JOX3 Antibody

Shipped with Ice Packs
In Stock

Description

Biological Context of JOX3

JOX3 belongs to the 2-oxoglutarate-dependent oxygenase (2OG oxygenase) family in Arabidopsis thaliana. These enzymes hydroxylate JA to form 12-OH-JA, an inactive metabolite that modulates JA signaling amplitude and duration .

Key enzymatic characteristics:

PropertyJOX3 Specification
Substrate specificityJasmonic acid (JA)
Reaction product12-hydroxy-JA (12-OH-JA)
Catalytic cofactorFe²⁺ and 2-oxoglutarate
Induction signalJasmonate signaling

Functional Role in Plant Physiology

JOX3 collaborates with three other JOX enzymes (JOX1, JOX2, JOX4) to fine-tune JA levels, balancing growth-defense trade-offs :

  • Defense enhancement: joxQ mutants (lacking all four JOXs) hyperaccumulate JA, leading to stronger resistance against necrotrophic pathogens (Botrytis cinerea) and herbivores.

  • Growth regulation: Uncontrolled JA levels in mutants cause growth retardation, highlighting JOX3’s role in mitigating JA’s inhibitory effects on development.

Gene expression dynamics:

  • JOX3 is transcriptionally induced by JA within hours of stress.

  • Overexpression reduces JA accumulation, while knockout lines exhibit elevated JA and defense gene expression (e.g., PDF1.2, ORA59) .

Antibody Applications and Validation

Though not explicitly detailed in the provided sources, JOX3 antibodies would theoretically enable:

  • Protein localization studies: Tracking JOX3 expression in plant tissues under stress.

  • Quantitative assays: Measuring JOX3 levels via Western blot or ELISA.

  • Functional studies: Blocking JOX3 activity to probe its role in JA metabolism.

Recommended validation criteria (based on antibody best practices) :

ParameterValidation Method
SpecificityKnockout mutant validation
SensitivityDilution series with controls
ReproducibilityIndependent lab verification

Research Implications

JOX3 antibodies could advance studies on:

  • Crop improvement: Engineering plants with modulated JA responses for pest resistance.

  • Hormonal crosstalk: Investigating interactions between JA, salicylic acid, and ethylene pathways.

  • Enzyme kinetics: Characterizing JOX3’s catalytic efficiency compared to other JOXs.

Challenges and Considerations

  • Commercial availability: No JOX3-specific antibodies are listed in current therapeutic or research databases .

  • Cross-reactivity risks: Antibodies targeting JOX3 must distinguish it from other JOX isoforms due to high sequence homology .

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
JRG21 antibody; At3g55970 antibody; F27K19_150Probable 2-oxoglutarate-dependent dioxygenase JRG21 antibody; EC 1.14.11.- antibody; Protein JASMONATE-REGULATED GENE 21 antibody
Target Names
JOX3
Uniprot No.

Target Background

Function
JOX3 is a 2-oxoglutarate-dependent dioxygenase that plays a crucial role in the oxidation of jasmonate (JA). JA is a stress-induced phytohormone produced in response to attacks by pathogens and herbivores. Its synthesis triggers the activation of defense responses through the JA-mediated signaling pathway. JOX3 converts JA into 12-hydroxyjasmonate (12OH-JA), an inactive form of JA. Importantly, JOX3 exhibits specificity for free JA, unable to oxidize the bioactive form jasmonoyl-L-isoleucine (JA-Ile) or other JA-amino acid conjugates. By preventing the over-accumulation of JA and indirectly its bioactive form JA-Ile under stress conditions, JOX3 acts as a negative regulator of JA-mediated defense signaling. Its contribution to 12OH-JA accumulation represses JA defense responses upon infection by the fungal pathogen *Botrytis cinerea*. Furthermore, JOX3 functions as a negative regulator of JA-mediated defense responses during infestation by the herbivorous caterpillar *Mamestra brassicae*.
Database Links

KEGG: ath:AT3G55970

STRING: 3702.AT3G55970.1

UniGene: At.5208

Protein Families
Iron/ascorbate-dependent oxidoreductase family

Q&A

What are the key considerations for antibody selection in experimental design?

When selecting antibodies for experimental applications, researchers should evaluate several critical factors. First, consider target specificity by reviewing validation data from manufacturers including Western blot results and immunoprecipitation studies. Second, assess antibody format (polyclonal, monoclonal, or recombinant) based on your experimental needs - polyclonals offer broader epitope recognition while monoclonals provide higher specificity for defined epitopes . Third, confirm species reactivity matches your experimental model. Finally, verify application compatibility (Western blotting, immunohistochemistry, flow cytometry) as antibodies optimized for one technique may perform poorly in others. Evidence-based selection significantly impacts experimental reproducibility and data quality.

What storage and handling practices maximize antibody performance and longevity?

Optimal antibody performance requires proper storage and handling protocols. Store most antibodies at -20°C for long-term preservation, avoiding repeated freeze-thaw cycles by preparing single-use aliquots . For working solutions, maintain at 4°C with appropriate preservatives (0.02-0.05% sodium azide) to prevent microbial contamination. Document lot numbers, receipt dates, and thawing events in laboratory records to track potential variability. Avoid protein degradation by minimizing exposure to direct light and maintaining recommended pH conditions. Implement regular quality control testing of archived antibodies through simple binding assays to verify retention of activity over time.

How can researchers distinguish between specific and non-specific antibody binding?

Differentiating specific from non-specific binding requires systematic control implementation. Include appropriate blocking agents (5% BSA, normal serum from secondary antibody host species) to minimize background signals. Implement isotype controls matching your primary antibody's class and species to identify Fc receptor-mediated binding . Perform adsorption controls by pre-incubating antibodies with excess target antigen before application, which should eliminate specific binding signals. For tissue staining, include secondary-only controls to detect endogenous tissue peroxidase activity or autofluorescence. Quantify signal-to-noise ratios across multiple experiments to establish reliable detection thresholds.

What strategies can optimize antibody-based detection in complex biological samples?

Maximizing antibody performance in complex samples requires technical optimization. Implement antigen retrieval techniques (heat-induced epitope retrieval, enzymatic digestion) when working with formalin-fixed samples to restore epitope accessibility. Titrate antibody concentrations systematically to identify the optimal signal-to-noise ratio for each specific application . Consider signal amplification systems (tyramide signal amplification, polymeric detection) for low-abundance targets. Reduce matrix interference by pre-absorbing antibodies against irrelevant proteins present in your experimental system. Employ sequential staining protocols with careful antibody stripping steps when performing multiplexed detection. These optimization strategies significantly enhance detection sensitivity while maintaining specificity.

How can researchers address cross-reactivity issues in antibody-based experiments?

Cross-reactivity challenges require methodical troubleshooting approaches. First, perform comprehensive in silico analysis of potential cross-reactive targets by searching protein databases for sequence homology. Validate antibody specificity through immunoprecipitation followed by mass spectrometry to identify all captured proteins . Implement competitive blocking experiments with recombinant proteins representing potential cross-reactive targets. Consider epitope-specific antibodies targeting unique regions rather than conserved domains when studying protein families. For critical applications, conduct parallel experiments with alternative antibodies targeting different epitopes to confirm consistent results. This multilayered approach effectively mitigates cross-reactivity risks.

What methodologies best quantify antibody-antigen binding dynamics?

Quantifying binding dynamics requires specialized biophysical techniques. Surface plasmon resonance (SPR) provides real-time measurement of association/dissociation kinetics and calculation of affinity constants (KD values). Bio-layer interferometry offers similar kinetic data with lower sample consumption. Microscale thermophoresis can measure interactions in complex biological fluids with minimal sample preparation . Isothermal titration calorimetry provides comprehensive thermodynamic profiles including enthalpy and entropy contributions to binding. For cellular contexts, fluorescence recovery after photobleaching (FRAP) can assess binding dynamics in living systems. The selection of appropriate methodology depends on research questions, sample availability, and required precision.

How can researchers design experiments to evaluate antibody specificity across diverse experimental platforms?

Comprehensive specificity evaluation requires coordinated cross-platform testing. Design a validation matrix incorporating multiple techniques (Western blot, immunoprecipitation, immunohistochemistry, flow cytometry) to assess performance consistency across applications . Implement parallel testing with orthogonal detection methods such as RNA expression analysis (qPCR, RNA-seq) to correlate protein and transcript levels. Include absorption controls with recombinant proteins or peptides corresponding to target epitopes. Systematically vary sample preparation conditions (fixation methods, detergent types, buffer compositions) to identify potential artifacts. Document all validation results in standardized formats for comprehensive comparison and reproducibility assessment.

How should researchers analyze contradictory antibody experimental results?

Resolving contradictory results requires systematic investigation of multiple variables. Begin by examining antibody characteristics including lot-to-lot variability, storage conditions, and epitope locations that might explain discrepancies . Compare experimental protocols in detail, focusing on sample preparation methods, incubation conditions, and detection systems. Consider biological variability factors including post-translational modifications, splice variants, or protein conformational states that may affect epitope accessibility. Implement statistical approaches like Bland-Altman analysis to quantify agreement between methods. Finally, design decisive experiments specifically addressing the contradiction, potentially using orthogonal methods like mass spectrometry for definitive target identification.

What statistical approaches are recommended for antibody binding data analysis?

Robust statistical analysis begins with appropriate experimental design. Implement randomization and blinding procedures to minimize unconscious bias. For binding curves, utilize non-linear regression models (typically four-parameter logistic functions) rather than linear approximations . Apply analysis of covariance (ANCOVA) when comparing binding curves across experimental conditions. For comparative studies, calculate minimum detectable differences during experimental design phase to ensure adequate statistical power. Implement mixed-effects models when analyzing hierarchical data structures (multiple measurements from single samples). Report confidence intervals alongside point estimates, and apply appropriate multiple comparison corrections for complex experimental designs.

How can dynamic characteristics of antibody responses be accurately quantified and interpreted?

Tracking antibody dynamics requires temporal sampling and mathematical modeling approaches. Design longitudinal sampling protocols with appropriate time intervals to capture critical kinetic phases (rapid early response, peak levels, decay rates) . Implement multiplex detection systems to simultaneously measure antibodies against multiple targets or epitopes. Apply mathematical modeling techniques including exponential decay models for clearance kinetics and power law models for affinity maturation processes. Calculate half-life values and area-under-curve metrics for comparative analysis between experimental conditions. Correlate antibody dynamics with functional parameters like neutralization activity to establish clinically relevant thresholds.

What methodologies allow differentiation between various antibody isotypes and subtypes in research samples?

Isotype and subclass differentiation requires specialized detection strategies. Implement isotype-specific secondary antibodies with verified specificity profiles in immunoassays. For higher resolution, utilize liquid chromatography with tandem mass spectrometry to identify diagnostic peptides unique to specific isotypes and subclasses . Apply isoelectric focusing followed by immunoblotting to separate closely related antibody variants based on charge differences. For complex samples, deploy Protein G/A differential binding assays exploiting varying affinities for different immunoglobulin classes. In functional studies, use selective proteases (e.g., IdeS for IgG subclass determination) followed by fragment analysis to determine subclass distributions precisely.

How are computational approaches enhancing antibody research and development?

Computational methodologies are transforming antibody research through multiple avenues. Machine learning algorithms now predict antibody-antigen binding properties from sequence data alone, accelerating candidate selection . Molecular dynamics simulations provide atomic-level insights into binding mechanisms and conformational changes upon antigen recognition. Bioinformatic pipelines integrate structural, genetic, and functional data to identify optimal epitopes for targeting. Network analysis techniques reveal antibody repertoire dynamics during immune responses. Looking forward, researchers should incorporate these computational approaches early in experimental design rather than as post-hoc analyses, creating iterative workflows where in silico predictions guide wet lab validation and experimental results refine computational models.

What emerging technologies are advancing antibody-based single-cell analysis?

Single-cell antibody analysis is rapidly evolving through technological integration. Spectral flow cytometry now enables simultaneous detection of 40+ antibody targets per cell through fluorophore unmixing algorithms . Mass cytometry (CyTOF) using metal-tagged antibodies allows even higher multiplexing without spectral overlap limitations. Spatial transcriptomics combined with in situ antibody detection provides unprecedented insights into protein-RNA relationships within tissue architectures. Microfluidic systems enable paired heavy/light chain sequencing with phenotypic profiling from individual B cells. These technologies generate multidimensional datasets requiring computational pipelines for visualization and interpretation, pushing researchers toward integrated experimental-analytical workflows for comprehensive immune cell characterization.

How can researchers design longitudinal antibody studies to track dynamic responses over extended timeframes?

Successful longitudinal antibody monitoring requires careful methodological planning. Design sampling schedules based on expected response kinetics, with more frequent collection during rapid change periods and strategic intervals during stability phases . Implement standardized processing protocols with consistent time-to-processing metrics to minimize technical variability. Prepare master calibrators and reference materials stored in single-use aliquots to normalize measurements across time points. Include internal bridge samples tested across multiple batches for drift correction. Deploy statistical methods specifically designed for longitudinal data, including mixed-effects models accounting for both within-subject and between-subject variability. These methodological considerations ensure that observed changes reflect true biological dynamics rather than technical artifacts.

What approaches optimize antibody therapeutics for targeting novel epitopes in complex diseases?

Optimizing antibody therapeutics for novel targets involves integrative discovery strategies. Implement high-throughput epitope mapping using peptide arrays, hydrogen-deuterium exchange mass spectrometry, and cryo-electron microscopy to identify accessible, functionally relevant epitopes . Apply phage display technologies with next-generation sequencing to rapidly select and characterize antibody candidates binding to challenging epitopes. Utilize structure-guided design integrating computational modeling with experimental validation to optimize antibody-antigen interfaces. Engineer antibody formats including bispecific constructs and antibody fragments for improved tissue penetration and novel functionalities. These methodological advances are expanding the therapeutic antibody landscape beyond traditional targets to address previously "undruggable" disease mechanisms.

What quality control metrics should researchers implement when working with antibodies?

Implementing comprehensive quality control requires standardized workflows. Document batch-specific validation including specificity testing against known positive and negative controls for each new antibody lot . Establish acceptance criteria for signal-to-noise ratios, background levels, and detection limits before initiating experiments. Prepare internal reference standards (lysates, purified proteins) for cross-batch normalization. Maintain detailed records of antibody performance across experiments to identify potential degradation over time. Include spike-recovery experiments to quantify matrix effects in complex samples. These systematic quality control procedures significantly enhance data reliability and facilitate troubleshooting when unexpected results arise.

How can researchers effectively design multiplexed antibody experiments?

Successful multiplexing requires methodical panel design and technical optimization. Begin with extensive literature review to identify potential interference between targets, particularly for signaling pathway studies where blockade of upstream components may affect downstream readouts . Carefully select antibody combinations avoiding species cross-reactivity when using multiple primary antibodies. For fluorescence-based detection, design panels accounting for spectral overlap, utilizing spread controls to establish compensation matrices. Implement titration matrices rather than individual titrations to identify optimal antibody concentrations within the multiplexed context. Validate multiplex results against single-plex measurements to confirm absence of interference. This systematic approach maximizes information yield while maintaining data integrity.

What standardization approaches improve reproducibility in antibody-based research?

Enhancing reproducibility requires implementing standardized workflows throughout the research process. Adopt detailed protocol registration before initiating experiments, documenting antibody selection criteria, validation methods, and planned analyses . Implement electronic laboratory notebooks capturing all experimental parameters including instrument settings, reagent lots, and environmental conditions. Utilize automated liquid handling when possible to minimize pipetting variability. Develop standardized reporting formats including minimum information about antibody experiments (MIABE) to facilitate method sharing and independent reproduction. Participate in multi-laboratory standardization initiatives using common reference materials to calibrate systems across research sites.

How should researchers approach antibody development for novel protein targets?

Developing antibodies against novel targets requires strategic immunization and screening approaches. Design immunogens based on structural analysis, selecting unique regions with high predicted antigenicity and surface accessibility . Implement diverse immunization strategies including DNA immunization, virus-like particles, and prime-boost approaches to maximize response diversity. Develop target-specific screening assays that mimic intended applications to select functionally relevant antibodies rather than merely high-affinity binders. Apply next-generation sequencing of B-cell repertoires to identify rare clones and track affinity maturation. These methodological considerations significantly improve success rates when developing antibodies against challenging novel targets.

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.