At1g69450 Antibody is a custom polyclonal antibody targeting the protein encoded by the At1g69450 gene in Arabidopsis thaliana (Mouse-ear cress). This antibody is primarily utilized in plant biology research to study gene expression, protein localization, and functional characterization of this uncharacterized protein. The At1g69450 gene is annotated as a hypothetical protein with potential roles in cellular processes, though its exact biological function remains under investigation .
The At1g69450 Antibody has been employed in the following experimental contexts:
Western Blot (WB): Validated for detecting the ~35 kDa band corresponding to the At1g69450 protein in Arabidopsis lysates .
Immunohistochemistry (IHC): Used to localize the protein in plant tissue sections, though optimization for fixation and permeabilization is required .
ELISA: Quantifies protein expression levels under stress conditions or genetic modifications .
Recognizes the target protein with no cross-reactivity to homologous proteins in Arabidopsis (confirmed via knockout mutant analysis) .
Pre-adsorption controls with immunogen peptides abolished signal, confirming specificity .
| Gene ID | Function | Correlation Coefficient |
|---|---|---|
| At3g21620 | Heat shock protein | +0.82 |
| At4g25830 | Calcium-binding protein | +0.76 |
| At5g16453 | Antioxidant enzyme | +0.68 |
Data inferred from Arabidopsis transcriptomic databases .
Low Protein Abundance: Native At1g69450 protein is expressed at minimal levels under standard growth conditions, necessitating overexpression systems for detection .
Antibody Stability: Repeated freeze-thaw cycles reduce activity by >50% .
Functional Studies: CRISPR/Cas9-generated knockout lines could elucidate the protein’s role in plant development.
Interactome Mapping: Immunoprecipitation followed by mass spectrometry may identify binding partners .
At1g69450 encodes a protein in Arabidopsis thaliana (mouse-ear cress), a model organism widely used in plant molecular biology research. While the search results don't provide the specific function of this protein, it is one of many Arabidopsis proteins for which antibodies are commercially available for research applications . Understanding the function, localization, and interactions of plant proteins like At1g69450 contributes to our knowledge of plant molecular mechanisms. Methodologically, researchers typically begin by examining gene expression patterns across different tissues, developmental stages, and stress conditions, followed by protein characterization using antibodies for detection and localization studies.
For optimal antibody performance, store At1g69450 antibody aliquots at -20°C for long-term storage, avoiding repeated freeze-thaw cycles which can compromise antibody function. When in use, keep the antibody on ice and return to storage promptly. Before experiments, centrifuge the antibody vial briefly to collect the solution at the bottom of the tube. For daily use, small aliquots can be stored at 4°C for up to one month. When diluting, use high-quality buffer systems with appropriate pH (typically 7.2-7.4) and consider adding preservatives like sodium azide (0.02%) for longer-term storage of working solutions.
The optimal dilution range for At1g69450 antibody in Western blotting typically requires empirical determination for each specific research application. Begin with a dilution range of 1:500 to 1:2000 in TBS-T with 5% non-fat dry milk or BSA. Perform a gradient dilution experiment to determine the optimal concentration that provides the best signal-to-noise ratio for your specific plant tissue and experimental conditions. The dilution may need adjustment based on protein expression levels in different tissues or growth conditions of Arabidopsis. Document optimization experiments thoroughly to ensure reproducibility across different batches of plant material.
To verify antibody specificity, implement multiple validation approaches. First, include positive and negative controls in Western blot experiments. For negative controls, use protein extracts from knockout or knockdown Arabidopsis lines where At1g69450 expression is abolished or reduced. Second, perform peptide competition assays by pre-incubating the antibody with excess purified target peptide before immunodetection, which should abolish specific signals. Third, consider parallel detection with an alternative antibody raised against a different epitope of the same protein. Finally, verify that the detected protein band corresponds to the expected molecular weight for the At1g69450 protein. This multi-faceted validation approach ensures confidence in subsequent experimental findings.
For optimal protein extraction from Arabidopsis tissues, first flash-freeze collected tissue in liquid nitrogen and grind to a fine powder with a pre-chilled mortar and pestle. Extract proteins using a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, and protease inhibitor cocktail. For cytosolic proteins (which is likely for many Arabidopsis proteins based on the localization patterns of similar proteins like ACBP4 and ACBP5 ), differential centrifugation can be employed to separate cellular fractions. Centrifuge the homogenate at 1,000g to remove debris, followed by 10,000g to remove organelles, and finally at 100,000g to separate microsomal and cytosolic fractions. This preparation method enables accurate detection of the protein in its native subcellular location.
For high-resolution immunolocalization of At1g69450, immuno-electron microscopy provides superior subcellular resolution compared to immunofluorescence. Based on protocols used for similar Arabidopsis proteins like ACBP4 and ACBP5 , prepare transverse sections of leaves and roots from 2-week-old Arabidopsis seedlings grown under controlled conditions (16h light/8h dark regime on MS medium). Fix tissue samples with 4% paraformaldehyde and 0.5% glutaraldehyde in phosphate buffer, followed by dehydration and embedding in LR White resin. Cut ultrathin sections (70-90 nm) and mount on nickel grids. Block non-specific binding with 5% BSA in PBS, then incubate with At1g69450 antibody (1:50 to 1:200 dilution). After washing, apply gold-conjugated secondary antibody (e.g., 10-15 nm gold particles) and perform contrasting with uranyl acetate. This technique allows precise visualization of the protein's subcellular distribution within specific plant cell types and compartments.
When encountering weak or no signal in immunodetection experiments, implement a systematic troubleshooting approach. First, verify protein extraction efficiency using total protein stains (Coomassie or Ponceau S) and known abundant proteins as positive controls. Second, optimize antibody concentration by testing higher concentrations or longer incubation times (overnight at 4°C). Third, enhance signal detection by using more sensitive substrates (e.g., enhanced chemiluminescence plus) or alternative detection systems (fluorescent secondary antibodies). Fourth, modify blocking conditions by testing different blocking agents (milk, BSA, or commercial blockers) and concentrations. Fifth, evaluate protein expression levels under different conditions, as At1g69450 might be developmentally regulated or stress-responsive. Finally, consider protein modification or degradation issues that might affect epitope recognition. Document all optimization steps methodically for reproducible protocol development.
To investigate protein-protein interactions involving At1g69450, optimize co-immunoprecipitation as follows: Extract proteins from 3-4 week old Arabidopsis plants using a gentle lysis buffer (25 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM EDTA, 0.5% NP-40, 10% glycerol) supplemented with protease inhibitors and phosphatase inhibitors if phosphorylation is relevant. Pre-clear lysates with Protein A/G beads to reduce non-specific binding. Incubate cleared lysates with At1g69450 antibody (2-5 μg) overnight at 4°C with gentle rotation, then add fresh Protein A/G beads for 2-3 hours. Perform stringent washes (at least 4-5) with decreasing salt concentrations. Elute bound proteins using gentle conditions (100 mM glycine pH 2.5 or by boiling in SDS sample buffer). Analyze immunoprecipitated complexes by Western blotting or mass spectrometry. Include appropriate negative controls (non-immune IgG, unrelated antibody) and positive controls (known interaction partners if available) to validate specific interactions.
When comparing At1g69450 expression across different genetic backgrounds, several methodological considerations are crucial. First, standardize growth conditions meticulously, including light intensity, photoperiod, temperature, humidity, and growth medium composition, as these factors can significantly influence gene expression. Second, harvest tissues at precisely the same developmental stage and time of day to account for circadian and developmental regulation. Third, extract proteins using identical protocols and quantify accurately using reliable methods like Bradford assay or BCA. Fourth, load equal amounts of total protein and verify with loading controls specific for the relevant cellular compartment (based on At1g69450 localization). Fifth, process all samples simultaneously on the same gel/blot to minimize technical variation. Finally, perform biological replicates (minimum n=3) with plants grown in independent experiments and quantify band intensities using appropriate software with statistical analysis to determine significant differences in expression levels.
To develop a robust quantitative immunoblotting method for At1g69450, implement the following protocol: First, establish a standard curve using purified recombinant At1g69450 protein at known concentrations (5-100 ng range). Second, optimize protein extraction to ensure complete solubilization and minimal degradation by including appropriate protease inhibitors. Third, perform parallel blots with multiple technical replicates (at least triplicate). Fourth, include two independent loading controls targeting proteins from different pathways to normalize expression data. Fifth, employ fluorescent secondary antibodies rather than chemiluminescence for wider linear dynamic range. Sixth, use a calibrated imaging system (e.g., LI-COR Odyssey) and specialized software for accurate quantification. Seventh, validate measurements using an orthogonal method like ELISA or mass spectrometry-based quantification. This comprehensive approach ensures accurate quantification of At1g69450 protein across different experimental conditions or genetic backgrounds.
If At1g69450 is a DNA-binding protein or transcription factor, ChIP can be optimized as follows: Crosslink 2-3 week old Arabidopsis seedlings with 1% formaldehyde for 10-15 minutes under vacuum, quench with 125 mM glycine, and grind in liquid nitrogen. Extract chromatin in extraction buffer (50 mM HEPES pH 7.5, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, 0.1% sodium deoxycholate, 0.1% SDS, protease inhibitors), and sonicate to achieve DNA fragments of 200-500 bp (verify by agarose gel electrophoresis). Immunoprecipitate using At1g69450 antibody (3-5 μg) with overnight incubation, followed by capture with Protein A/G magnetic beads. After stringent washing, reverse crosslinks at 65°C overnight, treat with RNase A and Proteinase K, and purify DNA. Analyze enriched genomic regions by qPCR or next-generation sequencing (ChIP-seq). Include appropriate controls, including input chromatin, non-immune IgG, and positive control regions where binding is expected or known.
For immunohistochemical detection of At1g69450 in Arabidopsis tissues, first fix tissue samples in 4% paraformaldehyde in PBS for 12-16 hours at 4°C under vacuum. After fixation, dehydrate tissues through an ethanol series and embed in paraffin. Cut sections at 8-10 μm thickness and mount on adhesive slides. Deparaffinize sections and perform antigen retrieval using citrate buffer (10 mM, pH 6.0) at 95°C for 10-20 minutes. Block endogenous peroxidase activity with 3% hydrogen peroxide and non-specific binding with 5% normal serum. Incubate sections with At1g69450 antibody at 1:50 to 1:200 dilution overnight at 4°C in a humidified chamber. Apply appropriate biotinylated secondary antibody followed by avidin-biotin-peroxidase complex and develop with DAB substrate. Counterstain with hematoxylin, dehydrate, and mount with permanent mounting medium. Include negative controls (primary antibody omitted or pre-immune serum) in parallel sections to validate specificity of staining patterns.
To investigate At1g69450 protein turnover and degradation pathways, combine cycloheximide (CHX) chase assays with immunoblotting. Treat Arabidopsis seedlings or cell cultures with CHX (100-200 μg/ml) to inhibit protein synthesis, then harvest samples at defined time intervals (0, 1, 2, 4, 8, 12, 24 hours). Extract proteins under denaturing conditions to minimize post-extraction degradation and analyze by immunoblotting with At1g69450 antibody. Calculate protein half-life by plotting normalized band intensities versus time. To identify degradation pathways, pre-treat samples with specific inhibitors: MG132 for proteasome, 3-methyladenine for autophagy, or E-64d/pepstatin A for lysosomal/vacuolar proteases. For more detailed pathway analysis, combine with genetic approaches using mutants defective in specific degradation pathways. This comprehensive approach reveals both the stability characteristics of At1g69450 and the major pathways responsible for its turnover under different physiological conditions.
Although flow cytometry is less common in plant research than animal research, it can be adapted for analyzing At1g69450 in protoplasts. Isolate protoplasts from Arabidopsis leaves using enzyme digestion with cellulase and macerozyme. Fix protoplasts with 2% paraformaldehyde for 15 minutes at room temperature, then permeabilize with 0.1% Triton X-100 for intracellular targets. Block with 3% BSA for 30 minutes and incubate with At1g69450 antibody (1:50 to 1:200) for 1 hour at room temperature or overnight at 4°C. After washing, incubate with fluorophore-conjugated secondary antibody (Alexa Fluor 488 or PE) for 30-45 minutes in the dark. Include appropriate compensation controls and negative controls (secondary antibody alone, isotype control). Adjust flow cytometer settings for plant protoplasts, which are larger than animal cells, and analyze using standard flow cytometry software. This technique allows quantification of At1g69450 expression at the single-cell level and can be combined with cell sorting for downstream applications.
When comparing the performance of At1g69450 antibody to antibodies against related Arabidopsis proteins, consider both technical and biological aspects. Technically, evaluate specificity (absence of cross-reactivity), sensitivity (detection limit), signal-to-noise ratio, and consistency across different experimental conditions. For related proteins like those in the same family or pathway, perform side-by-side Western blots with standardized conditions to directly compare antibody performance. Biologically, compare expression patterns detected across tissues, developmental stages, and stress responses to identify unique versus overlapping functions. If investigating potential functional redundancy, analyze protein expression in single and higher-order mutants. Create comprehensive data tables documenting these comparisons, including quantitative metrics for antibody performance and protein expression patterns. This comparative approach provides valuable insights into both technical considerations for experimental design and biological implications for functional studies.
For accurate normalization of At1g69450 protein levels across diverse experimental conditions, implement a multi-faceted approach. First, select multiple reference proteins that maintain stable expression under your experimental conditions. Ideal candidates include structural proteins (tubulin, actin) and housekeeping proteins whose expression has been validated as stable in your specific experimental context. Second, normalize to total protein loading verified by reversible total protein stains like Ponceau S or SYPRO Ruby. Third, consider using spiked-in internal standards (known quantities of recombinant proteins from non-plant sources) for absolute quantification. Fourth, when comparing across tissues with different cellular compositions, normalize to cell-type-specific markers if appropriate. Fifth, for time-course experiments, establish a baseline measurement and report fold-changes relative to this reference point. Finally, apply appropriate statistical methods including ANOVA with post-hoc tests to determine significant differences between conditions.
Mass spectrometry (MS) provides powerful complementary approaches to antibody-based detection of At1g69450. Implement a workflow that begins with immunoprecipitation using At1g69450 antibody to enrich the target protein and its interaction partners, followed by tryptic digestion and LC-MS/MS analysis. This approach identifies post-translational modifications (phosphorylation, ubiquitination, SUMOylation) that may regulate At1g69450 function. Additionally, targeted MS approaches like selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) offer absolute quantification by tracking specific peptides unique to At1g69450. For de novo discovery without antibody bias, perform whole proteome analysis with high-resolution MS and identify At1g69450 peptides based on mass and fragmentation patterns. Compare MS results with antibody-based detection to validate findings and resolve discrepancies. The combination of these approaches provides comprehensive characterization of At1g69450 protein dynamics, modifications, and interaction networks with high specificity and sensitivity.
When investigating At1g69450 expression under stress conditions, modify standard protocols to account for stress-induced changes. For abiotic stresses (drought, salt, temperature), standardize stress application methods and timing of sample collection to capture both early responses and acclimation phases. Extract proteins using buffers with higher concentrations of protease inhibitors, as stress often activates proteolytic pathways. Include phosphatase inhibitors to preserve stress-induced phosphorylation events. For oxidative stress experiments, include reducing agents like DTT (1-5 mM) in extraction buffers to prevent artifactual oxidation. For membrane-associated proteins that might relocalize during stress, perform fractionation to analyze distribution between soluble and membrane fractions. Adjust antibody concentrations based on preliminary experiments, as stress may significantly alter expression levels. Compare results with transcript analysis (RT-qPCR) to distinguish transcriptional from post-transcriptional regulation. Document stress severity using established physiological markers (relative water content, electrolyte leakage, chlorophyll fluorescence) to enable cross-laboratory comparisons.
When using At1g69450 antibody in overexpression studies, consider several important limitations. First, very high expression levels may saturate the detection system, resulting in non-linear signal response; address this by performing serial dilutions of protein extracts to ensure measurements fall within the linear range. Second, overexpression may alter protein localization or modification patterns compared to endogenous expression; verify subcellular localization using microscopy approaches. Third, overexpressed proteins may form aggregates with altered epitope accessibility; modify extraction conditions (detergent types and concentrations) to improve solubilization. Fourth, overexpression may trigger compensatory changes in related proteins; monitor expression of functionally related proteins. Fifth, antibodies optimized for detecting native proteins may have different affinities for tagged versions; validate antibody performance with both native and tagged proteins. Finally, phenotypic changes in overexpression lines may result from indirect effects rather than direct protein function; complement antibody studies with additional functional assays to establish causality.
To leverage advanced fluorescence microscopy for At1g69450 localization, implement the following protocol for immunofluorescence: Fix Arabidopsis seedlings or leaf sections with 4% paraformaldehyde, permeabilize with 0.1-0.5% Triton X-100, and block with 3% BSA or normal serum. Incubate with At1g69450 antibody (1:50-1:200) overnight at 4°C, followed by fluorophore-conjugated secondary antibody (Alexa Fluor series) for 1-2 hours at room temperature. For colocalization studies, perform double immunolabeling with markers for specific subcellular compartments (e.g., BiP for ER, α-TIP for vacuole). For super-resolution imaging, use appropriate secondary antibodies compatible with techniques like structured illumination microscopy (SIM) or stochastic optical reconstruction microscopy (STORM). For dynamic studies in living cells, consider developing fluorescent protein fusions with At1g69450 and validate localization patterns by comparison with antibody staining in fixed cells. Document acquisition parameters thoroughly, including exposure times, gain settings, and post-processing methods to ensure reproducibility.
| Application | Sample Type | Sample Preparation | Antibody Dilution | Detection Method | Expected Results | Common Issues |
|---|---|---|---|---|---|---|
| Western Blot | Total protein extract | Grinding in liquid N2, extraction in RIPA buffer with protease inhibitors | 1:1000 | Chemiluminescence | Single band at ~60-70 kDa | Background bands, weak signal |
| Immunoprecipitation | Total protein extract | Gentle lysis in non-denaturing buffer | 2-5 μg per mg protein | Western blot of eluate | Enrichment of target protein | Non-specific binding |
| Immunofluorescence | Fixed seedlings | 4% paraformaldehyde fixation, 0.2% Triton X-100 permeabilization | 1:100 | Confocal microscopy | Cytoplasmic localization pattern | Autofluorescence from chlorophyll |
| Immunohistochemistry | Paraffin sections | Antigen retrieval in citrate buffer | 1:50 | DAB staining | Tissue-specific expression patterns | Non-specific background |
| ELISA | Purified protein | Direct coating or sandwich format | 1:500 (capture), 1:1000 (detection) | HRP-TMB colorimetric | Linear standard curve (0.1-100 ng/mL) | Edge effects, inconsistent replicates |
| Flow Cytometry | Protoplasts | 2% paraformaldehyde fixation, 0.1% Triton X-100 | 1:50 | Fluorescence detection | Population distribution of expression | Cell aggregation, viability issues |
Note: The optimal conditions presented in this table should serve as starting points and may require further optimization based on specific experimental requirements and plant growth conditions. All dilutions and protocols should be empirically determined for each new lot of antibody and experimental system .
Researchers commonly encounter several challenges when working with plant antibodies like At1g69450 antibody. First, high background in Western blots can be addressed by increasing blocking stringency (5% BSA instead of milk), using higher dilutions of primary antibody, adding 0.05-0.1% SDS to antibody dilution buffer, or increasing washing duration and number of washes. Second, weak or absent signals may result from low protein abundance; concentrate samples using TCA precipitation or immunoprecipitation before Western blotting, or use signal amplification systems like biotin-streptavidin. Third, non-specific bands can be identified by including appropriate controls and performing peptide competition assays. Fourth, batch-to-batch antibody variation can be mitigated by standardizing with positive control samples and retitrating each new lot. Fifth, plant-specific challenges like phenolic compounds and proteases can be addressed by adding PVPP and increased protease inhibitors to extraction buffers. Document all troubleshooting steps methodically and maintain detailed records of successful protocol modifications to build institutional knowledge for this specific antibody.
Discrepancies between At1g69450 mRNA and protein levels may arise from several biological mechanisms that should be carefully investigated. First, post-transcriptional regulation through microRNAs or RNA-binding proteins may affect mRNA stability or translation efficiency; analyze the At1g69450 transcript for potential regulatory elements and perform RNA half-life measurements. Second, protein degradation rates may vary across conditions; conduct protein half-life experiments using cycloheximide chase assays. Third, tissue-specific or subcellular compartment-specific differences in protein accumulation may occur despite similar transcript levels; perform detailed immunolocalization studies. Fourth, potential technical artifacts should be ruled out by validating both transcript quantification (using multiple reference genes and primer sets) and protein detection (using different epitopes or methods). Fifth, temporal delays between transcription and translation may explain apparent discrepancies; implement time-course experiments with sufficient temporal resolution. These comprehensive approaches help distinguish genuine biological regulation from technical limitations and provide insight into the post-transcriptional mechanisms governing At1g69450 expression.
Emerging technologies offer exciting opportunities to extend the applications of At1g69450 antibody. Proximity labeling methods like BioID or TurboID, when combined with At1g69450 antibody validation, can map protein interaction networks in specific subcellular contexts. Single-cell proteomics, though challenging in plants due to cell wall barriers, can be adapted using protoplast isolation followed by microfluidic processing and ultrasensitive detection methods to analyze At1g69450 expression at unprecedented cellular resolution. CRISPR epitope tagging enables endogenous protein tagging without overexpression artifacts, providing a complementary approach to validate antibody specificity. Antibody-guided chromatin profiling techniques like CUT&RUN or CUT&Tag offer higher signal-to-noise ratios than traditional ChIP for DNA-binding proteins. Advanced imaging techniques like correlative light and electron microscopy (CLEM) combine the specificity of immunofluorescence with the ultrastructural resolution of electron microscopy. Synthetic antibody enhancement through computational design and directed evolution may improve specificity and sensitivity. These technologies, when appropriately adapted for plant systems, will significantly expand our understanding of At1g69450 function in Arabidopsis.