antibody specificity requires multiple orthogonal approaches. As demonstrated with NTD-specific antibodies, cross-reactivity testing is essential to ensure target specificity. Researchers should implement ELISA assays with both target and structurally similar proteins to assess potential cross-reactivity, similar to approaches used for differentiating SARS-CoV-2 NTD and RBD antibodies. Western blotting with positive and negative control samples, including knockdown/knockout systems, provides another critical validation layer. Immunoprecipitation followed by mass spectrometry analysis can further confirm target specificity and identify potential off-target interactions. For in situ applications, immunohistochemistry or immunofluorescence should be performed with appropriate blocking controls to verify specific staining patterns .

How should sample preparation be optimized for ERDJ2A antibody-based detection methods?

Optimal sample preparation is critical for reliable ERDJ2A antibody-based detection. Researchers should consider that protein conformation significantly impacts epitope accessibility. For fixed tissue or cell samples, fixation method and duration directly affect epitope preservation—paraformaldehyde generally maintains better epitope integrity compared to harsher fixatives like glutaraldehyde. Antigen retrieval methods (heat-induced or enzymatic) should be systematically optimized for each application. For protein extracts, the lysis buffer composition requires careful consideration, as detergents may disrupt protein conformation and epitope accessibility. Additionally, researchers should implement proper blocking procedures to minimize non-specific binding, typically using 3-5% BSA or serum from a species different from that in which the primary antibody was raised .

What controls are essential when using ERDJ2A antibody in immunoassays?

Comprehensive controls are fundamental for interpreting ERDJ2A antibody results. Every experiment should include positive controls (samples known to express the target) and negative controls (samples known not to express the target). Secondary antibody-only controls are necessary to assess background signal. For quantitative analyses, standard curves using purified recombinant protein should be included to establish detection limits and linear range. When feasible, genetic knockout/knockdown samples provide gold-standard negative controls. Similar to approaches used in coronavirus serology testing, isotype controls help distinguish specific binding from non-specific interactions. For multiplexed assays, single-stain controls are essential to establish spectral compensation and assess potential cross-reactivity among detection systems .

How can computational models predict ERDJ2A antibody binding specificity and cross-reactivity?

Advanced computational modeling has revolutionized our understanding of antibody specificity. For ERDJ2A and similar antibodies, biophysically interpretable models can disentangle multiple binding modes associated with specific ligands. These models, trained on experimentally selected antibodies, associate distinct binding modes with each potential ligand, enabling the prediction of specificity profiles beyond experimentally tested variants. Similar to the approach described by researchers working with phage display selections, a biophysics-informed model can identify contributions to binding from multiple epitopes even within a single experiment. This computational approach involves training on sequence-function relationships derived from high-throughput selection experiments against diverse combinations of related ligands. The resulting models can successfully predict binding profiles to new ligand combinations and even generate novel antibody variants with customized specificity profiles not present in the original training datasets .

What strategies can resolve contradictory ERDJ2A antibody binding data from different experimental platforms?

Resolving contradictory binding data requires systematic investigation of platform-specific variables. Researchers should first examine fundamental differences in antigen presentation across platforms—solution-phase methods (SPR, BLI) versus solid-phase assays (ELISA) can yield different results due to epitope accessibility and protein conformation differences. Similar to challenges faced with coronavirus antibodies, epitope masking or conformational changes may occur differentially between platforms. Concentration-dependent effects should be evaluated through careful titration experiments, as high antibody concentrations may reveal secondary, lower-affinity binding sites. Environmental factors (pH, ionic strength, temperature) should be standardized across platforms when possible. For truly contradictory data, orthogonal validation using functional assays or in vivo models becomes essential. Additionally, different antibody lots should be tested to rule out batch-to-batch variability as a source of discrepancy .

How can phage display technology be leveraged to engineer ERDJ2A antibody variants with enhanced specificity?

Phage display technology offers powerful approaches for engineering antibody specificity. For ERDJ2A and similar antibodies, researchers can implement selection strategies against multiple related ligands to identify variants with desired specificity profiles. As demonstrated in recent research, conducting parallel selections against different combinations of related ligands allows the identification of sequence features that confer specificity for particular epitopes. This approach can be enhanced through computational modeling that disentangles binding modes associated with different ligands. The process typically involves:

  • Creating a diverse antibody library with randomized complementarity-determining regions (CDRs)

  • Performing selections against the target of interest and structurally similar molecules

  • Using next-generation sequencing to analyze enriched sequences

  • Applying computational models to identify specificity-determining residues

  • Designing new variants with customized specificity profiles

This integrated experimental-computational approach enables the design of antibodies with either highly specific binding to a single target or controlled cross-reactivity across multiple targets .

What are the optimal experimental designs for characterizing ERDJ2A antibody binding kinetics and thermodynamics?

Comprehensive characterization of binding kinetics and thermodynamics requires complementary methodologies. Surface Plasmon Resonance (SPR) provides real-time, label-free measurement of association (kon) and dissociation (koff) rate constants, from which equilibrium dissociation constants (KD) can be derived. For ERDJ2A antibody, researchers should implement multi-cycle kinetics with at least five antibody concentrations spanning 0.1-10× the expected KD value. Bio-Layer Interferometry (BLI) offers similar kinetic data with potentially higher throughput. Isothermal Titration Calorimetry (ITC) provides direct measurement of binding thermodynamics (ΔH, ΔS, ΔG), offering insights into the energetic components driving interaction. Microscale Thermophoresis (MST) can be valuable for studying interactions in solution with minimal sample consumption. For all methods, researchers should carefully consider buffer conditions, antigen immobilization strategies, and potential mass transport limitations. Analysis should incorporate appropriate binding models (1:1, bivalent, heterogeneous ligand) based on the expected interaction mechanism .

How should ERDJ2A antibody be implemented in multiplex immunoassays to maintain specificity?

Implementing ERDJ2A antibody in multiplex assays requires careful optimization to preserve specificity. Cross-reactivity must be systematically evaluated against all other targets in the multiplex panel through single-antibody control experiments. Buffer composition should be optimized to minimize non-specific interactions while maintaining proper antibody folding and epitope recognition. Similar to approaches used in coronavirus serology testing, titration experiments should determine the optimal antibody concentration that maximizes specific signal while minimizing background. For fluorescence-based multiplex assays, spectral overlap must be addressed through proper compensation controls. The order of antibody addition can significantly impact results, especially for sequential staining protocols. When designing multiplex panels, researchers should consider potential steric hindrance between antibodies targeting epitopes in close proximity. Each new combination of antibodies requires validation to ensure that multiplexing doesn't alter individual antibody performance compared to single-target assays .

What factors influence ERDJ2A antibody performance in different tissue types and fixation methods?

Tissue-specific and fixation-dependent factors significantly impact ERDJ2A antibody performance. Different tissues possess unique extracellular matrices, lipid compositions, and endogenous peroxidase/phosphatase activities that can affect antibody penetration and background signal. The table below summarizes key considerations for optimizing antibody performance across common tissue preparations:

Fixation MethodEpitope PreservationRecommended Antigen RetrievalPenetration DepthOptimal Incubation
Fresh-frozenExcellentMinimal/NoneExcellentShorter (1-2h, 4°C)
Paraformaldehyde (4%)GoodHeat-induced (citrate buffer)GoodStandard (overnight, 4°C)
FormalinModerateHeat-induced (EDTA buffer)ModerateExtended (24-48h, 4°C)
GlutaraldehydePoorEnzymatic (proteinase K)LimitedExtended with higher concentration

Researchers should systematically evaluate these parameters for each new tissue type. Additionally, endogenous biotin blocking is essential for avidin-biotin detection systems, and endogenous peroxidase quenching is critical for HRP-based visualization. Tissue-specific autofluorescence should be addressed through appropriate quenching protocols for fluorescence-based detection .

How can ERDJ2A antibody be effectively incorporated into antibody-drug conjugate (ADC) development?

Incorporating ERDJ2A antibody into ADC development requires systematic evaluation of multiple parameters. Like trastuzumab deruxtecan and other clinically successful ADCs, researchers must first confirm that antibody binding doesn't significantly decrease after conjugation—a process requiring comparative binding assays with conjugated and unconjugated antibodies. The drug-to-antibody ratio (DAR) must be precisely controlled and characterized, typically using hydrophobic interaction chromatography or mass spectrometry. Conjugation site selection is critical, as random conjugation can impair binding and produce heterogeneous products. Site-specific conjugation technologies (engineered cysteines, non-natural amino acids, enzymatic approaches) enable more homogeneous ADCs with improved in vivo properties. Linker stability should be evaluated under physiological conditions to ensure proper drug release only at the target site. Cytotoxicity assays must include target-negative cell lines to confirm specificity, and in vivo pharmacokinetic studies should assess stability, tissue distribution, and therapeutic index .

What approaches can determine if ERDJ2A antibody-mediated effects are epitope-specific or Fc-mediated?

Distinguishing epitope-specific from Fc-mediated effects requires targeted experimental approaches. Researchers should generate F(ab')2 fragments by pepsin digestion or recombinant Fab fragments that lack the Fc region, then compare their activity to the intact antibody. If the biological effect persists with the Fab/F(ab')2, it is likely epitope-mediated. Fc receptor blocking experiments using anti-FcR antibodies or recombinant soluble FcR can further clarify the role of Fc-mediated functions. Engineering antibodies with mutations in the Fc region that selectively disable specific Fc functions (e.g., ADCC-null or CDC-null variants) provides another approach. For in vivo studies, comparing ERDJ2A antibody effects in wildtype versus FcR-knockout animals can definitively establish Fc-dependence. Additionally, comparing the original antibody with variants possessing identical variable regions but different Fc isotypes (IgG1, IgG2, IgG4) helps clarify which Fc-mediated functions contribute to observed effects, as these isotypes engage different Fc receptors with varying efficiencies .

How should researchers differentiate between specific and non-specific binding in ERDJ2A antibody applications?

Differentiating specific from non-specific binding requires rigorous analytical approaches. Competitive binding assays with excess unlabeled antibody or purified antigen can demonstrate binding specificity—specific binding should be competitively inhibited while non-specific binding remains unaffected. Researchers should analyze binding across a concentration range, as specific binding typically saturates while non-specific binding increases linearly with concentration. Similar to approaches used in coronavirus serology testing, binding to knockout/knockdown samples helps establish background signal levels attributable to non-specific interactions. Comparing binding patterns across multiple antibodies targeting different epitopes on the same protein can confirm specific recognition. For imaging applications, co-localization with other established markers provides additional validation. Statistical approaches including Scatchard analysis for equilibrium binding data help distinguish specific binding components from non-specific interactions. When analyzing ELISA data, researchers should calculate signal-to-background ratios and establish cutoff values based on negative control distributions .

What statistical approaches are most appropriate for analyzing ERDJ2A antibody binding across experimental replicates?

Robust statistical analysis of antibody binding data requires consideration of data structure and experimental design. For comparing binding across conditions, researchers should first assess data normality using Shapiro-Wilk or Kolmogorov-Smirnov tests. For normally distributed data, parametric tests (t-test, ANOVA with appropriate post-hoc tests) are appropriate. Non-normal distributions require non-parametric alternatives (Mann-Whitney, Kruskal-Wallis). When analyzing dose-response relationships, four-parameter logistic regression models typically provide better fits than linear models. For binding kinetics, model selection should be guided by Akaike Information Criterion (AIC) or Bayesian Information Criterion (BIC) to determine whether simple (1:1) or complex (heterogeneous ligand, bivalent analyte) binding models are most appropriate. Statistical power analysis should guide sample size determination, typically aiming for 80% power to detect biologically relevant effect sizes. For all analyses, researchers should report confidence intervals and effect sizes rather than just p-values. Hierarchical or mixed-effects models should be employed when analyzing data with nested structures (e.g., technical replicates within biological replicates) .

How can researchers integrate ERDJ2A antibody binding data with other -omics datasets for systems-level analysis?

Integrating antibody binding data with other -omics datasets requires sophisticated computational approaches. Correlation analysis between binding profiles and transcriptomic/proteomic data can reveal molecular mechanisms associated with antibody-target interactions. Network analysis tools (WGCNA, Bayesian networks) help place antibody binding in broader biological contexts by identifying co-regulated gene/protein modules. Researchers can employ dimensionality reduction techniques (PCA, t-SNE, UMAP) to visualize relationships between antibody binding and complex multi-dimensional -omics datasets. Pathway enrichment analysis using repositories like KEGG or Reactome can identify biological processes associated with differential antibody binding. Machine learning approaches, particularly supervised methods (random forests, support vector machines), can identify molecular signatures that predict antibody binding patterns. For temporal datasets, dynamic network analysis helps understand how antibody-target interactions evolve over time. Integration platforms like MultiAssayExperiment (R/Bioconductor) or similar Python frameworks facilitate the technical aspects of data integration. Researchers should implement proper batch correction methods when integrating datasets from different experiments or platforms .

What approaches can predict and mitigate immunogenicity risks with ERDJ2A antibody therapeutics?

Predicting and mitigating immunogenicity requires integrated computational and experimental strategies. In silico approaches using T-cell epitope prediction algorithms (NetMHCpan, IEDB) can identify potential immunogenic sequences within the antibody. These computational predictions should be validated through ex vivo T-cell assays using peripheral blood mononuclear cells (PBMCs) from diverse donors. Deimmunization strategies include removing T-cell epitopes through careful mutagenesis, framework "humanization," or using fully human antibody frameworks. Surface exposure analysis can identify regions accessible to immune surveillance that might benefit from deimmunization. Researchers should evaluate post-translational modifications, particularly non-human glycosylation patterns that may be immunogenic. Aggregation propensity should be assessed and minimized through stability engineering, as aggregates can enhance immunogenicity. Formulation optimization can further reduce aggregation risk during storage and administration. For clinical development, judicious immunogenicity monitoring protocols should include validated assays for anti-drug antibodies, with careful timing of sample collection to capture developing immune responses .

How can ERDJ2A antibody be engineered for optimized tissue penetration in solid tumors?

Engineering ERDJ2A antibody for enhanced tumor penetration requires addressing several biophysical barriers. Size-reduction strategies, including single-chain variable fragments (scFvs), Fabs, and nanobodies, can improve diffusion through tumor tissue compared to full IgG molecules. Affinity modulation presents a counterintuitive approach—extremely high-affinity antibodies often exhibit "binding site barrier" effects where strong binding to peripheral tumor cells prevents deeper penetration. Researchers have demonstrated that moderate-affinity variants can achieve more homogeneous tumor distribution. Charge modifications offer another strategy, as positively charged antibodies generally demonstrate better tissue penetration due to interactions with negatively charged extracellular components. Glycoengineering, particularly deglycosylation or specific glycoform selection, can influence pharmacokinetics and tissue distribution. Additionally, combination approaches targeting the tumor microenvironment (collagenase co-administration, TGF-β inhibition) can enhance antibody penetration by reducing stromal barriers. The table below summarizes engineering strategies and their impact:

Engineering ApproachMechanismPenetration ImpactTrade-offs
Size reduction (scFv, Fab)Enhanced diffusion+++++Shorter half-life, loss of Fc functions
Affinity modulation (lower KD)Reduced binding site barrier++++Potentially reduced target engagement
Positive charge engineeringImproved tissue interaction+++Increased clearance, kidney accumulation
GlycoengineeringAltered interactions with ECM++Potential immunogenicity
pH-sensitive bindingRecycling in tumor microenvironment+++Complex engineering required

Testing these approaches requires advanced imaging methods including intravital microscopy or MALDI imaging mass spectrometry to visualize antibody distribution in tumor tissues .

How might computational antibody design transform ERDJ2A antibody engineering for novel therapeutic applications?

Computational antibody design represents a transformative approach for ERDJ2A engineering. Recent advances in biophysically interpretable models enable the prediction and generation of antibody variants with tailored specificity profiles beyond what can be directly selected experimentally. As demonstrated in phage display experiments, these models can disentangle multiple binding modes associated with specific ligands, even when they are chemically very similar. Future applications include developing bispecific antibodies with precisely controlled affinity ratios for multiple targets, or creating antibody panels with graduated specificity across related antigens. Machine learning approaches trained on extensive antibody-antigen structural databases can now predict binding interfaces and engineer complimentary binding surfaces. Recent integration of protein language models like ESM-2 with structure prediction tools further enhances design capabilities by allowing sequence-based prediction of antibody properties. The computational approach can significantly reduce experimental screening requirements by enriching candidate libraries for likely successful variants. As these methods mature, they will enable rapid development of ERDJ2A variants with novel binding properties, unusual cross-reactivity profiles, or enhanced stability characteristics unattainable through traditional directed evolution approaches .

What emerging technologies will enhance our understanding of ERDJ2A antibody epitope recognition and binding dynamics?

Emerging technologies are revolutionizing antibody epitope characterization at unprecedented resolution. Single-molecule FRET (smFRET) enables direct observation of antibody-antigen binding dynamics, revealing transient conformational states invisible to bulk methods. Cryo-electron microscopy (cryo-EM) now achieves near-atomic resolution of antibody-antigen complexes without crystallization requirements, significantly expanding the range of complexes that can be structurally characterized. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) with improved temporal resolution can map epitopes while capturing conformational changes upon binding. Advanced surface plasmon resonance (SPR) implementations including SPR microscopy enable spatial mapping of binding events across heterogeneous surfaces. DNA-barcoded antibody libraries combined with next-generation sequencing allow massively parallel screening of binding properties across millions of variants simultaneously. Microfluidic systems integrating binding measurements with single-cell phenotyping connect antibody properties to functional outcomes. The integration of artificial intelligence with these experimental platforms is creating "closed-loop" systems that autonomously design, test, and refine antibodies through iterative learning. These technologies will provide unprecedented insights into the molecular basis of ERDJ2A binding specificity and its relationship to biological function .

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.