CYP714A2 Antibody

Shipped with Ice Packs
In Stock

Description

CYP714A2 Protein Overview

CYP714A2 is a member of the cytochrome P450 family in Arabidopsis thaliana, playing a critical role in GA deactivation and homeostasis. Key characteristics include:

PropertyDetails
Gene NameCYP714A2 (also termed ELA2)
FunctionCatalyzes oxidation reactions to deactivate bioactive GAs .
SubstratesGA12 (converted to 12α-hydroxy GA12 and GA53) .
Biological ImpactOverexpression reduces bioactive GA levels, causing semi-dwarfism in plants .
Tissue ExpressionFound in vascular tissues and reproductive organs .

Role of CYP714A2 in Gibberellin Regulation

CYP714A2 contributes to GA homeostasis through:

  • Deactivation: Converts active GAs (e.g., GA4) into less active or inactive forms via hydroxylation .

  • Redundancy: Functions alongside CYP714A1; double mutants exhibit increased GA levels and enhanced growth .

  • Regulatory Feedback: Expression is upregulated by bioactive GAs, ensuring tight control over GA signaling .

Applications of CYP714A2 Antibody in Research

Though not explicitly detailed in the provided sources, antibodies targeting CYP714A2 would enable:

  • Protein Localization: Visualizing CYP714A2 distribution in plant tissues via immunohistochemistry.

  • Expression Analysis: Quantifying CYP714A2 levels under different physiological conditions (e.g., stress, development) using Western blotting or ELISA.

  • Functional Studies: Blocking CYP714A2 activity to study GA accumulation effects on plant morphology.

Example Research Findings Enabled by Antibody Use

Study FocusHypothetical Application
GA Biosynthesis PathwaysCorrelating CYP714A2 expression with GA metabolite levels .
Plant Dwarfism MechanismsLinking CYP714A2 overexpression to reduced GA4 and stunted growth .
Hormonal Cross-TalkInvestigating interactions between GA and other hormones (e.g., auxin) .

Production and Validation of Plant-Targeting Antibodies

While the provided sources do not specify CYP714A2 antibody production, plant-derived monoclonal antibodies (mAbs) are typically generated using:

  • Transient Expression Systems: Viral vectors (e.g., bean yellow dwarf virus) in Nicotiana benthamiana enable rapid, high-yield antibody production .

  • Validation Steps:

    1. Specificity Testing: Ensure no cross-reactivity with related P450s (e.g., CYP714A1).

    2. Functional Assays: Confirm antibody efficacy in immunoprecipitation or neutralization .

Challenges and Future Directions

  • Epitope Conservation: CYP714A2’s structural similarity to other P450s may complicate antibody specificity.

  • Agricultural Relevance: Engineered antibodies could fine-tune GA levels to optimize crop yields .

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
CYP714A2 antibody; ELA2 antibody; At5g24900 antibody; F6A4.110Cytochrome P450 714A2 antibody; EC 1.14.-.- antibody; EUI-like P450 A2 antibody
Target Names
CYP714A2
Uniprot No.

Target Background

Function
CYP714A2 antibody is involved in the inactivation of early gibberellin (GA) intermediates.
Gene References Into Functions
  1. CYP714 family proteins play a crucial role in the biosynthesis of diverse gibberellin compounds. These proteins catalyze various oxidation reactions involving the C and D rings of gibberellins in both monocots and eudicots. PMID: 24009336
  2. CYP714A1 and CYP714A2 exhibit functional redundancy in regulating plant growth and development. Their primary function is likely to mediate gibberellin catabolism, contributing to the control of gibberellin levels within the plant. PMID: 21457373
Database Links

KEGG: ath:AT5G24900

STRING: 3702.AT5G24900.1

UniGene: At.30940

Protein Families
Cytochrome P450 family
Subcellular Location
Endoplasmic reticulum membrane; Single-pass type III membrane protein.
Tissue Specificity
Expressed in the shoot apical meristem (SAM) and petioles of young leaves, in the leaf margin and petiole vein of cotyledons, and at low levels in the filaments of developing flowers. Not detected in siliques.

Q&A

What is CYP714A2 and what is its function in plant biology?

CYP714A2 is a member of the cytochrome P450 family found in Arabidopsis thaliana. It functions as an oxidase enzyme involved in gibberellin (GA) metabolism, specifically catalyzing the conversion of ent-kaurenoic acid to steviol (ent-13-hydroxy kaurenoic acid). When using GA12 as a substrate, CYP714A2 primarily produces 12α-hydroxy GA12 (GA111) and, to a lesser extent, 13-hydroxy GA12 (GA53) . This enzymatic activity affects the balance between non-13-hydroxy GAs (like GA4) and 13-hydroxy GAs (like GA1), which influences plant growth. Overexpression of CYP714A2 in transgenic Arabidopsis plants leads to semi-dwarfism, demonstrating its role in modulating growth through GA metabolism pathways . The CYP714 family collectively contributes to the production of diverse GA compounds through various oxidations of C and D rings in both monocots and eudicots, with CYP714A2 being part of this broader regulatory network.

How do researchers generate polyclonal antibodies against CYP714A2?

Researchers typically generate polyclonal antibodies against CYP714A2 by first producing recombinant CYP714A2 protein or synthesizing unique peptide sequences from the protein. The standard methodology involves selecting antigenic regions based on computational analysis of the protein's secondary structure, hydrophilicity, and surface probability. For CYP714A2, researchers often focus on regions that are distinct from other CYP714 family members to enhance specificity. The selected antigen is then used to immunize animals (typically rabbits) over a period of several weeks with multiple booster injections to stimulate a robust immune response. Serum is collected and purified through affinity chromatography using the immobilized antigen. Validation involves Western blotting against plant tissue samples with known CYP714A2 expression patterns, comparing wild-type plants with CYP714A2 knockout or overexpression lines. Cross-reactivity with other CYP714 family members must be carefully assessed due to sequence homology between related cytochrome P450 enzymes .

What are the primary applications of CYP714A2 antibodies in plant science research?

CYP714A2 antibodies serve multiple critical functions in plant science research. First, they enable precise protein detection and quantification through Western blotting, allowing researchers to track expression levels across different tissues, developmental stages, or in response to environmental stressors. Immunohistochemistry and immunofluorescence microscopy with these antibodies reveal the spatial distribution of CYP714A2 within plant cells and tissues, providing insights into subcellular localization and potential functional domains. CYP714A2 antibodies are also valuable for co-immunoprecipitation experiments to identify interaction partners in gibberellin metabolism pathways, potentially uncovering regulatory mechanisms. Additionally, researchers employ these antibodies in chromatin immunoprecipitation (ChIP) assays when studying transcription factors that regulate CYP714A2 expression. For functional studies, antibodies can be used to deplete CYP714A2 in cell-free systems, allowing assessment of the consequences of enzyme absence on specific biochemical pathways related to gibberellin metabolism .

How can researchers design highly specific monoclonal antibodies for distinguishing CYP714A2 from closely related CYP714 family members?

Developing highly specific monoclonal antibodies for CYP714A2 requires sophisticated epitope mapping and selection strategies. Researchers should begin with comprehensive sequence alignment of all CYP714 family members to identify unique regions in CYP714A2. Computational approaches can predict surface-exposed epitopes that maximize structural differences between CYP714A2 and related proteins. For monoclonal antibody generation, researchers typically employ a phage display approach with large synthetic antibody libraries, where diversity is focused in the complementarity-determining regions (CDRs), particularly CDR3 . Multiple selection rounds with stringent washing steps can enrich for highly specific binders. Critically, negative selection steps should be included, where the library is pre-incubated with closely related proteins (CYP714A1, CYP714B1, etc.) to remove cross-reactive antibodies before selecting against CYP714A2 . Following selection, candidates must undergo rigorous validation through competitive ELISA, Western blotting, and immunoprecipitation assays using both recombinant proteins and plant extracts from wild-type, knockout, and overexpression lines. Advanced biophysical characterization techniques like surface plasmon resonance (SPR) should be employed to determine binding kinetics, with ideal candidates showing at least 100-fold higher affinity for CYP714A2 than for other family members .

What are the challenges in validating CYP714A2 antibody specificity in complex plant tissue samples?

Validating CYP714A2 antibody specificity in complex plant tissues presents several significant challenges. Plant tissues contain numerous cytochrome P450 enzymes with structural similarity to CYP714A2, creating potential for cross-reactivity. Additionally, post-translational modifications of native CYP714A2 may differ from the recombinant protein used for antibody generation, affecting epitope recognition. The relatively low natural expression level of CYP714A2 in many tissues requires highly sensitive detection methods while maintaining specificity. Researchers must develop a comprehensive validation strategy incorporating multiple controls: (1) side-by-side testing in wild-type, CYP714A2 knockout, and overexpression plant lines; (2) pre-absorption controls where antibody is pre-incubated with purified antigen before immunodetection; (3) peptide competition assays to confirm epitope specificity; and (4) parallel validation with orthogonal techniques like mass spectrometry or RNA expression analysis . Additionally, researchers should test the antibody across different tissue types and developmental stages to account for tissue-specific expression patterns and potential interfering compounds. For antibodies intended for immunolocalization studies, additional controls validating subcellular localization patterns against fluorescent protein fusion constructs are necessary to confirm accurate representation of CYP714A2 distribution patterns.

How can computational modeling improve CYP714A2 antibody design for superior specificity?

Advanced computational modeling can significantly enhance CYP714A2 antibody design through several approaches. Protein structure prediction tools can generate accurate three-dimensional models of CYP714A2 based on homology to crystallized cytochrome P450 proteins, revealing unique surface features for targeted antibody binding. Machine learning algorithms trained on existing antibody-antigen complexes can predict optimal binding interfaces and CDR configurations for high specificity and affinity to CYP714A2 . Molecular dynamics simulations can assess the stability of predicted antibody-antigen interactions across different conditions, ensuring robust performance in various experimental contexts. Researchers should employ biophysics-informed computational models that integrate experimental data from phage display selections to identify binding modes specific to CYP714A2 versus other cytochrome P450s . This enables computational design of antibodies with customized specificity profiles—either highly specific for CYP714A2 or intentionally cross-reactive with defined subsets of related proteins. The approach involves optimizing energy functions associated with each binding mode to minimize or maximize interaction with desired or undesired targets, respectively . For ultimate validation, computationally designed antibody candidates should be synthesized and experimentally tested against a panel of cytochrome P450 proteins to confirm the predicted specificity profile.

What is the optimal protocol for immunoprecipitation of CYP714A2 from Arabidopsis tissues?

The optimal immunoprecipitation protocol for CYP714A2 must address its membrane association and relatively low abundance. Begin with 5-10 grams of appropriate plant tissue (preferably seedlings or young leaves with verified CYP714A2 expression). Grind tissue in liquid nitrogen and extract in a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 10% glycerol, 0.5% Triton X-100, 1 mM EDTA, 5 mM DTT, and protease inhibitor cocktail. The inclusion of mild detergent is critical for solubilizing membrane-associated CYP714A2. After centrifugation at 14,000g for 15 minutes, pre-clear the supernatant with Protein A/G beads for 1 hour at 4°C. Incubate the pre-cleared lysate with validated anti-CYP714A2 antibody (5-10 μg/mL) overnight at 4°C with gentle rotation. Capture antibody-protein complexes with fresh Protein A/G beads for 3 hours, followed by at least five washes with decreasing salt concentration (300 mM to 150 mM NaCl). For each experiment, include appropriate controls: (1) immunoprecipitation from CYP714A2 knockout tissue to assess non-specific binding; (2) use of pre-immune serum or isotype control antibody; and (3) a spike-in control with recombinant CYP714A2 to confirm antibody function. For co-immunoprecipitation studies to identify interaction partners, consider using chemical crosslinking (0.5-1% formaldehyde) prior to cell lysis to stabilize transient protein-protein interactions within the gibberellin metabolism complex .

How can researchers develop a quantitative ELISA for measuring CYP714A2 protein levels in plant samples?

Developing a quantitative ELISA for CYP714A2 requires careful optimization of antibody pairs and extraction conditions. Begin by selecting a capture antibody (monoclonal if available) and a detection antibody (preferably from a different species or isotype) that bind to non-overlapping epitopes on CYP714A2. For optimal sensitivity, extract plant samples in a buffer containing 100 mM Tris-HCl (pH 8.0), 150 mM NaCl, 5% glycerol, 0.1% Triton X-100, 1 mM EDTA, 5 mM DTT, and protease inhibitors. The extraction buffer should be optimized to maintain CYP714A2 stability while effectively solubilizing the protein from membrane fractions. Coat high-binding ELISA plates with capture antibody (2-5 μg/mL) overnight at 4°C, followed by blocking with 3% BSA or 5% non-fat milk. Incubate extracted samples and standards (purified recombinant CYP714A2 at 0-100 ng/mL) for 2 hours at room temperature. After washing, add biotinylated detection antibody followed by streptavidin-HRP and appropriate substrate. For accurate quantification, generate standard curves using recombinant CYP714A2 produced in a eukaryotic expression system to mimic plant post-translational modifications. Validate the ELISA by comparing measurements across wild-type, CYP714A2 knockout, and overexpression plant lines. To ensure specificity, perform competition assays with purified CYP714A1 and other related cytochrome P450 proteins. The lower limit of detection should be determined and reported, along with inter- and intra-assay coefficients of variation to establish reproducibility .

What approaches can be used to characterize the epitope binding properties of anti-CYP714A2 antibodies?

Characterizing the epitope binding properties of anti-CYP714A2 antibodies requires a multi-faceted approach to define specificity, affinity, and the precise binding region. Begin with peptide array analysis, using overlapping 15-20 amino acid peptides spanning the entire CYP714A2 sequence to identify linear epitopes recognized by the antibody. For conformational epitopes, hydrogen-deuterium exchange mass spectrometry (HDX-MS) can identify regions protected from deuterium exchange when the antibody is bound. Surface plasmon resonance (SPR) or bio-layer interferometry (BLI) should be employed to determine binding kinetics (kon, koff) and affinity constants (KD), ideally comparing binding to CYP714A2 versus related proteins like CYP714A1 . X-ray crystallography or cryo-electron microscopy of the antibody-antigen complex provides the most detailed structural information about the epitope. For polyclonal antibodies, epitope mapping through phage display libraries expressing random peptides can identify immunodominant epitopes. Functional epitope mapping is also valuable—testing whether the antibody inhibits enzymatic activity of CYP714A2 in vitro provides insights into whether the epitope overlaps with functionally important regions. Cross-reactivity profiling against a panel of related cytochrome P450 enzymes, particularly other CYP714 family members, should be conducted using ELISA and Western blotting to create a comprehensive specificity profile for each antibody .

How should researchers interpret discrepancies between CYP714A2 antibody detection and mRNA expression levels?

Discrepancies between CYP714A2 protein detection using antibodies and mRNA expression levels are common and require careful analysis. These differences may reflect several biological phenomena: (1) post-transcriptional regulation through microRNAs or RNA-binding proteins affecting translation efficiency; (2) differential protein stability or degradation rates, particularly in response to hormonal or environmental signals; (3) tissue-specific translational control mechanisms; or (4) technical limitations in either protein or RNA detection methods. To systematically investigate such discrepancies, researchers should first validate both detection methods—confirming antibody specificity using knockout controls and RNA detection using multiple primer pairs or probes. Time-course experiments can reveal temporal dynamics, as protein levels often lag behind mRNA changes. Treating plants with proteasome inhibitors (e.g., MG132) can determine if protein degradation contributes to discrepancies. Polysome profiling can assess translational efficiency of CYP714A2 mRNA. For quantitative comparison, absolute quantification of both mRNA (using digital PCR) and protein (using recombinant protein standards in Western blots) provides more reliable data than relative methods. Researchers should also consider cell- or tissue-specific differences that might be masked in whole-organ analyses. When discrepancies persist despite thorough validation, they likely represent genuine biological regulation and should be reported as important findings regarding post-transcriptional control of CYP714A2 expression .

How can researchers effectively validate antibody specificity when genomic knockout of CYP714A2 is lethal or severely impacts plant development?

Validating antibody specificity when CYP714A2 knockout produces lethal or severely developmental phenotypes requires alternative approaches. Researchers should implement inducible silencing systems such as dexamethasone-inducible RNAi or estradiol-inducible CRISPR interference (CRISPRi) targeting CYP714A2, enabling timed downregulation while avoiding developmental lethality. Another approach is tissue-specific knockout using tissue-specific promoters driving Cas9 expression, allowing antibody validation in specific tissues while maintaining functional CYP714A2 in tissues critical for survival. Heterozygous knockout plants may show reduced CYP714A2 levels suitable for partial validation. Transient expression systems offer alternatives—virus-induced gene silencing (VIGS) can temporarily reduce CYP714A2 expression in specific tissues, or protoplast systems allow introduction of CYP714A2 siRNAs followed by antibody testing. For comprehensive validation, a biochemical approach using recombinant CYP714A2 and related proteins in competitive Western blots or ELISAs can establish specificity without requiring genetic manipulation. Additionally, mass spectrometry can verify antibody specificity by identifying proteins immunoprecipitated from plant extracts. If epitope-tagged CYP714A2 can be expressed without disrupting function, dual detection with anti-epitope and anti-CYP714A2 antibodies provides strong validation evidence. Finally, correlation with orthogonal measurements like CYP714A2 enzymatic activity provides functional validation of antibody specificity .

How can researchers design multiplex immunoassays to simultaneously detect CYP714A2 and other enzymes in the gibberellin biosynthesis pathway?

Designing multiplex immunoassays for simultaneous detection of CYP714A2 and other gibberellin biosynthesis enzymes requires careful antibody selection and assay optimization. Begin by selecting antibodies raised in different host species (e.g., rabbit anti-CYP714A2, mouse anti-GA20ox, goat anti-GA3ox) to enable simultaneous detection without cross-reactivity between secondary antibodies. For multiplex Western blotting, fluorescently labeled secondary antibodies with distinct emission spectra allow simultaneous imaging on systems with appropriate filter sets. Alternatively, if using chemiluminescence, sequential probing with HRP-conjugated secondaries can be performed after thorough stripping between rounds. For multiplex immunofluorescence microscopy, select fluorophores with minimal spectral overlap (e.g., Alexa Fluor 488, 568, and 647) and include appropriate single-staining controls to establish bleed-through parameters for image analysis. Bead-based multiplex assays represent an advanced approach—coupling antibodies to distinctly coded microbeads enables simultaneous capture and detection of multiple proteins in a single sample. Flow cytometry can then quantify each target based on bead identity and signal intensity. For all multiplex approaches, extensive validation is required to ensure antibodies maintain specificity and sensitivity in the multiplex format. Cross-reactivity matrices should be established by testing each primary antibody against all recombinant proteins in the panel. Dynamic range matching is also critical—detection methods must accommodate the potentially different expression levels of each target protein. Finally, data analysis requires appropriate normalization strategies to allow meaningful comparison of relative abundances across the gibberellin biosynthesis pathway .

What approaches enable investigation of post-translational modifications of CYP714A2 using specific antibodies?

Investigating post-translational modifications (PTMs) of CYP714A2 requires specialized antibody-based approaches. For phosphorylation analysis, researchers can use phospho-specific antibodies generated against predicted or known phosphorylation sites in CYP714A2, based on consensus motifs for plant kinases and phosphoproteomic data. These antibodies should be validated using recombinant CYP714A2 treated with phosphatases as negative controls. Alternatively, immunoprecipitate CYP714A2 using general anti-CYP714A2 antibodies followed by Western blotting with anti-phosphoserine/threonine/tyrosine antibodies. For glycosylation analysis, lectins coupled with CYP714A2 immunoprecipitation can identify specific glycan structures; treatment with glycosidases provides necessary negative controls. Ubiquitination can be studied through immunoprecipitation under denaturing conditions (to disrupt non-covalent interactions) followed by detection with anti-ubiquitin antibodies. For studying redox modifications, antibodies specific to oxidized cysteines or derivatization methods like biotin-switch assays combined with CYP714A2 immunoprecipitation are effective. Acetylation can be detected using anti-acetyllysine antibodies after CYP714A2 immunoprecipitation. Mass spectrometry remains the gold standard for comprehensive PTM analysis and should be used to validate antibody-based findings. Site-directed mutagenesis of putative modification sites can confirm their functional relevance—expressing mutant versions in CYP714A2 knockout backgrounds followed by phenotypic and biochemical analyses. Importantly, researchers should consider that PTMs may be transient and context-dependent, requiring analysis under various physiological conditions, developmental stages, and stress responses .

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.