AP1 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
Made-to-order (14-16 weeks)
Synonyms
AP1 antibody; AGL7 antibody; At1g69120 antibody; F4N2.9 antibody; Floral homeotic protein APETALA 1 antibody; Agamous-like MADS-box protein AGL7 antibody
Target Names
AP1
Uniprot No.

Target Background

Function
APETALA1 (AP1) is a transcription factor that plays a pivotal role in floral development in Arabidopsis thaliana. It functions in synergy with LEAFY to promote early floral meristem identity, and subsequently facilitates the transition of an inflorescence meristem into a floral meristem. AP1 is essential for the normal development of sepals and petals in flowers. It positively regulates the B class homeotic proteins APETALA3 and PISTILLATA in collaboration with LEAFY and UFO. AP1 interacts with SEPALLATA3 or AP3/PI heterodimer to form complexes that could be involved in gene regulation during floral meristem development. Additionally, AP1 positively regulates AGAMOUS in conjunction with LEAFY. It exhibits a redundant function with CAULIFLOWER in the up-regulation of LEAFY. Together with AGL24 and SVP, AP1 controls the identity of the floral meristem and regulates the expression of class B, C and E genes. Furthermore, AP1 represses flowering time genes AGL24, SVP and SOC1 in emerging floral meristems.
Gene References Into Functions
  1. APETALA1 establishes determinate floral meristem through regulating cytokinins homeostasis in Arabidopsis. PMID: 26359644
  2. Suppression of cytokinin biosynthesis and activation of cytokinin degradation mediates AP1 function in establishing determinate floral meristems. PMID: 24753595
  3. Co-expression analysis identifies CRC and AP1 as regulators of Arabidopsis fatty acid biosynthesis. PMID: 22676405
  4. Different action of the APETALA1 gene on the development of reproductive organs in flowers of the abruptus mutant of Arabidopsis thaliana. PMID: 21950056
  5. Results suggest distinct functions of AP1 during the initiation of flower development. PMID: 20360106
  6. The floral homeotic PISTILLATA (PI) protein and its interacting partner APETALA3 directly act, in combination with other factors, to restrict the expression of AP1 during early stages of floral development. PMID: 16640596
  7. AP1, AGL24 and SVP redundantly control floral meristem identity. PMID: 18694458
  8. Angiostatin K1-3 induced E-selectin expression via AP1 and Ets-1 binding to the proximal E-selectin promoter (-356/+1), which was positively mediated by JNK activation. PMID: 18761727

Show More

Hide All

Database Links

KEGG: ath:AT1G69120

STRING: 3702.AT1G69120.1

UniGene: At.10200

Subcellular Location
Nucleus.
Tissue Specificity
Expressed in young flower primordia, later becomes localized to sepals and petals.

Q&A

What is the AP-1 transcription factor complex and why is it important in research?

AP-1 is a dimeric transcription factor complex composed of proteins belonging to the Jun (cJUN, JUNB, JUND), Fos (cFOS, FRA1, FRA2), ATF and JDP families. The combinatorial assembly of these proteins creates context-specific transcriptional regulators that control diverse cellular processes.

AP-1 is particularly important in research because it represents a critical regulatory node in cellular signaling. The state of the AP-1 transcription factor network has been shown to play a unifying role in explaining diverse patterns of cellular plasticity, particularly in cancer models. For example, in melanoma, a regulated balance among AP-1 factors cJUN, JUND, FRA2, FRA1, and cFOS determines the intrinsic diversity of differentiation states and adaptive responses to MAPK inhibitors . Additionally, AP-1 activity is crucial for chromatin remodeling during T cell activation, making it a key factor in immune regulation .

What types of AP-1 antibodies are available for research applications?

Researchers can access several types of AP-1 antibodies:

  • Component-specific antibodies: Target individual AP-1 proteins (e.g., antibodies against cJUN, cFOS, FRA1)

  • Phospho-specific antibodies: Detect phosphorylated forms of AP-1 proteins (e.g., p-cJUN, p-FRA1)

  • Complex-specific antibodies: Recognize structural components of AP-1-associated proteins, such as AP-1 complex subunit mu-1 (AP1M1)

These antibodies are validated for various applications including Western blotting, immunohistochemistry, ChIP, and flow cytometry, depending on the specific product and epitope.

How do I select the appropriate AP-1 antibody for my research question?

Selection should be based on:

  • Target specificity: Determine which AP-1 component is relevant to your research (e.g., cJUN vs. FRA1)

  • Post-translational modifications: Consider whether you need to detect specific phosphorylated forms

  • Application compatibility: Verify validation for your specific application (WB, IHC, ChIP, etc.)

  • Species reactivity: Confirm reactivity with your experimental model organism

  • Clonality: Monoclonal antibodies offer higher specificity for single epitopes, while polyclonal antibodies may provide stronger signals through multi-epitope binding

When studying heterogeneous cell populations or dynamic processes like differentiation, consider antibodies targeting multiple AP-1 factors simultaneously, as studies have shown that combinations of AP-1 proteins (e.g., cFOS, FRA1, FRA2, cJUN, JUNB, JUND) collectively predict cellular states better than individual markers .

How can I effectively use AP-1 antibodies in ChIP-seq experiments?

For successful ChIP-seq with AP-1 antibodies:

  • Antibody selection: Choose ChIP-validated antibodies specific to your AP-1 component of interest

  • Crosslinking optimization: AP-1 factors bind DNA transiently; optimize formaldehyde crosslinking time (typically 10-15 minutes)

  • Sonication parameters: Aim for chromatin fragments of 200-300bp

  • Controls: Include:

    • Input control (non-immunoprecipitated chromatin)

    • IgG control (non-specific antibody)

    • Positive control regions (known AP-1 binding sites)

  • Validation: Confirm enrichment at known AP-1 target regions by qPCR before sequencing

Research has shown that AP-1 binds at >70% of newly opened chromatin regions within 5 hours of T cell activation, making timing a critical consideration for capturing dynamic binding events . Use fresh antibodies and optimize antibody concentration through titration experiments.

What is the best approach for detecting multiple AP-1 proteins in single cells?

Based on recent research methodologies, multiplexed approaches are most effective:

  • Iterative indirect immunofluorescence imaging (4i): This technique allows sequential staining with multiple antibodies against different AP-1 components. Studies have successfully used this to measure 11 AP-1 transcription factors and 6 phosphorylation states in melanoma cells .

  • Mass cytometry (CyTOF): Using metal-conjugated antibodies for simultaneous detection of multiple AP-1 proteins

  • Multiplex immunofluorescence: Using spectrally distinct fluorophores and/or antibodies from different host species

Protocol considerations:

  • Careful antibody validation for specificity

  • Sequential staining protocols with appropriate blocking between rounds

  • Image registration for accurate co-localization analysis

  • Single-cell segmentation algorithms for quantification

Recent research has demonstrated that patterns of AP-1 variation at single-cell protein levels strongly correlate with differentiation states, with factors like p-cFOS, FRA2, ATF4, cFOS, p-FRA1, and cJUN being particularly predictive .

How should I optimize Western blotting protocols for AP-1 antibodies?

ParameterRecommendationRationale
Lysis bufferRIPA with phosphatase inhibitorsPreserves phosphorylation status of AP-1 proteins
Protein amount20-50μg total proteinBalances signal strength with specificity
Gel percentage10-12% polyacrylamideOptimal for separating AP-1 proteins (35-60kDa)
Transfer conditions100V for 1 hour or 30V overnightEfficient transfer without heat-induced degradation
Blocking5% BSA in TBST (for phospho-antibodies)Prevents background without masking phospho-epitopes
Primary antibody dilution1:1000-1:4000Start with manufacturer recommendation
IncubationOvernight at 4°CMaximizes specific binding
DetectionECL or fluorescence-basedSelect based on expected abundance and dynamic range

For detecting multiple AP-1 proteins on the same blot, consider:

  • Sequential stripping and reprobing (risk of signal loss)

  • Multiplex fluorescence detection (requires antibodies from different host species)

  • Parallel blots from the same samples

Molecular weights for common AP-1 proteins: cJUN (~39kDa), cFOS (~62kDa), FRA1 (~40kDa), JUND (~35kDa), AP1M1 (~48.6kDa)

How do I interpret conflicting results when analyzing different AP-1 components?

Conflicting results between different AP-1 components are common due to:

  • Contextual activity: AP-1 functions as dimeric complexes with context-dependent compositions

  • Cell-type specificity: AP-1 expression patterns vary among cell types

  • Temporal dynamics: AP-1 components show different activation kinetics

  • Post-translational modifications: Phosphorylation status affects activity independently of abundance

Methodological approach to resolve conflicts:

  • Comprehensive profiling: Measure multiple AP-1 components simultaneously when possible

  • Time-course experiments: Capture dynamic changes over relevant timescales

  • Correlation analysis: Use multivariate statistical modeling to identify relationships

  • Single-cell approaches: Account for cellular heterogeneity

Research has shown that the predictivity of AP-1 patterns for cellular states (e.g., melanoma differentiation) can be captured at both the transcript and protein levels, but with component-specific variations. For example, ATF4 shows inconsistent correlations between transcript and protein measurements , highlighting the importance of multi-level analysis.

What are the common sources of false positives/negatives when using AP-1 antibodies?

IssuePossible CausesSolutions
False Positives
Non-specific bindingCross-reactivity with similar epitopesUse monoclonal antibodies; validate with knockout controls
High backgroundInsufficient blocking; too concentrated antibodyOptimize blocking conditions; titrate antibody
False Negatives
Epitope maskingProtein complex formation; post-translational modificationsTry multiple antibodies targeting different epitopes
Antibody incompatibilityBuffer conditions affecting antibody performanceTest alternative fixation/extraction methods
Low expressionDetection limitsUse signal amplification; longer exposure times

Validation approaches:

  • Use genetic knockdown/knockout controls

  • Compare results with multiple antibodies targeting different epitopes of the same protein

  • Correlate with mRNA expression data where applicable

  • Include positive control samples with known expression

Research shows that prediction accuracy for AP-1-based cellular state classification can vary significantly (from ~0.35 to ~0.75) depending on the specific cell lines and AP-1 components analyzed , highlighting the importance of proper controls and validation.

How can AP-1 antibodies be used to study chromatin remodeling dynamics?

AP-1 factors play crucial roles in chromatin remodeling. Methodological approaches include:

  • Integrated ChIP-seq and ATAC-seq:

    • Use AP-1 antibodies for ChIP-seq to map binding sites

    • Correlate with ATAC-seq to identify regions of chromatin accessibility

    • Perform time-course experiments to capture dynamic changes

  • CUT&RUN or CUT&Tag with AP-1 antibodies:

    • Higher resolution alternative to traditional ChIP

    • Requires fewer cells

    • Lower background signal

  • Sequential ChIP (re-ChIP):

    • First IP with one AP-1 component antibody

    • Second IP with another transcription factor antibody

    • Identifies co-occupancy at specific loci

Research has shown that broad inhibition of AP-1 activity prevents chromatin opening at AP-1 sites and reduces the expression of nearby genes. For example, in T cells, AP-1 directs most chromatin remodeling within 5 hours of activation, with newly opened regions strongly enriched for AP-1 motifs .

What approaches can be used to study the role of AP-1 in disease-relevant cellular plasticity?

Multiple approaches can be integrated:

  • Combinatorial perturbations:

    • RNAi-mediated knockdown of specific AP-1 components

    • CRISPR/Cas9 genome editing of AP-1 components or binding sites

    • Small molecule inhibitors of AP-1 activity

  • Single-cell multi-omics:

    • Combine multiplexed protein measurements with transcriptomics

    • Analyze cells before and after perturbations

    • Map trajectories of cellular state transitions

  • Patient-derived models:

    • Apply AP-1 profiling to patient samples

    • Correlate with treatment responses

    • Identify predictive biomarkers

Research has demonstrated that perturbing the balance of AP-1 factors through genetic depletion or pharmacological inhibition (e.g., MAPK inhibitors) shifts cellular heterogeneity in predictable ways. This has particular relevance for melanoma, where AP-1 states predict responses to therapy .

How can computational approaches enhance the interpretation of AP-1 antibody-based experiments?

Advanced computational methods improve data interpretation:

  • Machine learning classification:

    • Random forest models can predict cellular states from AP-1 measurements

    • SHAP (SHapley Additive exPlanations) values quantify contributions of specific AP-1 factors

    • Models trained on multiplexed AP-1 data achieve classification accuracies of ~0.74 for melanoma differentiation states

  • Network inference:

    • Reconstruct AP-1 regulatory networks from ChIP-seq and expression data

    • Identify key nodes and feedback mechanisms

    • Predict system responses to perturbations

  • Multi-modal data integration:

    • Correlate AP-1 binding with chromatin accessibility, histone modifications, and gene expression

    • Identify cooperative and antagonistic relationships with other factors

    • Generate testable hypotheses about regulatory mechanisms

Implementation requires:

  • Rigorous quality control and normalization

  • Appropriate feature selection methods

  • Cross-validation to assess model generalizability

  • Integration of domain knowledge about AP-1 biology

How can AP-1 antibodies be used to study the relationship between AP-1 and immune-related disease risk?

Recent research has identified substantial overlap between AP-1-dependent chromatin elements and risk loci for multiple immune diseases . Methodological approaches include:

  • Genetic-epigenetic integration:

    • Map AP-1 binding sites in disease-relevant cell types

    • Overlay with GWAS risk loci

    • Identify functional SNPs that alter AP-1 binding

  • Patient-stratified analyses:

    • Profile AP-1 component levels in patient samples

    • Correlate with disease subtypes or progression

    • Identify potential biomarkers

  • Functional validation:

    • Use CRISPR/Cas9 to edit disease-associated AP-1 binding sites

    • Assess impact on chromatin accessibility and gene expression

    • Measure functional consequences in cellular assays

Research has specifically highlighted connections between AP-1-dependent elements and risk loci for multiple sclerosis, inflammatory bowel disease, and allergic diseases , suggesting broad relevance across immune-mediated conditions.

What are the considerations for developing multiplex assays that include AP-1 antibodies?

Creating effective multiplex assays requires:

  • Antibody compatibility assessment:

    • Test for cross-reactivity between antibodies

    • Ensure fixation and permeabilization conditions work for all targets

    • Validate spectral separation or alternative detection methods

  • Panel design strategies:

    • Include major AP-1 components (cJUN, JUND, FRA1, FRA2, cFOS)

    • Add phospho-specific antibodies for activation status

    • Incorporate lineage markers and functional readouts

  • Quality control metrics:

    • Use single-stain controls

    • Include fluorescence-minus-one (FMO) controls

    • Apply compensation or unmixing algorithms

  • Data analysis considerations:

    • Dimensionality reduction (tSNE, UMAP)

    • Clustering algorithms

    • Trajectory analysis

Research using iterative indirect immunofluorescence imaging has successfully multiplexed measurements of 21 proteins, including 11 AP-1 transcription factors and 6 AP-1 phosphorylation states, demonstrating the feasibility of comprehensive AP-1 profiling .

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.