At2g43610 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

At2g43610 is a gene in Arabidopsis thaliana encoding a class-IV chitinase-like protein (KEGG ID: T00041) . Chitinases are hydrolytic enzymes that break down chitin, a structural polysaccharide in fungal cell walls and insect exoskeletons. The At2g43610 antibody is a specialized tool designed to detect and study this protein’s expression, localization, and functional roles in plant immunity .

Functional Role in Plant Immunity

At2g43610 is implicated in pathogen defense through chitin degradation, a key mechanism for disrupting fungal invaders . Its expression is induced during immune responses, as shown in studies on clubroot resistance in Brassica crops . The antibody facilitates investigations into:

  • Protein-protein interaction networks via immunoprecipitation-mass spectrometry (IP-MS) .

  • Subcellular localization during pathogen challenges.

  • Gene expression modulation under stress conditions (e.g., hydroponic systems with pathogen elicitors) .

Table 1: Experimental Insights into At2g43610

Study FocusMethodologyKey FindingsSource
Immune-related interactomeIP-MSIdentified interactors linked to defense signaling and stress response pathways.
Chitin degradationOligoarray analysisUpregulated 48 hours post-inoculation in resistant Arabidopsis lines.
Enzyme activity profilingqRT-PCR & SDS-PAGEValidated constitutive overexpression in pathogen-resistant cultivars.

Critical Observations:

  • At2g43610 contributes to basal immunity by degrading chitin from fungal pathogens .

  • Its antibody has been used to validate protein expression in transgenic plants, confirming its role in jasmonic acid signaling and systemic acquired resistance .

Applications in Biotechnology

The At2g43610 antibody is pivotal for:

  • Western blotting: Quantifying protein levels in mutant vs. wild-type plants .

  • Immunolocalization: Mapping tissue-specific expression during fungal infections .

  • Functional genomics: Screening knockout lines for altered disease susceptibility .

Challenges and Future Directions

While the antibody has enabled breakthroughs in plant immunity research, limitations include:

  • Cross-reactivity with homologous chitinases in related species .

  • Need for epitope-specific validation to avoid false positives in complex protein extracts .

Ongoing work focuses on engineering monoclonal variants of the antibody for improved specificity and high-throughput phenotyping .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At2g43610 antibody; F18O19.28 antibody; Endochitinase At2g43610 antibody; EC 3.2.1.14 antibody
Target Names
At2g43610
Uniprot No.

Q&A

What is the At2g43610 protein and why are antibodies against it important for plant research?

At2g43610 encodes a chitinase family protein in Arabidopsis thaliana that plays a critical role in plant defense mechanisms against fungal pathogens. Antibodies targeting this protein are valuable research tools for studying plant immunity, stress responses, and pathogen interactions. These antibodies enable researchers to detect, isolate, and characterize the protein's expression patterns, subcellular localization, and functional changes under various conditions. In experimental contexts, these antibodies facilitate immunoprecipitation, immunohistochemistry, and Western blot analyses that help elucidate the protein's role in plant defense signaling pathways and pathogen recognition mechanisms.

What are the optimal storage conditions for maintaining At2g43610 antibody activity?

To maintain optimal activity of At2g43610 antibodies, proper storage conditions are crucial. Most purified antibodies should be stored at -20°C for long-term stability, with working aliquots kept at 4°C to minimize freeze-thaw cycles. Storage buffers typically contain glycerol (50%) to prevent freezing damage, along with preservatives like sodium azide (0.02-0.05%) to inhibit microbial growth. Antibodies should be protected from direct light exposure, and repeated freeze-thaw cycles should be avoided as they can lead to protein denaturation and loss of binding activity. For monoclonal antibodies against At2g43610, manufacturer storage guidelines should be strictly followed, as some formulations may have specific requirements to maintain epitope recognition capabilities.

How should I design experiments to validate the specificity of new At2g43610 antibodies?

Validating the specificity of new At2g43610 antibodies requires a multi-faceted approach similar to those used in antibody development for therapeutic applications . First, perform Western blot analysis using protein extracts from wild-type Arabidopsis alongside At2g43610 knockout mutants. A specific antibody will detect a band of the expected molecular weight (approximately 30-35 kDa) in wild-type samples but not in knockout samples. Second, conduct immunoprecipitation followed by mass spectrometry to confirm that the antibody pulls down the target protein. Third, perform immunolocalization studies to verify that the observed subcellular localization matches the known distribution pattern of At2g43610. Additionally, heterologous expression systems can be used to express recombinant At2g43610 with epitope tags as positive controls. Cross-reactivity with other chitinase family proteins should be assessed through competitive binding assays. This comprehensive validation strategy ensures that experimental observations genuinely reflect At2g43610 biology rather than artifacts of non-specific binding.

What controls should be included when using At2g43610 antibodies for Western blot analysis?

When performing Western blot analysis with At2g43610 antibodies, several controls are essential to ensure reliable and interpretable results. At minimum, include:

  • Positive control: Protein extract from wild-type Arabidopsis thaliana tissues known to express At2g43610

  • Negative control: Protein extract from At2g43610 knockout mutants

  • Loading control: Detection of a constitutively expressed protein (such as actin or tubulin) to normalize protein loading

  • Molecular weight marker: To confirm the detected band is at the expected size

  • Secondary antibody-only control: To identify any non-specific binding of the secondary antibody

  • Peptide competition assay: Pre-incubation of the antibody with the immunizing peptide should abolish specific binding

Including these controls helps distinguish specific signals from background noise and enables accurate interpretation of experimental results. For quantitative Western blot analysis, standard curves using recombinant At2g43610 protein at known concentrations should be included to enable accurate quantification of the target protein levels across different samples.

How can I optimize immunoprecipitation protocols for At2g43610 protein interactions?

Optimizing immunoprecipitation (IP) protocols for studying At2g43610 protein interactions requires careful consideration of several parameters. Start by testing different lysis buffers to find one that effectively solubilizes the protein while preserving its interactions - typically, a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% NP-40 or Triton X-100, and protease inhibitors works well for plant proteins. The antibody-to-lysate ratio should be systematically optimized; typically starting with 2-5 μg of antibody per 500 μg of total protein. Pre-clearing the lysate with protein A/G beads can reduce non-specific binding.

For detecting transient or weak interactions, consider using crosslinking agents like formaldehyde or DSP (dithiobis(succinimidyl propionate)) before cell lysis. The incubation time should be optimized, with 2-4 hours at 4°C often providing a good balance between binding efficiency and background. Washing conditions are critical - start with less stringent washes and increase stringency as needed. For elution, both native (competitive elution with excess antigen) and denaturing (SDS buffer) methods should be tested to determine which best preserves the interactions of interest.

Active learning approaches similar to those described for antibody-antigen binding studies can be applied to systematically optimize these parameters with fewer experimental iterations .

How can I quantify At2g43610 expression levels using immunoblotting techniques?

Accurate quantification of At2g43610 expression levels using immunoblotting requires a standardized approach. Start with carefully optimized protein extraction and electrophoresis conditions to ensure consistent loading and transfer. For quantitative analysis, use a digital imaging system with a wide linear dynamic range rather than film-based detection. Always include a standard curve using purified recombinant At2g43610 protein at known concentrations (typically 5-7 points ranging from 0.1-100 ng) on each blot to enable absolute quantification.

Normalize target protein signals to a stable reference protein (such as actin or GAPDH) that shows minimal variation under your experimental conditions. For relative quantification between samples, the following formula can be applied:

Relative expression = (Intensity of At2g43610 band / Intensity of reference protein band) × 100%

When comparing expression across multiple blots, include a common reference sample on each blot as an inter-blot calibrator. Statistical analysis should account for technical replicates (multiple measurements from the same biological sample) and biological replicates (measurements from independent biological samples). A minimum of three biological replicates is recommended for robust statistical inference.

For temporal expression studies, consider displaying the data as shown in the table below:

Treatment Time (hours)Relative At2g43610 Expression (%)Statistical Significance
0 (Control)100 ± 8.5-
3142 ± 12.3p < 0.05
6215 ± 18.7p < 0.01
12273 ± 25.1p < 0.001
24196 ± 16.4p < 0.01

What approaches should I use to resolve contradictory results when using different At2g43610 antibodies?

When different At2g43610 antibodies yield contradictory results, a systematic troubleshooting approach is necessary. First, comprehensively characterize each antibody's properties, including the epitope recognized, production method (monoclonal vs. polyclonal), and validation history. Create a detailed comparison table documenting differences in experimental conditions, detection methods, and results for each antibody.

Consider the following strategies to resolve contradictions:

  • Epitope accessibility: Different antibodies may recognize distinct epitopes that vary in accessibility depending on protein conformation, post-translational modifications, or interaction partners. Map the epitopes recognized by each antibody and assess whether structural changes under your experimental conditions might affect epitope accessibility.

  • Validation in knockout lines: Test all antibodies in parallel using At2g43610 knockout mutants to assess specificity rigorously.

  • Orthogonal methods: Employ complementary techniques such as mass spectrometry, RNA-seq, or fluorescently tagged At2g43610 constructs to independently verify expression patterns or localization.

  • Antibody combinations: Similar to the approach used for SARS-CoV-2 antibodies , consider using antibody pairs that recognize different regions of the protein to increase specificity and reduce false results.

  • Cross-validation with multiple lots: Test different production lots of the same antibody to identify lot-to-lot variability issues.

Document all troubleshooting steps and their outcomes systematically. This approach not only helps resolve contradictions but can also yield valuable insights into At2g43610 biology, such as identifying previously unknown protein isoforms, post-translational modifications, or context-dependent conformational changes.

How can I use At2g43610 antibodies to study protein-protein interactions in planta?

Studying At2g43610 protein-protein interactions in planta requires sophisticated immunological approaches. Co-immunoprecipitation (co-IP) using At2g43610 antibodies is the foundation for such studies, allowing capture of the protein along with its interaction partners under near-native conditions. For optimal results, use a two-step cross-linking approach: first, a membrane-permeable crosslinker like DSP to stabilize protein complexes in intact cells, followed by a plant-specific extraction buffer (containing 100 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, protease inhibitors, and phosphatase inhibitors).

Proximity ligation assays (PLA) offer an alternative approach for visualizing protein interactions in fixed plant tissues. This technique uses pairs of antibodies (anti-At2g43610 and antibodies against suspected interaction partners) linked to oligonucleotides that, when in close proximity, enable rolling circle amplification and fluorescent detection of interaction sites with subcellular resolution.

Bimolecular fluorescence complementation (BiFC) can complement antibody-based approaches by expressing fusion proteins of At2g43610 and potential partners with split fluorescent protein segments. While not directly using antibodies, BiFC results can validate interactions detected by immunological methods.

For identifying novel interaction partners, antibody-based pull-downs followed by mass spectrometry provide a powerful discovery platform. This approach benefits from active learning methods similar to those described for antibody-antigen binding predictions , where machine learning algorithms can help prioritize candidate interactors for validation, reducing the experimental iterations needed to identify genuine biological interactions.

What strategies can improve detection sensitivity for low-abundance At2g43610 protein in plant tissues?

Detecting low-abundance At2g43610 protein in plant tissues presents significant technical challenges that require specialized strategies. Implement a multi-faceted approach to enhance sensitivity:

  • Sample preparation optimization: Use tissue-specific extraction methods with optimized buffers containing chaotropic agents (8M urea) to maximize protein solubilization, along with protease inhibitor cocktails to prevent degradation. Consider using subcellular fractionation to concentrate the protein from relevant cellular compartments.

  • Signal amplification techniques: Employ tyramide signal amplification (TSA) for immunohistochemistry and immunofluorescence applications, which can increase sensitivity by 10-100 fold compared to standard detection methods. This enzymatic amplification method uses the catalytic activity of HRP to generate multiple reporter molecules at the site of antibody binding.

  • Multiplex detection systems: Utilize multiple primary antibodies targeting different epitopes on At2g43610 simultaneously, each with distinguishable secondary antibodies, to increase the signal-to-noise ratio through signal convergence.

  • Advanced instrumentation: Use highly sensitive detection systems such as cooled CCD cameras for Western blot imaging or photomultiplier tubes for immunofluorescence microscopy. Consider single-molecule detection methods like ground state depletion microscopy followed by individual molecule return (GSDIM) for extremely low abundance proteins.

  • Antibody engineering: Consider using antibody fragments (Fab, scFv) which often provide better tissue penetration and reduced background compared to full IgG molecules. Alternatively, employ recombinant antibodies with enhanced affinity through directed evolution techniques.

A systematic comparison of detection limits for different methods is presented below:

Detection MethodMinimum Detectable At2g43610 (pg)Dynamic RangeSpecial Considerations
Standard Western blot100-500102High background with plant tissues
Chemiluminescent Western10-50103Requires optimization for plant extracts
Fluorescent Western5-25104Lower background, better quantification
TSA-enhanced immunodetection0.5-2103Complex protocol, potential artifacts
Capillary immunoassay1-5104Minimal sample requirement
Single-molecule detection0.05-0.1102Specialized equipment needed

How can I develop a high-throughput screening assay using At2g43610 antibodies?

Developing a high-throughput screening (HTS) assay using At2g43610 antibodies requires careful design to ensure sensitivity, specificity, and reproducibility across large sample numbers. Begin with assay format selection - sandwich ELISA typically offers the best combination of specificity and sensitivity for plant proteins. This requires two antibodies recognizing different epitopes: a capture antibody immobilized on a microplate and a detection antibody conjugated to an enzyme or fluorophore.

For optimal performance, the following parameters should be systematically optimized:

  • Antibody pair selection: Test multiple combinations of capture and detection antibodies to identify pairs with minimal interference and maximal signal-to-background ratio. Consider using active learning techniques similar to those described for antibody-antigen binding studies to efficiently identify optimal antibody combinations with fewer experimental iterations.

  • Buffer optimization: Develop plant-specific extraction and assay buffers that minimize matrix effects while preserving At2g43610 epitopes. Include plant-derived blocking proteins to reduce non-specific binding.

  • Miniaturization: Adapt the assay to 384-well or 1536-well formats to increase throughput and reduce reagent consumption. This requires careful optimization of liquid handling parameters and evaporation control.

  • Automation compatibility: Ensure all assay steps are compatible with liquid handling robotics, including optimized mixing parameters, incubation times, and washing procedures.

  • Quality control metrics: Implement comprehensive quality control metrics including Z'-factor calculation (aim for Z' > 0.5), coefficient of variation (target CV < 15%), and signal-to-background ratio (minimum 5:1).

For data analysis, develop machine learning algorithms to identify patterns in protein expression across different conditions. The table below outlines key assay performance metrics that should be achieved for a robust HTS assay:

Performance ParameterTarget ValueOptimization Strategy
Limit of detection≤ 50 pg/mLSignal amplification, antibody affinity maturation
Dynamic range≥ 3 log unitsOptimize antibody concentration and detection system
Z'-factor> 0.7Reduce variability through standardized protocols
Coefficient of variation< 10%Automation, standardized reagents, statistical outlier removal
Throughput≥ 10,000 samples/dayProcess optimization, parallel processing
Reagent stability≥ 6 months at 4°CBuffer optimization, stabilizing additives

What fixation and permeabilization methods are optimal for immunolocalization of At2g43610 in different plant tissues?

The choice of fixation and permeabilization methods significantly impacts the success of At2g43610 immunolocalization in plant tissues. Different plant tissues and subcellular compartments require tailored approaches to balance epitope preservation with adequate tissue penetration.

For whole-mount preparations of Arabidopsis seedlings, a shorter fixation (2 hours) in 2% paraformaldehyde with 0.1% Triton X-100 provides a good compromise between structural integrity and antibody penetration. Vacuum infiltration during fixation (3 cycles of 10 minutes at 15 inHg) significantly improves fixative penetration in dense tissues.

Permeabilization requirements vary by tissue type:

Tissue TypeRecommended PermeabilizationIncubation TimeSpecial Considerations
Leaf tissue0.5% Triton X-100 in PBS2 hoursCuticle may require additional treatment
Root tissue0.3% Triton X-100 in PBS1 hourGentle agitation improves penetration
Meristematic tissue0.2% Tween-20 + 1% DMSO3 hoursHigher detergent concentrations damage structural integrity
Mature stems0.5% Triton X-100 + cell wall digestion*4 hoursEnzymatic treatment essential for antibody access

*Cell wall digestion mix: 1% cellulase R10, 0.5% macerozyme R10, 0.4M mannitol, 20mM KCl, 10mM CaCl2, pH 5.7

Antigen retrieval methods should be tested when working with fixed tissues, as they can significantly improve antibody binding. Heat-induced epitope retrieval (HIER) using citrate buffer (10mM, pH 6.0) at 95°C for 20 minutes often enhances At2g43610 detection in recalcitrant tissues.

How can I troubleshoot non-specific binding when using At2g43610 antibodies in immunofluorescence?

Non-specific binding is a common challenge when using At2g43610 antibodies for immunofluorescence in plant tissues. A systematic troubleshooting approach targeting each potential source of non-specificity will help resolve these issues:

  • Blocking optimization: Plant tissues contain numerous compounds that can cause high background. Test different blocking agents including:

    • 5% normal serum from the same species as the secondary antibody

    • 3% BSA supplemented with 0.5% cold fish skin gelatin

    • Plant-specific blocking solution containing 2% non-fat dry milk and 0.5% polyvinylpyrrolidone (PVP)

    Extend blocking time to 2-3 hours at room temperature or overnight at 4°C for thick or dense tissues.

  • Antibody dilution optimization: Test a wider range of primary antibody dilutions than typically used (from 1:100 to 1:2000). Higher dilutions often reduce non-specific binding while maintaining specific signals if combined with longer incubation times (48-72 hours at 4°C).

  • Wash buffer modifications: Include additives in wash buffers to reduce non-specific interactions:

    • 0.05-0.1% Tween-20 to reduce hydrophobic interactions

    • 150-500 mM NaCl to disrupt ionic interactions

    • 0.1% Triton X-100 for more stringent washing of membrane-rich tissues

  • Pre-absorption controls: Pre-incubate the primary antibody with 10-100 fold excess of the immunizing peptide before application to tissues. Specific signals should be eliminated while non-specific binding will remain.

  • Secondary antibody optimization: Use highly cross-adsorbed secondary antibodies specifically tested for minimal reactivity with plant proteins. Consider using fragment antibodies (F(ab')₂) to reduce non-specific binding via Fc receptors.

  • Autofluorescence management: Plant tissues often exhibit significant autofluorescence. Implement:

    • Spectral unmixing during image acquisition

    • Sudan Black B treatment (0.1% in 70% ethanol) for 10 minutes after antibody incubation

    • Sodium borohydride treatment (0.1% in PBS) for 15 minutes before blocking

    • Selection of fluorophores with emission spectra distinct from plant autofluorescence (far-red dyes often work well)

By systematically addressing these factors and documenting the outcome of each modification, researchers can develop optimized protocols that maximize signal-to-noise ratios for At2g43610 immunodetection in diverse plant tissues.

What approaches can be used to generate and validate new At2g43610-specific monoclonal antibodies?

Generating and validating new At2g43610-specific monoclonal antibodies requires a comprehensive strategy that integrates computational prediction, experimental screening, and rigorous validation. The process should follow these key steps:

  • Epitope selection: Begin with in silico analysis of At2g43610 protein sequence to identify optimal epitopes based on:

    • Surface accessibility prediction

    • Sequence uniqueness compared to other chitinase family proteins

    • Evolutionary conservation analysis across plant species

    • Secondary structure predictions

    • Post-translational modification sites to avoid

  • Immunization strategy: Use multiple immunization approaches in parallel:

    • Synthetic peptides conjugated to KLH carrier protein

    • Recombinant protein fragments expressed in E. coli

    • DNA immunization with codon-optimized At2g43610 sequences

  • Hybridoma generation and screening: Implement a tiered screening approach:

    • Initial ELISA screening against immunizing antigens

    • Secondary screening against full-length recombinant At2g43610

    • Tertiary screening using Western blotting against plant extracts

    • Final validation in At2g43610 knockout plants

  • Clone selection and antibody production: Select 3-5 lead clones based on:

    • Affinity (measured by surface plasmon resonance)

    • Specificity in multiple applications

    • Stability during purification and storage

    • Recognition of native protein in plant extracts

  • Advanced validation: Apply comprehensive validation similar to therapeutic antibody development approaches :

    • Epitope mapping using peptide arrays or hydrogen-deuterium exchange

    • Cross-reactivity assessment against related chitinase family proteins

    • Performance evaluation in multiple applications (Western blot, IP, IF, IHC, ELISA)

    • Functional blocking assays if applicable

  • Quality control: Establish rigorous QC procedures:

    • Stability testing under various storage conditions

    • Lot-to-lot consistency monitoring

    • Application-specific performance standards

Active learning approaches for antibody selection, similar to those used in therapeutic antibody development , can significantly accelerate the process of identifying optimal antibody candidates. By applying a Hamming Average Distance method to analyze sequence diversity among antibody candidates, researchers can strategically select a diverse set of antibodies for validation with minimal experimental iterations. This approach has been shown to reduce the number of required experimental samples by 35% while maintaining comparable performance to exhaustive testing.

How can machine learning approaches enhance the use of At2g43610 antibodies in research applications?

Machine learning (ML) approaches are revolutionizing antibody applications in research, with several promising directions for enhancing At2g43610 antibody utilization. Based on recent advances in antibody-antigen prediction technologies , several ML-powered approaches can be implemented:

  • Epitope prediction and antibody design: Deep learning models similar to AbAgIntPre can predict optimal epitopes on At2g43610 protein and design antibodies with improved specificity and affinity. These models analyze sequence data to identify regions most likely to generate highly specific antibodies, potentially achieving ROC-AUC values of 0.82 or higher for binding prediction .

  • Image analysis automation: Convolutional neural networks (CNNs) can automatically analyze immunohistochemistry and immunofluorescence images to quantify At2g43610 expression levels, subcellular localization patterns, and co-localization with other proteins. This reduces analyst bias and enables high-throughput phenotyping in large-scale studies.

  • Active learning for experimental design: Similar to the approaches described for antibody-antigen binding studies , active learning algorithms can optimize experimental designs for At2g43610 antibody applications. These algorithms strategically select the most informative experiments to perform, reducing the number of experiments needed by up to 35% while maintaining comparable accuracy.

  • Antibody specificity prediction: Transformer-based models like ESM and attention-based networks similar to AttABseq can predict how mutations in the At2g43610 sequence might affect antibody binding, helping researchers anticipate cross-reactivity issues when studying protein variants or homologs. These models have demonstrated 120% better performance than traditional sequence-based methods .

  • Quality control automation: ML algorithms can analyze batch-to-batch variation in antibody performance, flagging potential quality issues before they impact experimental results. These systems learn from historical performance data to establish expected binding patterns and detect anomalies.

Implementation of these approaches requires interdisciplinary collaboration between plant biologists, immunologists, and computational scientists, but the potential benefits include significantly improved reproducibility, reduced experimental costs, and accelerated research timelines.

What are the considerations for using At2g43610 antibodies in multiplexed protein detection systems?

Multiplexed protein detection systems that incorporate At2g43610 antibodies enable simultaneous analysis of multiple proteins in the same sample, providing valuable insights into complex plant signaling networks. Successfully implementing such systems requires careful consideration of several technical factors:

  • Antibody compatibility: Select At2g43610 antibodies that are compatible with other antibodies in the multiplex panel. Key considerations include:

    • Species origin: Ideally, antibodies should be from different host species to enable species-specific secondary antibodies

    • Isotype diversity: When antibodies must be from the same species, use different isotypes (IgG1, IgG2a, IgG2b) to enable isotype-specific detection

    • Working concentration compatibility: All antibodies should perform optimally at similar working concentrations

  • Signal separation strategies: Implement robust methods to distinguish signals from different antibodies:

    • Spectral multiplexing: Use fluorophores with minimal spectral overlap and apply spectral unmixing algorithms

    • Sequential detection: For mass cytometry or cyclic immunofluorescence, optimize antibody stripping or quenching protocols between detection cycles

    • Spatial separation: For proximity-based techniques like PLA, carefully validate that signals represent genuine protein interactions rather than random proximity events

  • Cross-reactivity mitigation: Prevent unwanted antibody interactions:

    • Conduct extensive cross-reactivity testing between all components of the multiplex panel

    • Apply absorption steps to remove cross-reactive antibodies when necessary

    • Include blocking steps specific to plant tissues (1% PVP, 0.5% ovalbumin) to reduce non-specific binding

  • Quantification challenges: Address the unique challenges of quantifying multiple signals:

    • Establish protein-specific standard curves for each target in the multiplex

    • Apply computational methods to correct for signal spillover between channels

    • Validate quantification accuracy using samples with known ratios of target proteins

  • Data analysis complexity: Develop appropriate analytical frameworks:

    • Implement multivariate statistical methods to analyze correlation patterns

    • Apply dimensionality reduction techniques (PCA, t-SNE) to visualize complex relationships

    • Develop custom machine learning algorithms to classify cellular states based on protein expression patterns

The table below outlines compatibility considerations for different multiplexing platforms:

Multiplexing PlatformMax. ProteinsAt2g43610 Antibody RequirementsSpecial Considerations for Plant Tissues
Fluorescence multiplexing4-8Multiple epitopes, different speciesAutofluorescence mitigation critical
Mass cytometry (CyTOF)40+Metal-conjugated, high specificityCell wall digestion essential for metal penetration
Sequential immunofluorescence20-100Heat/chemical stability for multiple cyclesSignal loss with each stripping cycle
Proximity extension assay92+Oligonucleotide conjugation compatibilityPlant secondary metabolites may inhibit DNA polymerase
Microarray-based detection50+Retention of specificity when spottedBackground binding to plant lectins

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.