GSK3B antibodies are engineered to bind specifically to GSK3B protein or its phosphorylated forms. They are categorized into:
Monoclonal antibodies: High specificity, targeting defined epitopes (e.g., clones 12B2, 15C2, MAB2506).
Polyclonal antibodies: Broader reactivity, recognizing multiple epitopes (e.g., DF7231, 15113-1-AP).
Feature | Monoclonal | Polyclonal |
---|---|---|
Specificity | Single epitope | Multiple epitopes |
Sensitivity | Higher reproducibility | Broader detection |
Applications | Western blot, IP, IHC, IF/ICC | WB, IF/ICC, ELISA |
Examples | 12B2 (GSK3β-specific), MAB2506 | DF7231, 15113-1-AP |
GSK3B antibodies are instrumental in studying GSK3B’s activity, localization, and interactions.
Used to quantify GSK3B expression and phosphorylation status. For example:
MAB2506 (R&D Systems) detects GSK3B at ~46 kDa in human and rodent cell lysates .
12B2 (Frontiers) specifically identifies non-phosphorylated Serine 9 GSK3β, distinguishing it from GSK3α .
Isolates GSK3B for downstream analysis.
Maps GSK3B localization in tissues and cells.
ZG004 (Thermo Fisher) stains GSK3B in nuclei and cytoplasm of human and rodent cells .
12B2 reveals punctate staining in neurons and glioblastoma cells, indicating subcellular compartmentalization .
GSK3B hyperactivation is implicated in Alzheimer’s disease (AD) and tauopathies:
Phosphorylates Tau: GSK3B induces tau hyperphosphorylation, a hallmark of neurofibrillary tangles in AD .
Mitochondrial Dysfunction: GSK3B phosphorylates Drp1, promoting mitochondrial fission and oxidative stress .
GSK3B modulates immune checkpoints:
PD-1/PD-L1 Axis: Inhibition of GSK3B reduces PD-1 expression, enhancing cytotoxic T-cell activity against tumors .
CTLA-4 Blockade: GSK3B inhibition reverses CTLA-4-mediated T-cell exhaustion, improving immunotherapy efficacy .
GSK3B phosphorylates ULK1 (S405/S415), activating autophagy in nutrient-deprived conditions. Pharmacological inhibition reduces autophagic cell death .
Phosphospecific antibodies have faced scrutiny due to:
Cross-Reactivity: Some antibodies (e.g., anti-pY216 GSK3β) show nuclear staining, while others (anti-pS21/9) detect focal adhesion proteins .
Silencing Experiments: Knockdown of GSK3B reduces antibody signals, confirming specificity in immunoblotting and IF .
GSK3β function is primarily regulated through phosphorylation at specific residues. Commercially available antibodies can specifically detect distinct phosphorylation states that indicate different activation levels. Antibodies targeting phosphorylated Ser9 detect the inhibited form of GSK3β, as this phosphorylation inactivates the kinase . Conversely, antibodies detecting phosphorylated Tyr216 recognize the activated form of GSK3β . Some specialized antibodies can also specifically detect non-phosphorylated Ser9 GSK3β, which represents the active form . When designing experiments, researchers must carefully select antibodies based on which activation state they wish to investigate, as using total GSK3β antibodies alone provides insufficient information about kinase activity.
While GSK3α and GSK3β share significant sequence homology in their catalytic domains, they differ in size (GSK3α is 51 kDa while GSK3β is 46 kDa) and have distinct N-terminal domains. Some antibodies are isoform-specific, while others detect both isoforms . Research shows that these isoforms can have non-redundant functions, particularly in T-cell mediated immunity where GSK3β plays a more prominent role in tumor rejection compared to GSK3α . Isoform-specific antibodies should be prioritized when investigating phenotypes where differential expression or function of GSK3α versus GSK3β has been established, or when conducting mechanistic studies requiring distinction between the two isoforms.
Proper validation of GSK3β antibodies requires several controls. For phospho-specific antibodies, include samples treated with agents known to modulate GSK3β phosphorylation (e.g., insulin treatment increases Ser9 phosphorylation) . Phosphatase-treated lysates serve as negative controls for phospho-specific antibodies. For antibodies claiming isoform specificity, GSK3β knockout/knockdown samples provide critical negative controls. When possible, recombinant GSK3β protein or immunizing peptides should be used in blocking experiments to confirm specificity . For immunohistochemistry applications, include control tissues with known GSK3β expression patterns and compare staining with and without immunizing peptides .
For Western blot detection of GSK3β, sample preparation is critical. Cells or tissues should be lysed in buffers containing phosphatase inhibitors to preserve phosphorylation states . Most GSK3β antibodies perform optimally at dilutions between 1:500-1:2000 . For phospho-specific detection, rapid sample processing is essential as phosphorylation states can change quickly. Normalized loading requires detection of both phosphorylated and total GSK3β on separate blots or through stripping and reprobing. For visualization of both GSK3α and GSK3β, ensure adequate resolution in the 45-51 kDa range, typically using 10% acrylamide gels. When comparing treatment effects, include appropriate positive controls such as insulin-treated samples, which induce Ser9 phosphorylation .
Successful immunohistochemistry for GSK3β requires careful optimization of several parameters. Antigen retrieval methods significantly impact detection—heat-induced epitope retrieval in citrate buffer (pH 6.0) works well for most GSK3β antibodies. Antibody dilutions typically range from 1:200-1:1000 for optimal staining . For phospho-specific detection, fixation should be rapid to preserve in vivo phosphorylation states. To confirm specificity, perform parallel staining with and without blocking peptides . For paraffin-embedded tissues, antibodies like ab75745 have been validated at 1:50 dilution . Researchers should validate staining patterns by comparing with published literature and confirming cellular localization patterns (GSK3β can be cytoplasmic, nuclear, or both depending on cellular context).
For flow cytometry applications, researchers must optimize fixation and permeabilization protocols, as these significantly impact antibody accessibility to intracellular GSK3β. For phospho-specific detection, use methanol or paraformaldehyde/methanol fixation which better preserves phospho-epitopes. Antibody concentrations typically range from 1:200-1:400 for flow cytometry . Include proper compensation controls when using multiple fluorophores, and utilize fluorescence-minus-one controls to set gates accurately. Validation should include positive control samples (e.g., cell lines with known GSK3β expression) and negative controls (isotype antibodies or blocking with immunizing peptides). For analyzing GSK3β activation in specific cell populations, combine with appropriate surface markers .
Research has revealed significant crosstalk between GSK3β and STAT3 signaling networks. To investigate this relationship, researchers should employ antibodies against both total and phosphorylated forms of GSK3β alongside STAT3 detection reagents . Cancer phospho-antibody arrays have identified STAT3 as a target of GSK3β, with GSK3β inhibition reducing STAT3 phosphorylation . Experimental approaches should include pharmacological inhibition of GSK3β (e.g., with specific inhibitors), genetic modulation using siRNA knockdown, and overexpression of constitutively active GSK3β (S9A) . Co-immunoprecipitation experiments can detect direct interactions between these proteins. When analyzing downstream effects, researchers should examine changes in target gene expression using qRT-PCR with primers specific for GSK3β and related pathway components .
To investigate GSK3β in Wnt/β-catenin signaling, researchers should employ a multi-faceted approach. Western blotting using antibodies against total GSK3β, phospho-Ser9 GSK3β, and downstream targets like β-catenin and phospho-β-catenin provides information about pathway activation status. Immunoprecipitation experiments using GSK3β antibodies can help identify interactions with other components of the destruction complex. For functional studies, combine GSK3β antibody detection with TOPFlash reporter assays measuring β-catenin-mediated transcriptional activity. When studying pharmacological modulation, include appropriate controls for pathway activation (Wnt ligands, GSK3β inhibitors) and inhibition. Importantly, researchers should remember that GSK3β functions within large protein complexes in this pathway, so analysis of its phosphorylation state alone may not fully reflect its activity toward β-catenin.
Studies have revealed distinct roles for GSK3α and GSK3β isoforms in T cell-mediated immunity. When investigating these functions, researchers should utilize isoform-specific antibodies combined with genetic approaches using conditional knockout models (e.g., Gsk3a cKO, Gsk3b cKO, and double cKO) . Flow cytometry analysis using antibodies against T cell activation markers alongside GSK3 isoform detection can help correlate expression patterns with functional states. Research shows that GSK3β plays a more significant role in controlling PD-1 expression, as Gsk3b cKO models show reduced Pdcd1 expression compared to Gsk3a cKO models . For tumor immunity studies, analyze tumor-infiltrating lymphocytes using immunohistochemistry with antibodies against CD4, CD8, and PD-1, comparing findings across different GSK3 knockout models .
Research has established connections between GSK3β activity and cancer progression. When studying this relationship, researchers should compare both total GSK3β expression and phosphorylation states (particularly at Ser9) between normal and cancerous tissues . Immunohistochemistry using validated GSK3β antibodies can reveal expression patterns within tumor microenvironments. Higher GSK3β expression correlates with poorer differentiation, increased metastasis rates, and worse prognosis in certain cancers like ESCC . Experimental approaches should include pharmacological inhibition and siRNA knockdown in cancer cell lines, combined with analysis of proliferation, migration, and invasion. For mechanistic studies, examine the effects of GSK3β modulation on downstream targets like STAT3, detecting changes in phosphorylation status using specific antibodies .
In neurodegenerative disease research, GSK3β has been implicated in tau phosphorylation and amyloid processing. Researchers should utilize antibodies that detect both Ser9 phosphorylation (indicating inhibition) and Tyr216 phosphorylation (indicating activation) to fully characterize GSK3β activity states . For brain tissue analysis, optimize immunohistochemistry protocols with proper antigen retrieval methods and validate specificity with blocking peptides . When studying GSK3β-mediated tau phosphorylation, combine GSK3β antibodies with phospho-tau antibodies recognizing sites specifically phosphorylated by GSK3β. Consider subcellular fractionation before Western blotting, as GSK3β localization can shift between cytoplasmic and nuclear compartments in disease states. For in vivo models, correlate biochemical findings with behavioral outcomes to establish functional significance.
When evaluating GSK3β inhibitors, researchers must carefully assess both direct target engagement and downstream pathway modulation. Western blotting with antibodies against phospho-Ser9 GSK3β can verify inhibitor effects, though some inhibitors work through mechanisms independent of Ser9 phosphorylation . Validation should include measuring phosphorylation of established GSK3β substrates (e.g., glycogen synthase, β-catenin). Novel non-phosphorylated Ser9/21 GSK3β/α antibodies can provide valuable information about the pool of active GSK3β . For quantification, establish dose-response relationships using varying inhibitor concentrations, and include time-course analyses as phosphorylation states can change rapidly. Include genetic approaches (siRNA knockdown, dominant-negative constructs) as complementary validation methods. For in vivo studies, assess tissue-specific effects of inhibitors by analyzing GSK3β phosphorylation in multiple organs.
Inconsistent phospho-GSK3β detection often stems from technical challenges in preserving phosphorylation states. Researchers should implement rapid sample processing with immediate addition of phosphatase inhibitors to lysis buffers . For Western blotting, optimize transfer conditions for proteins in the 45-51 kDa range, and consider using PVDF membranes which may better retain phosphoproteins. Phosphorylation states can be labile, so standardize the time between sample collection and analysis. If inconsistencies persist, compare multiple phospho-specific antibodies targeting the same site but recognizing different epitopes . For cell-based studies, strictly control culture conditions, as factors like cell density, serum components, and metabolic state can dramatically affect GSK3β phosphorylation. Finally, include appropriate positive controls, such as insulin-treated samples which reliably increase Ser9 phosphorylation .
Cross-reactivity between GSK3 isoforms presents challenges for isoform-specific analysis. To address this, researchers should first validate antibody specificity using recombinant GSK3α and GSK3β proteins in Western blots or ELISAs . For ambiguous results, employ genetic approaches using isoform-specific knockdown or knockout samples to confirm antibody specificity . When Western blotting, optimize gel conditions to clearly separate the 46 kDa (GSK3β) and 51 kDa (GSK3α) bands, typically using 10% acrylamide gels with extended run times. If cross-reactivity persists, consider immunoprecipitation with one verified isoform-specific antibody followed by detection with a second antibody. For critical experiments requiring absolute isoform specificity, implement parallel approaches using both antibody detection and genetic manipulation of specific isoforms .
Discrepancies between total GSK3β expression and phosphorylation patterns are common and reflect the complex regulation of this kinase. When facing such conflicts, researchers should remember that GSK3β activity is primarily regulated by phosphorylation rather than expression levels . Consider that different phosphorylation sites have opposing effects—Ser9 phosphorylation is inhibitory while Tyr216 phosphorylation is activating—so measure both sites when possible . Subcellular fractionation may reveal that changes occur in specific cellular compartments rather than in total protein levels. For comprehensive analysis, combine antibody detection of phosphorylation states with direct kinase activity assays measuring phosphorylation of GSK3β substrates. Remember that other regulatory mechanisms including protein-protein interactions and subcellular localization can affect GSK3β function independently of detectable phosphorylation changes.
Single-cell analysis of GSK3β requires specialized approaches to detect phosphorylation states at the individual cell level. For single-cell Western blotting, researchers must optimize lysis conditions and antibody concentrations for the reduced protein content. In mass cytometry (CyTOF), metal-conjugated GSK3β antibodies can be integrated into panels with dozens of other markers to correlate GSK3β activity with cell phenotypes . For imaging mass cytometry, validate antibodies specifically for this application, as performance can differ from traditional immunohistochemistry. Single-cell RNA-seq can complement protein-level analyses by examining GSK3β transcript levels alongside genome-wide expression patterns. When implementing these techniques, include appropriate controls at the single-cell level and validate findings with population-based methods to ensure reliability.
Multiplexed detection of GSK3β and its interacting proteins provides valuable insights into signaling networks. Proximity ligation assays (PLAs) offer a powerful approach, requiring antibodies against GSK3β and potential binding partners raised in different host species . Co-immunoprecipitation followed by mass spectrometry can identify novel interacting partners, using GSK3β antibodies validated for immunoprecipitation applications. For imaging-based multiplexing, implement cyclic immunofluorescence or multiplexed ion beam imaging, optimizing GSK3β antibody concentration for each technique. When designing multiplexed panels, include antibodies against known GSK3β regulators (e.g., Akt, which phosphorylates GSK3β at Ser9) and substrates (e.g., β-catenin, STAT3) . Critical controls should include competitive blocking with immunizing peptides and isotype controls to confirm specificity in multiplexed systems.
Integrating GSK3β phosphorylation data into systems biology frameworks requires multimodal approaches. Researchers should quantify both total GSK3β and site-specific phosphorylation across experimental conditions using validated antibodies . These protein-level measurements can be combined with transcriptomic data measuring expression of GSK3β itself and its known targets. Phosphoproteomics approaches can provide a broader view of signaling networks, allowing researchers to position GSK3β within global phosphorylation cascades. For mathematical modeling, include quantitative data on GSK3β phosphorylation kinetics in response to various stimuli. Network analysis should incorporate known GSK3β interactions from public databases, supplemented with experimental validation using techniques like co-immunoprecipitation or proximity ligation assays. This integrated approach provides a comprehensive understanding of GSK3β function within complex cellular systems.
Glycogen synthase kinase-3 beta (GSK-3β) is a serine/threonine kinase that plays a crucial role in various cellular processes. It is one of the two isoforms of glycogen synthase kinase-3 (GSK-3), the other being GSK-3 alpha (GSK-3α). GSK-3β is encoded by the GSK3B gene in humans and the Gsk3b gene in mice . This enzyme is involved in numerous cellular functions, including glycogen metabolism, cell cycle regulation, apoptosis, embryonic development, cell differentiation, and inflammation .
GSK-3β plays a fundamental role in the inflammatory response induced by bacterial pathogens. Depending on the pathogen and its virulence factors, GSK-3β may either promote or inhibit inflammation . It modulates inflammatory signaling in monocytes/macrophages and epithelial cells at the transcriptional level, primarily through the regulation of nuclear factor-kappaB (NF-κB) activity .