GSK3B Human

Glycogen synthase kinase-3 beta Human Recombinant
Shipped with Ice Packs
In Stock

Description

GSK3B Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 420 amino acids (1-420) and having a molecular mass of 46.0 kDa. 

Product Specs

Introduction
GSK3B, a member of the glycogen synthase kinase family, is a serine-threonine kinase that plays a role in energy metabolism, neuronal cell development, and body pattern formation. Genetic variations in the GSK3B gene have been linked to an altered risk of Parkinson's disease. Furthermore, research using mice suggests that elevated GSK3B gene expression may contribute to the development of Alzheimer's disease.
Description
Recombinant human GSK3B, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 420 amino acids (residues 1-420) with a molecular weight of 46.0 kDa.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The GSK3B solution, provided at a concentration of 0.5 mg/ml, is prepared in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.4 M urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the solution can be kept at 4°C. For extended storage, freeze the solution at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity greater than 85%.
Synonyms
GSK3B, Glycogen synthase kinase-3 beta, GSK-3 beta.
Source
Escherichia Coli.
Amino Acid Sequence
MSGRPRTTSF AESCKPVQQP SAFGSMKVSR DKDGSKVTTV VATPGQGPDR PQEVSYTDTK VIGNGSFGVV YQAKLCDSGE LVAIKKVLQD KRFKNRELQI MRKLDHCNIV RLRYFFYSSG EKKDEVYLNL VLDYVPETVY RVARHYSRAK QTLPVIYVKL YMYQLFRSLA YIHSFGICHR DIKPQNLLLD PDTAVLKLCD FGSAKQLVRG EPNVSYICSR YYRAPELIFG ATDYTSSIDV WSAGCVLAEL LLGQPIFPGD SGVDQLVEII KVLGTPTREQ IREMNPNYTE FKFPQIKAHP WTKVFRPRTP PEAIALCSRL LEYTPTARLT PLEACAHSFF DELRDPNVKL PNGRDTPALF NFTTQELSSN PPLATILIPP HARIQAAAST PTNATAASDA NTGDRGQTNN AASASASNST

Q&A

Where is GSK3β localized within human cells and how does this impact experimental design?

Contrary to traditional views of GSK3β as primarily a cytosolic protein, research has confirmed its presence in multiple cellular compartments including the nucleus and mitochondria. This subcellular distribution has been verified through both microscopy and immunoblot experiments . When designing experiments, this multi-compartmental localization requires careful consideration of cell fractionation techniques. Researchers should implement protocols that effectively separate cytosolic, nuclear, and mitochondrial fractions when attempting to quantify GSK3β activity in specific compartments. The compartment-specific functions of GSK3β may vary significantly, so experimental designs should account for these potential differences when assessing GSK3β-mediated signaling or when targeting the protein with inhibitors.

How do GSK3β phosphorylation sites regulate its activity and what methods best measure this regulation?

GSK3β activity is primarily regulated through inhibitory phosphorylation at Ser9, which creates a pseudosubstrate that blocks access to the active site. To effectively measure GSK3β activity status, researchers should employ phospho-specific antibodies that distinguish between total GSK3β and its phosphorylated forms. Western blotting with antibodies specific to phospho-Ser9-GSK3β provides a reliable measure of inhibitory phosphorylation. Additionally, in vitro kinase assays using purified GSK3β and known substrates (such as glycogen synthase peptides) can quantify actual enzymatic activity. For comprehensive analysis, researchers should consider both the phosphorylation status and the direct measurement of kinase activity, as post-translational modifications beyond Ser9 phosphorylation may also affect function.

What are the recommended qPCR primers and protocols for measuring GSK3B expression in human samples?

For qPCR analysis of human GSK3B, validated primer pairs targeting the reference sequence NM_002093 are commercially available. The recommended forward primer sequence is CCGACTAACACCACTGGAAGCT and the reverse sequence is AGGATGGTAGCCAGAGGTGGAT . When conducting qPCR analysis, follow this validated protocol:

  • Prepare reactions using lyophilized qSTAR qPCR primer mix reconstituted to 10 μM final concentration

  • Implement the following PCR program:

    • Stage 1: Activation at 50°C for 2 minutes

    • Stage 2: Pre-soak at 95°C for 10 minutes

    • Stage 3: 40 cycles of denaturation (95°C for 15 seconds) followed by annealing/extension (60°C for 1 minute)

    • Stage 4: Melting curve analysis (95°C for 15 seconds, 60°C for 15 seconds, 95°C for 15 seconds)

To ensure reliable results, always include appropriate reference genes for normalization (such as GAPDH, ACTB, or TBP) and validate primers with efficiency tests using serial dilutions of template cDNA .

How should researchers design robust in vivo studies to investigate GSK3β function while adhering to 3Rs principles?

Designing robust in vivo studies for GSK3β function requires careful planning that balances scientific rigor with ethical considerations. The NC3Rs Experimental Design Assistant (EDA) provides a free online platform to guide this process . When designing such studies, implement these key principles:

Additionally, apply the PREPARE guidelines during peer review of experimental designs and maintain consistent standards across both in vitro and in vivo studies . Document all procedural details comprehensively to ensure reproducibility and facilitate accurate reporting.

What are the technical challenges in developing specific GSK3β inhibitors for research purposes, and how can these be addressed?

Developing specific GSK3β inhibitors presents several technical challenges due to the high sequence homology between GSK3α and GSK3β catalytic domains (97%) and similarity with other kinases. Researchers can address these challenges through:

  • Structure-based design: Utilize machine learning approaches to identify compounds that interact with unique features of the GSK3β ATP-binding pocket. Recent research demonstrates that machine learning software can identify novel small-molecule treatments with increased specificity .

  • Allosteric targeting: Focus on identifying compounds that bind to regions outside the catalytic domain where sequence divergence between GSK3α and GSK3β is greater.

  • Selectivity screening: Implement comprehensive kinase panels to assess inhibitor specificity across >100 kinases, ensuring selectivity ratios (IC₅₀ GSK3β vs. other kinases) exceed 50-fold.

  • Cellular validation: Confirm inhibitor specificity through multiple approaches:

    • Phosphorylation status of known GSK3β substrates (e.g., glycogen synthase, β-catenin)

    • Phenotypic rescue experiments comparing inhibitor effects with GSK3β siRNA/shRNA knockdown

    • CRISPR-engineered cell lines with inhibitor-resistant GSK3β mutations

  • Pharmacokinetic optimization: Develop compounds with adequate tissue distribution, particularly considering blood-brain barrier penetration for neurological applications.

How does GSK3β contribute to insulin resistance mechanisms, and what experimental models best demonstrate this relationship?

GSK3β plays a complex role in insulin resistance mechanisms primarily through its inhibitory phosphorylation of glycogen synthase, a key enzyme in glucose storage. Research has revealed that GSK3β activity is abnormally elevated in tissues of diabetic and insulin-resistant individuals . To effectively study this relationship, researchers can implement these experimental models:

  • Cellular models:

    • Primary human skeletal muscle cells treated with palmitate to induce insulin resistance

    • 3T3-L1 adipocytes with siRNA-mediated GSK3β knockdown to assess insulin-stimulated glucose uptake

    • Human hepatocytes with selective GSK3β inhibitors to evaluate glycogen synthesis rates

  • Animal models:

    • Tissue-specific GSK3β knockout mice (using Cre-loxP technology) to differentiate the contributions of liver, muscle, and adipose GSK3β to whole-body insulin sensitivity

    • Diet-induced obesity models with subsequent GSK3β inhibitor treatment to evaluate potential therapeutic applications

  • Measurement approaches:

    • Hyperinsulinemic-euglycemic clamp studies to quantify insulin sensitivity following GSK3β manipulation

    • Metabolic tracer studies using labeled glucose to trace glycogen synthesis pathways

Importantly, when designing such studies, researchers should consider the complex interplay between GSK3α and GSK3β, as compensatory mechanisms may mask phenotypes in single-isoform manipulations . Species differences in GSK3β functions should also be considered when translating findings from animal models to human applications.

What is the evidence linking GSK3B genetic variants to mood disorders, and how should researchers approach replication studies?

Genetic studies have revealed significant associations between GSK3B polymorphisms and mood regulation. Specifically, the functional polymorphism GSK3B rs12630592 has been shown to interact with FXR1 rs496250 in regulating mood and emotional processing . In patients with mood disorders, the level of mania (in both acute and stabilized periods) and depression in stabilized periods was positively associated with GSK3B rs12630592 T allele, but only in carriers of the FXR1 rs496250 A allele . This gene-gene interaction explained approximately 11% of mania variance and 5% of interepisode depression variance.

For researchers designing replication studies, consider these methodological approaches:

  • Comprehensive phenotyping:

    • Use validated instruments like the Comprehensive Assessment of Symptoms and History (CASH) to derive symptom dimensions

    • Assess symptoms during both acute episodes and stabilized interepisode intervals

    • Distinguish between mania, depression, and psychotic dimensions

  • Statistical analysis:

    • Implement linear mixed effect models that include both main effects and interaction terms

    • Account for polygenic effects through appropriate random effect terms

    • Apply Bonferroni correction for multiple testing while ensuring sufficient power

  • Sample considerations:

    • Calculate required sample sizes based on the expected effect sizes (11% variance for mania, 5% for depression)

    • Include family-based designs where possible to control for population stratification

    • Consider different diagnostic categories (bipolar disorder, recurrent depression, schizophrenia) to test specificity of effects

The interaction between GSK3B and mood regulation appears specific to affective dimensions rather than psychotic symptoms, as the association was not observed in schizophrenia patients or with the psychotic dimension in mood disorder patients .

What mechanisms link GSK3β to neurodegenerative diseases, and what cellular assays best capture these relationships?

GSK3β has been implicated in multiple neurodegenerative disease processes, particularly through its role in tau hyperphosphorylation in Alzheimer's disease and through interactions with α-synuclein in Parkinson's disease . The enzyme acts as a phosphorylating agent for numerous substrates involved in neuronal function and survival. To effectively study these mechanisms, researchers should employ these cellular assays:

  • Tau phosphorylation assays:

    • In vitro kinase assays using recombinant GSK3β and tau protein to quantify phosphorylation at disease-relevant sites

    • Primary neuronal cultures treated with GSK3β inhibitors to assess changes in tau phosphorylation state

    • Western blotting with phospho-specific antibodies targeting GSK3β-dependent tau epitopes (e.g., Ser396/Ser404)

  • Neurotoxicity models:

    • SH-SY5Y neuroblastoma cells expressing wild-type or mutant GSK3β to assess differential vulnerability to oxidative stress

    • Primary neuronal cultures with GSK3β modulation (overexpression, knockdown, inhibition) subjected to amyloid-β or other disease-relevant stressors

    • Multi-electrode arrays to measure functional neuronal network activity following GSK3β manipulation

  • Protein aggregation assays:

    • Cell-based assays using fluorescently-tagged proteins (tau, α-synuclein) to monitor aggregation kinetics under GSK3β modulation

    • Thioflavin T fluorescence assays to quantify amyloid formation rates with purified proteins and active/inactive GSK3β

  • Mitochondrial function assessment:

    • Seahorse XF analysis to measure oxygen consumption rates in cells with altered GSK3β activity

    • JC-1 staining to assess mitochondrial membrane potential as a measure of mitochondrial health

These assays should be conducted with appropriate controls, including pharmacological inhibitors of varying specificity and genetic approaches to modulate GSK3β levels and activity .

How do GSK3β and GSK3α exhibit compensatory mechanisms in knockout models, and what techniques can distinguish their unique functions?

GSK3β and GSK3α demonstrate complex compensatory relationships in knockout models that can confound experimental interpretations. Studies have observed increased inactive phosphorylation of GSK3α in skeletal muscle with specific deletion of GSK3β, suggesting compensatory regulatory mechanisms . To effectively distinguish their unique functions while accounting for compensation:

  • Conditional and inducible knockout approaches:

    • Implement tamoxifen-inducible Cre-loxP systems to delete GSK3β in adult tissues, minimizing developmental compensation

    • Use tissue-specific promoters to target deletion to relevant cell types while preserving expression elsewhere

    • Compare acute (short-term) vs. chronic (long-term) deletion to identify compensatory adaptations

  • Isoform-specific molecular tools:

    • Develop highly selective inhibitors with >100-fold selectivity for one isoform over the other

    • Design isoform-specific CRISPR-Cas9 guide RNAs targeting non-homologous regions

    • Create isoform-specific antibodies targeting unique epitopes for selective immunoprecipitation

  • Analytical approaches to differentiate isoform contributions:

    • Implement phospho-proteomics to identify differential substrate preferences

    • Perform chromatin immunoprecipitation sequencing (ChIP-seq) to map isoform-specific interactions with genomic regions

    • Conduct interactome studies via BioID or proximity labeling to identify unique protein interaction partners

  • Rescue experiments with specificity controls:

    • Reintroduce wild-type or kinase-dead mutants of each isoform in double-knockout backgrounds

    • Use chimeric constructs with swapped domains to identify regions responsible for functional specificity

The complexity of GSK3 biology is evidenced by the fact that global deletion of GSK3α in mice causes cardiac hypertrophy, contractile dysfunction, and early mortality - pathological phenotypes associated with activated mTORC1 and suppressed autophagy . Similarly, GSK3β has essential roles in cardiac homeostasis under stress conditions . These findings highlight that the effects of GSK3 isoforms are tissue-specific, strain-dependent, and potentially species-dependent.

What are the current methodological approaches for studying GSK3β in mitochondrial function, and how can these be optimized?

GSK3β has been identified in mitochondria in addition to its cytosolic and nuclear localizations, suggesting specialized functions in this compartment . To effectively study mitochondrial GSK3β:

  • Mitochondrial fractionation and localization:

    • Implement differential centrifugation with density gradient purification to isolate intact mitochondria

    • Validate fractionation purity using compartment-specific markers (e.g., VDAC for outer membrane, COX IV for inner membrane)

    • Employ proteinase K protection assays to distinguish outer membrane-associated vs. matrix-localized GSK3β

    • Use super-resolution microscopy with dual-labeled antibodies to visualize GSK3β relative to mitochondrial structures

  • Functional assessment of mitochondrial GSK3β:

    • Measure respiratory capacity using Seahorse XF analyzers following mitochondria-targeted GSK3β interventions

    • Assess mitochondrial membrane potential with JC-1 or TMRM dyes under GSK3β modulation

    • Quantify mitochondrial ROS production using MitoSOX in response to GSK3β activity changes

    • Measure mitochondrial calcium handling with Rhod-2 fluorescence

  • Mitochondrial GSK3β substrate identification:

    • Perform mitochondrial phosphoproteomics comparing wild-type vs. GSK3β inhibition

    • Implement BioID with mitochondria-targeted GSK3β to identify proximal interacting proteins

    • Validate candidate substrates with in vitro kinase assays using purified mitochondrial proteins

  • Targeting strategies for mitochondrial GSK3β:

    • Develop mitochondria-targeted GSK3β inhibitors using TPP+ or MitoQ-like targeting moieties

    • Create mitochondria-targeted GSK3β via fusion with mitochondrial targeting sequences

    • Implement optogenetic approaches for spatiotemporal control of mitochondrial GSK3β activity

Each approach should include appropriate controls to ensure specificity, such as mitochondria-targeted kinase-dead GSK3β mutants and selective inhibitors with demonstrated mitochondrial penetration .

How do post-translational modifications beyond Ser9 phosphorylation regulate GSK3β, and what techniques best characterize these modifications?

While Ser9 phosphorylation is the most studied regulatory mechanism for GSK3β, multiple other post-translational modifications (PTMs) significantly influence its activity, localization, and substrate specificity. To comprehensively characterize these regulatory mechanisms:

  • Mass spectrometry-based PTM mapping:

    • Implement immunoprecipitation of endogenous GSK3β followed by LC-MS/MS analysis

    • Use enrichment strategies for specific modifications (e.g., TiO₂ for phosphopeptides, antibody-based enrichment for acetylation)

    • Apply quantitative approaches such as SILAC or TMT labeling to compare PTM profiles across conditions

    • Develop targeted parallel reaction monitoring (PRM) assays for known modification sites

  • Site-specific mutational analysis:

    • Generate point mutations at candidate modification sites (alanine substitutions to prevent modification, glutamate substitutions to mimic phosphorylation)

    • Assess functional consequences through in vitro kinase assays and cellular readouts

    • Create knock-in cell lines expressing only the mutant forms using CRISPR-Cas9 technology

  • Modification-specific antibodies and biosensors:

    • Develop antibodies specific to key modifications beyond Ser9 phosphorylation

    • Create FRET-based biosensors to monitor real-time changes in modification status

    • Implement proximity ligation assays to detect specific modified forms of GSK3β in situ

  • Investigating the writers, readers, and erasers of GSK3β modifications:

    • Identify kinases, phosphatases, acetyltransferases, and deacetylases that regulate GSK3β using screening approaches

    • Perform co-immunoprecipitation studies to validate direct enzyme-substrate relationships

    • Use selective inhibitors of modifying enzymes to assess effects on GSK3β function

Notable PTMs beyond Ser9 phosphorylation include acetylation, ubiquitination, SUMOylation, and additional phosphorylation events that collectively form a complex regulatory network controlling GSK3β function in different cellular contexts.

What are common sources of data inconsistency in GSK3β studies, and how can researchers address these challenges?

Researchers frequently encounter data inconsistencies in GSK3β studies due to several factors. Understanding and controlling these variables is crucial for generating reproducible results:

  • Isoform specificity issues:

    • Problem: Many commercial antibodies and inhibitors show cross-reactivity between GSK3α and GSK3β

    • Solution: Validate antibody specificity using isoform-specific knockouts or knockdowns; test inhibitor selectivity against both purified isoforms; when reporting results, clearly specify which isoform(s) were targeted

  • Context-dependent functions:

    • Problem: GSK3β effects vary by cell type, tissue, and physiological state

    • Solution: Standardize experimental conditions including cell density, passage number, and serum starvation protocols; perform experiments across multiple cell lines or primary cells from different donors; clearly document all contextual variables

  • Activation state measurement challenges:

    • Problem: Phospho-Ser9 levels don't always correlate with actual kinase activity

    • Solution: Combine phosphorylation assessment with direct kinase activity assays using validated substrates; monitor multiple readouts of GSK3β activity (e.g., β-catenin levels, glycogen synthase phosphorylation)

  • Genetic background effects:

    • Problem: Effects of GSK3β manipulation are sensitive to genetic background in animal models

    • Solution: Use consistent strain backgrounds; consider backcrossing to standardized backgrounds; include appropriate genetic controls; when possible, test effects across multiple strains

  • Baseline activity variations:

    • Problem: Basal GSK3β activity fluctuates with cell cycle, metabolic state, and stress levels

    • Solution: Synchronize cells before experiments; standardize feeding/fasting protocols for animal studies; monitor and control for stress indicators

Implementing the EQIPD (European Quality in Preclinical Data) Quality Management System can further reduce inconsistencies through standardized protocols, comprehensive documentation, and rigorous statistical analysis .

How should researchers interpret conflicting results between in vitro and in vivo studies of GSK3β function?

Discrepancies between in vitro and in vivo GSK3β studies are common and present significant interpretative challenges. To navigate these conflicts effectively:

  • Systematic reconciliation approach:

    • Create a comprehensive comparison table listing experimental conditions, GSK3β assessment methods, endpoints, and results from both systems

    • Identify specific variables that differ between systems (concentration/dose, exposure duration, presence of compensatory mechanisms)

    • Design bridging experiments that systematically vary one parameter at a time to identify critical differences

  • Physiological context considerations:

    • In vitro limitations: Isolated systems lack feedback mechanisms, inter-tissue crosstalk, and physiological fluctuations

    • In vivo complexities: Multiple inputs regulate GSK3β activity including hormones, nutrients, and neural signals

    • Solution: Develop more physiologically relevant in vitro systems (co-cultures, organoids, perfused systems) that better recapitulate in vivo complexity

  • Pharmacological vs. genetic manipulation differences:

    • Problem: Acute inhibition (pharmacological) often yields different results than chronic inhibition (genetic)

    • Solution: Compare acute vs. chronic treatments in both systems; use inducible genetic systems to better mimic pharmacological timing

  • Dose/concentration relevance:

    • Problem: In vitro studies often use inhibitor concentrations far exceeding achievable in vivo levels

    • Solution: Include pharmacokinetic/pharmacodynamic analyses to ensure in vitro concentrations reflect achievable tissue levels; test multiple concentrations spanning physiologically relevant ranges

  • Integrative analysis frameworks:

    • Implement computational modeling approaches that incorporate data from both systems

    • Use pathway analysis tools to identify compensatory mechanisms activated in vivo but absent in vitro

    • Consider systems biology approaches to map broader network effects

When publishing results, explicitly discuss discrepancies between systems and propose testable hypotheses to explain the differences, rather than simply favoring one system over another .

What quality control measures should be implemented when using GSK3β inhibitors in research applications?

Implementing rigorous quality control measures for GSK3β inhibitors is essential for generating reliable research data. Researchers should establish the following comprehensive quality control framework:

  • Inhibitor characterization:

    • Verify chemical identity and purity (>95%) using NMR, mass spectrometry, and HPLC

    • Determine aqueous solubility and stability under experimental conditions

    • Establish dose-response curves against purified GSK3β protein with appropriate positive controls

    • Quantify selectivity by screening against a panel of related kinases, particularly GSK3α

  • Cellular target engagement validation:

    • Confirm target engagement using cellular thermal shift assays (CETSA)

    • Monitor phosphorylation status of direct GSK3β substrates (e.g., glycogen synthase, β-catenin)

    • Implement orthogonal approaches: compare effects with genetic knockdown/knockout models

    • Use inactive analogs or structurally distinct inhibitors with similar selectivity profiles as controls

  • Experimental design considerations:

    • Include concentration-response relationships spanning at least 2 orders of magnitude

    • Establish appropriate treatment durations based on inhibitor pharmacokinetics

    • Include positive controls (known GSK3β modulators) and negative controls

    • Document batch information and storage conditions

  • Biological validation:

    • Confirm expected functional consequences in well-characterized biological systems

    • Validate efficacy across multiple cell types/tissues where GSK3β function has been established

    • Assess cellular toxicity profiles to distinguish specific GSK3β inhibition from general cytotoxicity

  • Reporting standards:

    • Provide complete methodological details including inhibitor source, catalog number, lot, concentration/dose calculation methods

    • Disclose all quality control data including selectivity profiles

    • Include raw data representations rather than only normalized results

    • Document any observed off-target effects

These quality control measures align with the principles outlined in the Global Good Statistical Practice standards and should be comprehensively documented to ensure experimental reproducibility .

How can machine learning approaches advance the discovery of GSK3β inhibitors, and what datasets are most valuable for training such models?

Machine learning (ML) offers significant potential for accelerating the discovery of novel GSK3β inhibitors with improved specificity and efficacy. Recent research has demonstrated that ML software can identify novel small-molecule treatments for conditions like Alzheimer's disease by targeting GSK3β . To effectively implement ML approaches:

  • Optimal training datasets:

    • Structure-activity relationship (SAR) data from diverse chemical scaffolds tested against GSK3β

    • Structural data including co-crystal structures of GSK3β with inhibitors at various binding sites

    • Selectivity profiles against related kinases, particularly GSK3α

    • Pharmacokinetic/pharmacodynamic relationships for existing GSK3β inhibitors

    • Phenotypic screening results linked to compound structures

  • ML algorithm selection and implementation:

    • Deep neural networks for complex structure-activity relationship modeling

    • Graph convolutional networks for analyzing molecular structures

    • Reinforcement learning approaches for multi-parameter optimization

    • Transfer learning from related kinase inhibitor datasets to overcome limited GSK3β-specific data

  • Model validation strategies:

    • Implement cross-validation using temporally separated data (train on older compounds, validate on newer ones)

    • Conduct prospective validation through synthesis and testing of ML-predicted compounds

    • Compare ML predictions against experimentally determined binding modes via X-ray crystallography

    • Validate across multiple cell types and assay systems

  • Integration with other computational approaches:

    • Combine ML predictions with physics-based molecular dynamics simulations

    • Implement ML-guided docking for binding mode predictions

    • Use quantum mechanical calculations to refine ML predictions of binding energetics

By integrating diverse datasets including structural information, bioactivity data, and selectivity profiles, ML approaches can identify novel chemical scaffolds that selectively target GSK3β with reduced off-target effects . This interdisciplinary approach represents a promising direction for developing next-generation GSK3β modulators for both research and therapeutic applications.

What emerging technologies show promise for studying GSK3β protein interactions and signaling dynamics in real-time?

Several cutting-edge technologies are transforming our ability to study GSK3β interactions and signaling dynamics with unprecedented temporal and spatial resolution:

  • Advanced imaging approaches:

    • FRET/BRET biosensors: Genetically encoded sensors that report on GSK3β activity or substrate phosphorylation in living cells with real-time resolution

    • Optogenetic GSK3β tools: Light-controlled activation or inhibition of GSK3β with subcellular precision

    • Super-resolution microscopy: Techniques like PALM, STORM, or STED to visualize GSK3β localization and interactions below the diffraction limit

    • Lattice light-sheet microscopy: For long-term 3D imaging of GSK3β dynamics with minimal phototoxicity

  • Proximity labeling technologies:

    • TurboID/miniTurboID: Engineered biotin ligases fused to GSK3β to identify proximal proteins with temporal control

    • APEX2: Engineered peroxidase for electron microscopy-compatible proximity labeling of GSK3β interaction partners

    • Split-BioID: For detecting protein-protein interactions involving GSK3β in specific cellular compartments

  • Single-cell technologies:

    • Single-cell phosphoproteomics: To analyze GSK3β-mediated phosphorylation events at single-cell resolution

    • Single-cell RNA-seq: To correlate GSK3β activity with transcriptional responses

    • Mass cytometry (CyTOF): For simultaneous detection of multiple GSK3β pathway components across heterogeneous cell populations

  • Protein engineering approaches:

    • Kinase activity reporters: Engineered substrates that change localization or fluorescence properties upon phosphorylation by GSK3β

    • Degron-based sensors: Systems where protein stability is coupled to GSK3β activity

    • Nanobodies: Small antibody fragments for tracking endogenous GSK3β with minimal perturbation

  • Microfluidic systems:

    • Organ-on-a-chip platforms: For studying GSK3β function in physiologically relevant multicellular contexts

    • Droplet microfluidics: To perform high-throughput single-cell analysis of GSK3β signaling

These technologies enable researchers to move beyond static snapshots of GSK3β signaling to dynamic, spatiotemporally resolved understanding of its functions in complex cellular contexts, potentially revealing new therapeutic opportunities and biological insights.

How might GSK3β research inform personalized medicine approaches for diseases with heterogeneous molecular pathology?

GSK3β research has significant potential to advance personalized medicine strategies across multiple disease contexts, particularly given its involvement in diverse signaling pathways and disease mechanisms:

  • Genetic stratification approaches:

    • Identify patient subgroups based on GSK3B polymorphisms (such as rs12630592) that predict differential disease progression or treatment response

    • Develop targeted therapies for patients with specific GSK3B-related genetic profiles

    • Implement companion diagnostics that assess GSK3B genetic variants alongside other interacting genes (like FXR1 rs496250) to predict treatment efficacy

  • Pathway-based biomarker development:

    • Create multi-parameter biomarker panels assessing GSK3β activity status alongside related pathway components

    • Develop tissue-specific GSK3β activity assays to guide treatment selection

    • Implement longitudinal monitoring of GSK3β-regulated substrates to track treatment response

  • Systems biology integration:

    • Generate computational models integrating patient-specific GSK3β pathway alterations with broader -omics data

    • Predict individual patient responses to GSK3β-targeting therapies based on personalized network models

    • Identify optimal combination therapies that address individual GSK3β dysregulation patterns

  • Targeting tissue-specific GSK3β functions:

    • Develop tissue-selective GSK3β modulators that address pathology in affected tissues while sparing normal GSK3β function elsewhere

    • Implement patient-specific organoid models to test GSK3β inhibitor efficacy on patient-derived cells before in vivo treatment

    • Consider disease stage-specific interventions based on temporal changes in GSK3β signaling during disease progression

  • Translation to clinical applications:

    • Design clinical trials with patient stratification based on GSK3B genotypes and GSK3β activity biomarkers

    • Develop dosing algorithms that account for individual variations in GSK3β baseline activity

    • Create decision support tools that integrate GSK3β pathway status with other clinical factors

The interaction between GSK3B rs12630592 and FXR1 rs496250 in mood disorders provides a compelling example of how genetic information could inform personalized treatment approaches . Similarly, the varied roles of GSK3β in different tissues suggest that pathway-specific targeting could minimize side effects while maximizing therapeutic efficacy in conditions ranging from diabetes to neurodegenerative diseases .

Product Science Overview

Introduction

Glycogen Synthase Kinase-3 Beta (GSK3β) 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 (GSK3), the other being GSK3α. GSK3β is highly conserved and is involved in numerous signaling pathways that regulate essential cellular functions.

Structure and Isoforms

GSK3 exists in two major isoforms: GSK3α and GSK3β. These isoforms are encoded by two distinct genes, GSK3A and GSK3B, respectively. Despite their high homology within the kinase domains (approximately 98% identity), they have different functions. GSK3β is particularly important for its role in various signaling pathways and cellular processes .

Functions and Significance

GSK3β is involved in a wide range of cellular functions, including:

  • Glycogen Metabolism: It regulates glycogen synthesis by phosphorylating and inhibiting glycogen synthase.
  • Cell Cycle Control: GSK3β influences cell proliferation and differentiation.
  • Apoptosis: It plays a role in programmed cell death.
  • Embryonic Development: GSK3β is crucial for proper development during the embryonic stages.
  • Cell Differentiation and Motility: It affects how cells differentiate and move.
  • Microtubule Function: GSK3β is involved in the regulation of microtubules, which are essential for cell structure and transport.
  • Inflammation: It modulates inflammatory responses, particularly through the regulation of nuclear factor-kappaB (NF-κB) activity .
Role in Disease and Therapeutic Potential

GSK3β has been implicated in various diseases, including:

  • Diabetes: As a regulator of glucose homeostasis, GSK3β is a target for diabetes treatment.
  • Neurodegenerative Diseases: It is involved in the pathogenesis of diseases like Alzheimer’s due to its role in tau phosphorylation.
  • Cancer: GSK3β can act as both a tumor suppressor and promoter, depending on the context.
  • Inflammatory Diseases: Its role in modulating inflammation makes it a potential target for treating inflammatory conditions .
Recombinant GSK3β

Human recombinant GSK3β is produced using recombinant DNA technology, which allows for the production of large quantities of the protein for research and therapeutic purposes. This recombinant form retains the functional properties of the native protein and is used in various studies to understand its role in cellular processes and disease mechanisms .

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 2024 Thebiotek. All Rights Reserved.