At4g17830 Antibody

Shipped with Ice Packs
In Stock

Description

Target Protein: AT4G17830

Gene: AT4G17830
Protein: Peptidase M20/M25/M40 family protein
Function: Involved in hydrolytic enzymatic activity, potentially regulating peptide metabolism or protein turnover in Arabidopsis .

CharacteristicDetailsSource
Cysteine Residues8 residues
Post-Translational Modification (PTM)S-sulfenylation (S-SG) observed under oxidative stress
Subcellular LocalizationPredicted cytoplasmic (based on homology)

Antibody Development and Validation

The At4g17830 antibody was generated using recombinant protein technology, where a unique antigenic region of the protein was expressed, purified, and injected into host animals (e.g., sheep or rabbit) .

Key Validation Steps:

  • Specificity: Tested via Western blot against Arabidopsis wild-type and mutant lines (e.g., T-DNA insertion mutants) to confirm absence of cross-reactivity .

  • Sensitivity: Detects target protein in picogram quantities via dot blot .

  • Applications: Validated for immunolocalization in root tissues and Western blotting .

Protein Expression Analysis

  • Western Blot: Detects a single band at the expected molecular weight (~50 kDa, inferred from homologous proteins) .

  • Subcellular Localization: Used in immunofluorescence to map protein distribution in Arabidopsis tissues .

Oxidative Stress Studies

The S-sulfenylation modification (S-SG) on cysteine residues, detected using this antibody, indicates redox-sensitive regulation under hydrogen peroxide (H₂O₂) exposure .

ConditionModification ObservedBiological Implication
1 mM H₂O₂S-SGActivation under moderate stress
20 mM H₂O₂No dataPotential protein degradation

Technical Limitations and Advancements

  • Challenge: Low signal-to-noise ratio in immunolocalization due to epitope accessibility .

  • Solution: Affinity purification and signal amplification protocols improved detection .

  • Comparative Advantage: Recombinant antibodies offer batch-to-batch consistency over traditional hybridoma-derived antibodies .

Future Directions

  • Structural Studies: Engineering single-chain variable fragments (scFvs) for crystallography .

  • High-Throughput Screening: Integration with CRISPR-edited Arabidopsis lines to study knockout phenotypes .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At4g17830 antibody; dl4945w antibody; T6K21Acetylornithine deacetylase antibody; EC 3.5.1.16 antibody; N-acetylornithinase antibody; AO antibody; Acetylornithinase antibody; NAO antibody
Target Names
At4g17830
Uniprot No.

Q&A

What is AT4G17830 and why is it important to generate antibodies against it?

AT4G17830 encodes a functional acetylornithine deacetylase (NAOD) in Arabidopsis thaliana. This enzyme plays a crucial role in mediating a linear pathway for ornithine biosynthesis. Silenced lines of plants with this gene flower early but demonstrate reduced fertility (siliques do not develop properly) and reduced ornithine levels . Generating antibodies against AT4G17830 is important for various research applications including protein localization, expression level analysis, protein-protein interaction studies, and functional characterization of the protein in different plant tissues and developmental stages. Antibodies provide a powerful tool to study this protein's role in ornithine biosynthesis and how it affects plant development and reproduction.

What are the key considerations when designing an epitope for AT4G17830 antibody production?

When designing an epitope for AT4G17830 antibody production, researchers should consider several critical factors:

  • Sequence uniqueness: Perform BLAST searches to ensure the selected epitope is specific to AT4G17830 and doesn't share significant homology with other proteins in the model organism .

  • Structural accessibility: Select regions that are likely to be exposed on the protein surface rather than buried within the folded structure. These regions are more accessible to antibody binding.

  • Hydrophilicity and antigenicity: Prioritize hydrophilic regions as they are more likely to be surface-exposed and immunogenic.

  • Conservation across species: If the antibody needs to recognize orthologs in different plant species, select epitopes with high sequence conservation across these species .

  • Avoid post-translational modification sites: Unless specifically studying these modifications, avoid regions containing potential glycosylation, phosphorylation, or other modification sites that might interfere with antibody recognition.

  • Length considerations: For synthetic peptide antigens, optimal length is typically 10-20 amino acids, while recombinant protein fragments should be large enough to fold properly but manageable for expression.

Confirming the protein sequence using canonical databases is essential before epitope selection to ensure targeting the correct protein isoform .

How can researchers verify the specificity of an AT4G17830 antibody?

Verifying antibody specificity for AT4G17830 requires a multi-faceted approach:

  • Western blot analysis using:

    • Wild-type plants (positive control)

    • at4g17830 knockout or knockdown mutants (negative control)

    • Plants overexpressing AT4G17830 (enhanced signal control)

  • Immunoprecipitation followed by mass spectrometry to confirm the antibody pulls down the correct protein.

  • Pre-absorption tests where the antibody is pre-incubated with purified AT4G17830 protein or peptide before immunostaining. This should eliminate or significantly reduce the specific signal.

  • Cross-reactivity testing against related proteins, particularly other deacetylases that might share structural similarities.

  • Immunohistochemistry patterns should match known expression patterns from transcriptomic data or promoter-reporter studies.

  • ELISA assays to quantitatively measure binding affinity and specificity against purified target and potential cross-reactive proteins.

Using multiple validation methods provides stronger evidence for antibody specificity than relying on a single technique, ensuring reliable experimental results in AT4G17830 research.

How do post-translational modifications of AT4G17830 affect antibody recognition and experimental outcomes?

Post-translational modifications (PTMs) of AT4G17830 can significantly impact antibody recognition, leading to variable or misleading experimental results. These effects include:

  • Epitope masking: PTMs like phosphorylation, glycosylation, or acetylation can physically block antibody access to the epitope. For instance, if an antibody targets a region that becomes phosphorylated during specific cellular conditions, signal intensity may decrease despite constant protein levels.

  • Conformational changes: PTMs can induce structural changes in AT4G17830 that either expose or conceal epitopes. An antibody raised against the native conformation may not recognize the modified form, creating false negatives in certain physiological states.

  • Differential recognition: Some antibodies may preferentially bind to modified or unmodified forms of AT4G17830. This selectivity, while sometimes desired, must be characterized to avoid misinterpretation of results.

  • Experimental implications: When using antibodies for techniques like immunoprecipitation or chromatin immunoprecipitation, PTMs can significantly affect protein-protein interactions or DNA binding properties of AT4G17830.

Researchers should characterize their antibodies regarding PTM sensitivity by:

  • Testing antibody recognition using phosphatase-treated samples

  • Comparing recognition in samples from different developmental stages or stress conditions when PTM profiles may differ

  • Using phospho-specific or other PTM-specific antibodies in parallel experiments if particular modifications are suspected

When antibodies are generated using synthetic peptides, they typically recognize linear epitopes and may not be sensitive to conformational changes induced by PTMs, while antibodies raised against purified proteins may be more sensitive to such modifications .

What are the challenges in generating antibodies that can distinguish between AT4G17830 and closely related proteins in plant systems?

Generating antibodies with high specificity for AT4G17830 over related proteins presents several significant challenges:

  • Sequence homology: Proteins from the same family often share conserved domains with high sequence similarity. This makes finding unique epitopes for antibody generation difficult. Researchers should perform comprehensive sequence alignment of AT4G17830 with all related deacetylases to identify regions with minimal homology .

  • Structural similarity: Even when sequences differ, proteins from the same family may have similar tertiary structures, creating epitopes with similar spatial arrangements despite different primary sequences. This can lead to cross-reactivity.

  • Expression level variability: If AT4G17830 is expressed at significantly lower levels than related proteins, even minor cross-reactivity can cause false positives due to the abundance of the related protein.

  • Developmental and tissue-specific regulation: The expression profiles of AT4G17830 and related proteins may vary across tissues or developmental stages, making validation complex and requiring multiple control samples.

  • Isoform-specific targeting: Alternative splicing or gene duplication may create closely related isoforms that differ in only a few amino acids but have distinct functions.

To address these challenges:

  • Use phage display technology to select antibodies with high specificity and affinity

  • Validate antibodies against knockout mutants for both AT4G17830 and related proteins

  • Perform competitive binding assays with purified proteins

  • Consider subtraction strategies during antibody selection to eliminate clones that bind to related proteins

  • Use recombinant expression of unique domains rather than whole proteins for immunization

A thorough characterization using multiple related proteins as controls in Western blots, immunoprecipitation, and immunolocalization experiments is essential to confirm specificity.

How can structural analysis inform better AT4G17830 antibody design and optimization?

Structural analysis provides valuable insights for designing highly specific and effective antibodies against AT4G17830:

  • Epitope accessibility mapping: Detailed structural information through techniques like X-ray crystallography or cryo-EM can identify surface-exposed regions of AT4G17830 that are ideal antibody targets. These exposed regions are more likely to be recognized in native conditions across various experimental applications.

  • Conformational epitope identification: While many antibodies target linear epitopes, structural analysis can reveal conformational epitopes formed by amino acids that are distant in primary sequence but proximal in the folded protein. These conformational epitopes often provide greater specificity.

  • Functional domain targeting: Structural information reveals functional domains of AT4G17830, allowing researchers to design antibodies that either target or avoid these regions depending on whether they want to inhibit function or simply detect the protein.

  • Antigen design optimization: For recombinant protein antigen production, structural knowledge helps in designing constructs that fold properly and present epitopes similarly to the native protein.

  • Homology modeling: When experimental structures are unavailable, computational approaches like homology modeling can predict AT4G17830's structure based on related proteins with known structures.

  • Molecular dynamics simulations: These can predict how flexible different regions of AT4G17830 are, helping to identify stable epitopes that maintain consistent conformation.

  • Cross-reactivity analysis: Structural superimposition of AT4G17830 with related proteins can identify regions that are structurally distinct despite sequence similarity.

By integrating structural information with sequence analysis, researchers can design antigens that target unique structural features of AT4G17830, significantly improving antibody specificity and reducing cross-reactivity with related proteins .

What are the optimal protocols for using AT4G17830 antibodies in Western blotting experiments?

When using AT4G17830 antibodies for Western blotting, researchers should follow these optimization guidelines:

  • Sample preparation:

    • Extract proteins using a buffer containing 50mM Tris-HCl (pH 7.5), 150mM NaCl, 1% Triton X-100, and protease inhibitor cocktail

    • Include 10mM DTT or β-mercaptoethanol to reduce disulfide bonds

    • Heat samples at 95°C for 5 minutes in Laemmli buffer

  • Gel selection and electrophoresis:

    • Use 10-12% SDS-PAGE gels for optimal resolution of AT4G17830 (predicted molecular weight should be verified)

    • Run samples alongside molecular weight markers and positive/negative controls

  • Transfer conditions:

    • Transfer to PVDF membrane (preferred over nitrocellulose for plant proteins)

    • Use wet transfer at 100V for 1 hour or 30V overnight at 4°C

    • Verify transfer efficiency with reversible staining (Ponceau S)

  • Blocking:

    • Block with 5% non-fat dry milk in TBST (preferred over BSA for plant samples)

    • Block for 1 hour at room temperature or overnight at 4°C

  • Primary antibody incubation:

    • Dilute antibody in blocking solution (optimal dilution typically 1:1000 to 1:5000)

    • Incubate overnight at 4°C with gentle rocking

    • Include proper controls: wild-type extracts, knockout mutant extracts, and recombinant protein

  • Washing and secondary antibody:

    • Wash membranes 4-5 times with TBST, 5 minutes each

    • Incubate with HRP-conjugated secondary antibody (1:5000-1:10000) for 1 hour

    • Extensive washing (5-6 times) to reduce background

  • Detection:

    • Use enhanced chemiluminescence (ECL) substrate

    • For weak signals, consider ECL Plus or SuperSignal West Femto

    • Optimize exposure time to avoid saturation

  • Troubleshooting specific issues:

    • Multiple bands: Verify if they represent isoforms, degradation products, or cross-reactivity

    • High background: Increase washing times or antibody dilution

    • No signal: Check protein transfer and consider less stringent washing conditions

This protocol should be optimized specifically for AT4G17830 antibodies through preliminary experiments testing different conditions .

How should researchers validate AT4G17830 antibodies for immunoprecipitation experiments?

Validating AT4G17830 antibodies for immunoprecipitation (IP) requires a systematic approach:

  • Initial validation steps:

    • Confirm antibody functionality in Western blot before attempting IP

    • Test antibody specificity using knockout/knockdown plant lines

    • Determine if the antibody recognizes native or denatured epitopes

  • Pre-clearing optimization:

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

    • Optimize pre-clearing time (typically 1-2 hours at 4°C)

    • Include appropriate detergent concentration to maintain protein solubility without disrupting antibody binding

  • Critical controls:

    • No-antibody control (beads only) to assess non-specific binding

    • Isotype control (irrelevant antibody of same isotype)

    • Input sample (5-10% of starting material) for comparison

    • IP from knockout/knockdown plants as negative control

  • Binding conditions optimization:

    • Test different antibody amounts (typically 1-5 μg per 500 μg protein)

    • Optimize incubation time (4 hours to overnight at 4°C)

    • Test different binding buffers varying salt concentration (100-300 mM NaCl)

  • Washing stringency:

    • Establish a washing protocol that removes non-specific proteins while retaining specific interactions

    • Test increasing salt concentrations in wash buffers (150-500 mM)

    • Optimize detergent type and concentration

  • Elution methods comparison:

    • Compare glycine elution (pH 2.5-3.0) vs. SDS elution vs. boiling in sample buffer

    • Determine which method provides the cleanest specific elution

  • Verification methods:

    • Western blot of IP eluate to confirm target protein presence

    • Mass spectrometry analysis to confirm identity and identify interacting partners

    • Reciprocal IP using antibodies against known interacting partners

  • Quantitative assessment:

    • Calculate IP efficiency by comparing signal intensity between input and IP

    • Determine signal-to-noise ratio by comparing specific band to background

For co-immunoprecipitation experiments, researchers should also consider cross-linking conditions if the interactions are transient or weak. Validation should be performed using multiple biological replicates to ensure reproducibility .

What considerations are important when using AT4G17830 antibodies for immunolocalization studies in plant tissues?

When using AT4G17830 antibodies for immunolocalization studies in plant tissues, researchers should consider the following critical factors:

  • Tissue fixation and processing:

    • Test multiple fixatives (4% paraformaldehyde, glutaraldehyde combinations)

    • Optimize fixation time (typically 2-24 hours) to maintain antigen recognition while preserving tissue structure

    • Consider whether paraffin embedding, cryosectioning, or vibratome sectioning is most appropriate for AT4G17830 detection

  • Antigen retrieval methods:

    • Compare heat-induced epitope retrieval (citrate buffer, pH 6.0)

    • Test enzymatic retrieval (proteinase K, pepsin)

    • Determine if retrieval improves signal without increasing background

  • Tissue permeabilization:

    • Optimize detergent concentration (0.1-0.5% Triton X-100)

    • Balance permeabilization with tissue integrity

    • Test different incubation times to allow antibody penetration

  • Blocking parameters:

    • Test different blocking agents (BSA, normal serum, commercial blockers)

    • Determine optimal blocking time (1-2 hours)

    • Include blocking steps for endogenous peroxidase if using HRP detection

  • Antibody dilution and incubation:

    • Establish optimal primary antibody dilution through titration experiments

    • Compare overnight incubation at 4°C vs. shorter incubations at room temperature

    • Determine if signal amplification systems (tyramide, streptavidin-biotin) are needed

  • Essential controls:

    • Omission of primary antibody

    • Pre-absorption with immunizing peptide/protein

    • Tissues from knockout/knockdown plants

    • Competing peptide controls

    • Secondary antibody only controls

  • Counterstaining considerations:

    • Select counterstains that don't interfere with antibody detection

    • Consider nuclear stains (DAPI) or membrane stains (FM4-64) for co-localization

  • Confocal microscopy parameters:

    • Optimize laser power and detector gain to avoid saturation

    • Establish proper settings using controls

    • Capture Z-stacks for three-dimensional analysis

    • Include co-localization with organelle markers for precise subcellular localization

  • Quantification approaches:

    • Develop consistent methods for signal quantification

    • Use software tools for co-localization analysis

    • Perform statistical analysis across multiple samples and replicates

  • Common issues and solutions:

    • Autofluorescence: Test different wavelengths or use spectral unmixing

    • Non-specific binding: Increase blocking stringency or antibody dilution

    • Weak signal: Try signal amplification systems or alternative fixation methods

These considerations help ensure reliable and reproducible immunolocalization results for AT4G17830 in plant tissues .

How can researchers reconcile contradictory results with AT4G17830 antibodies across different experimental methods?

When faced with contradictory results using AT4G17830 antibodies across different methods, researchers should implement this systematic approach:

By systematically investigating these factors, researchers can determine whether contradictions reflect technical limitations, antibody characteristics, or genuine biological complexity in AT4G17830 expression or modification patterns .

What controls are necessary when using AT4G17830 antibodies in cross-species studies?

When using AT4G17830 antibodies across different plant species, implementing robust controls is essential to ensure reliable and interpretable results:

  • Sequence homology assessment:

    • Perform multiple sequence alignments of AT4G17830 orthologs across target species

    • Calculate percent identity and similarity in epitope regions

    • Predict cross-reactivity based on conserved epitopes

  • Genetic controls:

    • Include known AT4G17830 knockout/knockdown mutants from model species

    • When possible, use CRISPR-generated mutants in non-model species

    • Include overexpression lines as positive controls

  • Recombinant protein controls:

    • Express recombinant proteins of AT4G17830 orthologs from each species

    • Use these for Western blot positive controls and antibody validation

    • Create standard curves for quantitative comparisons

  • Pre-absorption controls:

    • Pre-incubate antibody with excess purified protein/peptide from target species

    • Compare signal with and without pre-absorption

    • Verify specific signal suppression after pre-absorption

  • Cross-species validation panel:

    • Create a standardized panel of tissue extracts from all species

    • Run parallel experiments using identical conditions

    • Document antibody performance metrics across species

  • Loading controls:

    • Use antibodies against highly conserved proteins (actin, tubulin)

    • Verify even loading with total protein stains (Ponceau S, Coomassie)

    • Consider species-specific differences in reference gene expression

  • Alternative detection methods:

    • Complement antibody studies with RT-qPCR to compare transcript levels

    • Use mass spectrometry for protein identification confirmation

    • Consider creating species-specific antibodies for key comparisons

  • Data interpretation framework:

    • Establish clear criteria for positive identification

    • Create a scoring system for antibody performance across species

    • Document species-specific caveats and limitations

  • Statistical considerations:

    • Increase biological replicates when working across species

    • Perform power analysis to determine adequate sample size

    • Use appropriate statistical tests that account for cross-species variation

How can researchers address batch-to-batch variability in AT4G17830 antibodies?

Addressing batch-to-batch variability in AT4G17830 antibodies requires a comprehensive quality control strategy:

  • Standardized characterization protocol:

    • Develop a consistent testing protocol for each new antibody batch

    • Create a performance checklist including specificity, sensitivity, and background

    • Document key performance metrics for each batch:

      • Western blot detection limit

      • Signal-to-noise ratio

      • Specific band intensity

      • Cross-reactivity profile

  • Reference sample library:

    • Maintain a frozen library of standardized samples:

      • Wild-type plant extracts

      • Knockout mutant extracts

      • Recombinant AT4G17830 protein

    • Use identical samples to test each new batch

  • Bridging studies:

    • Always test new and old batches side-by-side

    • Calculate relative performance metrics

    • Determine correction factors if necessary

  • Multiplex validation approach:

    • Test new batches in multiple applications (Western, IP, IHC)

    • Identify application-specific variations

    • Document any application limitations

  • Bulk purchasing strategy:

    • When possible, purchase larger antibody quantities

    • Aliquot and store according to manufacturer recommendations

    • Use consistent aliquots throughout a project

  • Validation with alternative detection methods:

    • Confirm key experimental findings with orthogonal techniques

    • Use tagged protein expression when possible

    • Consider MS-based validation for critical results

  • Antibody storage optimization:

    • Determine optimal storage conditions through stability testing

    • Avoid freeze-thaw cycles by using small aliquots

    • Monitor antibody performance over time

  • Detailed record-keeping system:

    • Create an antibody database documenting:

      • Batch number

      • Production date

      • Validation date and results

      • Optimal working dilutions

      • Application-specific performance

      • Observed limitations

  • Statistical adjustment approaches:

    • Develop normalization methods for cross-batch comparisons

    • Include internal controls in all experiments

    • Consider using pooled standards across experimental sets

  • Alternative approaches when variability is unavoidable:

    • Consider generating monoclonal antibodies using phage display technology

    • Develop recombinant antibodies for consistent performance

    • Express tagged versions of AT4G17830 for detection with commercial tag antibodies

This systematic approach helps researchers maintain experimental consistency despite inherent batch-to-batch variability in AT4G17830 antibodies .

How can phage display technology improve the development of highly specific AT4G17830 antibodies?

Phage display technology offers significant advantages for developing highly specific AT4G17830 antibodies:

  • Targeted selection process:

    • Permits in vitro selection against specific AT4G17830 epitopes

    • Allows precise control over selection conditions

    • Enables negative selection against related proteins to reduce cross-reactivity

    • Facilitates selection of antibodies that recognize native protein conformations

  • Antibody library diversity advantages:

    • Provides access to extremely large antibody repertoires (10^10-10^11 variants)

    • Allows screening of antibody libraries from various sources (naïve, immunized, synthetic)

    • Increases probability of finding high-affinity binders with desired specificity

  • Selection stringency control:

    • Permits gradually increasing washing stringency during biopanning

    • Enables selection of antibodies with desired off-rates

    • Allows optimization of binding conditions to improve specificity

    • Facilitates selection under conditions that mimic intended application

  • Customized elution strategies:

    • Offers various elution methods (pH change, competitive elution, enzymatic cleavage)

    • Allows selection of antibodies with specific binding characteristics

    • Permits recovery of high-affinity binders through optimized protocols

  • Format flexibility:

    • Enables direct selection of different antibody formats (scFv, Fab, VHH)

    • Permits reformatting selected antibodies into desired final formats

    • Facilitates engineering for specific applications (detection vs. functional studies)

  • Epitope mapping potential:

    • Can be combined with peptide arrays or truncation libraries

    • Allows identification of the precise binding epitope

    • Enables selection of antibodies targeting specific functional domains of AT4G17830

  • Next-generation sequencing integration:

    • Permits deep analysis of selected antibody populations

    • Allows identification of enriched antibody families

    • Reduces selection time through early identification of promising candidates

  • Reproducibility advantages:

    • Sequence information enables consistent antibody reproduction

    • Eliminates hybridoma stability issues

    • Ensures long-term availability of identical reagents

  • Post-selection optimization:

    • Allows affinity maturation through additional rounds of mutagenesis and selection

    • Permits engineering for improved stability, solubility, and expression

    • Enables humanization or other sequence modifications if needed

  • Implementation strategy for AT4G17830:

    • Express purified AT4G17830 protein or specific domains

    • Conduct parallel selections against full-length protein and specific peptides

    • Include negative selection steps against related deacetylases

    • Validate selected antibodies using knockout controls and specificity assays

Phage display technology provides researchers with powerful tools to develop highly specific AT4G17830 antibodies with reproducible performance characteristics, addressing many limitations of traditional antibody production methods .

What are the key considerations for using AT4G17830 antibodies in chromatin immunoprecipitation (ChIP) experiments?

When using AT4G17830 antibodies for chromatin immunoprecipitation (ChIP) experiments, researchers should address these critical considerations:

  • Antibody qualification requirements:

    • Confirm antibody specificity through Western blotting and IP

    • Verify nuclear localization using immunofluorescence

    • Test multiple antibodies targeting different epitopes

    • Validate with tagged AT4G17830 ChIP using anti-tag antibodies

  • Crosslinking optimization:

    • Test different formaldehyde concentrations (0.75-1.5%)

    • Optimize crosslinking time (10-20 minutes)

    • Consider dual crosslinking (formaldehyde plus DSG/EGS) for improved efficiency

    • Evaluate potential epitope masking during crosslinking

  • Chromatin preparation considerations:

    • Optimize sonication conditions for plant chromatin

    • Verify fragment size distribution (200-500 bp ideal)

    • Test different extraction buffers for optimal AT4G17830 recovery

    • Consider nuclear isolation before sonication

  • ChIP-specific controls:

    • Input chromatin (non-immunoprecipitated)

    • IgG control or pre-immune serum

    • ChIP in AT4G17830 knockout/knockdown plants

    • Positive control using antibodies against known chromatin factors

    • Non-crosslinked samples to identify non-specific binding

  • ChIP protocol optimization:

    • Test different antibody amounts (2-10 μg typically)

    • Optimize antibody incubation time (overnight at 4°C optimal)

    • Compare different bead types (protein A, G, or A/G)

    • Establish stringent washing conditions to reduce background

  • Data analysis considerations:

    • Design primers for expected and control regions

    • Include both positive and negative genomic regions

    • Calculate enrichment relative to input and IgG control

    • Perform biological replicates (minimum 3) for statistical validity

  • ChIP-seq specific considerations:

    • Ensure sufficient DNA recovery for library preparation

    • Include spike-in controls for normalization

    • Perform appropriate bioinformatic analyses

    • Validate key findings with ChIP-qPCR

  • Biological interpretation framework:

    • Correlate binding sites with gene expression data

    • Identify enriched motifs at binding sites

    • Integrate with other epigenomic datasets

    • Consider time course experiments to capture dynamic interactions

  • Technical challenges for AT4G17830 ChIP:

    • As a metabolic enzyme, AT4G17830 may have non-chromatin functions

    • Distinguish between direct DNA binding and protein-protein interactions

    • Consider low abundance in chromatin fraction

    • Address potential indirect chromatin associations

  • Validation strategies:

    • Confirm key targets by ChIP-qPCR using independent antibodies

    • Perform reciprocal ChIP using antibodies against interacting proteins

    • Use alternative methods (DAP-seq, CUT&RUN) for validation

    • Generate transgenic plants expressing tagged AT4G17830 for validation

This comprehensive approach ensures reliable and interpretable ChIP results when studying AT4G17830 chromatin interactions, which may reveal unexpected nuclear functions beyond its known role in ornithine biosynthesis .

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.