AGL31 Antibody

Shipped with Ice Packs
In Stock

Description

Antigen Overview: IL-31

Interleukin-31 (IL-31) is a 24 kDa cytokine belonging to the α-helical cytokine family. It is primarily secreted by activated Th2 cells and binds to a heterodimeric receptor (IL-31RA/OSMRβ), activating JAK/STAT, PI3K/AKT, and MAPK pathways . IL-31 is implicated in pruritus (itching) and inflammatory skin conditions like atopic dermatitis .

Key Features of IL-31:

PropertyDetail
GeneIL-31 (Accession # Q6EBC2)
Precursor164 amino acids (23 aa signal peptide, 141 aa mature protein)
ReceptorIL-31RA (GP130 family) + OSMRβ
FunctionsPruritus induction, Th2-mediated inflammation, keratinocyte signaling

Subcellular Localization

  • IL-31 staining localizes to cell surfaces and cytoplasm in stimulated PBMCs .

  • Colocalizes with DAPI (nuclear counterstain), confirming cytoplasmic expression .

Functional Insights

  • IL-31 overexpression in transgenic mice correlates with non-atopic dermatitis and pruritus .

  • Therapeutic targeting of IL-31/IL-31RA is under investigation for inflammatory skin diseases .

Clinical and Research Relevance

IL-31 antibodies like AF2824 are critical for:

  • Diagnostic assays: Quantifying IL-31 in autoimmune/inflammatory conditions.

  • Mechanistic studies: Elucidating IL-31's role in Th2 immunity and itch pathways .

  • Therapeutic development: Neutralizing IL-31 in preclinical models of dermatitis .

Comparative Analysis of Anti-Cytokine Antibodies

TargetAntibody CloneApplicationKey FindingSource
IL-31AF2824ICC/ImmunoassayDetects IL-31 in activated T cells
PLIN1Polyclonal IgGLipodystrophy DxBlocks ABHD5 binding, increases lipolysis
HBV-preS1Chimeric S/preS1Vaccine DevelopmentEnhances neutralizing antibody titers

Limitations and Future Directions

  • AF2824 Specificity: Cross-reactivity with IL-31 isoforms requires further validation.

  • Therapeutic Potential: Clinical trials of IL-31 inhibitors (e.g., nemolizumab) show promise but are not yet FDA-approved .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks lead time (made-to-order)
Synonyms
AGL31 antibody; MAF2 antibody; At5g65050 antibody; MXK3.30Agamous-like MADS-box protein AGL31 antibody; Protein MADS AFFECTING FLOWERING 2 antibody
Target Names
AGL31
Uniprot No.

Target Background

Function
AGL31 is a putative transcription factor that inhibits vernalization induced by short cold periods. It functions as a floral repressor.
Gene References Into Functions
  • FLM and MAF2 repress flowering concurrently under low-temperature conditions. [MAF2] PMID: 25955034
  • Research elucidates the molecular mechanism of MAF2 gene function and the role of alternative MAF2 transcripts in modulating flowering time. PMID: 23111501
  • Association studies indicate a correlation between MAF2-MAF3 gene rearrangements and flowering time variation. PMID: 19139056
Database Links

KEGG: ath:AT5G65050

STRING: 3702.AT5G65050.3

UniGene: At.49225

Subcellular Location
Nucleus.
Tissue Specificity
Expressed in most plant tissues, roots, seedlings, leaves, stems, inflorescences, pollen, siliques and flowers.

Q&A

What is AGL and what is its role in glycogen metabolism?

AGL (amylo-alpha-1,6-glucosidase, 4-alpha-glucanotransferase) functions as a multifunctional enzyme in glycogen metabolism pathways. It acts as both a 1,4-alpha-D-glucan:1,4-alpha-D-glucan 4-alpha-D-glycosyltransferase and amylo-1,6-glucosidase during glycogen degradation processes . This bifunctional nature enables AGL to perform two critical steps in glycogen debranching: (1) transferring maltotriose units from α-1,6 branch points to nearby α-1,4 chains, and (2) hydrolyzing the remaining α-1,6 glucosidic linkage at the branch point. These coordinated activities are essential for complete glycogen breakdown in metabolically active tissues.

How are AGL31 antibodies validated for research applications?

Validation of AGL31 antibodies typically involves a multi-parameter approach beginning with immunogen design and antibody production, followed by rigorous specificity testing. Researchers should confirm antibody specificity through Western blot analysis comparing wild-type samples with genetically modified models lacking the target protein. Immunoprecipitation coupled with mass spectrometry provides additional validation by confirming the identity of captured proteins. Cross-reactivity assessment should include testing against closely related proteins, particularly other glycosidases with structural similarities. For immunohistochemical applications, validation should include antibody titration experiments across multiple tissue types, comparing staining patterns with previously established expression profiles . Finally, functional blocking experiments can demonstrate the antibody's ability to inhibit known AGL enzymatic activities.

What are the principal applications of AGL antibodies in glycogen metabolism research?

AGL antibodies serve as essential tools for investigating glycogen metabolism disorders and normal physiological processes. In immunohistochemistry and immunofluorescence applications, these antibodies enable visualization of AGL localization within tissues and subcellular compartments, often revealing dynamic translocation patterns during metabolic changes. Western blotting with AGL antibodies allows quantification of protein expression levels across different physiological conditions. Researchers use AGL antibodies in immunoprecipitation experiments to study protein-protein interactions, particularly within glycogen metabolism complexes. Flow cytometry applications permit analysis of AGL expression in specific cell populations, while ELISA-based methods enable quantitative measurement of AGL protein levels in biological samples. Additionally, these antibodies can be employed in functional assays to inhibit specific enzymatic activities, helping elucidate the distinct roles of AGL's transferase versus glucosidase functions in physiological contexts .

How should I optimize immunofluorescence protocols with AGL31 antibodies?

Optimizing immunofluorescence protocols with AGL31 antibodies requires systematic adjustment of multiple parameters to achieve specific signal with minimal background. Begin with fixation optimization—compare paraformaldehyde (4%) with methanol fixation, as AGL epitopes may be sensitive to specific fixation methods. For antigen retrieval, test both heat-mediated (citrate buffer, pH 6.0) and enzymatic methods to determine which best exposes the AGL epitopes without compromising tissue architecture. Blocking solutions should contain both serum (5-10% from species unrelated to primary and secondary antibodies) and BSA (1-3%) to minimize non-specific binding.

For the primary antibody step, perform a titration series (1:100 to 1:2000) with overnight incubation at 4°C, comparing signal-to-noise ratios. When using conjugated antibodies like Alexa Fluor 555 Anti-AGL , test incubation times between 1-3 hours at room temperature versus overnight at 4°C. Include controls for autofluorescence (no antibody), secondary antibody specificity (no primary), and biological specificity (known AGL-negative tissue). For multiplex staining, select fluorophores with minimal spectral overlap and validate each antibody individually before combination.

What controls are essential when using AGL31 antibodies in Western blot applications?

Implementing comprehensive controls for Western blot experiments with AGL31 antibodies is fundamental to generating reliable and interpretable results. Essential controls include:

  • Positive control: Include lysate from tissues with known high AGL expression (liver, muscle) to confirm antibody reactivity.

  • Negative control: Use samples from AGL-knockout models or tissues known to lack AGL expression to verify specificity.

  • Loading control: Employ housekeeping proteins (β-actin, GAPDH) that are expressed consistently across your experimental conditions to normalize protein loading.

  • Molecular weight verification: Use the non-glycosylated (~24 kDa) and glycosylated (~29 kDa) forms of the protein as internal references, as observed with similar proteins in the search results .

  • Peptide competition: Pre-incubate AGL31 antibody with the immunizing peptide to demonstrate binding specificity; this should eliminate specific bands.

  • Secondary antibody control: Omit primary antibody to detect non-specific binding of the secondary antibody.

  • Denaturation comparison: Compare reducing (+DTT) versus non-reducing conditions to assess the antibody's epitope dependency on protein conformation .

  • Cross-reactivity assessment: Test antibody against recombinant related proteins to confirm specificity against similar enzyme families.

How can I determine the appropriate concentration of AGL31 antibodies for my experiment?

Determining the optimal antibody concentration requires a systematic titration approach tailored to your specific experimental system. Begin with a broad concentration range based on manufacturer recommendations, typically starting with dilutions from 1:100 to 1:3200 for most applications . For Western blotting, prepare a dilution series and process identical membrane strips with equal protein loading. Quantify signal-to-noise ratio at each concentration using densitometry, selecting the dilution that provides maximum specific signal with minimal background.

For immunofluorescence or immunohistochemistry, perform a similar titration on serial sections of the same tissue block. Evaluate specific staining intensity, background levels, and pattern consistency. For immunoprecipitation experiments, titrate both antibody amount (1-10 μg) and incubation time (1-16 hours) against a fixed amount of lysate, then analyze pull-down efficiency by Western blot.

Create a standardized reference table for your laboratory documenting optimal concentrations across applications, tissue types, and detection methods. Remember that freshly isolated tissues may require different antibody concentrations than cultured cells or fixed archived samples. Finally, validate your selected concentration across multiple experimental replicates to ensure reproducibility before proceeding with critical experiments.

Why might I observe inconsistent staining patterns with AGL31 antibodies?

Inconsistent staining patterns with AGL31 antibodies can arise from multiple technical and biological factors. From a technical perspective, variations in fixation protocols significantly impact epitope preservation; overfixation can mask AGL epitopes while underfixation may compromise cellular architecture. Inconsistent antigen retrieval, particularly with heat-mediated methods, can create temperature gradients across tissue sections leading to variable epitope exposure. Antibody degradation through repeated freeze-thaw cycles or improper storage may reduce binding efficiency over time.

From a biological standpoint, AGL expression and localization can vary based on metabolic status, with potential translocation between cytoplasmic and glycogen-associated pools depending on cellular energy demands. Consider that post-translational modifications like glycosylation may obscure epitopes in certain cellular contexts, as observed with similar proteins showing distinct non-glycosylated (~24 kDa) and glycosylated (~29 kDa) bands . Tissue heterogeneity, particularly in metabolically complex organs like liver, can present as patchy staining that reflects genuine biological variation rather than technical artifacts.

To address these challenges, standardize your protocols rigorously, maintain detailed records of reagent lots and preparation dates, and consider using automated staining platforms for improved consistency. Include both positive and negative control tissues in each experimental run to establish reliable baseline expectations for staining patterns.

How can I validate the specificity of my AGL antibody results?

Validating AGL antibody specificity requires a multi-faceted approach employing complementary techniques. Begin with molecular validation using siRNA or CRISPR-Cas9 to generate AGL-knockdown or knockout models; a genuine AGL31 antibody should show proportionally reduced or absent signal in these systems compared to controls. Peptide competition assays provide another specificity check—pre-incubation of the antibody with the immunizing peptide should abolish specific staining while leaving non-specific signals unchanged.

For immunohistochemical applications, orthogonal validation using multiple antibodies targeting different AGL epitopes should produce concordant staining patterns. RNA-protein correlation studies comparing immunohistochemistry results with mRNA expression data from in situ hybridization or RT-qPCR can provide additional confidence in antibody specificity.

Mass spectrometry analysis of immunoprecipitated material offers perhaps the strongest validation approach. Following pull-down with your AGL31 antibody, mass spectrometry identification should confirm AGL as the predominant captured protein . This approach has the additional benefit of identifying potential cross-reactive proteins and interacting partners.

Finally, functional validation demonstrating that your antibody can inhibit the known enzymatic activities of AGL—both its 4-alpha-D-glycosyltransferase and amylo-1,6-glucosidase functions—provides the ultimate confirmation of both specificity and epitope relevance .

What approaches can resolve cross-reactivity issues with AGL antibodies?

Resolving cross-reactivity issues with AGL antibodies requires a strategic combination of experimental modifications and analytical approaches. First, employ higher dilutions of primary antibody, as cross-reactivity often occurs at lower specificity thresholds. More stringent washing protocols with increased detergent concentration (0.1-0.3% Tween-20) and extended wash times can significantly reduce non-specific binding .

Consider modifying blocking solutions by incorporating proteins from the same species as your secondary antibody to prevent Fc receptor binding, or add specific blockers like avidin/biotin when using biotinylated detection systems. For Western blot applications, increasing the stringency of membrane blocking with 5% milk or BSA in TBST and performing antibody incubations at 4°C can enhance specificity.

Pre-absorption techniques can effectively eliminate cross-reactivity; incubate your antibody with lysates from tissues expressing potential cross-reactive proteins but lacking AGL. Monoclonal antibodies like EPR8880 typically offer better specificity than polyclonals , but even monoclonals can cross-react with structurally similar epitopes.

For advanced applications, consider immunodepletion strategies where your sample is pre-cleared with antibodies against known cross-reactive proteins before AGL detection. Epitope mapping can identify which specific region of your antibody causes cross-reactivity, allowing for more informed experimental design. Finally, computational approaches comparing your AGL epitope sequence against proteome databases can predict potential cross-reactive proteins, enabling proactive experimental controls.

How can AGL antibodies be utilized in studying glycogen storage diseases?

AGL antibodies serve as powerful tools for investigating glycogen storage diseases (GSDs), particularly GSD type III (Cori or Forbes disease) which results from AGL deficiency. These antibodies enable multiple investigative approaches critical for understanding disease mechanisms and developing therapeutic strategies. Immunohistochemical analysis using AGL antibodies can visualize abnormal glycogen accumulation patterns and quantify residual AGL expression in patient biopsies, helping distinguish between various GSD subtypes with overlapping clinical presentations.

Western blot analysis with AGL antibodies allows researchers to characterize mutant AGL proteins, determining whether specific mutations affect protein stability (reduced levels) or enzymatic function (normal levels but impaired activity). This distinction has important implications for designing mutation-specific therapeutic approaches. For functional studies, researchers can use AGL antibodies to immunoprecipitate the enzyme from patient samples for subsequent activity assays, correlating specific mutations with particular enzymatic deficits in the transferase or glucosidase functions .

In cellular models, fluorescently-conjugated AGL antibodies like Alexa Fluor 555 Anti-AGL enable live-cell imaging to track abnormal subcellular localization of mutant proteins. Combined with electron microscopy, these approaches reveal ultrastructural changes in glycogen particle morphology associated with specific AGL mutations. For therapeutic development, AGL antibodies allow monitoring of protein replacement or gene therapy approaches, quantifying the restoration of enzyme levels and localization patterns following intervention.

What are the considerations when using AGL antibodies in multiplex immunofluorescence?

Multiplex immunofluorescence with AGL antibodies requires careful planning to achieve reliable co-localization data without technical artifacts. Primary considerations include antibody compatibility—selected antibodies must originate from different host species or utilize distinct IgG subclasses to prevent cross-recognition by secondary antibodies. For directly conjugated antibodies like Alexa Fluor 555 Anti-AGL , verify that the fluorophore's spectral properties are compatible with your other selected markers, maintaining at least 30nm separation between emission peaks.

Sequential staining protocols are often superior to simultaneous approaches, particularly when combining membrane, cytoplasmic, and nuclear markers with AGL. Begin with the lowest abundance target (often AGL) and proceed to more abundant proteins. For each antibody combination, perform single-stain controls on serial sections to establish baseline staining patterns before multiplex experiments.

When studying AGL in the context of glycogen metabolism, special consideration should be given to epitope masking, as glycogen particles can physically obstruct antibody access to associated proteins. Consider enzymatic pretreatment with amylase to expose cryptic epitopes within glycogen-rich regions. Spectral overlap between fluorophores can be addressed through linear unmixing algorithms during image analysis, but these require appropriate single-fluorophore controls for calibration.

Finally, heterogeneous expression of AGL across different cell types may necessitate cell-specific markers in your multiplex panel to accurately interpret co-localization patterns. Include complementary markers such as GLUT4 (insulin-responsive tissues) or glycogen synthase to create a more comprehensive picture of glycogen metabolism dynamics.

How can I use AGL antibodies to study protein-protein interactions?

AGL antibodies enable sophisticated analysis of protein-protein interactions within glycogen metabolism complexes through multiple complementary approaches. Co-immunoprecipitation (co-IP) represents the foundation of such studies—using AGL antibodies to pull down the protein complex followed by Western blot analysis for potential interaction partners such as glycogen synthase, glycogen phosphorylase, or regulatory proteins. For enhanced specificity, consider crosslinking proteins in situ before lysis using membrane-permeable crosslinkers like DSP (dithiobis[succinimidyl propionate]).

Proximity ligation assays (PLA) offer a powerful alternative for visualizing protein interactions in their native cellular context. This technique uses AGL antibodies in combination with antibodies against suspected interaction partners, generating fluorescent signals only when proteins are within 40nm of each other. PLA provides spatial information about interactions while maintaining cellular architecture.

For dynamic interaction studies, fluorescently-labeled AGL antibodies like Alexa Fluor 555 Anti-AGL can be employed in Förster Resonance Energy Transfer (FRET) experiments when paired with complementary fluorophore-labeled antibodies against interaction partners. FRET signal occurs only when proteins are within 10nm, providing strong evidence of direct interaction.

Mass spectrometry-based approaches following AGL immunoprecipitation can identify novel interaction partners in an unbiased manner. To distinguish specific from non-specific interactions, compare results from stimulated versus basal conditions, as many glycogen metabolism protein interactions are regulated by hormones or energy status. Finally, bimolecular fluorescence complementation (BiFC) using epitope-tagged proteins can validate interactions identified through antibody-based methods.

What are the emerging applications of AGL antibodies in metabolic disease research?

AGL antibodies are increasingly being deployed in novel research contexts beyond classical glycogen storage diseases, particularly in broader metabolic disorder investigations. Recent applications include studying the role of aberrant glycogen metabolism in cancer cells, where altered AGL expression correlates with metabolic reprogramming that supports rapid proliferation. These antibodies are enabling spatial metabolomics approaches that map glycogen utilization patterns across tumor microenvironments, revealing metabolic heterogeneity that may influence treatment response.

In neurodegenerative disease research, AGL antibodies are helping characterize the recently recognized role of neuronal glycogen metabolism in conditions like Alzheimer's and Lafora disease. Immunofluorescence with these antibodies reveals abnormal glycogen accumulation patterns in specific neuronal populations, potentially linking energy metabolism disruption to neurodegeneration.

Single-cell proteomics applications represent another frontier, with AGL antibodies employed in mass cytometry (CyTOF) panels to analyze glycogen metabolism heterogeneity at the single-cell level across diverse tissue environments. This approach has revealed previously unappreciated metabolic phenotypes within seemingly homogeneous cell populations.

In diabetes research, AGL antibodies are being integrated into systems biology approaches that simultaneously monitor multiple enzymes involved in glucose homeostasis, creating comprehensive profiles of metabolic adaptation and failure. These multiparameter analyses are identifying novel regulatory relationships between glycogen metabolism and insulin signaling pathways that may offer new therapeutic targets.

How are modified AGL antibodies contributing to vaccine development?

Modified antibodies against antigenic loop (AGL) domains are making significant contributions to vaccine development strategies, particularly for hepatitis B virus (HBV). Researchers have developed chimeric proteins incorporating both the AGL domain of the S protein and immunogenic epitopes from the preS1 region, creating novel immunogens with enhanced ability to elicit neutralizing antibodies . These chimeric constructs trigger superior humoral and cellular immune responses compared to traditional vaccine designs based solely on the S protein.

The strategic modification approach includes inserting preS1 epitopes of varying lengths into the AGL domain between amino acids 126 and 127 of the S protein to optimize immunogenicity while maintaining proper protein folding and stability . For example, the S/preS1 16-42 chimeric antigen demonstrated significantly improved expression and secretion compared to previous constructs while preserving the ability to assemble into subviral particles (SVPs) .

Immunological analysis reveals that these modified constructs induce an earlier and stronger IgG immune response than standard S protein vaccines, with significantly higher IgG1 titers indicating effective activation of Th2 cells and consequently robust humoral immunity . Importantly, sera from mice immunized with these chimeric antigens demonstrate neutralizing activity against both wild-type HBV and vaccine escape mutants .

The success of this approach suggests a general strategy for developing improved vaccines against other pathogens by incorporating multiple immunologically relevant epitopes into a single construct. Similar approaches could potentially be applied to design vaccines against emerging viral threats or to improve existing vaccines with suboptimal efficacy.

What technological advances are improving AGL antibody specificity and sensitivity?

Recent technological advances have significantly enhanced both the specificity and sensitivity of AGL antibodies, expanding their utility across research applications. Recombinant antibody engineering has enabled the development of monoclonal antibodies like EPR8880 with precisely defined epitope recognition sites, reducing batch-to-batch variability compared to traditional polyclonal approaches. The implementation of phage display technologies allows screening of vast antibody libraries to select clones with optimal binding characteristics for specific AGL epitopes.

Conjugation chemistry improvements have produced directly labeled antibodies like Alexa Fluor 555 Anti-AGL with enhanced fluorophore-to-antibody ratios while maintaining native binding properties. These advances reduce background and increase signal-to-noise ratios in imaging applications. Fragment antibody technologies, including single-chain variable fragments (scFvs) and nanobodies, provide improved tissue penetration and reduced background binding in complex samples.

Multiplex validation pipelines using orthogonal methods (immunoblotting, immunoprecipitation, and mass spectrometry) have become standard in antibody characterization, ensuring target specificity across applications. Manufacturers now routinely test antibodies against panels of related proteins to detect and document potential cross-reactivity issues before release.

For quantitative applications, the development of calibrated antibody techniques using recombinant protein standards enables absolute quantification of AGL levels in biological samples. Finally, computational epitope mapping and structural biology approaches help predict antibody binding sites and potential cross-reactivities before experimental testing, guiding more rational antibody design and application-specific optimization.

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.