A3G2XYLT Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
A3G2XYLT antibody; UF3GT antibody; UFGT antibody; UGT79B1 antibody; At5g54060 antibody; MJP23.2 antibody; Anthocyanidin 3-O-glucoside 2'''-O-xylosyltransferase antibody; A3G2''XylT antibody; EC 2.4.2.51 antibody
Target Names
A3G2XYLT
Uniprot No.

Target Background

Function
This antibody contributes to the final stages of anthocyanin biosynthesis. It catalyzes the conversion of cyanidin 3-O-glucoside to cyanidin 3-O-xylosyl(1->2)glucoside. This enzyme can utilize 3-O-glucosylated anthocyanidins/flavonols and uridine diphosphate (UDP)-xylose as substrates.
Gene References Into Functions
  1. Cytokinins enhance anthocyanin content and the transcript levels of sugar inducible structural gene UDPglucose: flavonoid 3-O-glucosyl transferase and regulatory gene PRODUCTION OF ANTHOCYANIN PIGMENT 1. PMID: 22699753
  2. UGT79B1 exhibits substrate specificity for 3-O-glucosylated anthocyanidins/flavonols and uridine diphosphate (UDP)-xylose, but not for 3,5-O-diglucosylated anthocyanidins, indicating that UGT79B1 encodes anthocyanin 3-O-glucoside: 2''-O-xylosyltransferase. PMID: 21899608
Database Links

KEGG: ath:AT5G54060

STRING: 3702.AT5G54060.1

UniGene: At.49795

Protein Families
UDP-glycosyltransferase family

Q&A

What is A3G2XYLT and what cellular processes does it regulate?

A3G2XYLT (AT5G54060) is an enzyme involved in flavonoid and anthocyanin biosynthesis in plants, particularly Arabidopsis thaliana. It functions as a UDP-glucose:flavonoid 3-o-glucosyltransferase that can be induced by cytokinins. The enzyme plays a critical role in the glycosylation of flavonoids, a process essential for their proper accumulation, compartmentalization, and biological activity within plant cells. This transferase adds specific sugar moieties to flavonoid structures, altering their solubility, stability, and bioactivity. Understanding A3G2XYLT's function is crucial for research in plant secondary metabolism, stress responses, and developmental processes where flavonoids and anthocyanins serve important roles .

What are the recommended applications for A3G2XYLT antibodies in plant research?

A3G2XYLT antibodies serve multiple applications in plant research, particularly for investigating flavonoid biosynthesis pathways. These antibodies can be used for immunohistochemistry to localize A3G2XYLT in plant tissues, Western blotting to quantify expression levels, immunoprecipitation to study protein-protein interactions, and chromatin immunoprecipitation (ChIP) if studying transcriptional regulation of the A3G2XYLT gene. They are particularly valuable for examining how environmental stresses or hormone treatments affect flavonoid metabolism via changes in A3G2XYLT expression or activity. When selecting an application, researchers should consider the preservation of antigen epitopes and potential cross-reactivity with other glycosyltransferases that may share structural similarities .

How does A3G2XYLT expression vary across different plant tissues and developmental stages?

A3G2XYLT expression demonstrates tissue-specific and developmental stage-dependent patterns in plants. Expression is typically higher in tissues where anthocyanin biosynthesis is active, such as in developing flowers, fruits during ripening, and leaves under stress conditions. Research data indicates upregulation (10.912-fold) in response to cytokinin treatment, suggesting hormonal control of its expression. Developmental regulation appears to correlate with flavonoid accumulation patterns during plant growth. When designing experiments to study A3G2XYLT, researchers should consider these expression variations and select appropriate tissue types and developmental stages that align with their research questions. Comparing expression levels across multiple tissues provides valuable context for interpreting antibody-based detection results .

What controls should be included when using A3G2XYLT antibodies in Western blotting experiments?

For rigorous Western blotting with A3G2XYLT antibodies, multiple controls are essential. Positive controls should include plant tissue samples known to express A3G2XYLT at high levels, such as cytokinin-treated seedlings where expression has been documented to increase 10.912-fold. Negative controls should include either A3G2XYLT knockout mutant tissue (if available) or tissues where expression is minimal. To validate antibody specificity, pre-absorption controls where the antibody is pre-incubated with purified A3G2XYLT protein prior to immunoblotting should be performed. Loading controls using antibodies against constitutively expressed proteins like actin or tubulin are crucial for normalization. Additionally, when studying regulatory mechanisms, include samples from plants under various treatment conditions (e.g., different hormone treatments, stress conditions) to observe expression dynamics .

How should samples be prepared to preserve A3G2XYLT epitopes for immunodetection?

Optimal sample preparation for A3G2XYLT immunodetection requires careful consideration of protein extraction and preservation methods. For plant tissues, use a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, and protease inhibitor cocktail. When extracting from Arabidopsis, crushing tissue in liquid nitrogen before adding buffer improves yield while preserving protein integrity. For immunohistochemistry, aldehyde-based fixatives (4% paraformaldehyde) better preserve protein epitopes than alcohol-based fixatives. Since A3G2XYLT is involved in flavonoid metabolism pathways, consider adding antioxidants like 1 mM DTT and 1 mM PMSF to prevent oxidation of phenolic compounds that could interfere with antibody binding. Store samples at -80°C and avoid repeated freeze-thaw cycles that can degrade epitopes .

What are the recommended parameters for immunoprecipitation of A3G2XYLT from plant extracts?

For successful immunoprecipitation of A3G2XYLT from plant extracts, begin with 500-1000 μg of total protein extracted in a non-denaturing buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 0.5% NP-40, 1 mM EDTA, protease inhibitor cocktail). Pre-clear the extract using 50 μl of Protein A/G beads for 1 hour at 4°C to reduce non-specific binding. Incubate the pre-cleared extract with 2-5 μg of A3G2XYLT antibody overnight at 4°C with gentle rotation. For the immunoprecipitation itself, add 50 μl of fresh Protein A/G beads and incubate for 3-4 hours at 4°C. Wash the beads 4-5 times with wash buffer (same as extraction buffer but with 0.1% NP-40) to reduce background. When studying potential protein-protein interactions, consider using crosslinking agents like DSP (dithiobis(succinimidyl propionate)) at 2 mM for 30 minutes before extraction to stabilize transient interactions involving A3G2XYLT in flavonoid biosynthesis complexes .

How can A3G2XYLT antibodies be used to study protein-protein interactions in flavonoid biosynthesis pathways?

A3G2XYLT antibodies offer powerful tools for elucidating protein interaction networks in flavonoid biosynthesis. Co-immunoprecipitation (Co-IP) using A3G2XYLT antibodies followed by mass spectrometry analysis can identify proteins that physically interact with A3G2XYLT in vivo. For studying dynamic interactions, proximity-dependent biotin identification (BioID) can be employed by fusing a promiscuous biotin ligase to A3G2XYLT, followed by streptavidin pulldown and mass spectrometry. Bimolecular fluorescence complementation (BiFC) provides spatial information about interactions by fusing A3G2XYLT and potential partners to complementary fragments of fluorescent proteins. For confirming direct interactions, in vitro pull-down assays using recombinant A3G2XYLT and candidate interactors can be performed. When analyzing results, researchers should consider that interactions may be transient or dependent on specific cellular conditions such as cytokinin-induced state changes, as A3G2XYLT has been shown to be upregulated (10.912-fold) in response to cytokinin treatment .

What approaches can be used to investigate post-translational modifications of A3G2XYLT?

Investigating post-translational modifications (PTMs) of A3G2XYLT requires multi-faceted approaches. Immunoprecipitate A3G2XYLT using specific antibodies followed by Western blotting with antibodies against common PTMs like phosphorylation (anti-phosphoserine, anti-phosphothreonine), ubiquitination (anti-ubiquitin), and glycosylation (lectins or glycosylation-specific antibodies). For comprehensive PTM profiling, perform immunoprecipitation followed by mass spectrometry analysis, which can identify multiple PTM types and their exact locations. To study the functional relevance of identified PTMs, site-directed mutagenesis can be performed to create PTM-mimicking or PTM-preventing variants. Protein stability can be assessed through cycloheximide chase assays comparing wild-type A3G2XYLT with PTM site mutants. When analyzing results, consider that PTMs may change in response to cytokinin treatment, as A3G2XYLT expression is cytokinin-inducible with a documented 10.912-fold upregulation under such conditions .

How can ChIP-seq with transcription factor antibodies be used to study regulation of A3G2XYLT gene expression?

Chromatin immunoprecipitation sequencing (ChIP-seq) with transcription factor antibodies can elucidate the regulatory mechanisms controlling A3G2XYLT gene expression. First, identify candidate transcription factors by analyzing the A3G2XYLT promoter for known binding motifs or through literature searches for factors involved in flavonoid pathway regulation or cytokinin response. Perform ChIP-seq using antibodies against these candidate transcription factors in both normal and cytokinin-treated plant tissues, as A3G2XYLT shows a 10.912-fold upregulation in response to cytokinins. For optimal results, crosslink plant tissue with 1% formaldehyde for 10 minutes, followed by chromatin isolation and sonication to generate 200-300bp fragments. Immunoprecipitate with 5μg of transcription factor antibody per sample. After sequencing, analyze binding peaks near the A3G2XYLT locus using MACS2 peak calling algorithm. Validate findings with techniques like electrophoretic mobility shift assays (EMSAs) and reporter gene assays. This approach allows identification of direct regulators of A3G2XYLT and construction of gene regulatory networks controlling flavonoid metabolism .

What strategies can address cross-reactivity issues with A3G2XYLT antibodies?

Cross-reactivity can be a significant challenge when working with A3G2XYLT antibodies due to structural similarities with other glycosyltransferases in plant tissues. To address this, employ a multi-tiered approach beginning with epitope analysis to select antibodies raised against unique regions of A3G2XYLT that have minimal sequence homology with related proteins. Pre-absorb antibodies with recombinant proteins of related glycosyltransferases to reduce non-specific binding. Validate antibody specificity using tissues from A3G2XYLT knockout or knockdown plants as negative controls. For Western blotting applications, perform peptide competition assays by pre-incubating the antibody with excess of the immunizing peptide before probing membranes. When cross-reactivity cannot be eliminated, consider using orthogonal detection methods like RNA-based techniques (qRT-PCR, RNA-seq) or activity assays specific to A3G2XYLT's enzymatic function to complement antibody-based detection. Always include appropriate controls in experiments to account for potential cross-reactivity .

How can researchers optimize immunohistochemistry protocols for localization of A3G2XYLT in plant tissues?

Optimizing immunohistochemistry for A3G2XYLT localization in plant tissues requires careful protocol adaptation. Begin with fixation optimization: test both 4% paraformaldehyde (4-12 hours at 4°C) and ethanol:acetic acid mixtures (3:1, 24 hours) to determine which better preserves A3G2XYLT epitopes while maintaining tissue morphology. For antigen retrieval, compare heat-induced (citrate buffer, pH 6.0, 95°C for 10 minutes) and enzymatic methods (proteinase K, 20 μg/ml for 15 minutes) to establish which works better for A3G2XYLT detection. Block sections with 5% BSA and 0.3% Triton X-100 in PBS for 2 hours to reduce background. Test antibody dilutions ranging from 1:100 to 1:1000 to identify optimal signal-to-noise ratio. Include controls with secondary antibody only and with competing peptide to validate specificity. When analyzing results, correlate A3G2XYLT localization with known sites of flavonoid accumulation and consider tissue-specific variations in subcellular localization patterns that may reflect functional compartmentalization of flavonoid biosynthesis .

What are the recommended approaches for quantifying A3G2XYLT protein levels in different experimental conditions?

For precise quantification of A3G2XYLT protein levels across experimental conditions, a multi-method approach yields the most reliable results. Western blotting with A3G2XYLT-specific antibodies provides relative quantification when properly normalized to loading controls like actin or GAPDH. Use a standard curve of recombinant A3G2XYLT protein (5-100 ng range) to enable absolute quantification. For higher throughput, develop an ELISA using sandwich format with two different A3G2XYLT antibodies recognizing distinct epitopes. Complement protein-level measurements with qRT-PCR to track transcript levels, particularly important for cytokinin response studies where A3G2XYLT shows 10.912-fold upregulation. For single-cell resolution, consider immunofluorescence quantification using confocal microscopy with constant laser power and detector settings across samples. Process images with software like ImageJ for fluorescence intensity quantification. When analyzing data, account for tissue-specific expression patterns and normalize appropriately to avoid misinterpreting differences that reflect tissue composition rather than treatment effects .

What steps should be taken when A3G2XYLT antibodies show weak or no signal in Western blots?

When encountering weak or absent signals in Western blots using A3G2XYLT antibodies, a systematic troubleshooting approach is necessary. First, verify protein extraction efficacy by staining the membrane with Ponceau S to confirm successful transfer. If protein is present, optimize extraction conditions using different buffers; for A3G2XYLT, try RIPA buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS) with added protease inhibitors. Increase protein loading to 50-100 μg per lane, as A3G2XYLT may be expressed at low levels in some tissues unless induced by cytokinins. Test different membrane types (PVDF tends to work better than nitrocellulose for some antibodies). Adjust antibody concentration by testing dilutions from 1:500 to 1:2000. Extend primary antibody incubation to overnight at 4°C and increase secondary antibody incubation to 2 hours at room temperature. Consider using enhanced chemiluminescence detection systems with longer exposure times (up to 30 minutes). Finally, verify the tissue source is appropriate, prioritizing tissues with known high expression like cytokinin-treated samples where A3G2XYLT shows 10.912-fold upregulation .

How can researchers address inconsistent results in immunoprecipitation experiments with A3G2XYLT antibodies?

Inconsistent immunoprecipitation results with A3G2XYLT antibodies often stem from multiple factors that can be systematically addressed. Begin by assessing antibody quality through a pilot Western blot to confirm it recognizes A3G2XYLT in your lysate. Test different antibody amounts (2-10 μg per reaction) to determine optimal concentration. Since A3G2XYLT functions in flavonoid metabolism, plant tissue extracts may contain phenolic compounds that interfere with antibody binding; add polyvinylpolypyrrolidone (PVPP, 2% w/v) to extraction buffer to absorb these compounds. Compare protein A, protein G, and mixed A/G beads to identify which has highest affinity for your antibody isotype. Modify wash stringency by adjusting salt concentration (150-500 mM NaCl) and detergent levels (0.1-1% NP-40) to balance between maintaining specific interactions and reducing background. For proteins with weak or transient interactions, consider adding chemical crosslinkers like DSP (1-2 mM) before cell lysis. Document all variables between experiments, including plant growth conditions, as A3G2XYLT expression is highly responsive to environmental factors and shows 10.912-fold upregulation with cytokinin treatment .

What approaches can resolve high background issues in immunofluorescence staining using A3G2XYLT antibodies?

High background in immunofluorescence with A3G2XYLT antibodies can be systematically reduced through multiple optimizations. Start by improving fixation—test 4% paraformaldehyde for shorter periods (15-30 minutes) to prevent over-fixation while maintaining epitope accessibility. For plant tissues, which may contain autofluorescent compounds, photobleach samples with high-intensity light before immunostaining or use specific reagents like 0.1% Sudan Black B in 70% ethanol for 10 minutes to quench autofluorescence. Optimize blocking by extending duration to 2 hours and testing different blocking agents (5% normal serum from the same species as the secondary antibody, 3% BSA, or commercial blocking solutions). Implement more stringent washing steps using 0.1% Tween-20 in PBS with at least 5 washes of 10 minutes each. Dilute primary antibody further (test range from 1:200 to 1:2000) and reduce secondary antibody concentration (optimal range often 1:500 to 1:2000). Centrifuge antibody solutions before use (15,000 × g for 10 minutes) to remove aggregates that cause punctate background. When analyzing flavonoid-rich tissues, where A3G2XYLT is likely to be highly expressed (showing 10.912-fold upregulation with cytokinin treatment), consider using confocal microscopy with spectral unmixing to distinguish between specific signal and autofluorescence .

How should researchers interpret differential expression of A3G2XYLT across experimental conditions?

Interpreting differential expression of A3G2XYLT requires contextual analysis within flavonoid biosynthesis pathways and plant physiological states. Begin by normalizing A3G2XYLT expression to multiple reference genes (at least three) that show stability under your experimental conditions. Compare expression patterns with other flavonoid pathway enzymes to determine if changes are pathway-wide or A3G2XYLT-specific. Consider natural variation in expression across different tissues and developmental stages when interpreting results. The documented 10.912-fold upregulation in response to cytokinins provides a benchmark for evaluating induction magnitude in your experimental conditions. For stress response experiments, correlate A3G2XYLT expression changes with anthocyanin accumulation levels, as these often correspond in stress-induced flavonoid production. When comparing treatments, perform appropriate statistical analyses (ANOVA with post-hoc tests for multiple comparisons) and report both statistical significance and effect sizes. Finally, validate protein-level changes with orthogonal methods (Western blotting, enzyme activity assays) to confirm that transcriptional changes translate to functional alterations in the flavonoid biosynthesis pathway .

What statistical approaches are recommended for analyzing A3G2XYLT antibody-based experimental data?

For robust statistical analysis of A3G2XYLT antibody-based experimental data, implement a multi-tiered approach tailored to the specific experimental design. For Western blot densitometry data, use linear mixed-effects models that account for both technical replicates (multiple blots) and biological replicates (independent samples). Transform data if necessary to meet normality assumptions (log transformation often works well for protein expression data). For comparing multiple treatment groups, use one-way ANOVA followed by Tukey's HSD or Dunnett's test when comparing to a control. For time-course experiments, consider repeated measures ANOVA or growth curve analysis. Sample size calculations should aim for at least 80% power to detect biologically meaningful effect sizes; for A3G2XYLT studies, the known 10.912-fold change with cytokinin treatment can serve as a reference for substantial biological effects. Implement hierarchical clustering or principal component analysis to identify patterns across multiple experimental conditions. When integrating A3G2XYLT protein data with other measurements (transcript levels, enzyme activity, metabolite concentrations), use correlation analyses and path modeling to establish relationships between variables. Report effect sizes and confidence intervals alongside p-values to provide a complete statistical picture .

How can researchers integrate A3G2XYLT antibody data with transcriptomic and metabolomic datasets?

Integrating A3G2XYLT antibody data with transcriptomic and metabolomic datasets creates a comprehensive understanding of flavonoid biosynthesis regulation. Begin by normalizing datasets to allow direct comparisons—standardize protein quantification data from antibody experiments, normalize transcriptomic data using established methods like DESeq2 or EdgeR, and scale metabolomic data appropriately. Align time points or treatment conditions precisely across datasets. Calculate Pearson or Spearman correlation coefficients between A3G2XYLT protein levels, transcript abundance, and relevant metabolite concentrations (flavonoid glycosides). Implement pathway enrichment analysis to contextualize A3G2XYLT within broader metabolic networks. Create integrated visualizations using heat maps, network graphs, or pathway maps that overlay multiple data types. For causality assessment, perform time-resolved experiments and analyze with methods like Granger causality or dynamic Bayesian networks. When possible, validate key relationships through perturbation experiments, such as examining metabolite profiles in A3G2XYLT knockdown plants. Consider the 10.912-fold upregulation of A3G2XYLT with cytokinin treatment as a benchmark for comparing expression dynamics with related genes and metabolites. Present integrated data in multidimensional visualizations that capture the complexity of these relationships while highlighting patterns relevant to your research question .

What emerging technologies could enhance A3G2XYLT antibody-based research?

Several cutting-edge technologies promise to revolutionize A3G2XYLT antibody-based research in plant biology. Proximity-dependent labeling techniques like TurboID or APEX2 can be combined with A3G2XYLT antibodies to map dynamic protein interaction networks in vivo with temporal resolution. Single-cell proteomics using methods like mass cytometry (CyTOF) adapted for plant cells could reveal cell-type-specific variations in A3G2XYLT expression within complex tissues. Super-resolution microscopy techniques (STORM, PALM) paired with fluorescently-labeled A3G2XYLT antibodies can visualize subcellular localization with unprecedented detail, potentially revealing microdomains of flavonoid biosynthesis within cellular compartments. CRISPR-based technologies like CUT&Tag could provide more precise mapping of transcription factor binding sites in the A3G2XYLT promoter region than traditional ChIP approaches. Biosensors developed from A3G2XYLT antibody fragments could enable real-time monitoring of protein dynamics in response to environmental stimuli. The integration of these technologies with computational modeling approaches like agent-based models could lead to predictive frameworks for understanding how A3G2XYLT regulation (including the 10.912-fold upregulation with cytokinin) affects flavonoid metabolism under various environmental conditions .

What are the potential applications of A3G2XYLT antibodies in studying plant responses to environmental stresses?

A3G2XYLT antibodies offer powerful tools for investigating plant stress responses through flavonoid metabolism. In drought stress studies, these antibodies can track spatiotemporal changes in A3G2XYLT expression across different tissues, correlating protein levels with flavonoid glycoside accumulation and water retention. For UV stress research, immunohistochemistry with A3G2XYLT antibodies can reveal tissue-specific induction patterns in epidermal versus mesophyll cells, providing insights into localized protection mechanisms. In temperature stress scenarios, combining A3G2XYLT protein quantification with membrane fluidity assays can elucidate how flavonoid glycosylation contributes to membrane stabilization. For pathogen response studies, A3G2XYLT antibodies can track protein dynamics during defense responses, potentially revealing how flavonoid glycosylation patterns change during immune activation. Multi-stress experiments comparing A3G2XYLT expression under various stressors can identify common regulatory pathways, especially in relation to phytohormone signaling networks since A3G2XYLT shows 10.912-fold upregulation with cytokinin treatment. Time-course immunoprecipitation experiments following stress application can capture the dynamics of A3G2XYLT interaction partners, potentially revealing stress-specific regulation of enzyme complex formation .

How might A3G2XYLT antibodies contribute to understanding evolutionary conservation of flavonoid metabolism across plant species?

A3G2XYLT antibodies offer valuable tools for comparative evolutionary studies of flavonoid metabolism across diverse plant lineages. By testing cross-reactivity of A3G2XYLT antibodies across species, researchers can assess structural conservation of this enzyme from bryophytes to angiosperms. Immunoprecipitation followed by mass spectrometry can identify species-specific interaction partners, revealing evolutionary divergence in enzyme complex formation. Western blot analysis of A3G2XYLT in different plant lineages under standardized conditions can quantify expression level differences that may reflect adaptive specialization. Immunohistochemistry across species can reveal shifts in tissue-specific localization patterns that correlate with ecological adaptations. For deeper evolutionary insights, detailed epitope mapping of antibody binding sites across orthologs can pinpoint conserved functional domains. Researchers should design sampling strategies that include representatives from major plant lineages and species adapted to diverse ecological niches. When analyzing data, consider that the 10.912-fold cytokinin responsiveness observed in Arabidopsis may vary in magnitude across species, potentially reflecting differences in hormonal regulation of secondary metabolism. These approaches collectively can reconstruct the evolutionary history of flavonoid glycosylation mechanisms and their functional diversification across plants .

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.