AGL61 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
AGL61 antibody; DIA antibody; At2g24840 antibody; F27C12.24 antibody; Agamous-like MADS-box protein AGL61 antibody; Protein DIANA antibody
Target Names
AGL61
Uniprot No.

Target Background

Function
AGL61, also known as DIANA, is a probable transcription factor that plays a crucial role in regulating central cell differentiation during female gametophyte development in plants.
Gene References Into Functions
  • Regulates the expression of downstream genes essential for central cell development. PMID: 18599653
  • AGL61 (DIANA) and AGL80 function synergistically to differentiate the central cell in Arabidopsis. PMID: 18713950
  • DIANA (AGL61) forms a protein complex with AGL80 to specify central cell fate in Arabidopsis ovules. PMID: 18713950
Database Links

KEGG: ath:AT2G24840

STRING: 3702.AT2G24840.1

UniGene: At.52900

Subcellular Location
Nucleus. Note=The transport to the nucleus is dependent on dimerization with AGL80.
Tissue Specificity
Expressed exclusively in the central cell of the female gametophyte and in early endosperm.

Q&A

What is AGL and why are antibodies against it important in research?

AGL (amylo-alpha-1, 6-glucosidase, 4-alpha-glucanotransferase) is also known as glycogen debranching enzyme (GDE). This protein plays a crucial role in glycogen metabolism by facilitating the breakdown of glycogen through its dual catalytic activities. AGL antibodies are essential research tools for studying glycogen storage diseases, metabolic disorders, and normal glycogen metabolism pathways. The protein has a molecular weight of approximately 174.8 kilodaltons and is encoded by the AGL gene in humans. Antibodies targeting this protein enable researchers to investigate its expression patterns, subcellular localization, and functional characteristics in various experimental models .

How do I determine the most appropriate AGL antibody for my specific research application?

Selecting the optimal AGL antibody requires consideration of multiple experimental parameters. First, determine your intended application (Western blot, immunofluorescence, immunohistochemistry, etc.) as antibodies vary in their performance across different techniques. Next, consider species reactivity - many AGL antibodies are specifically designed for human samples, while others may cross-react with mouse, rat, or other species. Review the literature for validated antibodies in your specific application and tissue/cell type. Examine documentation for the epitope region recognized by the antibody, as this may affect detection of specific isoforms or processed forms of AGL. Finally, consider validation data including positive/negative controls and citation records to ensure reliability. For particularly demanding applications, testing multiple antibodies in parallel may be necessary to identify the most suitable reagent .

What are the most common applications for AGL antibodies in research?

AGL antibodies demonstrate utility across multiple experimental applications. The most frequently employed techniques include Western blot (WB) for protein expression quantification, immunofluorescence (IF) for subcellular localization studies, immunohistochemistry (IHC) for tissue distribution analysis, and enzyme-linked immunosorbent assay (ELISA) for protein quantification in solution. Flow cytometry (FCM) applications are also reported for certain AGL antibody preparations. Each application requires specific antibody characteristics - for example, Western blot applications typically benefit from antibodies recognizing denatured epitopes, while immunofluorescence applications require antibodies that recognize native protein conformations. The majority of commercially available AGL antibodies are validated for Western blot and immunofluorescence applications, with many also suitable for immunohistochemistry on paraffin-embedded (IHC-p) sections .

How should I validate the specificity of an AGL antibody for my experimental system?

Comprehensive validation of AGL antibody specificity should include multiple complementary approaches. Begin with positive controls using tissues or cells known to express AGL (such as liver samples) and negative controls where AGL expression is absent or minimal. For definitive validation, employ genetic knockdown (siRNA, shRNA) or knockout (CRISPR-Cas9) approaches to create AGL-depleted samples - a specific antibody will show reduced or absent signal in these samples. Additional validation steps include pre-absorption controls (pre-incubating the antibody with purified AGL protein before immunostaining), peptide competition assays, and Western blot analysis to confirm binding to a protein of the expected molecular weight (approximately 174.8 kDa for full-length AGL). For monoclonal antibodies, verifying reactivity against the immunizing peptide provides further confirmation of specificity. Document all validation experiments thoroughly, as these data will support the reliability of subsequent experimental findings .

What factors affect the stability and performance of AGL antibodies in storage?

Maintaining optimal antibody performance requires careful attention to storage conditions. AGL antibodies, like most protein reagents, are susceptible to degradation through multiple mechanisms. Temperature fluctuations can promote denaturation and aggregation - most antibodies should be stored at -20°C for long-term stability, with working aliquots maintained at 4°C to minimize freeze-thaw cycles. Bacterial contamination presents another threat to antibody integrity, so aseptic technique during handling is essential. Chemical degradation can occur through oxidation or hydrolysis, which may be mitigated by storage buffers containing appropriate stabilizers. For unconjugated antibodies, typical storage buffers contain PBS with glycerol and preservatives like sodium azide. Document lot numbers, receipt dates, and any unusual observations during experiments, as performance can vary between manufacturing lots. Finally, prepare small working aliquots to minimize freeze-thaw cycles, which can progressively reduce antibody activity through protein denaturation and aggregation .

How can I optimize AGL antibody performance for challenging samples with low expression levels?

Detecting low-abundance AGL expression requires systematic optimization of signal amplification and background reduction strategies. Begin by implementing antigen retrieval protocols - for tissue sections, test both heat-induced epitope retrieval (citrate or EDTA buffers at varying pH) and enzymatic retrieval methods to identify optimal conditions. For immunofluorescence applications, employ tyramide signal amplification (TSA) systems, which can enhance signal detection by 10-100 fold compared to conventional detection methods. Consider sample enrichment strategies such as immunoprecipitation before Western blot analysis to concentrate the target protein. Optimize primary antibody concentration using a systematic titration approach, testing concentrations ranging from 1:100 to 1:5000 to identify the optimal signal-to-noise ratio. Extend primary antibody incubation times (overnight at 4°C) to improve antigen binding. For Western blot applications, increase protein loading and utilize high-sensitivity chemiluminescent substrates. Background reduction strategies include thorough blocking steps (BSA, normal serum, or commercial blocking reagents), extensive washing protocols, and pre-adsorption of secondary antibodies against tissue samples to remove cross-reactive antibodies .

What approaches can resolve contradictory results when different AGL antibodies yield inconsistent data?

Resolving discrepancies between different AGL antibodies requires systematic investigation of multiple variables. First, characterize the epitope specificity of each antibody - antibodies recognizing different domains of AGL may yield divergent results if protein processing, post-translational modifications, or isoform expression varies across experimental conditions. Second, employ orthogonal detection methods such as mass spectrometry to provide antibody-independent verification of protein presence and abundance. Third, evaluate the influence of sample preparation techniques - fixation methods, antigen retrieval protocols, and protein extraction buffers can differentially affect epitope accessibility. Fourth, compare antibody performance across multiple experimental systems, including cell lines with confirmed AGL expression profiles and knockout/knockdown models. Finally, consider that true biological variability may explain apparently contradictory results - AGL expression and localization may genuinely differ across developmental stages, cell cycle phases, or in response to specific stimuli. Thorough documentation of experimental conditions, including buffer compositions, incubation times/temperatures, and image acquisition parameters, facilitates meaningful comparison between experiments .

How can computational and AI methods enhance AGL antibody-based research?

The integration of artificial intelligence and computational approaches offers significant advantages for antibody-based research. Structure prediction algorithms such as ABodyBuilder2, part of the ImmuneBuilder suite, can predict antibody structures with high accuracy, achieving root mean square deviation (RMSD) values of 2.81 Å, particularly valuable for understanding antigen-antibody interactions. Inverse folding models like AntiFold, AbMPNN, and IgDesign allow researchers to optimize antibody sequences for specific structural properties, facilitating the development of custom antibodies with improved target specificity. For developability assessment, machine learning models like SOLart can predict antibody solubility with a Pearson correlation of 0.65, while PfAbNet-viscosity predicts solution viscosity - critical parameters for antibody performance in experimental applications. Language model-based approaches, including NetSolP (an ensemble of fine-tuned ESM1b models), enable prediction of antibody solubility from sequence data alone. These computational tools can guide experimental design by identifying optimal epitopes for antibody generation, predicting cross-reactivity with related proteins, and assessing potential performance limitations before experimental implementation .

What strategies can minimize off-target effects when using AGL antibodies in multiplex imaging studies?

Multiplex imaging with AGL antibodies presents challenges that require careful experimental design. Implement sequential staining protocols that include complete elution of primary-secondary antibody complexes between detection cycles, verified through no-primary controls for each round. Utilize antibodies raised in different host species to enable species-specific secondary antibody detection without cross-reactivity. When this is not possible, employ direct conjugation of fluorophores to primary antibodies, eliminating the need for species-specific secondary detection. Alternative approaches include tyramide signal amplification methods that permanently deposit fluorophores, allowing antibody stripping without signal loss. Carefully evaluate spectral overlap between fluorophores using single-stained controls and implement appropriate compensation algorithms during image analysis. Advanced spectral unmixing algorithms can further distinguish overlapping signals. For panel development, begin with pairwise testing of antibodies to identify potential interactions before progressing to higher-level multiplexing. Throughout the optimization process, include appropriate controls: isotype controls to assess non-specific binding, biological negative controls (tissues known not to express AGL), and technical controls (secondary-only staining) .

What are the most effective epitope retrieval methods for AGL detection in formalin-fixed tissues?

Optimizing epitope retrieval for AGL detection requires systematic evaluation of heat-induced epitope retrieval (HIER) and enzymatic digestion methods. For HIER approaches, test multiple buffer systems including citrate buffer (pH 6.0), Tris-EDTA (pH 9.0), and Tris-HCl (pH 10.0) at varying incubation times (10-30 minutes). Pressure-based HIER methods (pressure cooker or pressure retrieval systems) often yield superior results compared to microwave or water bath heating. For enzymatic retrieval, evaluate proteinase K, trypsin, and pepsin at different concentrations and incubation times. The optimal retrieval method depends on both the specific epitope recognized by the antibody and the fixation conditions of the tissue. For highly challenging samples, consider dual retrieval approaches that combine HIER followed by mild enzymatic digestion. After retrieval, allow sections to cool gradually to room temperature within the retrieval solution to prevent tissue detachment and artifacts. Include positive control tissues with known AGL expression in all optimization experiments. Document successful protocols comprehensively, including equipment settings, precise buffer compositions, and incubation times, as these parameters significantly impact reproducibility .

How can I determine whether post-translational modifications affect AGL antibody recognition?

Post-translational modifications (PTMs) can significantly impact antibody epitope recognition, requiring specific investigative approaches. First, examine the antibody documentation to determine whether the immunizing antigen contained or lacked specific PTMs. Next, employ strategies to experimentally assess modification effects: compare antibody reactivity in samples treated with enzymes that remove specific modifications (phosphatases, glycosidases, deubiquitinases) to untreated samples - decreased recognition after modification removal indicates PTM-dependent epitope recognition. Alternatively, enhance specific modifications through treatment with phosphatase inhibitors, proteasome inhibitors, or glycosylation modulators and assess changes in antibody reactivity. For definitive analysis, perform immunoprecipitation followed by mass spectrometry to characterize modification status at the antibody binding site. Computational analysis using algorithms like NetPhos, NetOGlyc, or UbPred can predict potential modification sites that might interfere with antibody binding. Consider also acquiring antibodies specifically designed to recognize modified versions of AGL if particular PTMs are research targets. Document all modification-specific behaviors in your experimental system to ensure proper interpretation of results .

What strategies improve reproducibility when using AGL antibodies across different experimental batches?

Achieving consistent results across experimental batches requires implementation of standardized protocols and quality control measures. First, maintain detailed inventory records of antibody lots, documenting performance characteristics for each lot received. When possible, reserve sufficient quantities of well-performing lots for critical experiments. Implement standardized sample preparation workflows, including consistent fixation times for tissues, identical lysis buffers for protein extraction, and standardized protein quantification methods. Prepare master mixes of common reagents to minimize pipetting variations. Include calibration controls in each experiment - samples with known AGL expression levels that serve as reference points for normalization across batches. For quantitative applications, develop standard curves using recombinant AGL protein. Implement image acquisition standardization, including consistent exposure settings, objective magnifications, and microscope configurations. During analysis, perform batch normalization using reference standards. For long-term projects, consider bulk preparation and aliquoting of key reagents (buffers, blocking solutions) to minimize compositional variations. Finally, implement regular equipment calibration schedules for critical instruments like plate readers and microscopes to ensure consistent performance .

How can glycan recognition patterns influence AGL antibody binding specificity?

Glycan recognition represents a critical but often overlooked factor in antibody specificity. As demonstrated in the case of HIV-1 neutralizing antibodies, glycan recognition can be a primary binding mechanism rather than a hindrance to protein epitope access. For AGL antibodies, the potential for glycan recognition is significant given that glycosylation sites (particularly N-linked glycans) may be present within or adjacent to antibody epitopes. Antibodies raised against native AGL protein may develop specificity for both protein sequences and their associated glycan structures through somatic hypermutation processes. This dual recognition can enhance binding specificity but also introduces variability when the target protein's glycosylation pattern changes across different cell types or physiological conditions. To investigate glycan contributions to binding, compare antibody reactivity against enzymatically deglycosylated AGL (using PNGase F for N-linked or O-glycosidase for O-linked glycans) versus native protein. Significant changes in binding affinity following deglycosylation suggest glycan involvement in epitope recognition. Mass spectrometry glycoproteomic analysis can identify specific glycan structures present at different sites, enabling correlation with antibody binding characteristics .

How can structural biology approaches enhance the development of high-specificity AGL antibodies?

Integrating structural biology approaches with antibody engineering enables rational design of AGL antibodies with enhanced specificity and performance characteristics. X-ray crystallography and cryo-electron microscopy of antibody-antigen complexes provide atomic-resolution insights into binding interfaces, identifying critical residues for interaction. This information guides site-directed mutagenesis to enhance binding affinity or reduce cross-reactivity with structurally similar proteins. Computational approaches like molecular dynamics simulations can further evaluate binding stability and predict the effects of sequence modifications. AI-driven structural prediction tools such as ABodyBuilder2 achieve remarkable accuracy in predicting antibody structures, with root mean square deviation values of 2.81 Å, particularly valuable for complementarity-determining regions that determine antigen specificity. Phage display technologies coupled with structural data enable directed evolution of antibodies with optimized binding characteristics. Epitope mapping through hydrogen-deuterium exchange mass spectrometry or peptide arrays identifies specific binding regions, allowing selection of antibodies targeting distinct epitopes for multiplexed detection applications. For challenging epitopes, structure-based computational design tools can generate antibody variants with complementary binding surfaces to specific AGL regions .

What advanced validation approaches can definitively confirm AGL antibody specificity in complex experimental systems?

Comprehensive validation of AGL antibodies in complex systems requires integration of multiple orthogonal approaches. Genetic validation using CRISPR-Cas9 knockout models provides the most definitive specificity assessment - persistent antibody signal in knockout samples indicates non-specific binding. For human tissues where genetic manipulation is not feasible, implement RNAscope or similar in situ hybridization methods to correlate mRNA expression with antibody staining patterns at single-cell resolution. Mass spectrometry-based validation offers antibody-independent confirmation of protein identity; techniques like selective reaction monitoring can quantify AGL with high specificity. Super-resolution microscopy techniques (STORM, PALM) can reveal subcellular localization patterns consistent with known AGL biology, providing another layer of validation. Validation across multiple antibodies targeting different AGL epitopes should yield consistent results in unmanipulated samples. For challenging applications, proximity ligation assays between multiple AGL antibodies can confirm that signals originate from the same protein molecule rather than non-specific binding. Finally, comprehensive literature review to compare observed patterns with previously reported distributions provides contextual validation. Document all validation approaches in publications to establish confidence in specificity claims .

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.