CYCA2-3 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

CYCA2;3 is a mitotic cyclin belonging to the A2-type cyclin family, which modulates the G2-to-M phase transition and suppresses endoreduplication (repeated DNA replication without cell division) in plants . The CYCA2;3 antibody targets this protein, enabling researchers to investigate its expression, localization, and functional interactions.

Antibody Development and Validation

  • Epitope Specificity: The antibody is typically raised against conserved regions of CYCA2;3, such as its N-terminal domain or destruction box (D-box), which is critical for protein degradation via the anaphase-promoting complex/cyclosome (APC/C) .

  • Validation Methods:

    • Western Blotting: Detects CYCA2;3 at ~51 kDa in protein extracts, as demonstrated in studies using HA-tagged CYCA2;3 constructs .

    • Immunoprecipitation (IP): Validates interactions with partners like CDKB1;1 kinase and the APC/C activator CCS52A1 .

    • Localization Studies: Used in GFP-fusion experiments to track CYCA2;3 dynamics in nuclei and cytoplasm during the cell cycle .

Key Research Applications

ApplicationExperimental UseSource
Cell Cycle RegulationQuantifies CYCA2;3 levels during G2/M transition and mitotic exit in synchronized cells.
EndoreduplicationAssesses CYCA2;3 stabilization in mutants (e.g., samba) to study ploidy regulation.
Protein InteractionsIdentifies binding partners like CDKB1;1 and Bora-PLK1 complexes in cytoplasmic fractions.
Mutant PhenotypingDetects CYCA2;3 accumulation in cyca2;3 null mutants with enhanced endoreduplication.

Major Research Findings

  • Role in Mitotic Entry: CYCA2;3-CDK complexes are essential for activating Polo-like kinase 1 (PLK1) in the cytoplasm, driving G2-to-M progression .

  • Endoreduplication Suppression: CYCA2;3 destabilization by APC/C<sup>SAMBA</sup> limits DNA re-replication, maintaining proper ploidy levels .

    • Null mutants: Exhibit semidominant endocycle promotion, increasing ploidy by 25–50% in trichomes and leaves .

  • Tissue-Specific Regulation: CYCA2;3 expression is enriched in stomatal guard cells and trichomes, where it restricts ectopic divisions .

Regulatory Mechanisms

  • APC/C-Dependent Degradation: SAMBA, a plant-specific APC/C regulator, targets CYCA2;3 for proteasomal degradation via its D-box .

  • CDK Complex Formation: CYCA2;3 binds CDKB1;1 to inhibit endocycle entry by blocking relicensing of DNA replication origins .

Technical Considerations

  • Cross-Reactivity: Validated against other A2-type cyclins (e.g., CYCA2;1, CYCA2;2) due to sequence homology .

  • Stability Assays: Used in cycloheximide chase experiments to measure protein half-life (~2–4 hours in wild-type vs. >6 hours in samba mutants) .

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
CYCA2-3 antibody; CYC3C antibody; At1g15570 antibody; T16N11.8Cyclin-A2-3 antibody; Cyc3c-At antibody; Cyclin-3c antibody; G2/mitotic-specific cyclin-A2-3 antibody; CycA2;3 antibody
Target Names
CYCA2-3
Uniprot No.

Target Background

Function
CYCA2;3 antibody targets a protein that plays a crucial role in regulating cell division and ploidy levels. It negatively regulates endocycles, ensuring proper control of DNA replication and cell size. Furthermore, this antibody targets a protein that promotes divisions in guard cells (GCs) following the symmetric division of guard mother cells (GMCs).
Gene References Into Functions
  1. CYCA2;3 negatively regulates endocycles and acts as a key regulator of ploidy levels in Arabidopsis endoreduplication. PMID: 16415207
Database Links

KEGG: ath:AT1G15570

STRING: 3702.AT1G15570.1

UniGene: At.1679

Protein Families
Cyclin family, Cyclin AB subfamily
Subcellular Location
Nucleus.

Q&A

What is CYCA2-3 and what is its primary function in plant cells?

CYCA2-3 (Cyclin A2;3) is a plant-specific A-type cyclin that forms functional complexes with cyclin-dependent kinases, particularly CDKB1;1. The CDKB1;1-CYCA2;3 complex plays a crucial role in controlling the balance between cell proliferation and endoreduplication. Biochemical studies have demonstrated that this complex primarily drives the mitotic cell cycle and prevents cells from prematurely exiting the mitotic cell cycle . In terms of molecular mechanism, CYCA2-3 likely functions by repressing the relicensing of DNA replication during the S-to-G2 transition while also enhancing CDK activity at the G2-to-M transition point .

How does CYCA2-3 interact with other cell cycle regulators?

CYCA2-3 primarily interacts with CDKB1;1 as confirmed through both yeast two-hybrid screening and tandem affinity purification (TAP) techniques . This interaction has been biochemically validated through multiple experimental approaches. Additionally, CYCA2-3 may interact with E2F transcription factors, suggesting its potential role during S phase regulation . The protein contains a D-box motif that mediates its interaction with APC/C (Anaphase Promoting Complex/Cyclosome) components, particularly CCS52A1, which targets CYCA2-3 for destruction in a D-box-dependent manner . This regulated destruction is critical for proper cell cycle progression and entry into endoreduplication cycles.

What methods can be used to detect CYCA2-3 protein interactions in plant samples?

Several complementary techniques have proven effective for detecting CYCA2-3 protein interactions:

  • Yeast Two-Hybrid Screening: This technique successfully identified CYCA2-3 as an interactor of CDKB1;1. Notably, using a dominant negative allele (CDKB1;1.N161) as bait improved detection, likely due to stabilization of the kinase-cyclin complex .

  • Tandem Affinity Purification (TAP): This powerful approach involves creating CYCA2-3 fusion proteins with a TAP tag, expressing them in Arabidopsis cell cultures, and purifying the resulting immunological complexes. Subsequent mass spectrometry analysis can identify interacting proteins with high confidence .

  • Co-immunoprecipitation with Fluorescent Tags: Using GFP-tagged CYCA2-3 allows for immunoprecipitation with anti-GFP antibodies, enabling both visualization of localization and biochemical analysis of interaction partners .

How can CYCA2-3 kinase activity be measured in experimental systems?

Kinase activity assays for CYCA2-3 complexes can be performed using the following methodology:

  • Generate plant lines expressing CYCA2-3-GFP fusion proteins and prepare protein extracts from appropriate tissues (e.g., 7-day-old seedlings induced with β-estradiol).

  • Immunoprecipitate the CYCA2-3-GFP complexes using agarose-conjugated anti-GFP monoclonal antibodies (approximately 530 μg of protein extract with 30 μL of 50% gel slurry).

  • After washing with appropriate buffers, perform histone H1 kinase reactions by resuspending the immunoprecipitates in reaction buffer containing [γ-33P]ATP, histone H1 substrate, and appropriate cofactors.

  • Incubate the reaction at 30°C for 20 minutes, then stop by adding SDS-PAGE loading buffer.

  • Analyze the phosphorylated products by SDS-PAGE, Coomassie staining, and autoradiography to detect [γ-33P]ATP incorporation into histone H1 .

This method allows quantification of CDK activity associated with CYCA2-3 complexes, with appropriate controls (e.g., Col-0 background signal subtraction).

What are the key considerations when selecting a CYCA2-3 antibody for immunolocalization studies?

When selecting an antibody for CYCA2-3 immunolocalization, researchers should consider:

  • Specificity: The antibody should specifically recognize CYCA2-3 without cross-reactivity to other A-type cyclins, particularly CYCA2-1, CYCA2-2, and CYCA2-4, which share sequence homology. Validation through Western blotting against recombinant proteins and extracts from knockout/overexpression lines is essential.

  • Epitope Accessibility: Consider whether the antibody recognizes an epitope that remains accessible in fixed tissue. The C-terminal region of CYCA2-3 tends to be more accessible than regions involved in protein-protein interactions.

  • Compatibility with Fixation Methods: Test the antibody with different fixation protocols (aldehyde-based, methanol/acetone, etc.) as some epitopes may be masked or destroyed by certain fixatives.

  • Signal-to-Noise Ratio: Evaluate the antibody's ability to produce clean signals with minimal background in your specific plant tissue of interest, as background can vary significantly between developmental stages and tissue types.

  • Compatibility with Co-localization Studies: If performing co-localization with other cell cycle markers, ensure the CYCA2-3 antibody is raised in a species compatible with your other primary antibodies.

How should Western blot protocols be optimized for CYCA2-3 detection?

Optimizing Western blot protocols for CYCA2-3 detection requires addressing several critical factors:

  • Sample Preparation: Include phosphatase inhibitors (e.g., sodium fluoride, β-glycerophosphate, sodium vanadate) and protease inhibitors in extraction buffers to prevent degradation of CYCA2-3, which is subject to rapid turnover via the APC/C pathway.

  • Protein Extraction Buffer: Use a buffer containing 50 mM Tris-HCl (pH 7.2-7.6), 250 mM NaCl, 0.1% Nonidet P-40, 2 mM EDTA, and 10% glycerol for optimal CYCA2-3 extraction .

  • Gel Percentage: Utilize 10-12% acrylamide gels for optimal resolution of CYCA2-3, which has a molecular weight of approximately 47 kDa.

  • Transfer Conditions: For efficient transfer of CYCA2-3, use semi-dry transfer systems with 20% methanol or wet transfer systems with 10% methanol at lower amperage for longer periods.

  • Blocking Conditions: Test both milk-based and BSA-based blocking solutions, as CYCA2-3 antibody performance can vary between these blocking agents.

  • Signal Enhancement: Consider using enhanced chemiluminescence substrates with extended activity for detecting low-abundance CYCA2-3 in certain tissue types.

  • Controls: Always include positive controls (extracts from plants overexpressing CYCA2-3) and negative controls (cyca2-3 mutant extracts) to validate specificity.

How does CYCA2-3 expression correlate with ploidy levels in different plant tissues?

CYCA2-3 expression shows strong inverse correlation with endoreduplication and ploidy levels in plant tissues. The following table summarizes ploidy distribution in various genetic backgrounds related to CYCA2-3 expression:

PloidyCol-0Col-0 × CYCA2;3-GFPCDKA1;1 × CYCA2;3-GFPCDKB1;1 × CYCA2;3-GFPCDKB1;1.N161 × CYCA2;3-GFP
Without β-estradiol
2C28.1 ± 0.626.4 ± 0.326.6 ± 0.827.4 ± 0.911.8 ± 1.0
4C36.8 ± 0.430.6 ± 0.835.1 ± 0.730.2 ± 1.428.7 ± 1.2
8C29.8 ± 0.433.8 ± 0.733.3 ± 1.035.5 ± 0.442.2 ± 4.0
16C4.9 ± 0.79.1 ± 1.25.1 ± 0.36.9 ± 1.017.0 ± 4.4
+10 μm β-estradiol
2C26.9 ± 0.328.9 ± 1.928.0 ± 1.247.1 ± 1.412.1 ± 0.8
4C37.1 ± 0.237.1 ± 0.040.7 ± 1.235.2 ± 1.032.0 ± 2.0
8C30.2 ± 0.431.1 ± 1.027.8 ± 1.016.5 ± 2.048.4 ± 1.3
16C5.2 ± 0.12.8 ± 0.92.9 ± 1.01.0 ± 0.47.4 ± 1.8

This data demonstrates that overexpression of CYCA2-3 with CDKB1;1 significantly increases the 2C population (47.1% versus 26.9% in control) while decreasing higher ploidy levels (particularly 8C and 16C populations) . This confirms CYCA2-3's role in promoting mitotic division while inhibiting endoreduplication. Interestingly, when CYCA2-3 is co-expressed with the dominant negative CDKB1;1.N161, the opposite effect occurs with increased endoreduplication (higher 8C and 16C populations), suggesting that the kinase activity of the CDKB1;1-CYCA2-3 complex is essential for restricting endoreduplication .

What molecular mechanisms regulate CYCA2-3 protein stability?

The stability of CYCA2-3 protein is primarily regulated through targeted proteolysis mediated by the Anaphase Promoting Complex/Cyclosome (APC/C). Key regulatory mechanisms include:

  • D-box Dependent Degradation: CYCA2-3 contains a destruction box (D-box) motif that is essential for protein turnover. Mutation of this D-box results in protein stabilization, confirming its role in regulating protein half-life .

  • CCS52A1-Mediated Targeting: Among APC/C activators, CCS52A1 specifically targets CYCA2-3 for degradation. This was demonstrated by the stabilization of CYCA2-3-GFP in ccs52a1 loss-of-function plants .

  • Tissue-Specific Regulation: While CCS52A1 regulates CYCA2-3 levels, this regulation shows tissue specificity. For example, CYCA2-3-GFP is not stabilized in the roots of ccs52a2 knockout plants, despite both CCS52A1 and CCS52A2 regulating DNA ploidy in leaf cells .

  • Transcriptional Control: Beyond protein degradation, CYCA2-3 levels are also controlled at the transcriptional level by factors such as FLP/MYB124 and MYB88, which repress CYCA2-3 expression in guard cells .

  • Hormone Signaling: Auxin signaling has been implicated in regulating CYCA2-3 expression, providing another layer of control that links developmental cues to cell cycle regulation .

This multilayered regulation ensures precise control of CYCA2-3 levels, which is critical for proper cell cycle progression and developmental transitions.

How can CYCA2-3 antibodies be used in studying the guard cell differentiation pathway?

CYCA2-3 antibodies can be powerful tools for investigating guard cell differentiation pathways through several experimental approaches:

  • Developmental Time Course Analysis: Using CYCA2-3 antibodies in immunohistochemistry to track protein levels during stomatal lineage progression can reveal the precise timing of CYCA2-3 downregulation during guard cell differentiation.

  • Chromatin Immunoprecipitation (ChIP) Studies: Combining CYCA2-3 antibodies with ChIP can identify genomic targets of CYCA2-3-containing complexes, particularly in the context of FLP/MYB88 regulation, which directly links to guard cell development .

  • Co-Immunoprecipitation with Developmental Regulators: CYCA2-3 antibodies can be used to detect interactions with stomatal lineage regulators like SPEECHLESS, MUTE, and FAMA to understand how cell cycle regulation is coordinated with cell fate specification.

  • Quantitative Analysis in Mutant Backgrounds: Immunoblotting with CYCA2-3 antibodies in various stomatal development mutants (especially flp-7myb88, which shows five-fold higher CYCA2-3 expression) can provide quantitative insights into how developmental programming controls cell cycle exit .

  • Single-Cell Analysis: Using CYCA2-3 antibodies in combination with cell-type specific markers can reveal heterogeneity in CYCA2-3 expression within the stomatal lineage, potentially identifying transitional states during differentiation.

These approaches can help elucidate the molecular mechanisms linking cell cycle regulation with guard cell differentiation and stomatal patterning.

What are the challenges in using CYCA2-3 antibodies for studying protein degradation dynamics?

Studying CYCA2-3 degradation dynamics using antibodies presents several technical challenges that researchers should consider:

  • Rapid Turnover Rate: CYCA2-3 has a short half-life due to APC/C-mediated degradation, making it difficult to capture the native protein at sufficient levels for detection without using proteasome inhibitors, which can alter cellular physiology.

  • Cell Cycle Phase Dependence: Since CYCA2-3 degradation is cell cycle-dependent, synchronization of plant cells is often necessary, which presents technical difficulties and may introduce artifacts.

  • D-box Recognition: Standard fixation methods for immunohistochemistry may alter D-box conformation or accessibility, potentially affecting antibody recognition of degradation-targeted versus stable populations of CYCA2-3.

  • Distinguishing Phosphorylated Forms: CYCA2-3 may exist in different phosphorylation states, which can affect degradation rates. Standard antibodies may not distinguish between these forms, requiring phospho-specific antibodies for complete analysis.

  • Tissue-Specific Regulation: As demonstrated by the differential effects of ccs52a1 versus ccs52a2 mutations on CYCA2-3 stability in different tissues, degradation mechanisms may be tissue-specific, necessitating tissue-specific optimization of protocols .

  • Antibody Sensitivity: Detecting the low endogenous levels of CYCA2-3 in wild-type tissues requires highly sensitive antibodies and detection methods, particularly when studying tissues undergoing active degradation.

To overcome these challenges, researchers often combine antibody-based approaches with fluorescent protein fusions (like CYCA2-3-GFP) and real-time imaging to track protein dynamics.

How can researchers resolve inconsistent CYCA2-3 antibody signals between different plant tissues?

When encountering tissue-specific variations in CYCA2-3 antibody signal, researchers should consider implementing the following solutions:

  • Optimize Tissue-Specific Extraction: Different plant tissues contain varying compositions of interfering compounds. Adjust extraction buffers accordingly, adding polyvinylpolypyrrolidone (PVPP) for polyphenol-rich tissues or increasing detergent concentrations for tissues with high lipid content.

  • Adjust Fixation Protocols: If performing immunohistochemistry, test different fixation methods and durations. Some tissues may require longer fixation times or different fixatives to properly preserve CYCA2-3 epitopes while maintaining tissue morphology.

  • Consider Developmental Timing: CYCA2-3 expression varies dramatically with developmental stage. Ensure precise staging of samples, particularly when comparing different tissue types that may develop asynchronously.

  • Account for Protein Stability Differences: Remember that CYCA2-3 stability is regulated differently across tissues, with specific APC/C activators like CCS52A1 and CCS52A2 showing tissue-specific activities . Include proteasome inhibitors when appropriate to normalize for degradation rate differences.

  • Validate with Alternative Methods: Confirm antibody results with transcript analysis (RT-qPCR) or reporter constructs (CYCA2-3 promoter fusions) to determine whether differences reflect actual expression patterns or technical artifacts.

  • Use Internal Controls: Include internal loading controls appropriate for each tissue type, as standard housekeeping genes may show tissue-specific expression variations.

What controls are essential when performing co-immunoprecipitation experiments with CYCA2-3 antibodies?

When conducting co-immunoprecipitation (co-IP) experiments with CYCA2-3 antibodies, the following controls are essential:

  • Input Control: Always include an aliquot of the pre-IP lysate to confirm the presence of target proteins and potential interactors before immunoprecipitation.

  • Negative Control Antibody: Perform parallel IPs with isotype-matched non-specific antibodies (such as normal IgG from the same species as the CYCA2-3 antibody) to identify non-specific binding.

  • No-Antibody Control: Include a sample treated identically but without adding any antibody to identify proteins that bind non-specifically to the beads.

  • Genetic Controls: Whenever possible, include samples from cyca2-3 knockout/knockdown plants as negative controls and CYCA2-3 overexpression lines as positive controls.

  • Competing Peptide Control: Pre-incubate the CYCA2-3 antibody with excess antigen peptide before immunoprecipitation to confirm signal specificity.

  • Reciprocal IP: Confirm interactions by performing reverse co-IPs using antibodies against suspected interaction partners (e.g., CDKB1;1) to precipitate CYCA2-3.

  • Non-Denaturing Conditions Control: Since CYCA2-3 forms complexes with CDKs, test both native and more stringent buffer conditions to distinguish direct from indirect interactions.

  • Cell Cycle Phase Controls: If possible, use synchronized cell populations to account for cell cycle-dependent interactions, as CYCA2-3 interactions may vary throughout the cell cycle.

Implementing these controls will substantially increase confidence in co-IP results and help distinguish biologically relevant interactions from experimental artifacts.

How might CYCA2-3 antibodies be used to investigate stress responses in plants?

CYCA2-3 antibodies could significantly advance our understanding of stress-induced cell cycle changes through several innovative approaches:

  • Stress-Induced Proteome Remodeling: Using CYCA2-3 antibodies to track protein abundance changes during abiotic stresses (drought, salt, temperature) could reveal how environmental signals modulate cell cycle machinery to adapt growth patterns.

  • Post-Translational Modification Profiling: Developing modification-specific antibodies (phospho-CYCA2-3, ubiquitinated-CYCA2-3) could uncover how stress signaling cascades directly modify CYCA2-3 function and stability.

  • Subcellular Relocalization Studies: Immunolocalization with CYCA2-3 antibodies before and after stress treatments could identify stress-induced changes in protein localization that might modify function independently of expression changes.

  • Chromatin Association Dynamics: ChIP-seq approaches using CYCA2-3 antibodies could map stress-induced changes in genomic targets of CYCA2-3-containing complexes, potentially revealing mechanisms of stress-adaptive growth responses.

  • Tissue-Specific Response Analysis: Immunohistochemistry with CYCA2-3 antibodies across different tissues during stress could identify organ-specific cell cycle responses, explaining differential growth responses to environmental challenges.

  • Hormone-Stress Interaction Studies: Since auxin signaling influences CYCA2-3 expression , using CYCA2-3 antibodies to investigate how stress hormones (ABA, ethylene, jasmonates) interact with growth hormones could uncover integration points between stress signaling and cell cycle control.

What emerging technologies might enhance the utility of CYCA2-3 antibodies in plant cell cycle research?

Several cutting-edge technologies show promise for expanding CYCA2-3 antibody applications:

  • Proximity Labeling Approaches: Combining CYCA2-3 antibodies with BioID or TurboID proximity labeling could map the complete protein interaction neighborhood around CYCA2-3 in specific cell types or developmental contexts.

  • Single-Cell Proteomics: Emerging single-cell proteomic technologies could enable quantification of CYCA2-3 levels in individual cells within complex tissues, revealing heterogeneity in cell cycle regulation at unprecedented resolution.

  • Super-Resolution Microscopy: Techniques like STORM or PALM combined with CYCA2-3 immunolabeling could reveal nanoscale organization of CYCA2-3-containing complexes relative to chromatin and other subcellular structures.

  • Live-Cell Antibody Fragments: Developing cell-permeable antibody fragments (nanobodies) against CYCA2-3 could enable live imaging of endogenous protein dynamics without requiring genetic modification.

  • CRISPR-Based Tagging: CRISPR knock-in approaches to tag endogenous CYCA2-3 with epitopes for enhanced antibody detection could overcome expression artifacts associated with overexpression systems.

  • Spatial Transcriptomics Integration: Combining CYCA2-3 immunohistochemistry with spatial transcriptomics could correlate protein abundance with transcriptional states across tissues, revealing post-transcriptional regulatory mechanisms.

  • Cryo-Electron Tomography: This approach could utilize CYCA2-3 antibodies conjugated to gold particles to visualize native complexes at molecular resolution within cellular contexts.

These technologies promise to transform our understanding of how CYCA2-3 functions within the complex network of cell cycle regulation and developmental programming in plants.

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.