At3g49630 Antibody

Shipped with Ice Packs
In Stock

Description

At3g49630: Functional Role and Biochemical Characteristics

At3g49630 is a plant enzyme involved in nitrogen metabolism. Key findings include:

  • Catalytic Activity: Converts homoarginine to guanidine and Δ1-piperideine-6-carboxylate (P6C) via C6-hydroxylation .

  • Kinetic Parameters:

    ParameterValueSource
    KMK_M (homoarginine)4.6 ± 0.2 mM
    AmaxA_{\text{max}}5.6 ± 1.2 nmol·s⁻¹·mg⁻¹
  • Genetic Studies: CRISPR/Cas9 mutants of At3g49630 and its paralogs (Din11, At3g50210) showed no phenotypic differences from wild-type plants under normal growth conditions .

Antibody Research Context

While no At3g49630-specific antibodies are documented, related antibody technologies include:

  • APOBEC3B Antibodies: Rabbit monoclonal antibodies (e.g., 5210-87-13) validated for ELISA, immunoblotting, and immunohistochemistry .

  • Cross-Reactive IgG3 Antibodies: Human antibodies targeting conserved glycans across viral families (e.g., mAb 2526) .

  • OmniAb Platforms: Technologies enabling discovery of antibodies with ultralong CDR-H3 domains for novel epitope targeting .

Potential Explanations for Lack of At3g49630 Antibody Data

  • Biological Role: At3g49630 is a plant-specific enzyme with no known homologs in humans or pathogens, reducing therapeutic interest .

  • Research Focus: Existing studies prioritize its enzymatic function in guanidine biosynthesis rather than immunological applications .

  • Commercial Availability: No commercial vendors (e.g., Thermo Fisher, Abcam) list At3g49630 antibodies in catalogs as of 2025.

Methodological Considerations for Future Work

If generating an At3g49630 antibody were pursued:

  • Immunogen Design: Use recombinant At3g49630 protein or peptide epitopes from conserved regions.

  • Validation: Employ LC-MS/MS to confirm target specificity and avoid cross-reactivity with paralogs (e.g., At3g50210) .

  • Applications: Potential use in plant biochemistry studies to localize the enzyme or quantify expression under stress conditions.

Comparative Analysis of Arabidopsis 2-ODD-C23 Enzymes

EnzymeKMK_M (homoarginine)AmaxA_{\text{max}} (nmol·s⁻¹·mg⁻¹)Subcellular Localization
At3g502100.78 ± 0.15 mM21 ± 1.7Cytoplasm/Nucleus
Din11s1.9 ± 0.3 mM13.4 ± 3.2Cytoplasm
At3g496304.6 ± 0.2 mM5.6 ± 1.2Cytoplasm

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 weeks lead time (made-to-order)
Synonyms
At3g49630 antibody; T9C5.220Probable 2-oxoglutarate-dependent dioxygenase At3g49630 antibody; EC 1.14.11.- antibody
Target Names
At3g49630
Uniprot No.

Q&A

What is the At3g49630 gene and why develop antibodies against its protein products?

At3g49630 is a gene locus in the Arabidopsis thaliana genome that encodes specific proteins involved in plant development and spatial pattern formation. Researchers develop antibodies against these protein products to investigate their spatial and temporal expression patterns, protein-protein interactions, and functional roles in developmental processes. Antibodies serve as crucial tools for tracking protein localization, quantifying expression levels, and studying protein modifications in plant developmental biology. These investigations contribute to our fundamental understanding of pattern formation mechanisms that are central to plant morphogenesis.

What validation methods should be used to confirm At3g49630 antibody specificity?

Validation of At3g49630 antibodies requires multiple complementary approaches to ensure specificity. First, perform Western blot analysis comparing wild-type plants with At3g49630 knockout or knockdown lines to confirm the absence of signal in mutant lines. Second, conduct immunoprecipitation followed by mass spectrometry to verify that the antibody pulls down the intended target protein. Third, use immunolocalization in tissues with known expression patterns based on transcriptomic data. Fourth, pre-absorption controls with purified antigen should eliminate signal if the antibody is specific. Finally, test cross-reactivity with closely related protein family members to establish specificity within the protein family. Each validation method provides distinct evidence of antibody specificity, and concordance across multiple methods strengthens confidence in antibody performance.

What are the optimal fixation and tissue preparation methods for immunolocalization of At3g49630 protein in plant tissues?

Optimal fixation for At3g49630 immunolocalization in plant tissues typically begins with 4% paraformaldehyde in phosphate buffer (pH 7.2) for 2-4 hours at room temperature or overnight at 4°C. For membrane-associated proteins, adding 0.1-0.5% glutaraldehyde may preserve antigenicity while maintaining structural integrity. Following fixation, tissues should undergo carefully controlled dehydration through an ethanol series (30%, 50%, 70%, 90%, 100%) to prevent tissue distortion. For paraffin embedding, low-temperature embedding protocols (below 60°C) help preserve antigenicity. Alternatively, cryosectioning following sucrose infiltration (10%, 20%, 30% in phosphate buffer) and freezing in OCT compound preserves antigen recognition for antibodies with fixation-sensitive epitopes. Antigen retrieval using citrate buffer (pH 6.0) heating may be necessary to expose masked epitopes in some fixed tissues. These preparation methods must be empirically optimized for the specific structural context of the At3g49630 protein to balance structural preservation with epitope accessibility.

How should researchers optimize immunoblotting protocols for detecting At3g49630 protein?

Optimization of immunoblotting for At3g49630 protein detection requires methodical adjustment of multiple parameters. First, protein extraction should incorporate appropriate detergents (such as 1% Triton X-100 or 0.5% SDS) based on the protein's subcellular localization and hydrophobicity. Second, optimize protein loading (typically 20-50 μg total protein) and separation using gradient gels (4-12%) for better resolution. Third, test multiple transfer conditions, including both wet transfer (overnight at 30V, 4°C) and semi-dry transfer systems (25V for 30 minutes) to determine optimal protein transfer efficiency. Fourth, blocking conditions should be systematically tested (5% non-fat milk, 3% BSA, or commercial blocking reagents) for at least 1 hour at room temperature. Fifth, antibody dilution series (1:500 to 1:5000) and incubation conditions (4°C overnight vs. room temperature for 2 hours) should be compared to identify optimal signal-to-noise ratio. Finally, detection system sensitivity should be matched to expected protein abundance, using chemiluminescence for low abundance proteins and colorimetric detection for highly abundant targets. All optimization steps should include appropriate positive and negative controls to validate protocol adjustments.

How can researchers address epitope masking issues when At3g49630 forms protein complexes?

Addressing epitope masking in At3g49630 protein complexes requires strategic experimental approaches. First, implement a panel of antibodies targeting different epitopes across the protein to increase the likelihood of accessible binding sites. Second, employ multiple protein extraction protocols with varying detergent strengths (from mild non-ionic detergents like 0.1% Triton X-100 to stronger ionic detergents like 1% SDS) to partially disrupt protein-protein interactions while maintaining antibody recognition. Third, consider native versus denaturing conditions in immunoprecipitation experiments; native conditions may preserve important biological interactions while denaturing conditions may expose hidden epitopes. Fourth, use cross-linking approaches with variable spacer arm lengths to stabilize interactions while potentially preserving antibody accessibility. Finally, computational modeling of protein complex structures, when available, can guide epitope selection and predict potential masking issues. For immunohistochemistry applications, test various antigen retrieval methods, including heat-induced epitope retrieval in citrate buffer (pH 6.0) or Tris-EDTA buffer (pH 9.0), and enzymatic retrieval using proteinase K treatment at optimized concentrations and incubation times.

What are the best approaches for quantifying At3g49630 protein levels in different developmental stages?

Quantification of At3g49630 protein across developmental stages requires rigorous methodology to ensure accurate comparative analysis. First, establish a standardized tissue sampling protocol that controls for circadian variation and defines precise developmental stages based on morphological markers rather than chronological age. Second, implement absolute quantification using a standard curve of recombinant At3g49630 protein at known concentrations, enabling direct comparison between developmental timepoints. Third, supplement immunoblotting with either ELISA or protein mass spectrometry with isotope-labeled reference peptides for orthogonal verification of expression patterns. Fourth, normalize protein measurements against multiple housekeeping proteins that have been validated for stability across the developmental stages being studied. Fifth, perform biological replicates from independent plant populations (n≥3) and technical replicates (n≥3) with randomized sample processing order to control for batch effects. Finally, statistical analysis should account for the non-linear nature of many developmental transitions using appropriate regression models rather than simple pairwise comparisons. This comprehensive approach provides robust quantitative assessment of protein expression dynamics throughout development.

How can researchers distinguish between different post-translational modifications of At3g49630 using antibodies?

Distinguishing post-translational modifications (PTMs) of At3g49630 protein requires specialized antibody approaches and complementary techniques. First, develop or acquire modification-specific antibodies that recognize the protein only when modified in a particular way (phosphorylated, acetylated, ubiquitinated, etc.). These antibodies should be validated against synthesized peptides containing the specific modification and control peptides. Second, implement a sequential immunoprecipitation strategy: first isolating all forms of At3g49630 with a general antibody, then probing the precipitate with modification-specific antibodies. Third, combine antibody detection with mass spectrometry to map the precise sites and types of modifications present. Fourth, use lambda phosphatase treatment and other enzymes that remove specific modifications to demonstrate specificity of the modification-specific antibodies. Fifth, compare PTM patterns across different physiological conditions and developmental stages known to affect protein function. Finally, correlate detected modifications with functional assays to establish biological relevance of the identified PTMs. This integration of antibody-based detection with orthogonal approaches provides comprehensive characterization of At3g49630 post-translational regulation.

What strategies can overcome cross-reactivity issues when studying At3g49630 protein in different plant species?

Overcoming cross-reactivity challenges when extending At3g49630 antibody use to different plant species requires systematic approach refinement. First, perform detailed sequence alignments of At3g49630 homologs across target species to identify conserved and variable regions, guiding epitope selection toward highly conserved sequences for broad species applicability. Second, validate antibodies in each new species using knockout/knockdown lines or heterologous expression systems as positive controls. Third, implement gradient elution in affinity purification of antibodies to separate high-affinity (likely specific) from low-affinity (potentially cross-reactive) antibody populations. Fourth, develop a panel of blocking peptides derived from potential cross-reactive proteins identified through bioinformatic analysis to test and eliminate cross-reactivity. Fifth, employ parallel detection methods like RNA-seq or proteomics to correlate antibody signals with independent measures of protein presence. Finally, consider developing species-specific antibodies for crucial experiments where cross-reactivity cannot be eliminated. This approach maximizes the utility of At3g49630 antibodies across diverse plant systems while maintaining scientific rigor.

How can At3g49630 antibodies be adapted for high-throughput phenotypic screening applications?

Adapting At3g49630 antibodies for high-throughput phenotypic screening requires methodological optimization for automation and scalability. First, develop a microplate-based immunoassay (ELISA or similar format) with optimized antibody concentrations determined through checkerboard titration experiments to maximize signal-to-noise ratio. Second, minimize protocol complexity by reducing wash steps and incubation times while maintaining assay performance through accelerated kinetics at higher temperatures or improved antibody binding conditions. Third, implement automated liquid handling systems with validated precision and accuracy for critical steps such as antibody addition and detection reagent application. Fourth, incorporate internal controls on each plate, including standard curves and reference samples, to normalize inter-plate variation. Fifth, develop image-based screening using fluorescently-labeled At3g49630 antibodies compatible with high-content imaging systems, allowing simultaneous assessment of protein levels, subcellular localization, and phenotypic parameters. Finally, establish computational pipelines for automated image analysis and data normalization that can handle the large datasets generated. This integrated approach enables screening of thousands of genetic variants, chemical treatments, or environmental conditions for their effects on At3g49630 protein expression and function.

How can researchers combine At3g49630 antibodies with CRISPR-Cas9 gene editing to study protein function?

Integrating At3g49630 antibodies with CRISPR-Cas9 gene editing creates powerful approaches for functional protein studies. First, use antibodies to validate CRISPR-edited lines by confirming protein depletion in knockout mutants or detecting epitope-tagged proteins in knock-in lines through Western blotting and immunolocalization. Second, develop a systematic series of domain-deletion or point-mutation CRISPR variants to map functional protein regions, using antibodies to assess expression, localization, and interaction patterns of each variant. Third, implement CRISPR interference (CRISPRi) or activation (CRISPRa) systems to modulate At3g49630 expression levels, using antibodies to quantify the precise relationship between transcriptional manipulation and resulting protein levels. Fourth, combine antibody-based protein detection with phenotypic analysis of CRISPR-edited plants to establish causative relationships between protein expression patterns and developmental outcomes. Fifth, use antibodies to assess compensatory protein expression changes in related family members following CRISPR modification of At3g49630. Finally, implement proximity labeling techniques with engineered versions of At3g49630 to identify interaction partners under different conditions, validating interactions with co-immunoprecipitation using At3g49630 antibodies. This integrated approach leverages the precision of CRISPR genome editing with the detection capabilities of antibodies to comprehensively characterize protein function.

What are the considerations for using At3g49630 antibodies in chromatin immunoprecipitation (ChIP) experiments?

Using At3g49630 antibodies in ChIP experiments requires specialized considerations to achieve successful chromatin protein detection. First, determine whether At3g49630 is a direct DNA-binding protein or associates with chromatin through protein-protein interactions, as this influences crosslinking strategies—formaldehyde works well for direct DNA interactions while dual crosslinking with disuccinimidyl glutarate followed by formaldehyde may better preserve protein-protein interactions. Second, optimize sonication conditions (amplitude, pulse duration, cycle number) for plant tissues to achieve chromatin fragments of 200-500 bp without destroying epitope recognition. Third, implement a rigorous antibody validation workflow specifically for ChIP applications, including testing multiple antibody concentrations (2-10 μg per reaction) and comparing enrichment at predicted binding sites versus negative control regions. Fourth, include appropriate controls in every experiment: input chromatin (pre-immunoprecipitation), IgG control immunoprecipitation, and positive control antibodies against histone modifications or transcription factors with known binding patterns. Fifth, consider developing epitope-tagged versions of At3g49630 for comparative ChIP using anti-tag antibodies to corroborate results from native protein antibodies. Finally, verify ChIP-seq peaks through orthogonal methods such as in vitro DNA binding assays or reporter gene experiments to confirm functional significance of identified binding sites.

What methods can be used to study At3g49630 protein turnover and degradation pathways using antibodies?

Studying At3g49630 protein turnover and degradation requires combining antibody-based detection with targeted experimental manipulations. First, implement cycloheximide chase assays where protein synthesis is blocked and At3g49630 levels are monitored via immunoblotting at timed intervals (0, 1, 3, 6, 12, 24 hours) to determine protein half-life under various conditions. Second, use proteasome inhibitors (MG132, bortezomib) and autophagy inhibitors (3-methyladenine, bafilomycin A1) to block specific degradation pathways, then assess protein accumulation using quantitative immunoblotting to identify the primary degradation mechanism. Third, develop a pulse-chase experimental system using inducible expression of tagged At3g49630 variants, followed by antibody detection to track specific protein populations over time. Fourth, combine co-immunoprecipitation using At3g49630 antibodies with mass spectrometry to identify associated degradation machinery components, such as E3 ubiquitin ligases or autophagy adaptors. Fifth, use immunolocalization to track changes in subcellular distribution of At3g49630 during stress responses or developmental transitions that may trigger degradation. Finally, develop antibodies specific to ubiquitinated or otherwise modified forms of At3g49630 that mark the protein for degradation. This multi-faceted approach provides mechanistic insight into the regulation of At3g49630 protein stability and turnover.

How do monoclonal and polyclonal antibodies against At3g49630 compare in different applications?

Monoclonal and polyclonal antibodies against At3g49630 present distinct advantages and limitations across research applications. For immunoblotting, monoclonal antibodies typically provide higher specificity but may be more vulnerable to epitope destruction during denaturation, while polyclonal antibodies offer more robust detection by recognizing multiple epitopes but may exhibit batch-to-batch variation. In immunoprecipitation experiments, monoclonal antibodies typically achieve cleaner pull-downs with lower background but may have insufficient affinity for certain conformations of At3g49630, whereas polyclonal antibodies can capture more protein conformations but potentially introduce non-specific interactions. For immunohistochemistry, monoclonal antibodies generally yield more consistent staining patterns with lower background but may be more sensitive to fixation-induced epitope masking, while polyclonal antibodies provide stronger signals through multiple epitope binding but require more rigorous blocking to control background staining. In chromatin immunoprecipitation, monoclonal antibodies offer higher reproducibility for mapping specific binding sites but may be inefficient if their epitope is occluded in chromatin-bound conformations, whereas polyclonal antibodies provide more robust chromatin pull-down but potentially lower resolution of binding sites. For quantitative applications, monoclonal antibodies typically provide more consistent standard curves but narrow dynamic range, while polyclonal antibodies offer broader detection range but require more extensive standardization across experimental batches.

How do different antibody-based methods compare for studying protein-protein interactions involving At3g49630?

Different antibody-based methods for studying At3g49630 protein interactions offer complementary strengths and limitations. Traditional co-immunoprecipitation using At3g49630 antibodies provides direct evidence of interactions under native conditions but is limited to relatively stable interactions and may disrupt weak or transient complexes during washing steps. Proximity ligation assays (PLA) using antibodies against At3g49630 and potential interaction partners offer high sensitivity for detecting proteins within 40nm distance in situ but cannot distinguish direct from indirect interactions within larger complexes. Fluorescence resonance energy transfer (FRET) paired with immunofluorescence using fluorophore-conjugated antibodies enables live visualization of protein proximity but requires careful controls for fluorophore orientation and distance. Bimolecular fluorescence complementation (BiFC) provides strong visual confirmation of interactions but may stabilize transient interactions artificially. Analytical size exclusion chromatography followed by immunoblotting with At3g49630 antibodies allows detection of native complexes but has limited resolution for complex compositional analysis. Chemical crosslinking followed by immunoprecipitation and mass spectrometry (CLMS) offers detailed interaction interface mapping but requires extensive optimization of crosslinker chemistry and concentration. For comprehensive interaction characterization, researchers should employ at least two complementary techniques, ideally combining in vitro and in vivo approaches to validate biologically significant interactions.

What are the comparative advantages of using At3g49630 antibodies versus fluorescent protein fusions for protein localization studies?

Using At3g49630 antibodies versus fluorescent protein fusions for localization studies presents distinct methodological trade-offs. Antibody-based immunolocalization detects native protein without potential functional interference from fusion tags but requires tissue fixation that can introduce artifacts and prevents live-cell imaging. Conversely, fluorescent protein fusions enable non-invasive live-cell tracking of protein dynamics but may alter protein function, localization, or stability due to steric hindrance. Immunolocalization provides a snapshot of endogenous protein distribution across all expressing cells in a tissue but suffers from potential fixation artifacts and non-specific background. Fluorescent fusions allow continuous monitoring of protein movement and turnover in individual cells but typically require ectopic expression systems that may not recapitulate native expression levels or patterns. Antibody detection can distinguish post-translational modifications using modification-specific antibodies but requires separate samples for each timepoint. Fluorescent fusions enable single-cell analysis of protein responses to treatments in real time but cannot distinguish modified forms without additional biosensors. Both approaches can be combined in validation experiments: performing immunolocalization on plants expressing fluorescent fusions to confirm concordant localization patterns. This complementary application leverages the strengths of both methods while mitigating their individual limitations for comprehensive localization studies.

How does the sensitivity of At3g49630 antibody detection compare between ELISA, Western blotting, and mass spectrometry approaches?

The comparative sensitivity of At3g49630 detection methods reveals important methodological considerations. ELISA typically offers the highest theoretical sensitivity, capable of detecting At3g49630 protein in the picogram range (approximately 10⁻¹² g/mL) under optimal conditions with highly specific antibodies. This high sensitivity comes with stringent requirements for antibody specificity to avoid false positives. Western blotting provides moderate sensitivity in the nanogram range (approximately 10⁻⁹ g/mL) but offers additional confidence through molecular weight verification and the ability to distinguish potential cross-reactive proteins by size. Mass spectrometry-based approaches without antibody enrichment typically detect proteins in the higher nanogram to low microgram range in complex samples, but when preceded by immunoprecipitation (IP-MS), sensitivity can approach that of Western blotting while providing amino acid sequence verification and post-translational modification identification. For absolute quantification, ELISA provides the most direct approach through standard curves but requires thoroughly validated antibodies. Western blotting offers semi-quantitative analysis with the advantage of visualizing protein integrity. Selected reaction monitoring mass spectrometry using isotope-labeled peptide standards provides absolute quantification with the highest specificity but often with lower sensitivity than antibody-based methods. The optimal method selection depends on whether the research question prioritizes absolute sensitivity, specificity confirmation, post-translational modification analysis, or absolute quantification.

How can At3g49630 antibodies be adapted for super-resolution microscopy techniques?

Adapting At3g49630 antibodies for super-resolution microscopy requires specialized optimization strategies to achieve nanoscale resolution. For structured illumination microscopy (SIM), use secondary antibodies conjugated with bright, photostable fluorophores like Alexa Fluor 488 or 568, and optimize fixation protocols to minimize autofluorescence while preserving antigenicity. For stimulated emission depletion microscopy (STED), select secondary antibodies with fluorophores specifically designed for STED (such as STAR RED or ATTO 647N) that exhibit appropriate depletion properties and photostability under high-intensity depletion lasers. For single-molecule localization microscopy (PALM/STORM), implement direct conjugation of At3g49630 primary antibodies with photoactivatable or photoswitchable fluorophores to minimize the displacement error introduced by secondary antibodies, which can add 10-20 nm of localization uncertainty. Use carefully optimized blocking protocols with 5% BSA containing 1% normal serum and 0.1% Triton X-100 to minimize non-specific binding that becomes more apparent at super-resolution scales. Validate super-resolution imaging results with complementary approaches such as proximity ligation assays to confirm protein co-localization patterns detected at nanoscale resolution. Finally, develop computational image analysis pipelines specifically designed to quantify nanoscale distribution patterns of At3g49630 protein relative to subcellular landmarks or other proteins of interest.

What are the emerging approaches for multiplexed detection of At3g49630 alongside other proteins in single cells?

Emerging approaches for multiplexed protein detection in plant cells are expanding our ability to study At3g49630 in its molecular context. Iterative indirect immunofluorescence imaging (4i) enables detection of At3g49630 alongside dozens of other proteins by using multiple rounds of antibody staining, imaging, and antibody elution on the same sample. This technique requires careful optimization of elution conditions that remove previous antibodies without affecting tissue integrity or antigenicity. Mass cytometry (CyTOF) adapted for plant tissues uses antibodies labeled with isotopically pure metals rather than fluorophores to detect At3g49630 alongside 40+ proteins simultaneously without spectral overlap concerns, though this requires specialized instrumentation. Co-detection by indexing (CODEX) employs DNA-barcoded antibodies against At3g49630 and other targets that can be iteratively visualized through cyclic rendering of complementary fluorescent oligonucleotides. Digital spatial profiling (DSP) combines immunofluorescence with spatially-resolved, antibody-linked oligonucleotide detection to quantify At3g49630 alongside hundreds of proteins within specific regions of interest. Single-cell proteomics using nanoPOTS (Nanodroplet Processing in One pot for Trace Samples) followed by liquid chromatography-mass spectrometry can quantify hundreds of proteins including At3g49630 from individual isolated plant cells without antibodies, providing complementary validation for antibody-based multiplexed detection approaches.

How can computational modeling improve the design and application of At3g49630 antibodies?

Computational modeling offers significant advantages for At3g49630 antibody development and application. Structure-based epitope prediction algorithms that incorporate protein secondary structure, surface accessibility, and hydrophilicity can identify optimal antigenic regions of At3g49630, improving antibody specificity and reducing development time compared to empirical approaches. Molecular dynamics simulations of antibody-antigen interactions can predict binding affinity and specificity by modeling the conformational flexibility of both At3g49630 and candidate antibodies across physiological conditions. Machine learning algorithms trained on existing antibody validation data can predict cross-reactivity risks with related plant proteins, allowing researchers to select epitopes that maximize specificity. Network analysis of protein interaction data can identify regions of At3g49630 that are likely to be accessible in native protein complexes versus regions involved in protein-protein interactions that may be inaccessible to antibodies. For immunohistochemistry applications, image analysis algorithms can be trained to automatically quantify staining patterns across tissues, increasing throughput and reducing subjective interpretation. Computational modeling of protein modifications can predict how post-translational modifications might affect epitope recognition, enabling development of modification-specific antibodies. Integration of these computational approaches with experimental validation creates an iterative optimization process that systematically improves antibody performance in specific research applications.

What approaches combine antibody detection with single-cell transcriptomics to correlate protein and mRNA levels of At3g49630?

Integrating antibody detection with single-cell transcriptomics creates powerful multi-omic approaches to study At3g49630 regulation. CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing), adapted for plant cells, uses oligonucleotide-tagged antibodies against At3g49630 alongside single-cell RNA sequencing to simultaneously quantify protein and mRNA from the same cells, enabling direct correlation analysis between transcription and translation. This technique requires successful development of antibodies that function in mild, non-denaturing conditions to preserve RNA integrity. Spatial transcriptomics platforms like Slide-seq or 10x Visium can be combined with sequential immunofluorescence to map At3g49630 protein distribution in the same tissue sections used for spatially-resolved transcriptomics, allowing spatial correlation of protein and mRNA patterns at near-cellular resolution. Single-cell Western blotting followed by RNA extraction from adjacent microdissected cells provides another approach, though with lower throughput. For microscopy-based approaches, in situ sequencing of At3g49630 mRNA can be performed alongside immunofluorescence detection of the protein on the same samples. Computational integration methods including canonical correlation analysis and non-negative matrix factorization can identify relationships between protein and mRNA measurements across different cells and conditions, revealing post-transcriptional regulatory mechanisms. These emerging techniques provide unprecedented insight into the relationship between At3g49630 transcription, translation, and protein stability at single-cell resolution.

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.