PDF1.1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to PDFAntibody

PDF1.1 Antibody (AS16 3973) is a rabbit-derived polyclonal antibody targeting Arabidopsis thaliana PDF1.1, a member of the plant defensin family. Defensins are antimicrobial peptides critical for plant innate immunity, and PDF1.1 is specifically implicated in pathogen response pathways . This antibody is widely used in plant biology research to study defensin localization, expression dynamics, and stress responses.

Key Properties

PropertyDetails
Host SpeciesRabbit
ClonalityPolyclonal
ReactivitySolanum lycopersicum (tomato), Arabidopsis thaliana
ImmunogenKLH-conjugated synthetic peptide from A. thaliana PDF1.1 (UniProt: P30224-1, TAIR: At1g75830)
Cross-ReactivityConserved in A. thaliana isoforms PDF1.2c, PDF1.2b, PDF1.2A, PDF1.3
PurityAffinity-purified serum in PBS (pH 7.4)
StorageLyophilized or reconstituted at -20°C; avoid repeated freeze-thaw cycles

Antibody Validation

  • The immunogen sequence is conserved across multiple A. thaliana PDF isoforms but distinct from PDF2, ensuring specificity .

  • Validation typically involves western blotting and immunohistochemistry, though users must optimize protocols for specific applications .

Mechanistic Studies

  • Pathogen Response: PDF1.1 Antibody helps elucidate defensin upregulation during fungal or bacterial challenges, particularly in S. lycopersicum .

  • Localization: Used to map PDF1.1 distribution in plant tissues under stress conditions.

Technical Considerations

  • Dilution: Recommended starting concentration for assays like immunohistochemistry (IHC) or western blot is 1:500–1:2,000, depending on the system .

  • Controls: Include knockout plant lines or pre-immune serum to confirm signal specificity .

Comparative Analysis of Plant Defensin Antibodies

While PDF1.1 Antibody focuses on A. thaliana and tomato, other defensin-targeting antibodies (e.g., PDF2-specific tools) exhibit distinct reactivity profiles. For example:

TargetAntibody ClonalityHostKey Applications
PDF1.1PolyclonalRabbitPathogen response, localization
PDF2MonoclonalMouseIsoform-specific studies
Drosophila PDFMonoclonal (PDF C7)MouseNeuropeptide signaling

Limitations and Best Practices

  • Cross-Reactivity: Despite sequence conservation, off-target binding to unrelated defensins may occur without rigorous validation .

  • Storage Stability: Lyophilized formulations reduce degradation but require careful reconstitution (50 µl sterile water per 50 µg vial) .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
PDF1.1 antibody; AFP1 antibody; LCR67 antibody; At1g75830 antibody; T4O12.7Defensin-like protein 13 antibody; Anther-specific protein S18 homolog antibody; Cysteine-rich antifungal protein 1 antibody; AFP1 antibody; Low-molecular-weight cysteine-rich protein 67 antibody; Protein LCR67 antibody; Plant defensin 1.1 antibody
Target Names
PDF1.1
Uniprot No.

Target Background

Function
Demonstrates broad-spectrum antimicrobial activity, including antifungal properties. In vitro studies indicate sensitivity of antifungal activity to inorganic cations.
Database Links

KEGG: ath:AT1G75830

STRING: 3702.AT1G75830.1

UniGene: At.128

Protein Families
DEFL family
Subcellular Location
Secreted.
Tissue Specificity
Expressed predominantly in siliques and dry seeds.

Q&A

What is PD-1 and how do antibodies targeting this pathway function in research?

PD-1 is an immune checkpoint receptor expressed primarily on activated T cells that plays a critical role in regulating immune responses. The PD-1/PD-L1 axis represents a major immunosuppressive mechanism exploited by tumors to evade immune surveillance. Antibodies targeting this pathway can be used to block these interactions, thereby restoring T cell function and enhancing anti-tumor immunity.

In research settings, PD-1 antibodies serve multiple functions:

  • Detection of PD-1 expression on immune cells

  • Blocking PD-1/PD-L1 interactions in functional assays

  • Studying downstream signaling pathways

  • Developing and evaluating potential immunotherapeutic approaches

Approximately 20% of breast cancer patients suffer from distant metastasis, making immune checkpoint inhibitors an emerging treatment area for these patients with otherwise poor outcomes .

How should researchers validate PD-1 antibodies before experimental use?

Antibody validation is critical before using PD-1 antibodies in research applications. The "five pillars" approach to validation includes:

  • Genetic validation: Use PD-1 knockout (KO) cell lines as negative controls to confirm antibody specificity. This approach is considered superior to other control methods, especially for Western blot and immunofluorescence applications.

  • Orthogonal validation: Compare results obtained with antibody-dependent techniques (e.g., immunohistochemistry) with antibody-independent methods (e.g., mass spectrometry or mRNA expression).

  • Independent antibody validation: Use different antibodies targeting distinct epitopes of PD-1 and compare staining patterns.

  • Recombinant expression validation: Overexpress PD-1 in cells that don't normally express it to confirm specific detection.

  • Immunocapture MS validation: Use mass spectrometry to identify proteins captured by the antibody.

This comprehensive validation approach is essential considering that approximately 50% of commercial antibodies fail to meet basic standards for characterization, resulting in financial losses of $0.4-1.8 billion per year in the United States alone .

What controls should be included when using PD-1 antibodies in research?

When using PD-1 antibodies, the following controls are essential:

Essential negative controls:

  • PD-1 knockout cell lines (gold standard)

  • Cell types known not to express PD-1 (confirmed by orthogonal methods)

  • Isotype-matched control antibodies

  • Secondary antibody-only controls

Required positive controls:

  • Samples with confirmed PD-1 expression (e.g., activated T cells)

  • PD-1-transfected cell lines

  • Known positive tissue sections (for IHC)

Application-specific controls:

  • For IHC: Include tissue-specific controls as antibody performance can vary between tissue types

  • For flow cytometry: Fluorescence-minus-one (FMO) controls

  • For blocking experiments: Include competitive inhibition controls

Recent studies have shown that an average of approximately 12 publications per protein target included data from antibodies that failed to recognize the relevant target protein, highlighting the critical importance of proper controls .

What are the main immunoassay methods for detecting PD-1 and PD-L1 expression?

Several immunoassay methods are commonly used to detect PD-1 and PD-L1 expression:

MethodFrequency of UseKey AdvantagesCommon Antibody ClonesMajor Limitations
Immunohistochemistry92.9%Preserves tissue context22C3 (30.8%), SP142 (19.2%), SP263Subjective scoring, fixation variability
Flow CytometryCommon for cell suspensionsMulti-parameter analysisVariousLimited to single-cell suspensions
Western BlottingCommon for specificity validationMolecular weight confirmationVariousLoses spatial information
ELISAPrimary for soluble formsQuantitativeVariousLimited to soluble proteins
ImmunofluorescenceGrowing in popularityMultiplexing capabilityVariousAutofluorescence issues

The YCharOS group's comprehensive analysis demonstrated that knockout cell lines are superior controls for Western blots and even more crucial for immunofluorescence imaging, highlighting the importance of proper validation for each method .

How does antibody format affect PD-1 detection performance?

Antibody format significantly impacts PD-1 detection in research applications:

Isotype/subtype effects:

  • Different isotypes (IgG, IgM, IgA) and IgG subtypes (IgG1, IgG2, IgG3, IgG4) possess distinct properties

  • Human IgG1 engages Fc receptors efficiently, while IgG4 has minimal effector function

  • These differences affect background binding in tissues with high Fc receptor expression

Fragment formats:

  • Fab fragments: Eliminate Fc-mediated background, improving signal-to-noise ratio

  • F(ab')₂: Retain bivalent binding but reduce Fc-related artifacts

  • scFv (single-chain variable fragments): Smaller size enables better tissue penetration

Species considerations:

  • For diagnostic applications, species switching allows the creation of human variants that avoid HAMA (human anti-mouse antibody) responses

  • Chimeric antibodies containing human constant domains and mouse variable domains reduce non-specific binding to heterophilic antibodies

  • This makes them useful for diagnostic assay development with batch-to-batch reproducibility

What are the challenges in interpreting PD-1/PD-L1 expression data across different assay platforms?

Interpreting PD-1/PD-L1 expression across platforms presents several challenges:

Technical variability sources:

  • Different antibody clones demonstrate varying staining patterns and intensities

  • Pre-analytical variables (fixation time, tissue processing) significantly impact staining

  • Different detection systems and amplification methods alter sensitivity

Scoring system disparities:

  • Various cutoff values define PD-L1 positivity across studies

  • Inconsistent inclusion of immune cell vs. tumor cell staining

  • Qualitative vs. quantitative assessment methods

Biological complexities:

  • Dynamic expression influenced by tumor microenvironment

  • Spatial heterogeneity in expression, particularly in large tumors

  • Treatment-induced changes in expression patterns

Standardization efforts:

  • Harmonization studies show incomplete concordance between platforms

  • Blueprint PD-L1 Assay Comparison Project revealed significant inter-assay variability

  • Lack of universal reference standards complicates cross-study comparison

These challenges underscore the importance of standardized reporting, including detailed methodological documentation, antibody specifications, and context-dependent validation for each specific application .

How do chimeric and humanized PD-1 antibodies differ in research applications?

Chimeric and humanized PD-1 antibodies represent different engineering approaches with distinct research applications:

CharacteristicChimeric AntibodiesHumanized Antibodies
StructureMouse variable domains with human constant regionsOnly CDRs from mouse, framework regions human
ImmunogenicityModerateMinimal
Production costLowerHigher
ApplicationsEarly research, diagnosticsTherapeutics, advanced preclinical models
AdvantagesGood batch-to-batch consistency, cost-effectiveMinimal immunogenicity, better in vivo performance
ExamplesHistorical therapeutics like infliximabMost modern therapeutic antibodies

For research purposes, chimeric antibodies often provide sufficient humanization to avoid false positive results due to heterophilic antibodies while maintaining cost-effectiveness. For diagnostic applications, chimeric antibodies reduce the risk of non-specific binding to human anti-mouse antibodies (HAMA) that can cause false positive assay results .

Humanized antibodies are critical for therapeutic development derived from non-human sources, involving the transfer of critical non-human amino acids to a human antibody framework, reducing immunogenicity while preserving binding specificity .

What methodologies exist for characterizing novel PD-1 antibodies?

Characterization of novel PD-1 antibodies requires a multi-faceted approach:

Specificity assessment:

  • Knockout validation: Testing against PD-1 knockout cell lines (gold standard)

  • Knockdown validation: siRNA or shRNA-mediated depletion as alternative

  • Cross-reactivity testing: Evaluation against related proteins (other CD28 family members)

  • Orthogonal validation: Correlation with mRNA expression or mass spectrometry data

Binding properties characterization:

  • Surface plasmon resonance (SPR): Determines binding kinetics (kon, koff) and KD

  • Bio-layer interferometry (BLI): Alternative for real-time binding analysis

  • Isothermal titration calorimetry (ITC): Provides thermodynamic binding parameters

Epitope mapping:

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS)

  • X-ray crystallography for atomic-level resolution

  • Competition binding with antibodies of known epitopes

  • Alanine scanning mutagenesis

Functional characterization:

  • PD-1/PD-L1 blockade assays to confirm mechanism

  • T cell activation assays to assess functional impact

  • In vivo tumor models for therapeutic candidates

Manufacturability assessment:

  • Expression yield quantification in relevant systems

  • Size-exclusion chromatography for aggregation analysis

  • Stability testing under various conditions

  • Glycosylation profiling for effector function prediction

A comprehensive characterization approach combining multiple validation methods provides the most reliable antibody assessment, as demonstrated by the YCharOS group's analysis of 614 antibodies targeting 65 proteins .

How does antibody engineering affect the performance of PD-1 detection?

Antibody engineering significantly impacts PD-1 detection through several mechanisms:

Variable domain modifications:

  • Humanization affects binding characteristics and may alter specificity

  • Affinity maturation enhances detection of low-abundance PD-1 but may increase non-specific binding

  • CDR optimization improves specificity for particular epitopes

Constant region engineering:

  • Fc engineering reduces background in tissues with high Fc receptor expression

  • Isotype selection influences stability in specific buffers and fixation conditions

  • Glycoengineering affects Fc receptor interactions and non-specific binding

Format diversification:

  • Bispecific formats enhance sensitivity by co-targeting PD-1 and another marker

  • Multivalent designs increase avidity for detecting low-expressing cells

  • Fragment formats improve tissue penetration and reduce background

Recombinant antibodies consistently outperform both monoclonal and polyclonal antibodies across multiple assays. A case study demonstrated that properly humanized antibodies showed 2-4 fold higher expression yields and reduced aggregation (>99.5% monomer content) compared to chimeric versions, highlighting the impact of engineering on performance .

What are the best practices for troubleshooting non-specific binding with PD-1 antibodies?

Non-specific binding challenges require systematic troubleshooting approaches:

Fundamental validation first:

  • Confirm antibody specificity using PD-1 knockout controls

  • If non-specific binding persists in knockout samples, test alternative antibody clones

  • Compare recombinant vs. traditional monoclonal antibodies (recombinants typically show lower non-specific binding)

Application-specific optimization:

Western Blotting optimization:

  • Increase blocking stringency (5% BSA or milk)

  • Add 0.1-0.5% detergents to reduce hydrophobic interactions

  • Implement more stringent washing conditions

  • Reduce primary antibody concentration systematically

  • Test alternative membrane types

Immunohistochemistry/Immunofluorescence optimization:

  • Optimize antigen retrieval methods

  • Employ additional blocking steps

  • Include Fc receptor blocking reagents

  • Test alternative fixation methods

  • Implement peptide competition controls

Flow Cytometry optimization:

  • Include dead cell exclusion dyes

  • Implement Fc receptor blocking

  • Optimize permeabilization for intracellular staining

  • Compare different fluorophore conjugates

  • Use proper compensation controls

Sample-specific considerations:

  • For human samples, use human serum in blocking buffer

  • For tissues with high endogenous peroxidase, include quenching steps

  • Consider using Fab fragments to eliminate Fc-mediated interactions

The YCharOS group's comprehensive antibody validation approach revealed that many commercially available antibodies require application-specific optimization to reduce non-specific binding .

How can researchers evaluate PD-1 antibody performance across different tissue types?

Evaluating PD-1 antibody performance across diverse tissues requires a methodical approach:

Comprehensive tissue panel development:

  • Include lymphoid tissues (tonsil, lymph node) as positive controls

  • Select disease-relevant tissues matching research focus

  • Include tissues known for technical challenges (high autofluorescence, etc.)

  • Prepare multi-tissue arrays for consistent processing

Systematic optimization strategy:

  • Customize fixation based on tissue characteristics

  • Adjust antigen retrieval conditions for each tissue

  • Modify blocking protocols to address tissue-specific background

  • Titrate antibody concentration individually for challenging tissues

Multi-parameter assessment:

  • Quantify signal-to-noise ratio across tissue types

  • Compare staining with multiple PD-1 antibody clones

  • Correlate with mRNA expression from matched samples

  • Co-localize with cell type-specific markers

Advanced validation approaches:

  • Laser capture microdissection followed by mass spectrometry

  • Multiplexed immunofluorescence for cell type specificity

  • Spatial transcriptomics correlation with protein expression

  • Fresh tissue validation compared to fixed tissues

Research has shown that antibody performance is context-dependent, requiring characterization for each specific tissue type and application. The Alpbach Workshops on Affinity Proteomics emphasized that characterization data are potentially cell or tissue type specific, necessitating tailored validation approaches .

What considerations are important when developing PD-1 antibodies for therapeutic applications?

Developing PD-1 antibodies for therapeutic applications involves several critical considerations:

Humanization and immunogenicity assessment:

  • Full humanization minimizes immunogenicity

  • Use of humanized germline sequences associated with fewer adverse events

  • Assessment of potential T-cell epitopes that could trigger immune responses

  • Deimmunization strategies for problematic sequences

Manufacturability optimization:

  • Expression yield must be sufficient for large-scale production (>2-3 g/L)

  • Aggregation propensity should be minimal (>99.5% monomer content)

  • Stability under various storage conditions must be robust

  • Post-translational modifications should be consistent

Functional property engineering:

  • Binding affinity optimization for efficacy without excessive off-target effects

  • Epitope selection affects mechanism of action

  • Half-life engineering for appropriate dosing regimen

  • Tissue penetration properties for target distribution

Effector function tuning:

  • Isotype selection based on desired effector functions

  • Fc engineering to enhance or silence specific functions

  • Glycosylation profile optimization for effector control

A case study highlighted in the research demonstrated that an antibody being developed for glioblastoma treatment showed poor manufacturability in its chimeric form, but after humanization screening, several variants achieved 2-4 fold higher expression levels with minimal aggregation (>99.5% monomer content) .

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.