The antibody is validated for multiple techniques, with dilutions varying by application:
Protocols emphasize stringent controls to minimize nonspecific binding, particularly in mitotic cells where cross-reactivity with spindle pole proteins has been reported .
While designed for phospho-Ser9 specificity, challenges persist:
Phospho-GSK3B (Ser9) is a hallmark of inactivated GSK3B, which stabilizes β-catenin in the Wnt pathway. Recombinant antibodies enable precise detection of this regulatory state across models:
Wnt/β-Catenin Pathway: Phosphorylation at Ser9 inhibits GSK3B, promoting β-catenin nuclear translocation .
Metabolic Regulation: Linked to insulin signaling and glycogen synthesis via GYS1 inhibition .
Disease Relevance: Altered Ser9 phosphorylation implicated in Alzheimer’s disease, Parkinson’s, and cancer .
This recombinant monoclonal antibody against Phospho-GSK3B (Ser9) was developed using a synthesized peptide derived from human Phospho-GSK3B (Ser9). The process involved immunizing an animal with the peptide, isolating positive splenocytes, extracting RNA, obtaining DNA through reverse transcription, sequencing and screening for the GSK3B antibody gene, and amplifying the heavy and light chain sequences by PCR before cloning them into plasma vectors. Subsequently, the vector clones were transfected into mammalian cells for antibody production. The final product is a purified recombinant GSK3B antibody obtained using affinity chromatography. This antibody exhibits reactivity with GSK3B protein from humans and finds applications in ELISA, Western blotting, immunohistochemistry, and immunofluorescence assays.
The protein encoded by the GSK3B gene is a serine-threonine kinase belonging to the glycogen synthase kinase subfamily. It acts as a negative regulator of glucose homeostasis and plays a crucial role in various cellular pathways, including energy metabolism, inflammation, ER-stress, mitochondrial dysfunction, and apoptosis. Studies have revealed several notable characteristics of GSK3B:
GSK3B (Glycogen synthase kinase-3 beta) is a serine-threonine kinase belonging to the glycogen synthase kinase subfamily. It functions as a negative regulator of glucose homeostasis and is involved in energy metabolism, inflammation, endoplasmic reticulum stress, mitochondrial dysfunction, and apoptosis pathways . Phosphorylation at Serine 9 (Ser9) is a key regulatory mechanism that inhibits GSK3B activity. This post-translational modification is crucial for controlling numerous cellular processes including metabolism, cell proliferation, and neuronal function. When GSK3B is phosphorylated at Ser9, its kinase activity is significantly reduced, leading to activation of downstream pathways that were previously suppressed by active GSK3B .
Phospho-GSK3B (Ser9) antibodies are generated through several sophisticated biotechnological approaches. For recombinant monoclonal antibodies, the process involves immunizing animals with synthesized peptides containing the phosphorylated Ser9 residue derived from human GSK3B. The positive splenocytes are isolated, and RNA is extracted and reverse transcribed to DNA. The GSK3B antibody gene is then sequenced, screened, and the heavy and light chain sequences are amplified by PCR and cloned into plasma vectors. These vector clones are subsequently transfected into mammalian cells for production of the recombinant antibody . For polyclonal antibodies, rabbits are immunized with human GSK3β-derived peptide sequences surrounding the phosphorylation site (T-T-S(p)-F-A). The antibodies are purified using affinity-chromatography with epitope-specific phosphopeptides to ensure high specificity for the phosphorylated form of GSK3B .
GSK3 exists in two distinct isoforms: GSK-3α and GSK-3β. While these isoforms share high sequence homology in their kinase domains (approximately 98%), they differ in their N- and C-terminal regions, resulting in unique functional properties. Research demonstrates that these isoforms have non-redundant activities in T cells and differ in their impact on various cellular processes. GSK-3β appears to play a more significant role in T cell-mediated anti-tumor immunity compared to GSK-3α. Studies using conditional gene targeting have shown that deletion of GSK-3β alone suppresses tumor growth to the same degree as double knockout of both isoforms, whereas GSK-3α knockout mice behave similarly to wild-type . Furthermore, the individual isoforms differentially affect PD-1, IFNγ, and granzyme B expression, with GSK-3β having a more pronounced effect on reducing PD-1 expression .
For Western Blotting applications using Phospho-GSK3B (Ser9) antibodies, the following methodological approach is recommended:
Sample Preparation: Prepare protein lysates from cells or tissues of interest, ensuring phosphatase inhibitors are included in the lysis buffer to preserve phosphorylation status.
Antibody Dilution: For recombinant monoclonal antibodies, use dilutions ranging from 1:500 to 1:5000 . For polyclonal antibodies, use dilutions of 1:500 to 1:1000 .
Detection System: Use an appropriate secondary antibody conjugated to HRP or fluorescent tags, followed by standard detection methods.
Controls: Include both positive controls (samples known to contain phosphorylated GSK3B) and negative controls (samples where GSK3B is dephosphorylated or where phosphatases have been used to remove phosphorylation).
A validated example from research literature involves HT22 cells pretreated with Tat-C3 (1 μg/ml) for 1 hour, followed by treatment with fAβ (1 μM or 10 μM) for 24 hours. The protein samples were then analyzed by western blotting using Phospho-GSK3B (Ser9) antibody to assess changes in phosphorylation status following experimental treatments .
For optimal results in immunohistochemistry (IHC) and immunofluorescence (IF) applications:
Immunohistochemistry Protocol:
Tissue Preparation: Fix tissues in appropriate fixative (commonly 4% paraformaldehyde) and embed in paraffin or prepare frozen sections.
Antigen Retrieval: Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0).
Antibody Dilution: For recombinant antibodies, use dilutions of 1:50-1:200 . For polyclonal antibodies, use 1:50-1:100 .
Blocking: Block with 5-10% normal serum from the species of the secondary antibody.
Detection: Use appropriate detection systems (e.g., HRP-DAB).
Immunofluorescence Protocol:
Cell/Tissue Preparation: For cells, fix in 4% paraformaldehyde and permeabilize with 0.3% saponin .
Antibody Dilution: For recombinant antibodies, use 1:20-1:200 . For polyclonal antibodies, use 1:100-1:200 .
Staining: Stain with primary antibody in buffer containing 0.3% saponin for 2 hours at 4°C, followed by appropriate fluorophore-conjugated secondary antibody .
Counterstaining: Use DAPI or other nuclear stains for orientation.
These protocols should be optimized based on the specific sample type and experimental requirements.
For flow cytometry applications detecting phosphorylated GSK3B, the following protocol has been validated in research settings:
Cell Preparation: Suspend 10^6 cells in 100μl PBS.
Surface Marker Staining: If analyzing specific cell populations, first stain with surface markers (e.g., anti-CD8α, anti-CD4, anti-CD3) at 1:100 dilution for 2 hours at 4°C.
Fixation and Permeabilization: Fix cells in 4% paraformaldehyde (PFA), then permeabilize with 0.3% saponin.
Intracellular Staining: Stain with Phospho-GSK3B (Ser9) antibody in saponin-containing PBS for 2 hours at 4°C.
Secondary Antibody: If primary antibody is not directly conjugated, incubate with appropriate secondary antibody.
Analysis: Analyze cell staining on a flow cytometer (e.g., Beckman Coulter CytoFLEX S) .
This method allows for simultaneous analysis of phosphorylated GSK3B levels alongside other cellular markers, enabling correlation of GSK3B phosphorylation with specific cell phenotypes or activation states.
The phosphorylation status of GSK3B at Ser9 plays a critical role in regulating T cell function within tumor microenvironments through several interconnected mechanisms:
PD-1 Expression Regulation: GSK-3β is a positive regulator of PD-1 expression in CD8+ T cells. Deletion of GSK-3β or inhibition of GSK-3 activity significantly reduces PD-1 expression, potentially enhancing anti-tumor immunity .
T Cell Infiltration: Studies using conditional knockout models have demonstrated that GSK-3β deletion results in significantly increased infiltration of both CD4+ and CD8+ T cells into tumors. This effect appears to be specific to the β isoform, as GSK-3α deletion does not produce the same outcome .
Effector Function Enhancement: GSK-3β deletion increases the expression of effector molecules in tumor-infiltrating CD8+ T cells, including:
Isoform-Specific Effects: While both GSK-3α and GSK-3β isoforms contribute to T cell function, they do so to different degrees. GSK-3β appears to have a dominant role in controlling T cell-mediated anti-tumor immunity, as evidenced by:
These findings highlight the potential therapeutic value of targeting GSK-3β phosphorylation to enhance T cell-mediated anti-tumor responses.
To investigate the non-redundant functions of GSK3 isoforms, researchers employ several sophisticated experimental approaches:
Conditional Gene Targeting: Creating conditional knockout models where GSK-3α, GSK-3β, or both are selectively deleted in specific cell types (e.g., T cells) allows for the precise examination of isoform-specific functions. This approach has revealed that GSK-3β plays a more significant role in T cell-mediated anti-tumor immunity than GSK-3α .
Isoform-Specific Inhibitors: Using chemical compounds that preferentially inhibit one isoform over the other can help discriminate between their functions without genetic manipulation.
Phosphorylation Site-Specific Antibodies: Utilizing antibodies like Phospho-GSK3B (Ser9) that recognize specific phosphorylation sites enables the monitoring of isoform-specific activity regulation in different cellular contexts.
Tumor Models: Employing different tumor models (e.g., flank models, intravenous B16 melanoma models) to assess how deletion of specific GSK3 isoforms affects:
Multiparameter Flow Cytometry: Analyzing multiple parameters simultaneously (e.g., surface markers, intracellular cytokines, transcription factors) to determine how GSK3 isoforms differentially affect various aspects of T cell phenotype and function.
Immunohistochemistry: Examining tissue sections to quantify differences in immune cell infiltration, PD-1 expression, and regulatory T cell (Foxp3+) populations in tumors from mice lacking specific GSK3 isoforms .
These complementary approaches provide a comprehensive understanding of the distinct roles played by GSK-3α and GSK-3β in various biological processes.
GSK3B phosphorylation status has significant implications for neurological disease progression through multiple mechanisms:
Parkinson's Disease: GSK3B polymorphisms alter transcription and splicing patterns and interact with Tau haplotypes to modify disease risk in Parkinson's disease. Phosphorylation at Ser9, which inhibits GSK3B activity, appears to be protective against certain neurodegenerative processes .
Alzheimer's Disease and Frontotemporal Dementia: GSK3B plays a crucial role in dementia susceptibility. Phosphorylated (inactive) GSK3B levels are often decreased in Alzheimer's disease, leading to hyperphosphorylation of tau protein and formation of neurofibrillary tangles .
Neuronal Development and Survival: GSK3B is a multifunctional kinase that regulates diverse early events of neuronal development, including neurogenesis, neuronal migration, differentiation, and survival in the immature brain. Proper regulation of its phosphorylation status is essential for normal brain development .
Amyloid-Beta Signaling: Experimental evidence using HT22 cells treated with fibrillar amyloid-beta (fAβ) demonstrates that GSK3B phosphorylation status changes in response to Aβ signaling, potentially linking this kinase to Alzheimer's disease pathogenesis .
Monitoring GSK3B phosphorylation at Ser9 using specific antibodies provides valuable insights into disease mechanisms and potential therapeutic strategies for neurological disorders.
When designing experiments with Phospho-GSK3B (Ser9) antibodies, including appropriate controls is essential for valid data interpretation:
Essential Controls:
Phosphatase Treatment Control: Treat a portion of your sample with lambda phosphatase to remove phosphorylation modifications. This should eliminate or significantly reduce signal with the phospho-specific antibody, confirming specificity for the phosphorylated form.
Total GSK3B Control: Probe parallel samples with an antibody recognizing total GSK3B (phosphorylation-independent) to normalize phospho-signal to total protein levels.
Positive Control: Include samples known to contain high levels of phosphorylated GSK3B (Ser9), such as cells treated with insulin, serum, or other stimuli known to activate the PI3K/Akt pathway, which phosphorylates GSK3B at Ser9.
Negative Control: Include samples where GSK3B phosphorylation is minimized, such as serum-starved cells or cells treated with PI3K/Akt inhibitors.
Isotype Control: Use an antibody of the same isotype but irrelevant specificity to assess non-specific binding, particularly important for flow cytometry and immunohistochemistry applications.
Cross-Reactivity Assessment: When examining both GSK3 isoforms, confirm that your Phospho-GSK3B (Ser9) antibody doesn't cross-react with phosphorylated GSK3A (Ser21), which has a similar surrounding sequence.
Implementation of these controls ensures reliable interpretation of experimental results and helps troubleshoot technical issues.
When encountering discrepancies in GSK3B phosphorylation data across different experimental techniques, researchers should systematically address these inconsistencies through the following approaches:
Technique-Specific Considerations:
Western Blotting: Phosphorylation signals can be affected by sample preparation methods, particularly the effectiveness of phosphatase inhibitors during lysis.
Immunohistochemistry/Immunofluorescence: Fixation methods and antigen retrieval procedures significantly impact phospho-epitope detection.
Flow Cytometry: Cell permeabilization conditions may differentially affect accessibility to phospho-epitopes.
Antibody Validation Strategy:
Confirm antibody specificity using phosphatase treatments in each experimental system.
Validate results with multiple antibody clones recognizing the same phospho-epitope.
Consider using alternative methods to detect GSK3B activity (e.g., kinase assays, downstream substrate phosphorylation).
Temporal Dynamics:
GSK3B phosphorylation is highly dynamic and can change rapidly. Ensure that samples across different techniques are collected under identical temporal conditions.
Perform time-course experiments to map the kinetics of phosphorylation changes.
Spatial Resolution Differences:
Immunostaining techniques provide spatial information that may reveal cell-specific or subcellular localization patterns not detectable by western blotting.
Flow cytometry allows analysis of phosphorylation at the single-cell level, potentially revealing heterogeneity masked in population-based assays.
Quantification Approaches:
Standardize quantification methods across techniques.
For western blotting, always normalize phospho-signals to total GSK3B levels.
For imaging techniques, employ consistent thresholding and analysis parameters.
By systematically addressing these factors, researchers can reconcile apparent discrepancies and develop a more comprehensive understanding of GSK3B phosphorylation dynamics in their experimental systems.
Reproducibility in GSK3B phosphorylation studies is influenced by several critical factors that researchers must carefully control:
Sample Handling and Preparation:
Phosphatase Inhibition: Immediate and effective inhibition of phosphatases during sample collection is crucial, as phosphorylation status can change rapidly post-collection.
Temperature Control: Maintain samples at appropriate temperatures throughout processing to prevent artifactual changes in phosphorylation.
Standardized Lysis Conditions: Use consistent buffer compositions, incubation times, and mechanical disruption methods.
Experimental Timing and Conditions:
Cell Culture Variables: Control for cell density, passage number, and growth conditions, as these affect baseline phosphorylation.
Treatment Timing: Standardize the duration of treatments and the time between treatment and sample collection.
Circadian Factors: For animal studies, control for time of day, as GSK3B phosphorylation may follow circadian patterns.
Antibody Factors:
Lot-to-Lot Variability: Validate new antibody lots against previous ones before use in critical experiments.
Storage and Handling: Follow manufacturer recommendations for antibody storage and avoid repeated freeze-thaw cycles.
Optimal Dilutions: Determine optimal antibody dilutions for each application through titration experiments.
Technical Parameters:
Protocol Standardization: Maintain detailed protocols and minimize variations in experimental procedures.
Instrument Calibration: Regularly calibrate and maintain equipment used for detection and analysis.
Quantification Methods: Use consistent approaches for signal quantification and normalization.
Biological Variables:
Genetic Background: Control for genetic differences in model organisms or cell lines.
Age and Sex Effects: Account for age and sex as potential variables affecting GSK3B regulation.
Health Status: Ensure consistent health status of research subjects or cell cultures.
Controlling these factors will significantly enhance reproducibility in GSK3B phosphorylation studies and improve the reliability of research findings.
Phospho-GSK3B (Ser9) antibodies offer valuable tools for exploring cancer immunotherapy resistance mechanisms through several innovative applications:
Checkpoint Inhibitor Resistance Assessment:
GSK3B is a positive regulator of PD-1 expression in CD8+ T cells, and its phosphorylation status may predict responsiveness to PD-1/PD-L1 inhibitors .
Analyzing phospho-GSK3B levels in tumor-infiltrating lymphocytes before and during checkpoint inhibitor therapy can help identify resistance mechanisms related to T cell exhaustion.
Biomarker Development:
Phospho-GSK3B (Ser9) levels in tumor or immune cells could serve as predictive biomarkers for immunotherapy response.
Multi-parameter analysis combining phospho-GSK3B with other immune markers could create more robust prediction models for treatment outcomes.
Combination Therapy Strategies:
Since GSK3B inhibition enhances T cell function and tumor control , measuring phospho-GSK3B can help identify patients who might benefit from combining GSK3B inhibitors with existing immunotherapies.
Phospho-GSK3B antibodies can be used to monitor on-target effects of GSK3B inhibitors in clinical samples.
Isoform-Specific Targeting:
Tumor Microenvironment Characterization:
Multiplexed imaging using phospho-GSK3B antibodies alongside other markers can map the spatial distribution of active/inactive GSK3B in different immune cell populations within the tumor microenvironment.
This approach provides insights into the heterogeneity of GSK3B signaling within tumors and its relationship to immunotherapy resistance.
These applications demonstrate how phospho-GSK3B antibodies can advance our understanding of immunotherapy resistance mechanisms and potentially inform more effective treatment strategies.
GSK3B phosphorylation status has profound implications for metabolic reprogramming in immune cells, representing an emerging frontier in immunometabolism research:
T Cell Activation and Differentiation:
GSK3B inhibition (increased Ser9 phosphorylation) promotes glycolytic metabolism in activated T cells, supporting their proliferation and effector function.
The metabolic shift associated with GSK3B inhibition influences CD8+ T cell differentiation, potentially favoring the development of memory precursors over terminal effectors.
Metabolic Pathway Regulation:
Integration with Nutrient-Sensing Pathways:
GSK3B interacts with other metabolic regulators like mTOR and AMPK, creating a signaling network that coordinates immune cell metabolism with nutrient availability.
Phospho-GSK3B serves as an integration point for various external signals that influence metabolic reprogramming during immune responses.
Impact on Tumor Microenvironment:
The metabolic competition between tumor cells and immune cells is partially regulated by GSK3B activity.
Modulating GSK3B phosphorylation may help overcome metabolic barriers to effective anti-tumor immunity in nutrient-depleted tumor microenvironments.
Therapeutic Opportunities:
Targeting GSK3B to alter immune cell metabolism represents a potential strategy for enhancing immunotherapy efficacy.
Monitoring phospho-GSK3B levels could help predict metabolic fitness of tumor-infiltrating lymphocytes and their potential for sustained anti-tumor activity.
Understanding these metabolic implications provides new avenues for therapeutic intervention and biomarker development in cancer immunotherapy and autoimmune diseases.
Spatial analysis of GSK3B phosphorylation within tumor microenvironments provides unprecedented insights into complex signaling networks that influence cancer progression and treatment response:
Cellular Heterogeneity Mapping:
Advanced immunohistochemistry using phospho-GSK3B (Ser9) antibodies can reveal the distribution of GSK3B activity across different cell types within tumors .
This approach identifies regions with differential GSK3B regulation, potentially correlating with tumor progression or treatment resistance zones.
Immune-Tumor Cell Interactions:
Signaling Pathway Integration:
Multiplexed imaging combining phospho-GSK3B with other phospho-proteins can map interconnected signaling networks within the spatial context of the tumor.
This reveals how GSK3B regulation correlates with other pathways (e.g., PI3K/Akt, Wnt/β-catenin) in specific tumor regions.
Microenvironmental Influence Assessment:
Correlating phospho-GSK3B patterns with hypoxic regions, nutrient availability, or extracellular matrix components provides insights into how the microenvironment modulates GSK3B activity.
This helps explain regional differences in tumor cell behavior and immune function within heterogeneous tumors.
Therapeutic Response Prediction:
Pre- and post-treatment spatial analysis of phospho-GSK3B can identify regions resistant to therapy.
Changes in the spatial pattern of GSK3B phosphorylation following treatment may predict long-term outcomes better than bulk measurements.
Technology Implementation:
Emerging spatial technologies like imaging mass cytometry, multiplexed ion beam imaging, or spatial transcriptomics combined with phospho-proteomics can provide unprecedented resolution of GSK3B signaling networks within the intact tumor architecture.
This spatial perspective transforms our understanding from a bulk average view to a detailed map of signaling heterogeneity, dramatically enhancing our ability to understand and target cancer-relevant pathways.