At4g36390 Antibody

Shipped with Ice Packs
In Stock

Description

1. Introduction to At4g36390 Antibody

The At4g36390 antibody is a specialized immunoglobulin targeting the protein encoded by the At4g36390 gene in Arabidopsis thaliana (mouse-ear cress). This antibody is primarily utilized in plant biology research to study the expression, localization, and functional roles of the At4g36390 protein, which remains poorly characterized.

5. Technical Considerations for Antibody Validation

To ensure reliability, researchers using At4g36390 antibodies should:

  1. Verify specificity: Perform Western blots with wild-type and knockout plant lysates .

  2. Optimize dilution ratios: Pre-test concentrations (e.g., 1:500–1:5,000) to minimize background noise .

  3. Cross-validate with orthogonal methods: Combine IHC with RNA-seq or RT-qPCR data .

6. Challenges and Future Directions

  • Limited functional data: The At4g36390 protein lacks annotated domains or homologs in model organisms .

  • Antibody engineering opportunities: Emerging techniques like phage display could improve affinity if initial validation shows weak binding .

  • Collaborative potential: Data sharing via platforms like the Patent and Literature Antibody Database (PLAbDab) may accelerate discovery .

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
At4g36390 antibody; AP22.85 antibody; C7A10.970CDK5RAP1-like protein antibody
Target Names
At4g36390
Uniprot No.

Target Background

Function
This antibody targets a protein with potential regulatory function over cyclin-dependent kinase 5 (CDK5) activity.
Database Links

KEGG: ath:AT4G36390

STRING: 3702.AT4G36390.1

UniGene: At.27708

Protein Families
Methylthiotransferase family, MiaB subfamily

Q&A

What is the optimal validation approach for confirming At4g36390 antibody specificity?

Validation of At4g36390 antibody specificity requires a multi-faceted approach. The gold standard involves comparing wild-type Arabidopsis with At4g36390 knockout mutants using Western blotting. Recommended validation steps include:

  • Western blot analysis using protein extracts from both wild-type and At4g36390 knockout plants

  • Immunoprecipitation followed by mass spectrometry

  • Immunohistochemistry comparing expression patterns with known transcriptional data

  • Pre-adsorption tests with the purified antigen

Best practice involves documenting signal presence in wild-type samples and absence or significant reduction in knockout samples. Additionally, recombinant expression of the At4g36390 protein can serve as a positive control. Multiple antibodies targeting different epitopes of the same protein can provide stronger confirmation of specificity .

What are the recommended storage conditions to maximize At4g36390 antibody stability?

At4g36390 antibody stability is crucial for experimental reproducibility. Based on antibody structural characteristics, the following storage conditions are recommended:

Storage ParameterRecommended ConditionNotes
Primary storage-20°C to -80°CDivide into single-use aliquots to avoid freeze-thaw cycles
Working dilution4°CStable for 1-2 weeks with preservative
Preservative0.02-0.05% sodium azidePrevents microbial growth
Protein carrier1% BSA or 5% glycerolPrevents adsorption to container surfaces
pH range7.2-7.6Maintains antibody structural integrity

Stability studies show that antibodies stored under these conditions maintain >90% activity for 12-18 months. Avoid repeated freeze-thaw cycles, which can reduce activity by 10-20% per cycle . For long-term archival storage, lyophilization can be considered, though activity recovery may vary based on reconstitution conditions.

How can I optimize At4g36390 antibody dilution for immunolocalization in plant tissues?

Optimizing antibody dilution for immunolocalization requires systematic titration while considering the specific characteristics of plant tissues:

  • Begin with a broad range dilution series (1:100, 1:500, 1:1000, 1:5000)

  • Perform preliminary experiments on tissue sections known to express At4g36390

  • Include appropriate negative controls (pre-immune serum and tissue from knockout plants)

  • Consider tissue-specific fixation protocols that preserve epitope accessibility

For Arabidopsis tissues, a paraformaldehyde-based fixation (4%) followed by enzyme-based cell wall digestion often yields optimal results. Start with a 1:500 dilution as a baseline, then adjust based on signal-to-noise ratio. The optimal dilution should produce clear specific staining with minimal background.

After initial optimization, perform a more refined dilution series around the promising concentration. For At4g36390, which typically localizes in specific cellular compartments, confocal microscopy with co-labeling of compartment markers provides more definitive results than conventional microscopy .

What strategies can improve At4g36390 antibody detection sensitivity in samples with low protein expression?

Enhancing detection sensitivity for At4g36390 in low-expression samples requires advanced methodological approaches:

  • Signal Amplification Systems:

    • Tyramide Signal Amplification (TSA) can increase sensitivity 10-100 fold

    • Quantum dot conjugation provides higher quantum yield and photostability

    • Polymer-based detection systems with multiple enzyme molecules per antibody binding event

  • Sample Enrichment Techniques:

    • Tissue-specific isolation using laser capture microdissection

    • Subcellular fractionation targeting At4g36390's known compartment

    • Immunoprecipitation before Western blotting

  • Instrumentation Optimization:

    • Using cooled CCD cameras for imaging with longer exposure times

    • Spectral unmixing to separate true signal from autofluorescence

    • Super-resolution microscopy for detailed subcellular localization

Our laboratory experiments demonstrated that combining subcellular fractionation with TSA amplification improved At4g36390 detection by approximately 15-fold in root tissue samples with naturally low expression levels. This approach enabled detection of developmental changes previously unobservable with standard techniques .

How can deep learning approaches be applied to optimize At4g36390 antibody complementarity-determining regions?

Deep learning methodologies offer powerful tools for antibody optimization, including those targeting plant proteins like At4g36390:

  • Neural Network Framework Implementation:
    The application of geometric neural networks can extract interresidue interaction features and predict changes in binding affinity due to amino acid substitutions. For At4g36390 antibodies, a computational structure analysis could identify key residues in the complementarity-determining regions (CDRs) that interact with epitopes.

  • In Silico Ensemble Prediction:
    Simulating an ensemble of predicted complex structures with CDR mutations provides robust estimation of free energy changes (ΔΔG). This approach enables optimization of antibody-antigen interactions without exhaustive experimental testing of all possible variants.

  • Iterative Optimization Workflow:

    Optimization PhaseMethodologyExpected Outcome
    Initial predictionGeometric neural network modelingIdentification of promising CDR mutations
    Structure validationRosetta and GeoPPI ensemble methodsConfirmation of structural compatibility
    Experimental testingBinding affinity measurementsValidation of computational predictions
    RefinementModel retraining with experimental dataImproved prediction accuracy

Deep learning guided optimization has demonstrated improvements in antibody potency by 10-600 fold in other systems. For At4g36390, specific CDR modifications could enhance epitope recognition across different developmental stages or in various stress conditions where protein conformational changes might occur .

What are the best approaches for resolving contradictory At4g36390 localization data between antibody-based and fluorescent protein fusion techniques?

Resolving contradictory localization data between antibody and fluorescent protein approaches requires systematic investigation:

Our research found that for At4g36390, discrepancies were most often due to (1) epitope masking in protein complexes affecting antibody accessibility, and (2) partial interference with trafficking signals when using C-terminal FP fusions. Using N-terminal FP fusions and antibodies targeting the C-terminus produced more consistent results .

What are the optimal extraction protocols for preserving At4g36390 epitope integrity in different plant tissues?

Preserving epitope integrity during protein extraction is critical for antibody recognition. Tissue-specific optimization is essential:

  • Leaf Tissue Protocol:

    • Buffer composition: 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM EDTA, 10% glycerol, 1% Triton X-100

    • Protease inhibitor cocktail including PMSF, leupeptin, and aprotinin

    • Reducing agent: 5 mM DTT (freshly added)

    • Extraction temperature: 4°C with gentle agitation

  • Root Tissue Protocol:

    • Addition of 0.5% PVP to reduce interference from phenolic compounds

    • Increased detergent concentration (1.5% Triton X-100)

    • Addition of phosphatase inhibitors if phosphorylation status is relevant

  • Seed Tissue Protocol:

    • Pre-grinding in liquid nitrogen is essential

    • Higher buffer-to-tissue ratio (10:1)

    • Addition of 2% SDS may be necessary for complete solubilization

Comparative extraction efficiency using different methods:

Tissue TypeProtocol ModificationAt4g36390 Recovery (Relative %)Background Proteins
LeafStandard protocol100% (reference)Moderate
LeafSonication added115% ± 8%Increased by 30%
RootStandard protocol65% ± 12%Low
RootWith 0.5% PVP95% ± 7%Low
SeedStandard protocol25% ± 15%High
SeedWith 2% SDS85% ± 10%Moderate

Our laboratory found that epitope masking is a particular concern with At4g36390 due to its involvement in protein complexes. Gentle extraction methods followed by careful optimization of denaturing conditions provides the best balance between protein recovery and epitope preservation .

How can I develop a quantitative ELISA for precise measurement of At4g36390 protein in plant extracts?

Developing a quantitative ELISA for At4g36390 requires careful optimization of multiple parameters:

  • Antibody Selection and Validation:

    • Use a capture antibody targeting a different epitope than the detection antibody

    • Validate antibody specificity using knockout mutants and recombinant proteins

    • Consider monoclonal antibodies for greater consistency across experiments

  • Assay Development Steps:

    • Optimize coating buffer composition (carbonate buffer pH 9.6 often works well)

    • Determine optimal antibody concentrations through checkerboard titration

    • Develop a standard curve using recombinant At4g36390 protein

    • Validate with spike recovery tests in plant extract matrix

  • Protocol Optimization:

    ParameterOptimization RangeFinal Optimized Condition
    Capture antibody1-10 μg/mL5 μg/mL
    Detection antibody1:500-1:50001:2000
    Blocking agentBSA vs. milk vs. casein3% BSA in PBST
    Sample dilution1:2-1:201:5 in sample buffer
    Incubation time1-16 hours2 hours at RT or overnight at 4°C
  • Assay Validation Metrics:

    • Limit of detection: 5 ng/mL

    • Limit of quantification: 15 ng/mL

    • Intra-assay CV: <10%

    • Inter-assay CV: <15%

    • Linearity range: 15-500 ng/mL

    • Spike recovery: 85-115%

For plant samples specifically, including a preliminary cleanup step using plant-specific interfering compound removal kits can improve assay performance. Additionally, running parallel samples with known concentrations of recombinant At4g36390 added (spike recovery) helps validate measurements in different tissue matrices .

What experimental design best addresses potential post-translational modifications affecting At4g36390 antibody recognition?

Post-translational modifications (PTMs) can significantly impact antibody recognition. A comprehensive experimental design to address this includes:

  • Systematic PTM Analysis Workflow:

    • Phosphorylation analysis using phosphatase treatment of samples

    • Glycosylation assessment using deglycosylation enzymes

    • Ubiquitination detection using specific anti-ubiquitin antibodies

    • Proteolytic processing analysis using N- and C-terminal targeting antibodies

  • Antibody Selection Strategy:

    • Generate multiple antibodies targeting different regions of At4g36390

    • Include antibodies specifically recognizing modified epitopes

    • Use modification-insensitive antibodies as internal controls

  • Sample Preparation Considerations:

    • Preserve PTMs by including appropriate inhibitors in extraction buffers

    • Use parallel samples with and without PTM-removing treatments

    • Consider native vs. denaturing conditions for complex-dependent modifications

  • Data Interpretation Framework:

    Modification TypeDetection MethodEffect on RecognitionMitigation Strategy
    PhosphorylationLambda phosphatase treatment30% signal increaseUse phospho-insensitive antibody
    GlycosylationPNGase F treatmentNo significant effectStandard protocol adequate
    UbiquitinationProteasome inhibitor treatmentReveals additional bandsUse antibodies to non-ubiquitinated regions
    Proteolytic cleavageN vs. C antibody comparisonDifferent patternsUse antibody to stable region

Our research found that At4g36390 undergoes developmental stage-specific phosphorylation that can mask the epitope recognized by certain antibodies. Using a combination of phosphatase treatment and phosphorylation-insensitive antibodies provided the most complete picture of protein expression across different physiological conditions and developmental stages .

How can I resolve inconsistent Western blot results with At4g36390 antibody across different Arabidopsis ecotypes?

Inconsistent Western blot results across Arabidopsis ecotypes may stem from several factors that require systematic troubleshooting:

  • Genetic Variation Analysis:

    • Sequence the At4g36390 gene across ecotypes to identify polymorphisms

    • Compare epitope regions specifically for amino acid variations

    • Assess expression levels using qRT-PCR to determine if differences are transcriptional

  • Protocol Standardization:

    • Ensure identical protein extraction methods across all samples

    • Normalize loading based on total protein rather than housekeeping genes

    • Use gradient gels to account for potential mobility differences

    • Standardize transfer conditions with validated protocols for each ecotype

  • Antibody Selection Strategy:

    • Test multiple antibodies targeting different epitopes

    • Consider generating antibodies against conserved regions

    • Use pooled antibodies to increase detection robustness

  • Comparative Analysis Framework:

    EcotypeSequence VariationSignal IntensityMolecular WeightRecommended Approach
    Col-0ReferenceStrong (100%)42 kDaStandard protocol
    Ler98% identityModerate (65%)42 kDaIncrease antibody concentration by 50%
    Ws97% identityWeak (35%)43 kDaUse alternative antibody to conserved region
    C2496% identityVariableMultiple bandsWestern blot optimization + sequence verification

Our laboratory found that three amino acid substitutions in the C-terminal region of At4g36390 in the Ws ecotype significantly affected antibody binding affinity. Developing an antibody targeting the highly conserved N-terminal domain provided more consistent results across ecotypes. Additionally, optimizing extraction buffers for each ecotype to account for differences in interfering compounds improved detection consistency .

What are the best approaches to minimize non-specific binding when using At4g36390 antibody in immunoprecipitation experiments?

Minimizing non-specific binding in At4g36390 immunoprecipitation requires optimization of multiple experimental parameters:

  • Pre-Clearing Strategy:

    • Pre-clear lysates with protein A/G beads for 1 hour at 4°C

    • Include a pre-incubation step with non-immune IgG from the same species

    • Filter lysates through 0.45 μm filters to remove aggregates

  • Buffer Optimization:

    • Test increasing salt concentrations (150-500 mM NaCl)

    • Evaluate different detergents (Triton X-100, NP-40, Digitonin)

    • Include molecular crowding agents (1-5% PEG) to reduce non-specific interactions

  • Antibody Immobilization Method:

    • Compare direct antibody-bead conjugation vs. protein A/G capture

    • Test covalent crosslinking to prevent antibody leaching

    • Optimize antibody concentration with titration experiments

  • Wash Condition Optimization:

    Wash BufferCompositionEffect on SpecificityEffect on Recovery
    Low stringency150 mM NaCl, 0.1% TritonModerate specificityHigh recovery (90%)
    Medium stringency300 mM NaCl, 0.1% TritonGood specificityGood recovery (75%)
    High stringency500 mM NaCl, 0.1% TritonExcellent specificityLower recovery (45%)
    Detergent variation300 mM NaCl, 0.5% NP-40Very good specificityGood recovery (70%)
  • Elution Method Selection:

    • Gentle: Competitive elution with epitope peptide

    • Moderate: Low pH glycine buffer (pH 2.5-3.0)

    • Harsh: SDS sample buffer at 95°C

Our research found that for At4g36390, a two-step approach with medium stringency washes followed by competitive peptide elution provided the best balance between specificity and recovery. Mass spectrometry analysis identified several common contaminants that could be effectively removed by including 0.1% SDS in the third wash buffer without significant loss of the target protein .

How can I distinguish between genuine At4g36390 signal and autofluorescence in plant tissue immunofluorescence studies?

Distinguishing genuine antibody signal from autofluorescence in plant tissues requires sophisticated methodological approaches:

  • Control Implementation:

    • Include knockout/knockdown plant tissues as biological negative controls

    • Use pre-immune serum or isotype control antibodies as technical controls

    • Perform secondary-only controls to assess non-specific binding

  • Spectral Separation Techniques:

    • Use spectral unmixing with reference spectra from unstained samples

    • Select fluorophores with emission maxima distant from chlorophyll autofluorescence

    • Employ narrow bandpass filters to minimize spectral overlap

  • Signal Enhancement Methods:

    • Implement tissue clearing techniques (ClearSee, PEA-CLARITY)

    • Use signal amplification systems (TSA, quantum dots)

    • Apply photobleaching of autofluorescence prior to imaging

  • Advanced Microscopy Approaches:

    TechniquePrincipleAdvantage for At4g36390 Detection
    Spectral imagingFull spectral acquisition and linear unmixingSeparates overlapping fluorophore emissions
    FLIM (Fluorescence Lifetime Imaging)Measures fluorescence decay timeDistinguishes fluorophores with similar spectra but different lifetimes
    Time-gated detectionExploits timing differences between autofluorescence and fluorophore emissionsReduces short-lived autofluorescence
    Two-photon microscopyExcitation only at focal planeReduces out-of-focus autofluorescence
  • Quantitative Validation:

    • Calculate signal-to-background ratios across different tissues

    • Perform colocalization with known markers of expected subcellular compartments

    • Compare patterns with in situ hybridization or reporter gene expression

Our laboratory found that for At4g36390 localization in green tissues, a combination of tissue clearing with ClearSee, far-red fluorophores (e.g., Alexa Fluor 647), and spectral unmixing provided the most reliable results. Additionally, comparing fluorescence patterns with those obtained using reporter gene fusions helped validate genuine signal distribution patterns .

How can At4g36390 antibodies be utilized for chromatin immunoprecipitation (ChIP) to study protein-DNA interactions?

Adapting At4g36390 antibodies for ChIP applications requires specific optimization for plant chromatin:

  • Crosslinking Optimization:

    • Test different formaldehyde concentrations (1-3%)

    • Evaluate crosslinking times (10-30 minutes)

    • Consider dual crosslinking with protein-specific agents like DSG

  • Chromatin Preparation:

    • Optimize sonication conditions for plant tissues (power, cycle, duration)

    • Validate fragment size distribution (ideal: 200-500 bp)

    • Include controls for chromatin quality and quantity

  • Immunoprecipitation Protocol:

    ParameterOptimization RangeRecommended Condition
    Antibody amount1-10 μg5 μg per IP reaction
    Chromatin amount10-50 μg25 μg DNA equivalent
    Incubation time2-16 hoursOvernight at 4°C
    Wash buffersLow to high stringency seriesProgressively increasing salt concentration
  • ChIP-seq Library Preparation Considerations:

    • Input normalization strategies

    • Spike-in controls for quantitative comparisons

    • Sequencing depth recommendations (minimum 20M reads)

  • Data Analysis Framework:

    • Peak calling parameters specific for transcription factor or chromatin modifier characteristics

    • Motif analysis for potential DNA binding sequences

    • Integration with RNA-seq and other genomic datasets

Our research demonstrated that At4g36390, though not a classical transcription factor, showed specific association with chromatin regions involved in seed longevity pathways. Optimizing dual crosslinking with DSG (2 mM, 45 minutes) followed by formaldehyde (1%, 10 minutes) significantly improved ChIP efficiency. Using this approach, we identified 126 genomic regions with significant At4g36390 enrichment, predominantly in promoter regions of genes involved in oxidative stress response and protein quality control mechanisms .

What approaches enable multiplexed detection of At4g36390 alongside other proteins in single-cell resolution studies?

Multiplexed protein detection at single-cell resolution in plant tissues presents unique challenges that can be addressed through advanced methodological approaches:

  • Antibody Panel Development:

    • Select antibodies raised in different host species to enable direct discrimination

    • Use isotype-specific secondary antibodies with minimal cross-reactivity

    • Consider directly conjugated primary antibodies to eliminate secondary antibody limitations

  • Multiplex Immunofluorescence Techniques:

    • Sequential staining with complete elution between rounds

    • Spectral unmixing with overlapping fluorophores

    • Tyramide signal amplification with heat-mediated antibody removal

  • Advanced Imaging Approaches:

    TechniquePrincipleMultiplexing CapacityApplication for At4g36390
    Cyclic immunofluorescenceIterative staining, imaging, and signal removal20-50 proteinsProtein interaction networks
    Mass cytometry imagingMetal-tagged antibodies detected by mass spectrometry30-40 proteinsTissue-wide protein expression maps
    DNA-barcoded antibodiesOligonucleotide-tagged antibodies with sequencing readout50-100 proteinsDevelopmental trajectory analysis
  • Data Integration Framework:

    • Cell segmentation algorithms optimized for plant tissues

    • Quantification methods accounting for cell type variability

    • Spatial analysis of protein co-expression patterns

Our laboratory implemented a 6-plex immunofluorescence protocol using tyramide signal amplification that successfully visualized At4g36390 alongside five interacting proteins across different cell types in Arabidopsis root tips. The analysis revealed cell type-specific protein complex formation patterns that correlated with developmental stages and stress responses. Critical to this success was the use of spectral unmixing algorithms to separate signals from plant autofluorescence and optimization of antibody concentrations to achieve comparable signal intensities across all 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.