At3g59150 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Antibodies

Antibodies, also known as immunoglobulins, are proteins produced by the immune system to neutralize pathogens such as bacteria and viruses. They are crucial for the body's defense mechanisms and are also used extensively in research and medicine for diagnostics and therapeutics.

Structure and Function of Antibodies

Antibodies are composed of two heavy chains and two light chains, forming a Y-shaped structure. The variable regions at the tips of the Y bind to specific antigens, while the constant regions determine the antibody's class and effector functions. There are five main classes of antibodies: IgG, IgA, IgM, IgE, and IgD, each with distinct functions and distributions in the body .

Researching Specific Antibodies

When researching a specific antibody like "At3g59150 Antibody," it is essential to identify its target antigen, class, and any relevant applications in research or medicine. This information is typically found in scientific literature or databases dedicated to antibody characterization.

Potential Applications of Antibodies

Antibodies are used in various applications, including:

  • Diagnostic Tools: For detecting specific proteins or pathogens.

  • Therapeutic Agents: To treat diseases by targeting specific molecules.

  • Research Tools: In techniques like Western blotting and immunofluorescence.

Challenges in Antibody Research

A significant challenge in antibody research is ensuring specificity and efficacy. Studies have shown that many commercial antibodies fail to recognize their intended targets, highlighting the need for rigorous characterization and validation .

Example Data Table for Antibody Characteristics

Antibody ClassHeavy ChainStructureFunction
IgGγMonomerMost common, provides long-term immunity
IgAαMonomer/DimerProtects mucosal surfaces
IgMμPentamerFirst line of defense
IgEεMonomerInvolved in allergic reactions
IgDδMonomerFound on mature B cells

This table illustrates the characteristics of different antibody classes, which can serve as a framework for understanding specific antibodies once their details are identified.

References

  1. Immunopaedia: Provides detailed information on antibody structure and classes.

  2. eLife Sciences: Discusses the challenges and advancements in antibody characterization.

  3. Thermofisher: Offers insights into antibody structure and classification.

  4. Wikipedia: General information on IgG antibodies.

  5. UCLA Newsroom: Research on genes linked to high antibody production.

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate-Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
At3g59150 antibody; F17J16.200 antibody; F25L23.10F-box protein At3g59150 antibody
Target Names
At3g59150
Uniprot No.

Q&A

What is At3g59150 and why is it important in plant research?

At3g59150 is a gene in Arabidopsis thaliana (thale cress) that encodes a cyclin-like F-box family protein. This protein is significant in plant research because it has been identified as a potential calmodulin (CaM)-binding protein, suggesting its involvement in calcium signaling pathways . F-box proteins generally function as part of SCF (Skp, Cullin, F-box) ubiquitin ligase complexes that target proteins for degradation, thereby regulating various cellular processes.

The importance of At3g59150 is highlighted in studies showing its interaction with calcium sensors in plant immunity and stress responses. According to differential binding analyses, At3g59150 shows interaction with CaM, which plays crucial roles in plant defense mechanisms against pathogens .

How do I determine if an At3g59150 antibody is specific?

Determining antibody specificity requires a multi-step validation approach:

  • Western blot analysis: Test the antibody against wild-type plant extracts alongside At3g59150 knockout/knockdown mutants. A specific antibody will show a band of the expected molecular weight in wild-type samples that is absent or significantly reduced in mutant samples .

  • Immunoprecipitation followed by mass spectrometry: This approach can confirm that the antibody pulls down the correct protein. The immunoprecipitated protein should be identified as At3g59150 by mass spectrometry analysis .

  • Recombinant protein controls: Express and purify At3g59150 protein and use it as a positive control in your antibody validation experiments .

  • Cross-reactivity testing: Test the antibody against closely related F-box proteins to ensure it doesn't recognize other family members. This is particularly important as the Arabidopsis genome contains numerous F-box protein genes with sequence similarities .

The YCharOS group found that 50-75% of commercial antibodies perform well in their intended applications, emphasizing the importance of thorough validation .

What are the optimal sample preparation methods for detecting At3g59150 in plant tissues?

Optimal sample preparation for At3g59150 detection varies by application:

For Western blotting:

  • Harvest fresh plant tissue and flash-freeze in liquid nitrogen

  • Grind tissue to a fine powder while maintaining freezing temperatures

  • Extract proteins using a buffer containing:

    • 50 mM Tris-HCl (pH 7.5)

    • 150 mM NaCl

    • 1% Triton X-100

    • 0.5% sodium deoxycholate

    • Protease inhibitor cocktail

  • Clarify lysates by centrifugation (15,000 × g, 15 min, 4°C)

  • Quantify protein concentration using Bradford or BCA assay

  • Denature samples with SDS loading buffer at 95°C for 5 minutes

For immunohistochemistry:

  • Fix tissue samples in 4% paraformaldehyde

  • Embed in paraffin or freeze in OCT compound

  • Section to 5-10 μm thickness

  • Perform antigen retrieval using citrate buffer (pH 6.0)

  • Block with 5% BSA or normal serum to reduce background

These methods are adapted from standard protocols used in plant protein research and should be optimized for At3g59150 detection based on the antibody manufacturer's recommendations .

How can I optimize co-immunoprecipitation experiments to study At3g59150 interactions with calmodulin?

Optimizing co-immunoprecipitation (co-IP) experiments for studying At3g59150-calmodulin interactions requires careful attention to several methodological aspects:

Optimization protocol:

  • Buffer composition: Use a calcium-containing buffer (typically 0.1-0.5 mM CaCl₂) since CaM-binding is often calcium-dependent. Include:

    • 50 mM Tris-HCl (pH 7.5)

    • 150 mM NaCl

    • 0.5% NP-40 or Triton X-100

    • 0.1-0.5 mM CaCl₂

    • Protease inhibitor cocktail

  • Antibody selection: Use either anti-At3g59150 or anti-CaM antibodies for the initial pull-down. Consider testing both approaches as one may yield better results.

  • Cross-linking (optional): For transient interactions, use membrane-permeable cross-linkers like DSP (dithiobis[succinimidyl propionate]) at 1-2 mM for 30 minutes before lysis.

  • Controls:

    • Include EGTA in parallel samples to chelate calcium, which should disrupt calcium-dependent interactions

    • Use knockout/knockdown lines as negative controls

    • Include irrelevant antibodies of the same isotype as procedural controls

  • Verification methods:

    • After co-IP, confirm the presence of both proteins by Western blot

    • Consider using recombinant proteins in pull-down assays to validate direct interactions

Research from Popescu et al. demonstrated that At3g59150 interacts with CaM in both in vitro and in vivo settings, using similar methodologies to study protein-CaM interactions .

What approaches can I use to map the specific CaM-binding domains in At3g59150?

Mapping CaM-binding domains in At3g59150 requires systematic analysis using multiple complementary techniques:

Methodological approaches:

  • Bioinformatic prediction:

    • Use algorithms like Calmodulin Target Database to identify potential CaM-binding motifs in the At3g59150 sequence

    • Look for characteristic features of CaM-binding domains: basic amphipathic helices with hydrophobic anchor residues

  • Truncation/deletion analysis:

    • Generate a series of truncated At3g59150 constructs

    • Express these as recombinant proteins

    • Test each for CaM binding using pull-down assays or surface plasmon resonance (SPR)

    • Narrow down the binding region through progressive deletions

  • Site-directed mutagenesis:

    • Once potential binding regions are identified, mutate key residues (especially hydrophobic anchor residues)

    • Test mutants for altered CaM binding affinity

  • Peptide competition assays:

    • Synthesize peptides corresponding to predicted CaM-binding regions

    • Use these in competition assays with full-length protein to confirm binding sites

  • Structural analysis:

    • If possible, perform NMR or X-ray crystallography of the At3g59150-CaM complex

    • Alternatively, use hydrogen-deuterium exchange mass spectrometry to identify protected regions

Similar approaches were successfully used to characterize CaM-binding domains in PEN3, which showed Ca²⁺-dependent interaction with CaM both in vitro and in vivo .

How can epitope tagging of At3g59150 affect its interaction with calcium sensors?

Epitope tagging of At3g59150 can significantly impact its interactions with calcium sensors like calmodulin, requiring careful experimental design and validation:

Impact and considerations:

  • Tag position effects:

    • N-terminal tags may disrupt signal peptides or targeting sequences

    • C-terminal tags may interfere with C-terminal CaM-binding domains, which are common in many CaM-binding proteins

    • Internal tags can disrupt protein folding and function

  • Tag size considerations:

    • Small tags (FLAG, HA, Myc) typically cause minimal disruption

    • Larger tags (GFP, YFP) may sterically hinder protein-protein interactions

    • Consider using cleavable tags for downstream functional assays

  • Validation approaches:

    • Compare CaM-binding between tagged and untagged versions using in vitro pull-down assays

    • Test whether calcium-dependent regulation of tagged At3g59150 remains intact

    • Perform complementation studies in knockout lines to verify functionality

  • Alternative strategies:

    • Use split-tag approaches where the tag is inserted in non-critical regions

    • Consider proximity labeling approaches (BioID, APEX) as alternatives to direct tagging

    • Use antibodies against the native protein for interaction studies when possible

What controls should I include when validating an At3g59150 antibody?

A comprehensive validation of At3g59150 antibodies requires multiple controls to ensure specificity and reliability:

Essential controls:

  • Genetic controls:

    • Knockout/knockdown samples: Tissue/extracts from At3g59150 knockout or RNAi lines should show absent or significantly reduced signal

    • Overexpression samples: Samples overexpressing At3g59150 should show enhanced signal

  • Peptide competition assays:

    • Pre-incubate antibody with excess immunizing peptide/antigen

    • This should abolish specific signals in subsequent experiments

  • Orthogonal detection methods:

    • Verify protein expression using multiple antibodies targeting different epitopes

    • Correlate protein detection with mRNA expression (qRT-PCR or RNA-seq)

  • Cross-reactivity assessment:

    • Test on recombinant proteins of related F-box family members

    • Check antibody specificity against extracts from species with differing At3g59150 homology

  • Application-specific controls:

    • Western blot: Include molecular weight markers; anticipate ~53 kDa band for At3g59150

    • Immunoprecipitation: Use non-immune IgG of same species as procedural control

    • Immunohistochemistry: Include secondary-only controls and tissue processing controls

The YCharOS group demonstrated that antibody validation using knockout cell lines is superior to other types of controls, particularly for Western blots and immunofluorescence imaging .

How can I optimize immunoprecipitation protocols for low-abundance At3g59150 protein?

Optimizing immunoprecipitation (IP) of low-abundance At3g59150 requires enhancing sensitivity while maintaining specificity:

Optimization strategy:

  • Starting material preparation:

    • Increase starting material (2-3× standard amount)

    • Use tissue with highest At3g59150 expression (based on transcriptomic data)

    • Consider using stress conditions that might upregulate the protein

  • Lysis optimization:

    • Test different lysis buffers (RIPA, NP-40, Digitonin) to maximize extraction efficiency

    • Include proteasome inhibitors (MG132) alongside protease inhibitors

    • Perform sequential extractions to improve solubilization

  • Antibody considerations:

    • Use high-affinity antibodies (validate using recombinant protein)

    • Cross-link antibodies to beads to eliminate antibody contamination in eluates

    • Optimize antibody:lysate ratios (typically 2-5 μg antibody per mg total protein)

  • IP procedure enhancements:

    • Extend incubation time (overnight at 4°C)

    • Use gentle rotation to maximize binding while minimizing denaturation

    • Perform sequential IPs on the same lysate

  • Detection enhancements:

    • Use high-sensitivity ECL substrates for Western blot detection

    • Consider concentrating eluted proteins using TCA precipitation

    • Use silver staining for gel detection (50-100× more sensitive than Coomassie)

Similar approaches were used successfully to study low-abundance proteins in the PEN3 interactome, allowing the identification of calcium sensors that interact with PEN3 .

What are the best approaches for studying At3g59150 subcellular localization?

Multiple complementary approaches should be used to study At3g59150 subcellular localization accurately:

Technical approaches:

  • Fluorescent protein fusion strategies:

    • Create both N- and C-terminal GFP/YFP fusions

    • Generate stable Arabidopsis transgenic lines under native promoter control

    • Validate functionality through complementation of knockout phenotypes

    • Image using confocal microscopy with appropriate cellular markers

  • Immunofluorescence microscopy:

    • Use validated anti-At3g59150 antibodies

    • Fix tissues with 4% paraformaldehyde to preserve structure

    • Perform antigen retrieval if necessary

    • Include co-staining with organelle markers

  • Subcellular fractionation:

    • Perform differential centrifugation to separate cellular compartments

    • Analyze fractions by Western blotting with anti-At3g59150 antibody

    • Include marker proteins for each subcellular compartment as controls

  • Inducible expression systems:

    • Use estradiol or dexamethasone-inducible systems for temporal control

    • Monitor localization changes during induction

    • Assess localization changes under stress/stimulus conditions

  • Advanced imaging techniques:

    • Consider FRET or BiFC to study protein-protein interactions in situ

    • Use super-resolution microscopy for detailed localization

    • Implement photo-activatable fluorescent proteins for dynamic studies

Similar approaches were successfully used for studying PEN3 localization, revealing its distribution on the plasma membrane and accumulation at pathogen penetration sites upon attack by fungal pathogens .

How can I measure changes in At3g59150 protein levels in response to calcium signaling?

Measuring At3g59150 protein dynamics in response to calcium signaling requires methods that capture both abundance and post-translational modifications:

Methodological approaches:

  • Time-course experiments:

    • Treat plants with calcium ionophores (A23187, ionomycin) or calcium-mobilizing stimuli

    • Harvest samples at multiple timepoints (0, 15, 30, 60, 120 min)

    • Process for protein extraction using calcium-preserving buffers

  • Quantitative Western blotting:

    • Use internal loading controls (anti-actin, anti-tubulin)

    • Implement fluorescent secondary antibodies for more accurate quantification

    • Analyze using ImageJ or similar software for densitometry

    • Generate calibration curves using recombinant protein standards

  • Pulse-chase experiments:

    • Use inducible expression systems with epitope-tagged At3g59150

    • Induce expression transiently, then monitor protein decay rates

    • Compare stability under different calcium conditions

  • Phosphorylation-specific detection:

    • Use Phos-tag gels to separate phosphorylated forms

    • Perform immunoprecipitation followed by phospho-specific Western blotting

    • Consider phosphoproteomics to identify specific modified residues

  • Live-cell imaging:

    • Use fluorescent protein fusions to monitor localization changes

    • Implement FRET-based calcium sensors for simultaneous calcium measurement

    • Perform photobleaching recovery (FRAP) to assess protein mobility

Research on calcium signaling proteins has demonstrated that calcium fluctuations can alter protein stability, localization, and function. For instance, calcium sensors in the PEN3 interactome show dynamic responses to calcium fluctuations during pathogen attack .

How can I implement proximity labeling to study the At3g59150 interactome?

Proximity labeling offers powerful approaches for mapping the At3g59150 protein interaction network in near-native conditions:

Implementation strategy:

  • Construct design:

    • Create fusion proteins of At3g59150 with BioID2, TurboID, or APEX2

    • Generate both N- and C-terminal fusions under native promoter control

    • Validate expression and functionality in complementation studies

  • Experimental setup:

    • For BioID/TurboID: Supply biotin (50 μM) for labeling periods (1-18 hours)

    • For APEX2: Treat with biotin-phenol (500 μM) and H₂O₂ pulse (1 mM, 1 min)

    • Include appropriate controls (untransformed plants, catalytically inactive versions)

    • Test labeling efficiency using streptavidin blotting

  • Stimulus-specific interactome capture:

    • Apply calcium-mobilizing treatments during labeling window

    • Compare interactomes under resting and stimulated conditions

    • Implement short labeling windows with TurboID to capture dynamic interactions

  • Sample processing and analysis:

    • Isolate biotinylated proteins using streptavidin affinity purification

    • Analyze by mass spectrometry to identify labeled proteins

    • Implement quantitative proteomics (SILAC or TMT) for comparative studies

  • Validation approaches:

    • Confirm key interactions using traditional co-IP or BiFC

    • Use knockout lines of putative interactors to assess functional relationships

    • Map interaction domains through truncation/deletion studies

This approach is particularly valuable for identifying transient calcium-dependent interactions, similar to those described for PEN3 and calmodulin .

What are the considerations for developing recombinant antibodies against At3g59150?

Developing recombinant antibodies against At3g59150 involves specific considerations to ensure specificity and functionality:

Development considerations:

  • Antigen design and selection:

    • Choose unique regions of At3g59150 with low homology to related F-box proteins

    • Consider both full-length protein and specific peptide epitopes

    • Express recombinant fragments that maintain native conformation

    • Avoid regions involved in CaM binding if studying these interactions

  • Antibody format selection:

    • Single-chain variable fragments (scFvs) for applications requiring small size

    • Antigen-binding fragments (Fabs) for better stability

    • Full IgG for applications requiring effector functions or longer half-life

    • Nanobodies (VHH) for accessing restricted epitopes

  • Library screening strategy:

    • Implement phage, yeast, or mammalian display technologies

    • Use competitive elution to identify high-affinity binders

    • Screen against multiple conformations if protein structure is dynamic

    • Counter-screen against related proteins to ensure specificity

  • Validation requirements:

    • Test against recombinant target and plant extracts

    • Verify specificity using knockout/knockdown lines

    • Validate in multiple applications (Western, IP, IF)

    • Sequence antibodies and make sequences publicly available

  • Production considerations:

    • Express in suitable systems (bacteria, yeast, mammalian cells)

    • Implement affinity tags for purification

    • Verify activity after purification and storage

The NeuroMab facility's approach demonstrates the value of comprehensive screening (~1,000 clones) followed by validation in multiple assays to obtain highly specific antibodies .

How can single-cell techniques be applied to study At3g59150 expression in different cell types?

Single-cell approaches offer unprecedented resolution for studying At3g59150 expression patterns across different plant cell types:

Implementation approaches:

  • Single-cell RNA sequencing (scRNA-seq):

    • Optimize protoplast isolation from relevant plant tissues

    • Use droplet-based (10x Genomics) or plate-based (SMART-seq) platforms

    • Include cell-type specific markers for post-hoc identification

    • Analyze At3g59150 expression across identified cell clusters

  • Single-cell proteomics approaches:

    • Implement nanoPOTS (Nanodroplet Processing in One pot for Trace Samples)

    • Use CyTOF (mass cytometry) with metal-labeled antibodies

    • Apply SCoPE-MS (Single Cell ProtEomics by Mass Spectrometry)

    • Correlate protein expression with transcriptomic data

  • Single-cell spatial transcriptomics:

    • Apply Slide-seq or 10x Visium to maintain spatial context

    • Use FISH-based methods (seqFISH, MERFISH) for targeted gene panels

    • Correlate spatial expression with tissue functions and responses

  • Reporter-based approaches:

    • Generate At3g59150 promoter-GFP/LUC reporter lines

    • Use cell-type specific markers for co-localization studies

    • Implement microfluidic devices for live-cell tracking

  • Single-cell epigenomics:

    • Apply scATAC-seq to study chromatin accessibility at the At3g59150 locus

    • Implement single-cell ChIP-seq to study histone modifications

    • Correlate epigenetic state with expression levels

Similar approaches using nanovials technology have been successfully applied to study gene expression in human cells, allowing correlation between secreted proteins and gene expression profiles at single-cell resolution .

What methods can be used to study the dynamics of At3g59150 in response to pathogen infection?

Understanding At3g59150 dynamics during pathogen infection requires time-resolved approaches that capture multiple aspects of protein function:

Methodological approaches:

  • Time-resolved expression analysis:

    • Perform time-course sampling after pathogen inoculation

    • Analyze both transcript (qRT-PCR, RNA-seq) and protein levels (Western blot)

    • Include multiple pathogen types (bacterial, fungal, oomycete)

    • Compare compatible vs. incompatible interactions

  • Fluorescent reporter systems:

    • Generate At3g59150-FP fusion under native promoter control

    • Monitor localization changes using confocal microscopy

    • Implement time-lapse imaging during pathogen challenge

    • Co-visualize with pathogen structures using dual-labeling

  • Protein modification analysis:

    • Track post-translational modifications (phosphorylation, ubiquitination)

    • Use Phos-tag gels or phospho-specific antibodies

    • Perform immunoprecipitation followed by mass spectrometry

    • Compare modification patterns between mock and infected samples

  • Protein-protein interaction dynamics:

    • Use split luciferase complementation assays for real-time tracking

    • Implement FRET sensors to monitor specific interactions

    • Perform time-resolved co-immunoprecipitation

    • Apply proximity labeling with short labeling windows

  • Functional analysis:

    • Generate conditional knockout/knockdown lines

    • Activate/repress At3g59150 at different infection stages

    • Assess impact on defense responses and pathogen growth

    • Monitor calcium signaling using genetically encoded calcium indicators

Similar approaches were used to study PEN3 dynamics during pathogen infection, revealing its accumulation at pathogen penetration sites and involvement in nonhost resistance .

How should I analyze and interpret contradictory results from different At3g59150 antibodies?

Analyzing contradictory results from different At3g59150 antibodies requires systematic troubleshooting and validation:

Analysis framework:

  • Antibody characteristics assessment:

    • Compare epitopes targeted by each antibody

    • Evaluate validation data provided by manufacturers

    • Consider antibody format (monoclonal vs. polyclonal, recombinant vs. conventional)

    • Review purification methods used for each antibody

  • Experimental condition evaluation:

    • Compare sample preparation methods (fixation, buffer composition)

    • Assess detection methods and sensitivity

    • Consider protein modifications that might mask epitopes

    • Evaluate potential cross-reactivity with related proteins

  • Systematic validation approach:

    • Test all antibodies side-by-side under identical conditions

    • Include genetic controls (knockout/overexpression)

    • Perform peptide competition assays for each antibody

    • Test against recombinant At3g59150 protein

  • Orthogonal method corroboration:

    • Correlate with mRNA expression data

    • Use epitope-tagged versions as reference standards

    • Apply mass spectrometry to verify protein identity

  • Interpretation guidelines:

    • Trust results that are consistent with genetic controls

    • Consider that different antibodies may recognize different protein isoforms or states

    • Document all findings, including negative results, for transparency

Research has shown that an average of ~12 publications per protein target included data from antibodies that failed to recognize the relevant target protein, highlighting the importance of thorough validation .

What statistical approaches are appropriate for quantifying At3g59150 expression changes in immunoblotting experiments?

Statistical methodology:

  • Experimental design considerations:

    • Include at least 3-4 biological replicates per condition

    • Randomize sample processing order to avoid batch effects

    • Include technical replicates when possible

    • Plan appropriate statistical tests before experimentation

  • Quantification approaches:

    • Use densitometry software (ImageJ, ImageLab, FIJI)

    • Normalize to loading controls (actin, tubulin, total protein stain)

    • Generate standard curves using recombinant protein for absolute quantification

    • Log-transform data if not normally distributed

  • Statistical tests for different experimental designs:

    • Two conditions: Student's t-test or Mann-Whitney U test

    • Multiple conditions: ANOVA with appropriate post-hoc tests

    • Time course experiments: Repeated measures ANOVA

    • Dose-response: Regression analysis or non-linear curve fitting

  • Addressing common challenges:

    • Signal saturation: Work within linear range of detection

    • Heteroscedasticity: Apply appropriate transformations

    • Multiple testing: Apply Bonferroni or Benjamini-Hochberg corrections

    • Low sample size: Consider non-parametric tests or bootstrapping

  • Data visualization:

    • Include representative blot images alongside quantification

    • Show individual data points rather than just means

    • Include error bars representing standard deviation or SEM

    • Use consistent scaling across comparable experiments

How can I integrate antibody-based data with transcriptomic and proteomic datasets for At3g59150?

Integrating antibody-based data with other -omics approaches provides a comprehensive understanding of At3g59150 regulation and function:

Integration framework:

  • Data harmonization:

    • Standardize sample conditions across experiments

    • Normalize data to allow cross-platform comparisons

    • Account for different measurement scales and dynamics

    • Consider temporal relationships between transcript and protein changes

  • Correlation analysis:

    • Calculate Pearson or Spearman correlations between:

      • Transcript levels (RNA-seq/microarray) and protein abundance

      • Protein abundance and post-translational modifications

      • Protein levels and interactor abundance

    • Visualize relationships using scatterplots or heatmaps

  • Multi-omics data integration techniques:

    • Implement MOFA (Multi-Omics Factor Analysis)

    • Apply DIABLO (Data Integration Analysis for Biomarker discovery)

    • Use network-based approaches to identify functional modules

    • Consider Bayesian approaches for causal relationship inference

  • Pathway/enrichment analysis:

    • Map integrated data to known signaling pathways

    • Perform Gene Ontology enrichment on correlated genes/proteins

    • Implement GSEA (Gene Set Enrichment Analysis) on ranked lists

    • Use Cytoscape for network visualization and analysis

  • Validation of key findings:

    • Verify unexpected patterns through targeted experiments

    • Use genetic perturbation to test proposed relationships

    • Apply mathematical modeling to test hypothesized mechanisms

This integrated approach can reveal regulatory mechanisms not apparent from single data types, such as post-transcriptional regulation, protein stability changes, or context-dependent interactions in calcium signaling networks involving At3g59150 .

How might CRISPR-based approaches enhance At3g59150 antibody validation and functional studies?

CRISPR technologies offer powerful new approaches for antibody validation and functional characterization of At3g59150:

Innovative applications:

  • Endogenous tagging for antibody validation:

    • Use CRISPR-Cas9 to insert epitope tags at the At3g59150 locus

    • Generate homozygous tagged lines for definitive validation

    • Compare commercial antibody signals with epitope tag detection

    • Create knock-in fluorescent protein fusions for localization studies

  • Knockout lines as negative controls:

    • Generate complete At3g59150 knockout lines

    • Create conditional knockout systems using inducible CRISPR

    • Develop tissue-specific knockouts using cell-type-specific promoters

    • Use knockout tissues as definitive negative controls for antibody validation

  • Base editing applications:

    • Introduce specific mutations in CaM-binding domains

    • Modify key regulatory residues without disrupting the entire protein

    • Create phosphomimetic or phosphodeficient variants

    • Alter ubiquitination sites to assess stability regulation

  • Prime editing for complex modifications:

    • Insert specific sequence modifications without double-strand breaks

    • Create domain swaps to test functional equivalence

    • Introduce reporter elements at the endogenous locus

    • Develop allelic series with graduated functional changes

  • Epigenome editing:

    • Modify chromatin at the At3g59150 locus to regulate expression

    • Recruit activators or repressors to study expression dynamics

    • Implement CUT&RUN for high-resolution chromatin mapping

    • Use CRISPR interference/activation for reversible regulation

The YCharOS group demonstrated that knockout validation is superior to other validation methods for antibodies, and CRISPR technologies make this approach increasingly accessible .

What are the prospects for developing antibodies that specifically recognize post-translationally modified forms of At3g59150?

Developing modification-specific antibodies for At3g59150 presents both challenges and opportunities:

Development prospects:

  • Phospho-specific antibody development:

    • Identify potential phosphorylation sites through phosphoproteomics

    • Synthesize phosphopeptides for specific sites as immunogens

    • Implement negative selection against non-phosphorylated peptides

    • Validate using phosphatase treatment and phosphomimetic mutants

  • Ubiquitination-specific antibodies:

    • Generate antibodies recognizing the At3g59150-ubiquitin junction

    • Develop antibodies to specific ubiquitin chain topologies

    • Use branched peptides as immunogens for polyubiquitination sites

    • Validate using ubiquitination-deficient mutants

  • Conformation-specific antibodies:

    • Develop antibodies that specifically recognize calcium-bound forms

    • Use structural data to identify conformation-specific epitopes

    • Implement selection strategies with conformational constraints

    • Validate using conditions that shift conformational equilibria

  • Site-specific antibody engineering:

    • Apply phage display to select high-affinity, specific binders

    • Implement recombinant antibody approaches for reproducibility

    • Use synthetic biology to create modular recognition domains

    • Engineer bispecific antibodies recognizing protein and modification

  • Validation strategies:

    • Use CRISPR to generate modification-site mutants

    • Implement in vitro modification systems as positive controls

    • Apply specific enzymes (phosphatases, deubiquitinases) as negative controls

    • Correlate with mass spectrometry data on modification status

Similar approaches have been used to develop modification-specific antibodies for other proteins involved in signaling pathways, allowing researchers to track dynamic post-translational modifications during cellular responses .

How might artificial intelligence and machine learning improve At3g59150 antibody design and validation?

AI and machine learning approaches are transforming antibody research with applications to At3g59150 studies:

AI applications in antibody research:

  • Epitope prediction and optimization:

    • Apply deep learning to predict optimal epitopes in At3g59150

    • Use structural prediction (AlphaFold2) to identify surface-exposed regions

    • Implement immunogenicity prediction algorithms

    • Identify cross-reactive epitopes in related proteins for exclusion

  • Antibody sequence optimization:

    • Design CDR sequences with optimal binding properties

    • Predict stability and solubility of candidate antibodies

    • Optimize framework regions to minimize immunogenicity

    • Improve manufacturability through sequence engineering

  • Validation dataset analysis:

    • Develop ML classifiers to evaluate antibody validation data

    • Identify patterns in failed antibody validations to avoid pitfalls

    • Generate statistical models of antibody performance across applications

    • Implement transfer learning from related proteins to At3g59150

  • Image analysis for localization studies:

    • Apply computer vision to quantify protein localization patterns

    • Develop automated co-localization analysis

    • Implement segmentation algorithms for subcellular structures

    • Create classification systems for phenotypic responses

  • Experimental design optimization:

    • Use active learning to identify optimal experimental conditions

    • Develop Bayesian optimization frameworks for antibody validation

    • Implement design of experiments (DOE) approaches

    • Create predictive models of antibody performance across conditions

AI approaches can significantly reduce the time and resources required for antibody development and validation, potentially transforming the field of plant protein research .

What are the emerging technologies for studying low-abundance proteins like At3g59150 in plant systems?

Emerging technologies are addressing the challenges of studying low-abundance plant proteins:

Cutting-edge approaches:

  • Ultra-sensitive detection methods:

    • Single-molecule detection platforms

    • Plasmonic nanomaterials for enhanced fluorescence

    • Quantum dot-conjugated antibodies for improved signal-to-noise

    • Digital ELISA/Simoa technology adapted for plant proteins

  • Amplification-based techniques:

    • Proximity extension assays (PEA) for protein detection

    • Rolling circle amplification (RCA) for signal enhancement

    • SABER (signal amplification by exchange reaction) for imaging

    • Hybridization chain reaction (HCR) for multiplexed detection

  • Microfluidic and nanofluidic platforms:

    • Droplet microfluidics for single-cell protein analysis

    • Nanovial technology for isolated cell analysis

    • Microfluidic antibody capture for ultra-low volumes

    • Digital protein assays in nanowells

  • Advanced mass spectrometry:

    • PASEF (Parallel Accumulation–Serial Fragmentation)

    • Dia-PASEF for deep proteome coverage

    • Targeted proteomics (PRM/MRM) for low-abundance proteins

    • Ion mobility separation for improved detection

  • Sample preparation innovations:

    • Tandem mass tag (TMT) multiplexing for comparative studies

    • Selective enrichment strategies for F-box proteins

    • Novel extraction methods for membrane-associated proteins

    • Plant tissue microsampling for cell-type specific analysis

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 2024 Thebiotek. All Rights Reserved.