ADF3 Antibody

Shipped with Ice Packs
In Stock

Description

Contextual Overview of ADF3 Antibody

The term "ADF3" may refer to Actin-Depolymerizing Factor 3 (ADF3) or relate to antibodies targeting the DF3 antigen, a mammary epithelial glycoprotein.

Key Distinctions:

  • ADF3: Associated with actin cytoskeleton regulation. The Destrin/ADF antibody (Sigma-Aldrich D8815) binds to G-actin and filamentous actin, influencing cellular motility and remodeling .

  • DF3 Antigen: A high-molecular-weight glycoprotein (300–400 kDa) detected by murine monoclonal antibody DF3, used as a biomarker in breast cancer diagnostics .

Table 1: Comparative Antibody Profiles

ParameterDestrin/ADF Antibody (D8815) DF3 Antigen-Targeting Antibody
TargetActin-depolymerizing factor (Destrin)DF3 glycoprotein (MUC1 epitope)
Molecular Weight~19 kDa (Destrin protein)300,000–400,000 Da (DF3 antigen)
ApplicationsWestern blot, immunofluorescenceCancer biomarker assays (RIA, EIA, transblot)
Clinical RelevanceCytoskeletal dynamics in cancer cellsMetastatic breast cancer monitoring
Gene AssociationHuman DSTN, mouse DstnMUC1 gene-encoded antigen

A. Destrin/ADF Antibody (D8815)

  • Mechanism: Binds stoichiometrically to G-actin and filamentous actin, disrupting helical twist and promoting cytoskeletal remodeling .

  • Functional Role: Critical for processes like endocytosis, phagocytosis, and cytokinesis.

  • Experimental Use: Detected in models including human A-431 epidermoid carcinoma and rat PC-12 cells at 1:1,500 dilution (western blot) .

B. DF3 Antigen-Targeting Antibodies

  • Diagnostic Utility: Elevated DF3 antigen levels (>150 U/ml via RIA) correlate with metastatic breast cancer (76% sensitivity) .

  • Specificity: Distinguishes breast cancer from gastrointestinal/pancreatic malignancies but cross-reacts with hepatoma (27%) and ovarian carcinoma (47%) .

  • Therapeutic ADC Potential: DF3-targeting antibody-drug conjugates (ADCs) are under exploration for solid tumors .

Emerging Applications in Oncology

  • ADCs Targeting DF3: Third-generation ADCs (e.g., fam-trastuzumab deruxtecan) use site-specific conjugation for homogeneous drug-antibody ratios (DAR 2–4), enhancing tumor penetration and reducing off-target toxicity .

  • Clinical Trials: Phase 3 LOTIS-5 trial (NCT06092455) evaluates ADCs like loncastuximab tesirine in relapsed diffuse large B-cell lymphoma, with results expected by 2025 .

Data Gaps and Future Directions

  • ADF3 Specificity: No direct studies on "ADF3 Antibody" were identified; further validation is required to clarify its target and therapeutic potential.

  • Technical Advances: Next-generation ADCs prioritize miniaturized antibodies (e.g., scFv-Fc formats) for improved solid tumor targeting .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
ADF3 antibody; At5g59880 antibody; MMN10.12 antibody; Actin-depolymerizing factor 3 antibody; ADF-3 antibody; AtADF3 antibody
Target Names
ADF3
Uniprot No.

Target Background

Function
ADF3 is an actin-depolymerizing protein. It severs actin filaments (F-actin) and binds to actin monomers.
Gene References Into Functions
  1. ADF3-dependent defense mechanism is linked to the transcriptional up-regulation of PHYTOALEXIN-DEFICIENT4, which is an important regulator of defenses against the green peach aphid. PMID: 29133373
Database Links

KEGG: ath:AT5G59880

STRING: 3702.AT5G59880.1

UniGene: At.24339

Protein Families
Actin-binding proteins ADF family
Subcellular Location
Cytoplasm, cytoskeleton.

Q&A

What are the key considerations for standardized production of monoclonal antibodies in research settings?

Standardized production of monoclonal antibodies requires careful attention to several critical factors. First, the bovine IgG must be stripped from fetal bovine serum (FBS) to prevent contamination, followed by thorough quality control to ensure purity. Hybridoma cell expansion protocols must be strictly followed for consistent antibody production, with careful monitoring of culture conditions to maintain cell viability and productivity .

Following production, rigorous purification steps using affinity chromatography techniques are essential. Quality control testing should include validation of antibody specificity, assessment of binding kinetics, and confirmation of functional activity. Additionally, endotoxin level detection and mycoplasma screening are critical safety checks that must be performed before using the antibodies in experimental settings .

How should researchers determine appropriate antibody concentrations for different experimental applications?

Determining optimal antibody concentrations requires systematic titration experiments tailored to each application. For immunoassays like ELISA, researchers should perform serial dilutions (typically 2-fold) of the antibody starting from 10 μg/ml down to 0.01 μg/ml against a fixed antigen concentration. The optimal concentration is where you observe strong specific signal with minimal background.

For cell-based assays, particularly those studying receptor-mediated signaling like the Dsg3 receptor system, concentration optimization is crucial as both insufficient and excessive antibody concentrations can compromise results . When antibodies act as receptor activators, as seen with the AK23 antibody in pemphigus vulgaris research, researchers should establish dose-response curves to identify concentrations that effectively trigger signaling cascades without causing off-target effects .

What quality control measures are essential when preparing antibodies for research use?

Quality control for research antibodies involves multiple validation steps. First, conduct SDS-PAGE analysis to confirm purity and integrity, looking for characteristic heavy and light chain bands with minimal contaminants. Immunoreactivity testing against target antigens using methods like ELISA or Western blotting should show specific binding with low background .

Particularly important are endotoxin level measurements using the Limulus Amebocyte Lysate (LAL) assay, as endotoxin contamination can invalidate experimental results, especially in immunological studies. Mycoplasma detection is another critical quality control step, accomplished through PCR-based methods to ensure cultures are not contaminated . For research applications studying signal transduction, functional validation assaying the antibody's ability to activate or inhibit specific signaling pathways is essential before conducting comprehensive experiments.

How are artificial intelligence approaches transforming antibody design and generation?

Artificial intelligence has revolutionized antibody engineering through novel computational frameworks that can generate antibodies with specific binding properties de novo. Recent advances include Pre-trained Antibody generative large Language Models (PALM-H3) that can generate heavy chain complementarity-determining region 3 (CDRH3) sequences with desired antigen-binding specificity . This approach reduces reliance on natural antibody isolation, which traditionally requires resource-intensive screening of serum samples.

The AI methodology combines pre-training on unpaired antibody sequences followed by fine-tuning on antigen-antibody affinity datasets. Models like the Roformer architecture provide interpretable insights into antibody design principles through attention mechanisms . When tested against SARS-CoV-2 antigens, AI-generated antibodies demonstrated high binding affinity and potent neutralization capability against multiple viral variants, including emerging variants like XBB .

What approaches enable the prediction of antibody-antigen binding affinity prior to experimental validation?

Predicting antibody-antigen binding affinity has been significantly enhanced through computational models like A2binder, which pairs antigen epitope sequences with antibody sequences to forecast binding specificity and affinity . This model employs a sophisticated architecture that extracts sequence features from both antigens and antibodies using large-scale pre-trained models.

The prediction pipeline utilizes Multi-Fusion Convolutional Neural Networks (MF-CNN) for feature fusion and affinity prediction, enabling accurate assessments even for previously unknown antigens . The model integrates information from ESM2-based antigen encoders and antibody Roformer decoders pre-trained on extensive antibody sequence databases. This computational approach allows researchers to prioritize antibody candidates before committing resources to expensive and time-consuming experimental validation, substantially accelerating the antibody development workflow .

How can researchers effectively mine large antibody repertoire datasets to identify therapeutic antibody candidates?

Mining large antibody repertoire datasets requires sophisticated computational approaches to identify patterns in the immense sequence space. Researchers should first compile comprehensive antibody sequence datasets from public repositories—currently, some databases contain up to 4 billion productive human heavy variable region sequences with 385 million unique CDR-H3s .

What are the optimal approaches for validating antibody specificity for receptor-targeted research?

Validating antibody specificity for receptor-targeted research requires multiple complementary approaches. First, perform binding assays using purified receptor proteins with techniques like surface plasmon resonance (SPR) to determine affinity constants and binding kinetics. This should be followed by cell-based assays using both receptor-positive and receptor-negative cell lines to confirm specificity in the cellular context .

For receptor-mediated signaling studies, such as those involving the Dsg3 receptor in pemphigus vulgaris research, validation should include downstream signaling assays to confirm the antibody's ability to activate or block specific pathways . Competition assays with known ligands or other antibodies with established binding sites can help confirm epitope specificity. Additionally, knockout/knockdown models provide definitive validation—the antibody should show significantly reduced binding in cells where the target receptor has been deleted or suppressed .

How should researchers interpret contradictory results between in vitro and in vivo antibody studies?

When facing contradictory results between in vitro and in vivo antibody studies, researchers should systematically analyze several factors. First, examine pharmacokinetic differences—antibodies may have different half-lives, tissue distribution, and target accessibility in vivo compared to controlled in vitro environments .

Second, consider the microenvironment effects present in vivo but absent in vitro, including interaction with the extracellular matrix, stromal cells, and immune components that can modulate antibody function . Third, assess antibody effector functions that may be active in vivo but not in vitro, such as antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) .

To resolve contradictions, design bridging studies using ex vivo systems or organoids that better recapitulate in vivo complexity while maintaining some experimental control. Additionally, comprehensive examination of antibody-induced signaling pathways across both systems can identify context-dependent molecular mechanisms explaining the discrepancies .

What methodology should be employed to analyze antibody-induced receptor signaling pathways?

Analysis of antibody-induced receptor signaling requires a multifaceted approach. Begin with phosphorylation state analysis of key signaling molecules using phospho-specific antibodies in Western blotting or phospho-flow cytometry to detect rapid signaling events. This should be followed by temporal analysis of signaling cascades, capturing both immediate (seconds to minutes) and delayed (hours) responses to antibody-receptor binding .

For comprehensive pathway mapping, combine targeted inhibitor studies with phosphoproteomic analysis. This approach involves treating cells with specific pathway inhibitors before antibody addition, then using mass spectrometry-based phosphoproteomics to identify altered phosphorylation sites . Functional validation through genetic approaches (siRNA knockdown or CRISPR-Cas9 editing of pathway components) helps confirm the significance of identified pathways.

This methodology has been successfully applied to understand pathological signaling in pemphigus vulgaris, where the AK23 antibody targeting Dsg3 induces specific pathological signaling cascades that can be pharmacologically modulated .

How can researchers design antibody drug conjugates (ADCs) with optimal therapeutic windows?

Designing antibody drug conjugates with optimal therapeutic windows requires careful optimization of multiple parameters. The selection of target antigen is crucial—ideal candidates are highly expressed on tumor cells but minimally expressed on normal tissues. Currently approved ADCs target specific overexpressed proteins including HER2, Trop2, Nectin4, and EGFR in solid tumors, and CD19, CD22, CD33, CD30, BCMA, and CD79b in hematological malignancies .

The antibody component requires careful engineering for appropriate affinity—moderate affinity often provides better tumor penetration than extremely high affinity, which can cause a "binding site barrier" where ADCs remain trapped near blood vessels rather than penetrating deeper into solid tumors . Antibody isotype selection impacts effector functions and pharmacokinetics, with IgG1 and IgG4 typically preferred over IgG2 (prone to aggregation) and IgG3 (rapid clearance) .

The linker chemistry between antibody and payload must balance stability in circulation with efficient release in target cells. The cytotoxic payload selection should consider potency, offering cell-killing capability at nanomolar or picomolar concentrations to accommodate the limited drug delivery capacity of antibodies .

What approaches enhance antibody penetration in challenging research models such as solid tumors?

Enhancing antibody penetration in solid tumors requires addressing several physiological barriers. First, consider antibody size optimization—smaller formats like Fab fragments, single-chain variable fragments (scFvs), or nanobodies can achieve superior tissue penetration compared to full IgG molecules .

Affinity modulation is critical, as counterintuitively, extremely high-affinity antibodies often show poorer tumor penetration due to the "binding site barrier" phenomenon, where strong binding to antigens near blood vessels prevents deeper penetration . Researchers should test antibodies with moderate affinity (KD in the 10^-8 to 10^-7 M range) alongside high-affinity variants.

Combination strategies that normalize tumor vasculature, such as co-administration with anti-angiogenic agents, can improve antibody delivery by reducing interstitial pressure and vascular permeability . Additionally, targeting components of the tumor microenvironment, such as the extracellular matrix or stromal cells, offers an alternative approach to conventional tumor cell targeting, potentially improving therapeutic efficacy by disrupting tumor-supporting structures .

What patterns emerge when comparing naturally occurring versus engineered therapeutic antibody sequences?

Analysis of naturally occurring versus engineered therapeutic antibody sequences reveals intriguing patterns relevant to antibody design. Large-scale data mining of human antibody repertoires shows that despite the theoretical diversity exceeding 10^15 antibodies, only approximately 270,000 unique CDR-H3s (0.07% of the total) are "highly public," appearing across multiple individuals .

Remarkably, about 6% of therapeutic antibody CDR-H3 sequences have direct matches in this small public set, suggesting therapeutic antibodies often inhabit a constrained sequence space that overlaps with naturally occurring antibodies . This pattern extends beyond just CDR-H3 to include matching variable gene usage, indicating convergent evolution toward certain sequence solutions.

Public CDR-H3s tend to have distinctive features compared to the general repertoire—they are typically shorter and exhibit lower sequence diversity (entropy), yet maintain sufficient diversity for therapeutic applications . These findings suggest that focusing antibody engineering efforts on naturally occurring public sequences may provide a productive strategy for therapeutic antibody development, potentially enhancing developability and reducing immunogenicity risks.

How is large-scale data mining of antibody sequences transforming antibody engineering approaches?

Large-scale data mining of antibody sequences has fundamentally transformed engineering approaches by identifying patterns within the vast theoretical antibody space. By analyzing repositories containing billions of sequences (such as the AbNGS database with 4 billion productive human heavy variable region sequences), researchers have discovered that despite the enormous potential diversity, only a small fraction of sequences (0.07% of unique CDR-H3s) are "highly public" and shared across multiple individuals .

This insight enables more targeted antibody engineering by focusing on the constrained sequence space where therapeutic antibodies are more likely to be found. The observation that 6% of therapeutic antibody CDR-H3s have direct matches in the public repertoire suggests that mining natural antibody sequences can accelerate therapeutic discovery . This approach helps overcome traditional challenges in antibody engineering by providing evolutionary-tested sequence templates that can be further optimized for specific applications, offering a more efficient alternative to traditional random library approaches.

What methodological advances are enabling the development of antibodies against traditionally challenging targets?

Development of antibodies against challenging targets has been revolutionized by complementary technological advances. AI-driven approaches like the PALM-H3 model can generate antibody sequences de novo with specific binding properties, bypassing traditional immunization or display methods . This approach has successfully produced antibodies against conserved epitopes that are typically difficult to target, such as the HR2 peptide of SARS-CoV-2 spike protein .

For targets with limited exposed surfaces or high homology to self-proteins, computational epitope mapping combined with structure-guided design helps identify unique antigenic regions. The integration of large antibody sequence databases with structural prediction algorithms enables identification of naturally occurring antibodies that may recognize challenging epitopes .

Additionally, advanced binding prediction tools like A2binder can rapidly screen candidate antibodies against difficult targets in silico before experimental validation . These computational approaches, combined with high-throughput experimental platforms, create a more efficient development pathway for antibodies against targets previously considered "undruggable."

How are antibody engineering approaches evolving to address emerging infectious diseases and pandemic preparedness?

Antibody engineering approaches for pandemic preparedness have evolved significantly with several key methodological advances. AI-driven antibody generation, exemplified by models like PALM-H3, now enables rapid development of antibodies against emerging pathogens without requiring patient samples or animal immunization . This technology has successfully generated antibodies against SARS-CoV-2 variants, including emerging XBB variants, demonstrating high binding affinity and neutralization capability .

Data mining of natural antibody repertoires offers another powerful approach by identifying conserved antibody sequences that may provide cross-reactive protection against related pathogens . This strategy leverages the observation that certain antibody sequences appear repeatedly across individuals, suggesting evolutionary selection for broadly protective functions.

Computational binding prediction tools like A2binder accelerate the screening process by accurately forecasting antibody-antigen interactions before experimental validation . This integrated approach—combining AI generation, natural repertoire mining, and computational prediction—creates a rapid response capability for emerging infectious diseases, potentially reducing antibody development timelines from months to weeks.

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.