Fact-checked by Grok 2 weeks ago

Glycogen synthase kinase-3 beta

Glycogen synthase kinase-3 beta (GSK-3β) is a constitutively active serine/ encoded by the GSK3B gene on human chromosome 3q13, belonging to the family alongside the isoform GSK-3α. Originally discovered in the for its ability to phosphorylate and inhibit —the rate-limiting in —GSK-3β plays a central role in glucose and insulin signaling by modulating metabolic pathways in response to nutrient availability. As a highly conserved present in all eukaryotes, it is ubiquitously expressed across tissues, with a molecular weight of approximately 47 kDa, and localizes to the , , and mitochondria to influence a broad array of cellular functions beyond . Structurally, GSK-3β features a catalytic kinase domain that shares 97% sequence identity with GSK-3α, enabling it to recognize and phosphorylate over 100 substrates, many of which require prior priming phosphorylation at a motif four residues C-terminal to the target serine or threonine (consensus: S/T-X-X-X-S/T(P)). The enzyme's N-terminal and C-terminal regions differ from GSK-3α, conferring isoform-specific functions, such as GSK-3β's essential role in embryonic development—knockout studies in mice demonstrate that GSK-3β deficiency is embryonic lethal, with no compensation by GSK-3α. Alternative splicing produces isoforms of GSK-3β that localize to specific cellular compartments, like growing neurites in neurons, highlighting its adaptability in specialized contexts. In addition to glycogen regulation, GSK-3β is a key integrator of multiple signaling cascades, including the Wnt/β-catenin pathway—where it phosphorylates β-catenin for degradation, thereby suppressing and gene transcription—and the insulin/PI3K/AKT pathway, which inhibits GSK-3β to promote anabolic processes like protein synthesis via . It also modulates neuronal functions, such as through AMPA receptor trafficking, and influences , , and mitochondrial by phosphorylating substrates like CREB, , and components of the . Dysregulation of GSK-3β activity is implicated in diverse pathologies: hyperactivation contributes to neurodegenerative diseases like Alzheimer's (via tau hyperphosphorylation) and (via impaired insulin sensitivity), while inhibition underlies aspects of and certain cancers, positioning GSK-3β as a promising yet challenging therapeutic target with inhibitors like and under investigation.

Molecular Structure and Expression

Protein Domains and Isoforms

Glycogen synthase kinase-3 beta (GSK-3β) is a serine/threonine encoded by the GSK3B gene, with the canonical human isoform consisting of 420 , though an alternative splice variant (GSK-3β2) results in a 433- protein containing a 13- insertion within the (around residues 296-308 in the longer form). The protein features a modular structure comprising an N-terminal (residues 1-134), a central (residues 135-343), and a C-terminal regulatory tail (residues 344-433). The adopts a bilobal fold typical of eukaryotic protein kinases, with an N-lobe containing a glycine-rich loop (residues 62-67) involved in ATP binding and a C-lobe that includes the activation loop (residues 200-216). A conserved lysine residue at position 85 (Lys85) within the is crucial for coordinating the phosphate groups of ATP, enabling nucleophilic attack by the substrate's hydroxyl group during . Structural insights from , such as the PDB entry 1J1B depicting GSK-3β bound to AMPPNP and Mg²⁺, reveal the bilobal architecture with a cleft between the lobes forming the ; the glycine-rich loop flexes to accommodate ATP, while the activation loop in its active conformation (autophosphorylated at Tyr216) positions catalytic residues for substrate phosphorylation. The N-terminal domain forms a β-barrel that contributes to substrate specificity, particularly for primed substrates, and the C-terminal tail modulates autoinhibition and interactions with regulatory proteins. These domains are evolutionarily conserved, with GSK3B orthologs found across eukaryotes from to mammals, reflecting its fundamental role in cellular signaling. The alternative GSK-3β2 isoform is predominantly expressed in neuronal tissues and localizes to growing neurites, contributing to growth and neuronal development. GSK-3β shares significant structural similarity with its paralog GSK-3α, encoded by the GSK3A gene on ; the two isoforms exhibit 98% identity within their domains but diverge in their N- and C-termini, with only 36% identity in the C-terminal 76 residues. Notably, GSK-3α possesses a glycine-rich N-terminal extension of 36 absent in GSK-3β, which may influence isoform-specific regulation and localization. While both isoforms are ubiquitously expressed, GSK-3β demonstrates broader tissue distribution, including higher levels in brain, heart, and compared to the more neuron-enriched GSK-3α. The GSK3B gene is located on human chromosome 3q13.33, spanning approximately 273 kb with 12 exons.

Gene Location and Tissue Expression

The GSK3B gene, encoding glycogen synthase kinase-3 beta, is located on the long arm of chromosome 3 at cytogenetic band 3q13.33, spanning approximately 273 kilobases from position 119,821,321 to 120,094,447 (GRCh38.p14 assembly). The gene consists of 12 exons, with the coding sequence distributed across these exons to produce a primary transcript that undergoes processing to yield the mature mRNA. Transcriptional regulation of GSK3B is mediated by binding sites for key transcription factors in its promoter and upstream regions, including and AP-1, which respond to inflammatory and stress-related signals to modulate expression levels. Alternative splicing of the GSK3B pre-mRNA generates multiple transcript variants, with two primary isoforms identified in human tissues: the canonical isoform (420 ) and the alternative isoform GSK-3β2 (433 ) containing a 13-amino-acid insertion in the catalytic domain, though over 70 transcripts have been annotated, most representing minor variants with limited functional divergence. GSK3B exhibits ubiquitous expression across human tissues at the mRNA and protein levels, reflecting its broad role in cellular signaling, but with notable variations in abundance. Highest levels are observed in the central nervous system, particularly in neurons where cytoplasmic expression predominates, exceeding that in astrocytes, while substantial expression also occurs in testis; in contrast, expression is lower in skeletal muscle and notably reduced in pancreas compared to neural tissues. Moderate expression is detected in heart and other organs, supporting its involvement in metabolic and developmental processes. During development, GSK3B expression is upregulated in the embryonic , peaking from late embryonic stages through early postnatal periods to facilitate , neuronal , and polarity establishment. Quantitative analysis from the GTEx database reveals mRNA levels (measured in transcripts per million, TPM) are 2- to 5-fold higher in neural tissues such as (median ~20-30 TPM) and compared to liver (median ~5-10 TPM), underscoring its enriched role in brain function.

Enzymatic Activity and Substrates

Kinase Mechanism

Glycogen synthase kinase-3 beta (GSK-3β) is a classified under EC 2.7.11.26. It catalyzes the transfer of the γ-phosphate from ATP to the hydroxyl groups of serine or residues on target proteins through a Mg²⁺-dependent mechanism: first, the enzyme binds ATP and Mg²⁺ to form a complex, followed by nucleophilic attack by the substrate's hydroxyl group on the γ-phosphate, resulting in phosphotransfer and ADP release. A hallmark of GSK-3β's catalytic specificity is its reliance on primed substrates, where prior by another at the +4 position (N+4) relative to the target residue is typically required for optimal activity. This preference is dictated by the consensus motif Ser/Thr-XXX-Ser/Thr, with the primed group at the C-terminal Ser/Thr interacting with a basic pocket in the enzyme's to properly orient the . Although GSK-3β exhibits low basal activity toward unprimed substrates, it can phosphorylate select targets without priming, such as β-catenin at specific sites. Kinetic analyses indicate that GSK-3β has a high for ATP, with a Michaelis constant (Km) of approximately 0.7–1 μM, allowing efficient at physiological concentrations. The maximum velocity (Vmax) is influenced by inhibitors that compete for the ATP-binding site or alter access. structures of GSK-3β, resolved at resolutions up to 2.1 Å, demonstrate that the backbone binds along a deep hydrophobic groove in the C-terminal lobe, while the primed phosphate coordinates with conserved basic residues (Arg96, Arg180, Lys205) in an adjacent pocket, stabilizing the . GSK-3β maintains constitutive activity without obligatory of its activation loop at Tyr216, unlike many kinases, though Tyr216 modestly enhances Vmax. Allosteric features include an intrinsic auto-inhibitory conformation where a segment near the activation loop partially occludes the substrate site in the apo form; substrate priming allosterically relieves this by engaging the P+4 phosphate pocket, promoting domain closure and efficient catalysis.00374-9)

Primary Substrates and Phosphorylation Sites

Glycogen synthase kinase-3 beta (GSK-3β) phosphorylates over 100 known substrates across diverse cellular processes, with proteomic studies suggesting up to 500 potential targets based on motif predictions. These substrates are typically recognized through specific consensus motifs, where GSK-3β preferentially acts on or residues. The enzyme exhibits a unique "primed" specificity for most substrates, requiring prior by another at a position four residues C-terminal to the GSK-3β target site (S/T-X-X-X-pS/pT, where X is any and pS/pT denotes /phosphothreonine). In contrast, non-primed sites are less common and often involve direct at Ser/Thr-Pro motifs or sites with an acidic residue immediately C-terminal, allowing GSK-3β to function without upstream priming. Mass spectrometry-based phosphoproteomics has identified novel targets, expanding the substrate repertoire beyond classical examples. The functional consequences of GSK-3β phosphorylation are predominantly inhibitory or promotive of , thereby modulating protein stability, activity, or localization, though rare cases promote nuclear export without degradation. Core substrates illustrate this breadth:
SubstrateKey Phosphorylation SitesPriming RequirementFunctional Outcome
Ser641 (also Ser652, Ser648, Ser644, Ser640)Primed (e.g., by CKII at Ser656)Inhibits enzymatic activity, reducing synthesis and storage.
β-CateninN-terminal Ser33, Ser37, Thr41Primed (by CK1 at Ser45)Marks for ubiquitination and proteasomal degradation, preventing transcriptional activation.
Ser396 (among 9 sites, e.g., Ser202, Thr205, Ser396, Ser404)Mixed (some primed, others non-primed)Promotes hyper, detachment from , and aggregation into neurofibrillary tangles.
Thr286Non-primedInduces nuclear export and proteasomal degradation, halting cell cycle progression at G1/S.
c-MycThr58 (primed at Thr62)Primed (by ERK1/2)Accelerates turnover via ubiquitination, limiting oncogenic transcriptional activity.
Mcl-1Ser159Primed (by JNK at Thr163)Facilitates ubiquitination and , sensitizing cells to .
IRS-1Ser302, Ser318PrimedInhibits signaling by promoting IRS-1 degradation and blocking PI3K activation, as identified in studies.
NFATSer259 (in SRR domain)PrimedTriggers nuclear export and inhibits DNA binding/transcriptional activity, an example of localization control without degradation.
These examples highlight GSK-3β's role as a central regulator, where often serves as a "priming" event in hierarchical signaling cascades, enabling precise control over substrate fate.

Regulation of Activity

Phosphorylation-Dependent Control

The activity of glycogen synthase kinase-3 beta (GSK-3β) is primarily regulated through events that either inhibit or activate its catalytic function. The most well-characterized inhibitory modification occurs at serine 9 (Ser9), where by upstream kinases such as (PKB/Akt), (PKA), and (PKC) significantly inhibits GSK-3β activity. This introduces a negative charge at the N-terminal extension, which acts as a pseudosubstrate that competitively blocks access to the enzyme's , particularly for primed substrates that require a prior four residues C-terminal to the GSK-3β target site. As a result, Ser9-phosphorylated GSK-3β exhibits dramatically diminished kinase activity toward its physiological substrates, such as glycogen synthase and β-catenin. In contrast, at 216 (Tyr216) in the activation loop of GSK-3β promotes its enzymatic activity, particularly in a manner that supports basal of non-primed substrates. This site is often autophosphorylated in the mature enzyme, but external kinases including and proline-rich 2 (PYK2) can also mediate Tyr216 , enhancing catalytic efficiency by stabilizing the active conformation of the kinase domain. Tyr216 is essential for GSK-3β's intrinsic activity independent of priming mechanisms, allowing it to engage certain substrates even in the absence of upstream priming kinases, though its impact is more pronounced on non-primed motifs compared to fully primed ones. Dephosphorylation of GSK-3β at Ser9 by protein phosphatases such as PP2A and PP1 restores its full catalytic activity, counteracting inhibitory signals and enabling rapid reactivation in response to cellular needs. PP2A preferentially targets GSK-3α but also dephosphorylates Ser9 on GSK-3β, while PP1 more specifically acts on the β isoform, with both phosphatases facilitating the removal of the inhibitory group through direct interaction with the . The of this allow for dynamic toggling of GSK-3β activity, with Ser9 removal leading to near-complete restoration of kinase function within physiological timescales. These phosphorylation events are integrated into broader signaling cascades that modulate GSK-3β in response to extracellular cues. In the insulin/PI3K pathway, insulin stimulation activates PI3K, which in turn recruits and activates Akt/PKB to phosphorylate Ser9, thereby inhibiting GSK-3β and promoting glycogen synthesis. Similarly, in the Wnt signaling pathway, activation leads to Dishevelled-mediated dissociation of the Axin-GSK-3β complex, indirectly reducing GSK-3β's access to β-catenin and mimicking inhibitory effects without direct Ser9 phosphorylation.

Subcellular Localization and Other Modifiers

Glycogen synthase kinase-3 beta (GSK-3β) exhibits a primarily cytoplasmic localization in unstressed, proliferating cells, where it maintains constitutive activity toward various substrates. Upon cellular stress, such as DNA damage or oxidative insult, GSK-3β translocates to the nucleus via its bipartite nuclear localization sequence, enabling it to phosphorylate nuclear targets like p53 and promote apoptotic gene expression. In the context of Wnt pathway activation, GSK-3β undergoes cytoplasmic sequestration into multivesicular bodies, spatially restricting its access to cytosolic substrates like β-catenin. During apoptosis, GSK-3β associates with mitochondria in neuronal and other cell types, where its activation facilitates Bax translocation, cytochrome c release, and caspase activation, exacerbating cell death. Scaffold-mediated sequestration represents a key non-covalent regulatory mechanism for GSK-3β. Binding to the scaffold protein Axin within the β-catenin destruction complex positions GSK-3β proximal to primed substrates, enhancing phosphorylation efficiency by over 20,000-fold through multimeric assembly with APC and casein kinase I. Conversely, binding to FRAT (GSK-3 binding protein) competes for the same hydrophobic groove on GSK-3β's C-terminal domain, displacing it from the Axin complex and inhibiting its activity toward β-catenin by blocking substrate access without altering intrinsic kinase catalysis. Allosteric and redox-based modifiers provide additional layers of indirect control over GSK-3β activity. Small molecules such as AR-A014418 bind within the ATP-competitive pocket of the domain, selectively inhibiting GSK-3β (IC50 = 104 nM) and mimicking the pathway-suppressive effects of Ser9 by preventing substrate binding in Wnt and insulin signaling contexts. Feedback loops and environmental modifiers fine-tune GSK-3β function. Autophosphorylation at Tyr216 in the loop constitutively enhances activity approximately fivefold, independent of upstream signals.

Involvement in Cellular Signaling Pathways

Wnt/β-Catenin Pathway

In the canonical Wnt/β-catenin signaling pathway, glycogen synthase kinase-3 beta (GSK-3β) functions as a central negative regulator by participating in the β-catenin destruction complex, which comprises Axin, (APC), and (CK1). In the absence of Wnt ligands, this multiprotein complex facilitates the of β-catenin at specific N-terminal residues—serine 33 (Ser33), serine 37 (Ser37), and 41 (Thr41)—following a priming at Ser45 by CK1. These events create a recognition motif for the E3 β-TrCP, leading to β-catenin polyubiquitination and subsequent proteasomal degradation, thereby preventing its accumulation and nuclear translocation. This process maintains low levels of cytosolic and nuclear β-catenin, suppressing transcription of Wnt target genes. Upon activation by Wnt ligands binding to receptors and co-receptors /6, the pathway undergoes a profound reconfiguration. (Dvl) is recruited to the plasma membrane, where it promotes the translocation of Axin to the receptor complex, resulting in disassembly of the destruction complex and inhibition of GSK-3β's phosphorylative activity toward β-catenin. Consequently, β-catenin evades degradation, accumulates in the , translocates to the , and interacts with TCF/LEF transcription factors to activate expression of target genes involved in , , and . This inhibition of GSK-3β occurs through multiple mechanisms, including sequestration of the kinase away from β-catenin and direct interference at the complex level, ensuring rapid . Beyond its role in β-catenin regulation, GSK-3β contributes to Wnt signal amplification via non-canonical actions at the receptor level. Specifically, GSK-3β phosphorylates the intracellular domain of at residues such as serine 1490 (Ser1490), which enhances Axin recruitment to the membrane-bound receptor complex and facilitates disassembly of the destruction complex. This phosphorylation event is essential for propagating the , highlighting GSK-3β's dual functionality in both suppressing and promoting pathway activity depending on subcellular context. Dysregulation of GSK-3β in the Wnt pathway has profound implications for development and disease. Hyperactive GSK-3β, often due to reduced inhibitory , excessively degrades β-catenin, suppressing Wnt signaling and impairing processes such as embryonic patterning and . In , mutations in destruction complex components like or β-catenin itself render GSK-3β's phosphorylative activity ineffective, leading to β-catenin stabilization, constitutive pathway activation, and tumorigenesis through unchecked proliferation of intestinal stem cells. These alterations underscore GSK-3β's critical role in maintaining pathway fidelity.

PI3K/Akt and Insulin Signaling

In the PI3K/Akt branch of insulin signaling, binding of insulin to its receptor activates the receptor tyrosine kinase, leading to recruitment and activation of phosphoinositide 3-kinase (PI3K). PI3K phosphorylates phosphatidylinositol-4,5-bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3), which recruits protein kinase B (PKB/Akt) to the plasma membrane via its pleckstrin homology domain. Activated Akt then phosphorylates GSK-3β at serine 9 (Ser9), resulting in its inhibition. Inhibition of GSK-3β by Akt-mediated relieves its suppressive effects on downstream targets, including (GS). Active GSK-3β normally GS at sites such as Ser641 and Ser645, maintaining GS in an inactive state; upon GSK-3β inhibition, these sites undergo dephosphorylation by protein phosphatase 1 (PP1), activating GS and promoting glycogen synthesis in response to insulin. Additionally, GSK-3β contributes to a loop by insulin receptor substrate-1 (IRS-1) on serine residues, such as Ser332 (with priming at Ser336), which attenuates IRS-1 phosphorylation and impairs insulin signal propagation. Physiologically, GSK-3β inhibition in insulin-sensitive tissues like liver and skeletal muscle enhances glucose uptake via GLUT4 translocation (indirectly through pathway crosstalk) and stimulates hepatic glycogen storage, thereby lowering blood glucose levels. Dysregulation of this pathway, characterized by elevated GSK-3β activity and reduced Ser9 phosphorylation, contributes to insulin resistance in type 2 diabetes; for instance, skeletal muscle from type 2 diabetic patients exhibits approximately twofold higher GSK-3β activity, correlating with impaired GS activation and reduced insulin-stimulated glucose disposal. Liver-specific GSK-3β knockout models demonstrate improved glucose tolerance and insulin sensitivity, underscoring its role in metabolic homeostasis.

Additional Pathways

GSK-3β integrates into the (Hh) signaling pathway by transcription factors, particularly GLI2 and GLI3, which promotes their processing into repressor forms in the absence of Hh ligands. This occurs sequentially with (PKA) and (CK1), marking GLI proteins for partial proteolysis and inhibiting their activator function to suppress target . Upon Hh ligand binding to Patched (PTCH), (SMO) activation inhibits the kinase complex including GSK-3β, preventing GLI and allowing full-length GLI activators to translocate to the . In the NF-κB pathway, GSK-3β positively regulates signaling by phosphorylating components of the (IKK) complex, particularly IKKγ/NEMO, which enhances and subsequent ubiquitination-dependent degradation. This promotes nuclear translocation and transcriptional activation of pro-inflammatory genes. TNF-α signaling can indirectly inhibit GSK-3β activity through Akt-mediated at Ser9, thereby modulating output in inflammatory contexts. GSK-3β contributes to through direct of at Ser315 and Ser376, which stabilizes , enhances its transcriptional activity, and facilitates its mitochondrial translocation to induce pro-apoptotic gene expression. Additionally, GSK-3β the anti-apoptotic protein Mcl-1, creating a recognition site for β-TrCP , leading to Mcl-1 proteasomal degradation and sensitization of cells to mitochondrial outer membrane permeabilization. Under cellular stress, GSK-3β translocates to mitochondria in a kinase-dependent manner, interacting with voltage-dependent anion channel 2 (VDAC2) to amplify release and activation. In regulation, GSK-3β phosphorylates the clock protein PER2, influencing its stability and nucleocytoplasmic shuttling to fine-tune the timing of clock gene oscillations. This phosphorylation, often in concert with CK1 and AMPK, promotes PER2 degradation during specific phases, thereby shortening or modulating the in response to metabolic cues.

Protein-Protein Interactions

Core Binding Partners

Glycogen synthase kinase-3 beta (GSK-3β) engages in direct interactions with several key proteins, primarily through specific domains on its structure, including the C-terminal helical region for and the N-terminal extension for other associations. One of the primary interactors is Axin, which binds to the C-terminal domain of GSK-3β (residues 262–299) via its GSK-3 interaction domain (GID), an amphipathic α-helix encompassing residues 383–401 of Axin. This interaction has a dissociation constant (Kd) of approximately 65 nM, as determined by GST pull-down assays, and facilitates the of the β-catenin destruction complex. β-Catenin serves as a direct substrate that binds transiently to GSK-3β, primarily following priming by CK1 at Ser45, which enhances affinity for subsequent GSK-3β phosphorylation sites (Ser33/37/Thr41); this interaction is low-affinity without scaffolding and is confirmed by co-immunoprecipitation in cellular extracts. FRAT (also known as GBP) binds to the same C-terminal hydrophobic channel on GSK-3β as Axin, utilizing a bipartite helical motif (FRATtide residues) that competes with Axin for binding, thereby inhibiting GSK-3β access to primed substrates in the absence of Wnt signaling; structural studies reveal overlapping interfaces with conformational plasticity in the GSK-3β loop (residues 285–299). Protein phosphatase 2A (PP2A) interacts directly with GSK-3β to the inhibitory Ser9 site, with enhanced association observed upon C-terminal truncation of GSK-3β, as shown in co-immunoprecipitation and assays; this modulates GSK-3β autoinhibition without requiring activity. p53 forms a complex with GSK-3β through the N-terminal region of GSK-3β (residues 78–92), promoting p53 acetylation at Lys373/382 independent of GSK-3β activity, as verified by co-immunoprecipitation and assays with deletion mutants. High-throughput screens, including and affinity purification, have identified over 100 interactors for GSK-3β, with motif preferences such as the AXS-like sequences in Axin facilitating high-affinity scaffold recruitment.

Scaffold Complexes and Networks

GSK-3β plays a central role in the Wnt destruction complex, a multi-protein assembly comprising Axin, (APC), (CK1), and β-catenin, which regulates β-catenin levels through and subsequent degradation in the absence of Wnt signaling. This complex dynamically assembles and disassembles; Wnt stimulation leads to disassembly by recruiting , inhibiting GSK-3β activity and stabilizing β-catenin for transcriptional activation. Axin acts as the scaffold, facilitating GSK-3β's of β-catenin at key sites (Ser33, Ser37, Thr41), while APC enhances processivity and CK1 primes the substrate. Recent studies highlight as a mechanism driving complex formation, with GSK-3β's localization modulated by these interactions. In insulin signaling, GSK-3β integrates into PI3K scaffolds centered on insulin receptor substrate-1 (IRS-1), where IRS-1 recruits PI3K to generate PIP3, activating Akt, which in turn GSK-3β at Ser9 to inhibit its activity. This -like assembly coordinates metabolic responses, with GSK-3β inactivation promoting activation and ; disruptions in this scaffold, such as IRS-1 serine phosphorylation, can lead to . The IRS-1-Akt-GSK-3β network exemplifies a modular signaling , allowing rapid signal propagation from receptor kinases. GSK-3β also participates in microtubule-associated complexes with and 1B (MAP1B), influencing neuronal transport and cytoskeletal dynamics during growth. of MAP1B by GSK-3β at multiple sites reduces its microtubule-binding affinity, promoting dynamic instability essential for neurite branching and . similarly serves as a , with GSK-3β-mediated hyperphosphorylation detaching it from , potentially impairing vesicular transport in neurons. These complexes highlight GSK-3β's role in integrating activity with cytoskeletal regulation. Network analyses, such as those from the database, underscore GSK-3β's centrality, positioning it as a hub in over 20 signaling pathways including Wnt, insulin, and , with strong interactions to partners like Akt (combined score >0.9). This connectivity reflects its average degree of approximately 8.5 in high-confidence s, emphasizing its integrative across cellular processes. In pathological contexts like , GSK-3β incorporates into neurofibrillary tangles alongside hyperphosphorylated , contributing to aggregate formation.

Physiological Roles

Metabolic Regulation

Glycogen synthase kinase-3 beta (GSK-3β) plays a central role in hepatic metabolism by phosphorylating and thereby inhibiting , the rate-limiting enzyme in synthesis. This inhibition reduces storage and promotes , contributing to elevated blood glucose levels during fasting states. In the liver, inhibition of GSK-3β suppresses the expression of gluconeogenic enzymes such as glucose-6-phosphatase, thereby enhancing . Liver-specific of GSK-3β in mice results in viable animals with improved glucose tolerance and insulin sensitivity, underscoring its role in maintaining metabolic balance. In , GSK-3β regulates through of sterol regulatory element-binding protein-1 (SREBP-1) at serine 73, which targets the mature form for ubiquitination and proteasomal degradation, thereby limiting . In the fasting state, this mechanism limits . In the fed state, insulin-mediated inhibition of GSK-3β stabilizes SREBP-1 to promote lipogenic . GSK-3β also influences oxidation indirectly via crosstalk with (ACC); its activity counteracts AMPK-mediated of ACC at serine 79, which would otherwise inhibit production and facilitate mitochondrial entry. Inhibition of GSK-3β thus enhances ACC , reduces levels, and boosts β-oxidation, helping to prevent ectopic accumulation in tissues like the liver.00456-2) As an energy sensor, GSK-3β engages in bidirectional crosstalk with (AMPK), a key regulator of cellular . Active GSK-3β inhibits AMPK by phosphorylating its α subunit, thereby dampening AMPK's activation in response to low ATP levels and limiting catabolic processes such as and . Conversely, GSK-3β inhibition activates the LKB1-AMPK pathway, mimicking the metabolic adaptations seen in caloric restriction by enhancing mitochondrial respiration, increasing NAD(P)H flux, and elevating . These effects promote efficient energy utilization and protect against metabolic stress. Systemically, GSK-3β modulates insulin secretion in pancreatic β-cells by restraining and function; conditional of GSK-3β in these cells expands β-cell mass and enhances glucose-stimulated insulin release, improving overall glucose tolerance. In peripheral tissues, GSK-3β inhibits adipocyte differentiation by phosphorylating β-catenin in the , thereby suppressing PPARγ expression and preadipocyte commitment to mature . This regulatory role helps maintain and prevents excessive fat accumulation. As part of insulin signaling, GSK-3β's inhibition by Akt promotes these anabolic effects in metabolic tissues.

Developmental and Cellular Processes

Glycogen synthase kinase-3 beta (GSK-3β) plays a critical role in embryonic development, particularly through its involvement in the , where it negatively regulates β-catenin stability to ensure proper axis formation and patterning. In mice, homozygous of Gsk3b results in embryonic lethality during mid-gestation, characterized by severe liver degeneration due to massive apoptosis and cardiac defects including double outlet right ventricle, ventricular septal defects, and with ventricular cavity obliteration. These phenotypes highlight GSK-3β's essential function in maintaining cellular survival and organ during embryogenesis, as its absence leads to dysregulated and in key developmental tissues. In cellular processes, GSK-3β modulates the by at Thr-286, which promotes its nuclear export, ubiquitination, and proteasomal degradation, thereby facilitating the G1/S phase transition and preventing aberrant progression. Additionally, GSK-3β c-Myc at Thr-58, enhancing its ubiquitination and degradation via the Fbw7 , which contributes to the maintenance of cellular quiescence and inhibits uncontrolled in response to signals. These events underscore GSK-3β's role as a key regulator of , ensuring orderly progression and preventing oncogenic transformation. GSK-3β influences neuronal development by phosphorylating collapsin response mediator protein-2 (CRMP2) at Thr-514, which inactivates CRMP2's ability to promote assembly and thus restricts growth and neuronal during . In synaptic processes, GSK-3β regulates the synaptic expression and of N-methyl-D-aspartate (NMDA) receptors by phosphorylating 4-kinase type IIα (PI4KIIα), stabilizing a pool of GluN2B-containing NMDA receptors essential for long-term depression () and overall . These mechanisms position GSK-3β as a modulator of neuronal architecture and adaptability, balancing growth and refinement in neural circuits.00030-4) Regarding apoptosis and survival, GSK-3β exhibits a dual role: it promotes by myeloid cell leukemia-1 (Mcl-1) at Ser-159, leading to Mcl-1 ubiquitination and degradation, which derepresses Bax activation and mitochondrial outer membrane permeabilization. Conversely, under cellular stress, GSK-3β supports survival by facilitating activation through of its inhibitors, thereby inducing anti-apoptotic and protecting against extrinsic death signals. This bifunctional regulation allows GSK-3β to fine-tune cellular fate in response to developmental and environmental cues, integrating pro- and anti-survival signals within the same pathway.00111-0)

Pathological Implications

Neurodegenerative and Psychiatric Disorders

In , hyperactive glycogen synthase kinase-3 beta (GSK-3β) plays a central role in tau hyperphosphorylation, targeting multiple serine and residues, including at least nine key sites that promote the detachment of from and facilitate the formation of neurofibrillary tangles. This aberrant disrupts and neuronal integrity, contributing to the cognitive decline observed in affected individuals. Furthermore, GSK-3β enhances amyloid-beta (Aβ) production by modulating the γ-secretase-mediated cleavage of amyloid precursor protein (APP), thereby increasing Aβ peptide generation and plaque deposition in the . Preclinical studies in transgenic models demonstrate that selective inhibition or silencing of GSK-3β reduces Aβ plaque load, tau pathology, and associated neuronal loss, highlighting its pathological significance. GSK-3β dysregulation also contributes to pathology, particularly through promotion of α-synuclein aggregation and subsequent loss of neurons in the . Elevated GSK-3β activity exacerbates α-synuclein phosphorylation and accumulation, leading to formation and impaired that drives neurodegeneration. This further impairs mitochondrial quality control, including mitophagy, which compounds and cell death in preclinical models of the disease. In , genetic variations in the GSK3B gene, such as single nucleotide polymorphisms rs334558 and rs3755557, influence susceptibility to mood episodes and response to therapy, a key that inhibits GSK-3β. Individuals with the TT at rs334558 exhibit poorer responsiveness, potentially due to altered GSK-3β . Hyperactivity of GSK-3β is associated with mood instability, as it disrupts and signaling pathways that maintain emotional , with elevated kinase levels observed in manic states. GSK-3β has emerging roles in other neurodegenerative and psychiatric conditions. In , inhibition of GSK-3β ameliorates mutant aggregation and associated cellular toxicity in neuronal models, suggesting it exacerbates polyglutamine-mediated pathogenesis. In , GSK-3β dysregulation intersects with hyperdopaminergic signaling, particularly via D2 receptor pathways, where excessive kinase activity inhibits function and contributes to cognitive and psychotic symptoms.

Cancer, Diabetes, and Other Conditions

GSK-3β exhibits a context-dependent in cancer, functioning as both a tumor suppressor and promoter through its regulation of β-catenin in the . In , GSK-3β typically acts as a tumor suppressor by phosphorylating β-catenin within the Axin/ destruction complex, marking it for ubiquitin-mediated degradation and thereby inhibiting Wnt/β-catenin signaling to suppress , , and . Mutations in , common in , disrupt this complex and inactivate GSK-3β, leading to β-catenin stabilization, nuclear translocation, and hyperactivation of oncogenic Wnt target genes such as c-Myc and , which drive tumorigenesis. Conversely, in , GSK-3β promotes tumor progression as an by sustaining and survival; its inhibition stabilizes c-Myc, which in this context induces and sensitizes cells to therapies like , highlighting GSK-3β's pro-tumorigenic effects via β-catenin-independent mechanisms in addition to its Wnt modulatory role. In diabetes, GSK-3β contributes to insulin resistance by phosphorylating insulin receptor substrate-1 (IRS-1) on serine residues, such as Ser332, which inhibits IRS-1 tyrosine phosphorylation and impairs downstream insulin signaling through the PI3K/Akt pathway. This serine phosphorylation also promotes IRS-1 ubiquitination and proteasomal degradation, exacerbating insulin resistance in high-glucose conditions. GSK-3β further drives β-cell dysfunction by suppressing proliferation and enhancing apoptosis in pancreatic islets, leading to reduced β-cell mass and impaired insulin secretion, as evidenced by β-cell-specific overexpression models that accelerate diabetes onset. Notably, Gsk3b haploinsufficiency in insulin-resistant mouse models, such as Ir^{+/-} and Irs2^{-/-}, protects against type 2 diabetes by improving glucose homeostasis, preserving β-cell mass through increased proliferation and decreased apoptosis, and enhancing insulin sensitivity without altering peripheral insulin resistance. GSK-3β promotes and autoimmunity primarily through activation of the pathway, where its activity (via Tyr216 ) enhances nuclear translocation and transcription of pro-inflammatory s such as IL-6 and TNF-α in response to stimulation. In , elevated GSK-3β in fibroblast-like synoviocytes drives production of inflammatory mediators including IL-1β, IL-6, and MMP-9 via , JNK, and p38 MAPK signaling, contributing to synovial and joint destruction; inhibition with compounds like TDZD-8 suppresses these responses and attenuates disease severity in collagen-induced arthritis models. Similarly, in , GSK-3β blockade reduces chronic intestinal in models by limiting -driven production and immune cell infiltration, mitigating disease progression. In cardiovascular pathologies, GSK-3β suppresses cardiac by inhibiting TGF-β1/SMAD-3 signaling in fibroblasts; its inhibition (via Ser9 ) enhances SMAD-3 and transcriptional activity, promoting , extracellular matrix deposition, and adverse post-ischemia. GSK-3β inhibition also confers protection against myocardial ischemia/ by reducing infarct size, , and , as demonstrated in preclinical models where pharmacological or genetic suppression limits damage through stabilization and modulation. Recent reviews (2020–2025) emphasize GSK-3β's role in post-myocardial remodeling, linking its dysregulation to and dysfunction while highlighting isoform-specific inhibition as a cardioprotective strategy.

Therapeutic Targeting

Inhibitor Classes and Mechanisms

GSK-3β inhibitors are broadly classified into ATP-competitive, substrate-competitive, allosteric, and natural or indirect categories, each targeting distinct molecular sites to modulate the enzyme's activity. ATP-competitive bind directly to the ATP-binding pocket within the , preventing ATP from associating with GSK-3β and thereby blocking of substrates. These compounds often exhibit high potency but suffer from limited selectivity due to the conserved nature of the ATP-binding site across kinases. For instance, 6-bromoindirubin-3'- (BIO), a prototypical ATP-competitive , forms hydrogen bonds with key residues such as Asp133 and Val135 in the , achieving an of approximately 5 nM against GSK-3β. Similarly, maleimides like SB-216763 bind in the same pocket, interacting with Glu97 and Lys85, but frequently cross-react with other kinases, including (CDK5), due to structural homology.01104-2) Substrate-competitive inhibitors, in contrast, target the substrate-binding groove adjacent to the , mimicking the primed motif required for GSK-3β recognition and thus preventing substrate docking without interfering with ATP binding. This approach enhances selectivity by exploiting the enzyme's unique substrate specificity. A representative example is L803-mts, a myristoylated cell-permeable derived from the β-catenin pseudosubstrate sequence, which binds near residues Arg96, Arg180, and Lys205, effectively blocking primed substrates like and with an in the low micromolar range. Another compound, TDZD-8 (4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione), operates via a similar non-ATP-competitive mechanism, forming a covalent that inhibits GSK-3β with an of 2 μM while showing minimal activity against CDKs, , or PKC at concentrations up to 100 μM. Allosteric inhibitors induce conformational changes in GSK-3β by outside the catalytic , often at interfaces involved in protein-protein interactions, which can lead to more subtle and potentially isoform-specific modulation. For example, BIO has also been implicated in allosteric effects at the Axin-binding interface, stabilizing an inactive conformation, though its primary action remains ATP-competitive. More dedicated allosteric agents, such as certain thiadiazolidinones, bind to pockets like the glycine-rich loop or distant allosteric sites (e.g., pockets 4 or 5), altering the enzyme's flexibility and substrate access with values around 1-3 μM, offering advantages in reducing off-target effects compared to ATP-site binders. Natural and indirect inhibitors encompass compounds that do not directly compete at the but modulate GSK-3β through upstream pathways or unique binding modes. , a cornerstone therapeutic for , inhibits GSK-3β indirectly by depleting levels via inhibition of inositol monophosphatase, which enhances inhibitory Ser9 and reduces activity, with effective concentrations around 1-2 mM. , a thiadiazolidinone derivative, acts as an irreversible non-ATP-competitive inhibitor, forming a with Cys199 in the substrate-binding region, thereby locking the enzyme in an inactive state with an of 60 nM and selectivity over kinases lacking a homologous . Selectivity remains a major challenge across inhibitor classes, as GSK-3β shares 85% sequence identity with GSK-3α, often resulting in dual that complicates therapeutic targeting. ATP-competitive are particularly prone to off-target effects on CDK5 and other proline-directed kinases due to overlapping ATP pockets, potentially contributing to or cytoskeletal disruption. Substrate-competitive and allosteric agents mitigate this by engaging unique epitopes, but achieving brain and avoiding isoform still demands refined chemical optimization.

Preclinical and Clinical Developments

Preclinical studies have demonstrated the therapeutic potential of GSK-3β inhibition in neurodegenerative disease models. In transgenic mouse models of Alzheimer's disease (AD) expressing human tau with missense mutations, chronic lithium treatment reduced mutant tau protein aggregation and phosphorylation, thereby decreasing tau lesions through enhanced ubiquitination. Similarly, lithium arrested the development of neurofibrillary tangles in mice with advanced tau pathology, highlighting its role in mitigating tau hyperphosphorylation. For Parkinson's disease (PD), the GSK-3β inhibitor tideglusib improved motor symptoms and protected dopaminergic neurons in MPTP-induced mouse models by reducing neuronal death, with evidence from conditional GSK-3β knockout mice confirming the isoform's specific contribution to parkinsonian pathology. In amyotrophic lateral sclerosis (ALS) models, GSK-3β inhibitors like NP031112 (also known as tideglusib) have shown promise in preventing motor neuron degeneration and delaying disease progression, as supported by studies indicating reduced neuroinflammation and oxidative stress in preclinical rodent models. Clinical trials targeting GSK-3β have yielded mixed results across neurological and oncological indications. Lithium remains the standard of care for bipolar disorder, where its GSK-3β inhibitory effects contribute to mood stabilization and long-term maintenance therapy, with extensive evidence from randomized controlled trials confirming its efficacy in preventing manic and depressive episodes. In progressive supranuclear palsy (PSP), a phase II randomized trial of tideglusib (NCT01049399) conducted from 2010 to 2012 failed to meet its primary endpoint of improvement on the PSP rating scale after 52 weeks but demonstrated reduced brain atrophy progression, particularly in parietal and occipital regions, prompting renewed interest in GSK-3β inhibitors for tauopathies. In oncology, the selective GSK-3β inhibitor 9-ING-41 (elraglusib) has advanced to phase I/II trials for refractory cancers, showing a favorable toxicity profile and clinical benefit when combined with chemotherapy, including partial responses and stable disease in advanced pancreatic and other solid tumors; as of 2025, phase II trials report improved survival in metastatic pancreatic cancer when combined with gemcitabine/nab-paclitaxel. Biomarker development for GSK-3β-targeted therapies faces challenges, particularly in applications. (CSF) measurements of GSK-3β activity have been explored as potential endpoints in clinical trials, with some studies linking reduced CSF phosphotau levels to GSK-3β inhibition, though results vary due to inconsistent target engagement. A key hurdle is penetration, as many GSK-3β inhibitors exhibit poor blood-brain barrier crossing, limiting efficacy in neurodegenerative disorders and necessitating optimized formulations for CNS delivery. Emerging developments include isoform-selective inhibitors and combination strategies. Patents filed in 2024 and 2025 describe novel isoform-selective GSK-3β inhibitors, such as N-(pyridin-3-yl)-2-amino-isonicotinamide derivatives, aimed at enhancing specificity and reducing off-target effects for and other conditions; a 2025 highlights novel scaffolds for selective GSK-3β inhibitors, including advancements in brain-penetrant compounds. In cancer, preclinical and early support elraglusib's role in enhancing /T-cell effector stimulation and anti-tumor responses, with ongoing exploration of combinations with immunotherapies.

References

  1. [1]
    Glycogen synthesis and beyond, a comprehensive review of GSK3 ...
    Glycogen synthase kinase‐3 (GSK3) is a highly evolutionarily conserved serine/threonine protein kinase first identified as an enzyme that regulates glycogen ...
  2. [2]
    Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases
    Glycogen synthase kinase-3 (GSK3) may be the busiest kinase in most cells, with over 100 known substrates to deal with.
  3. [3]
    GSK3B - Glycogen synthase kinase-3 beta | UniProtKB - UniProt
    Several potent GSK3 (GSK3A and GSK3B) inhibitors have been identified and characterized in preclinical models for treatments of type 2 diabetes (PubMed:19366350) ...
  4. [4]
    Identification of the Axin and Frat Binding Region of Glycogen ...
    Low expression. Open table in a new tab. Residue Lys85, which forms part of the ATP binding site and is commonly mutated to generate kinase-dead variants ...
  5. [5]
    Crystal Structure of Glycogen Synthase Kinase 3β - ScienceDirect.com
    The visible N-terminal domain (35–134) of GSK3β consists of a seven-stranded β sheet curved over on itself to form a closed orthogonal β barrel (Figure 1). This ...
  6. [6]
    The Structure of Phosphorylated GSK-3β Complexed with a Peptide ...
    The core of the kinase domain is elaborated in GSK-3 by unique structural elements at the N and C termini. At the N terminus a unique β strand, βLO, (residues ...
  7. [7]
    GSK-3: Functional Insights from Cell Biology and Animal ... - Frontiers
    The kinase is negatively regulated by N-terminal phosphorylation of serine residues of the enzyme (Ser21 for GSK-3α and Ser9 for GSK-3β; Sutherland et al., 1993 ...Missing: 56-320 | Show results with:56-320
  8. [8]
    GSK-3: tricks of the trade for a multi-tasking kinase - PMC
    Although highly homologous within their kinase domains (98% identity), the two gene products share only 36% identity in the last 76 C-terminal amino acid ...
  9. [9]
    Interactomes of Glycogen Synthase Kinase-3 Isoforms
    Feb 13, 2023 · The kinase is highly conserved across species, with two protein isoforms encoded by distinct genes in mammals, GSK-3α and GSK-3β. In humans ...
  10. [10]
    Gene: GSK3B (ENSG00000082701) - Summary - Homo_sapiens
    Chromosome 3: 119,821,321-120,094,994 reverse strand. This gene has 71 transcripts (splice variants), 232 orthologues, 1 paralogue and is associated with 85 ...
  11. [11]
    GSK3B glycogen synthase kinase 3 beta [ (human)] - NCBI
    Sep 21, 2025 · It is a negative regulator of glucose homeostasis and is involved in energy metabolism, inflammation, ER-stress, mitochondrial dysfunction, and apoptotic ...Missing: 3p21. | Show results with:3p21.
  12. [12]
    Tissue expression of GSK3B - Summary - The Human Protein Atlas
    Summary of GSK3B expression in human tissue. Cytoplasmic expression in most tissues, most abundant in CNS and testis.
  13. [13]
    Brain tissue expression of GSK3B - The Human Protein Atlas
    Summary of GSK3B expression in human brain tissue. Cytoplasmic expression in most tissues, most abundant in CNS and testis.
  14. [14]
    Developmental expression and localization of glycogen synthase ...
    GSK-3β expression is highest from 18 days embryonic to 10 days postnatal, strongly in developing neurons, and found in pericarya and dendrites. Axonal tracts ...Missing: GSK3B CNS
  15. [15]
    GTEx Portal
    - **mRNA Expression Levels**: Data from GTEx Analysis Release V10 (dbGaP Accession phs000424.v10.p2) shows GSK3B expression in TPM (Transcripts Per Million) across tissues.
  16. [16]
    glycogen synthase kinase 3 beta | GSK subfamily
    The IUPHAR/BPS Guide to Pharmacology. glycogen synthase kinase 3 beta - GSK subfamily. Detailed annotation on the structure, function, physiology, ...
  17. [17]
  18. [18]
    [PDF] ProQinase™ GSK3-beta - Reaction Biology
    2.0 µg GST-GSK3-beta. 250. 150. 75. 50. 25. 15. 37. 100. 20. GSK3-beta Lot 003: Determination of Vmax and KM value for ATP. Determination of KM value & Specific ...
  19. [19]
    What Are the bona fide GSK3 Substrates? - PMC - PubMed Central
    Nearly 100 proteins are proposed to be substrates for GSK3, suggesting that this enzyme is a fundamental regulator of almost every process in the cell.<|control11|><|separator|>
  20. [20]
    Glycogen synthase kinase 3β induces apoptosis in cancer cells ...
    Dec 8, 2008 · Most GSK3β is cytoplasmic if cells are unstressed and growing, although a low level remains detectable in the nucleus. Nuclear levels of GSK3β ...
  21. [21]
    Protein S-Nitrosylation Controls Glycogen Synthase Kinase 3β ...
    May 25, 2018 · GSK-3β has a well-characterized bipartite nuclear localization sequence which can direct its translocation to the nucleus under certain ...
  22. [22]
    Wnt Signaling Requires Sequestration of Glycogen Synthase Kinase ...
    Dec 23, 2010 · We report that Wnt signaling triggers the sequestration of GSK3 from the cytosol into multivesicular bodies (MVBs), so that this enzyme becomes separated from ...
  23. [23]
    Involvment of Cytosolic and Mitochondrial GSK-3β in Mitochondrial ...
    Using immunocytochemistry and subcellular fractionation techniques, we showed that GSK-3β partially localized within mitochondria in both neuronal cell models.
  24. [24]
    Structural basis for recruitment of glycogen synthase kinase 3β to ...
    Detailed comparison of the axin and FRAT GSK3β complexes allows the generation of highly specific mutations, which abrogate binding of one or the other.
  25. [25]
  26. [26]
    Mechanisms for redox-regulation of protein kinase C - ResearchGate
    Aug 7, 2025 · ... oxidative. modifications at this site reduce substrate-binding affinity. Second, Cys199 oxidation may inhibit enzyme activity through. an ...
  27. [27]
    In vivo regulation of glycogen synthase kinase 3β activity in neurons ...
    Aug 17, 2017 · GSK3β is thought to be constitutively activated by autophosphorylation at Tyr216 and inactivated by phosphorylation at Ser9.
  28. [28]
    Cardioprotection by GSK-3 inhibition: role of enhanced glycogen ...
    Our study indicates that reduction in H + production by GSK-3 inhibition is an early and upstream event that lessens Ca 2+ i overload during ischaemia and ...
  29. [29]
    Identification of targets of the Wnt pathway destruction complex in ...
    Mar 31, 2009 · The targeting of β-catenin for degradation is mediated by phosphorylation of its N-terminal serine residues by glycogen synthase kinase 3 (GSK3) ...
  30. [30]
    Control of β-Catenin Phosphorylation/Degradation by a Dual-Kinase ...
    β-catenin degradation is initiated upon amino-terminal serine/threonine phosphorylation, which is believed to be performed by glycogen synthase kinase-3 (GSK-3) ...
  31. [31]
    Induction of Gsk3β-β-TrCP Interaction Is Required for Late Phase ...
    In the absence of Wnt, β-catenin is phosphorylated by Gsk3 on serine 33 and 37 and threonine 41 (which requires priming phosphorylation of β-catenin by CK1) (3); ...
  32. [32]
    Wnt/β-catenin signalling: function, biological mechanisms, and ...
    Jan 3, 2022 · The canonical Wnt pathway, also known as the Wnt/β-catenin pathway, involves the nuclear translocation of β-catenin and activation of target ...
  33. [33]
    Destruction complex dynamics: Wnt/β-catenin signaling alters Axin ...
    The central regulator of the Wnt/β-catenin pathway is the Axin/APC/GSK3β destruction complex (DC), which, under unstimulated conditions, targets cytoplasmic β- ...
  34. [34]
    β-Catenin stabilization stalls the transition from double-positive to ...
    Jun 15, 2007 · Activation of the Wnt/β-catenin cascade results in inhibition of the constitutive activity of GSK-3β5 by the cytoplasmic protein Dishevelled ( ...
  35. [35]
    Rapid, Wnt-Induced Changes in GSK3β Associations that Regulate ...
    Wnt stimulation rapidly disrupts GSK3β/Axin complexes, leading to β-catenin stabilization. Gα proteins contribute to this disruption and β-catenin ...
  36. [36]
    Multifaceted roles of GSK-3 and Wnt/β-catenin in hematopoiesis and ...
    In this review, we will discuss the roles that GSK-3 plays in hematopoiesis and leukemogenesis as how this pivotal kinase can interact with multiple signaling ...
  37. [37]
    GSK3: a multifaceted kinase in Wnt signaling - PMC - PubMed Central
    Upon FZD activation, Goα rapidly dissociates from FZD; this process correlates with the dissociation of GSK3β from AXIN. ... GSK-3 inhibitors and insulin receptor ...Gsk3: A Multifaceted Kinase... · Gsk3 In Wnt-Induced... · Wnt-Gsk3 Signaling Beyond...
  38. [38]
    control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled ...
    Jan 15, 2008 · Canonical Wnt/β-catenin signaling has central roles in development and diseases, and is initiated by the action of the frizzled (Fz) ...<|separator|>
  39. [39]
    [PDF] Glycogen Synthase Kinase 3 in Wnt Signaling Pathway and Cancer
    At the protein level, GSK-3α and GSK-3β are highly homologous in their kinase domain (98%), but outside this region, their sequences differ substantially. GSK- ...
  40. [40]
    Wnt signaling in colorectal cancer: pathogenic role and therapeutic ...
    Jul 14, 2022 · MicroRNA-520e targets AEG-1 to suppress the proliferation and invasion of colorectal cancer cells through Wnt/GSK-3β/β-catenin signalling.
  41. [41]
    Signaling pathways involved in colorectal cancer: pathogenesis and ...
    Oct 7, 2024 · Wnt pathway dysregulation in colorectal cancer. Enhanced Wnt signaling is a key driver of CRC development and progression. The Wnt pathway ...
  42. [42]
    Inhibition of glycogen synthase kinase-3 by insulin ... - Nature
    Dec 28, 1995 · GSK3 is inhibited by serine phosphorylation in response to insulin or growth factors 3,9–11 and in vitro by either MAP kinase-activated protein (MAPKAP) kinase ...
  43. [43]
    Phosphorylation and inactivation of glycogen synthase kinase 3 by ...
    Glycogen synthase kinase 3 (GSK-3) is implicated in multiple biological processes including metabolism, gene expression, cell fate determination, ...
  44. [44]
    Serine 332 Phosphorylation of Insulin Receptor Substrate-1 by ...
    Here, we show that GSK-3 phosphorylates Ser332 of IRS-1 and that mutation at this site enhances insulin-induced tyrosine phosphorylation of IRS-1. We bring ...
  45. [45]
    Phosphorylation of insulin receptor substrate 1 by glycogen ... - PNAS
    Here we report that glycogen synthase kinase 3 (GSK-3) is capable of phosphorylating IRS-1 and that this modification converts IRS-1 into an inhibitor of IR ...
  46. [46]
    Tissue-Specific Role of Glycogen Synthase Kinase 3β in Glucose ...
    Liver-specific GSK-3β knockout mice are viable and glucose and insulin tolerant and display “normal” metabolic characteristics and insulin signaling. Mice ...
  47. [47]
    A highlight on Sonic hedgehog pathway
    Mar 20, 2018 · GSK3β, can act as a positive regulator of Shh signaling by phosphorylating SUFU and promoting the release of SUFU from Gli, at least when the ...
  48. [48]
    Hedgehog/GLI Signaling Pathway: Transduction, Regulation ... - MDPI
    GLI transcription factors are sequestered by SUFU and phosphorylated by PKA, CK1, and GSK3β, marking them for proteolytic cleavage. The cleavage of the C- ...2. Hh/gli Signaling Pathway... · 2.2. Receptor Complex · 2.3. The Gli Family Of...
  49. [49]
    GSK-3β controls NF-kappaB activity via IKKγ/NEMO - PMC
    Dec 8, 2016 · Our data suggest that GSK-3β is critically important for ordered NF-κB signalling through modulation of NEMO phosphorylation.
  50. [50]
    GSK-3β: A Bifunctional Role in Cell Death Pathways - PMC
    Active GSK-3β also positively regulates NFκB by activating IKK, IκB, and p65, leading to the inhibition of TNF-mediated apoptosis. These actions inhibit the ...
  51. [51]
    Degradation of Mcl-1 by β-TrCP Mediates Glycogen Synthase ... - NIH
    In an attempt to understand the molecular mechanism of GSK-3β-induced apoptosis, we found that GSK-3β physically associated with and phosphorylated Mcl-1 and ...
  52. [52]
    Translocation of Glycogen Synthase Kinase-3β (GSK-3β), a ... - NIH
    These results demonstrate that GSK-3β translocates from the cytosol to mitochondria in a kinase activity- and VDAC2-dependent manner.
  53. [53]
    A chemical biology approach reveals period shortening of ... - PNAS
    Dec 30, 2008 · These results clarify the role of GSK-3β in the period regulation of the mammalian clockworks and highlight the effectiveness of chemical ...
  54. [54]
  55. [55]
    Wnt-induced dephosphorylation of Axin releases β-catenin from the ...
    Phosphorylation of Axin with GSK-3β increases Axin's affinity for β-catenin. ... We then tested directly whether the phosphorylation of Axin by GSK-3β regulates ...
  56. [56]
    C‐terminal truncation of GSK‐3β enhances its dephosphorylation by ...
    Mar 7, 2017 · Truncation of GSK-3β promoted its nuclear translocation and enhanced its interaction with PP2A, leading to its Ser9 dephosphorylation by PP2A.
  57. [57]
    Caveolin-1 Expression Inhibits Wnt/β-Catenin/Lef-1 Signaling by ...
    GSK-3β Is Excluded from Caveolae Membranes and Caveolin-1 Expression Blocks the Interaction between β-Catenin and GSK-3β. Glycogen synthase kinase-3β is a key ...
  58. [58]
    GSK3β N-terminus binding to p53 promotes its acetylation - PMC
    Mar 5, 2009 · In this study we identified the GSK3β N-terminal amino acids 78–92 as necessary for its association with p53. Inhibitors of GSK3 impaired the ...Missing: core PP2A Caveolin-
  59. [59]
    The β-Catenin Destruction Complex - PMC - PubMed Central - NIH
    Axin is a key control point for β-catenin destruction by virtue of its ability to promote efficient phosphorylation of β-catenin and APC; indeed, the GSK-3 and ...
  60. [60]
    A novel GSK3-regulated APC:Axin interaction regulates Wnt ... - eLife
    Sep 22, 2015 · A new dynamic mechanistic model explains how the destruction complex negatively regulates Wnt signaling in development and oncogenesis.
  61. [61]
    Liquid–liquid phase separation drives the β‐catenin destruction ...
    Aug 21, 2021 · Wnt stimulation induces the assembly of the receptor-associated signalosome and the inactivation of the destruction complex, leading to β- ...
  62. [62]
    Role of insulin receptor substance-1 modulating PI3K/Akt insulin ...
    In this paper, we review the links between IRS-1 and the PI3K/Akt insulin signaling pathway, and highlight phosphorylated IRS-1 which negatively regulated by ...Irs And The Pi3k/akt Insulin... · Akt/gsk3 · Table 2Missing: scaffold hubs
  63. [63]
    Akt phosphorylates insulin receptor substrate to limit PI3K-mediated ...
    Jul 13, 2021 · We show that Akt itself engages this negative feedback by phosphorylating insulin receptor substrate (IRS) 1 and 2 on a number of residues.Missing: hubs | Show results with:hubs
  64. [64]
    Glycogen synthase kinase-3beta phosphorylation of MAP1B at ...
    GSK-3beta phosphorylation of MAP1B appears to act as a molecular switch regulating the control that MAP1B exerts on microtubule dynamics in growing axons and ...Missing: Tau complex transport
  65. [65]
    The GSK3–MAP1B pathway controls neurite branching and ...
    MAP1B is highly expressed in differentiating neurons and in regions of the adult nervous system that retain neuronal plasticity or regenerate after injury ( ...
  66. [66]
    MAP1B regulates microtubule dynamics by sequestering EB1/3 in ...
    Apr 9, 2013 · MAP1B, a structural microtubule (MT)‐associated protein highly expressed in developing neurons, plays a key role in neurite and axon extension.
  67. [67]
    None
    Nothing is retrieved...<|control11|><|separator|>
  68. [68]
    Tau phosphorylation by GSK-3β promotes tangle-like filament ...
    We have found that phosphorylation by GSK-3β is sufficient to cause tau filaments to coalesce into tangle-like aggregates similar to those isolated from ...
  69. [69]
    Inhibition of GSK-3 selectively reduces glucose-6-phosphatase and ...
    We find that selectively reducing GSK-3 activity strongly reduces the expression of both gluconeogenic genes.
  70. [70]
    Genetic Deficiency of Glycogen Synthase Kinase-3β Corrects ...
    We demonstrated that genetically reducing the levels of Gsk-3β in the insulin-resistant mouse improved glucose homeostasis.
  71. [71]
    GSK-3β Function in Bone Regulates Skeletal Development, Whole ...
    Pronounced hypoglycemia is apparent in KO pups prior to first feeding (data not shown) and might contribute to the 30% mortality at P0. Adult female (but not ...
  72. [72]
    Glycogen synthase kinase-3-mediated phosphorylation of serine 73 ...
    Nov 20, 2015 · We propose that in the fasting state induction of GSK-3β leads to phosphorylation and accelerated degradation of SREBP-1c. GSK-3β has also ...
  73. [73]
    Inhibition of AMPK Catabolic Action by GSK3 - ScienceDirect.com
    May 9, 2013 · AMPK phosphorylates and inhibits both ACC and Raptor (Gwinn et al., 2008), stimulating fatty acid β-oxidation (Merrill et al., 1997) and ...
  74. [74]
    Gsk-3β Controls Autophagy by Modulating LKB1-AMPK Pathway in ...
    GSK-3β Inhibition Leads to AMPK Activation. As a major energy sensor, AMPK is activated once cells suffer energy crisis [4]. Because GSK-3β inhibition ...
  75. [75]
  76. [76]
    GSK3β Regulates Brain Energy Metabolism - PMC - PubMed Central
    We demonstrate that GSK3β inhibition increases mitochondrial respiration and membrane potential and alters NAD(P)H metabolism. These metabolic effects are ...
  77. [77]
    Conditional ablation of Gsk-3β in islet beta cells results in expanded ...
    On a normal diet β-Gsk-3β−/− mice were found to have mild improvement of glucose tolerance and glucose-induced insulin secretion, and increased beta cell mass ...
  78. [78]
    Inhibition of DYRK1A and GSK3B induces human β-cell proliferation
    Oct 26, 2015 · Insufficient pancreatic β-cell mass or function results in diabetes mellitus. While significant progress has been made in regulating insulin ...
  79. [79]
    Adipogenic Differentiation of Muscle Derived Cells is Repressed by ...
    Jun 11, 2018 · Results: The application of lithium chloride to muscle derived cells significantly increased the phosphorylation of GSK-3β and thus inhibited ...
  80. [80]
    Role and Regulation of Glycogen Synthase Kinase-3 in Obesity ...
    GSK-3α and GSK-3β has emerged as a critical player in obesity-associated metabolic pathologies such as type 2 diabetes (T2D), and cardiovascular diseases (CVDs ...Missing: patterns | Show results with:patterns
  81. [81]
    Requirement for glycogen synthase kinase-3beta in cell survival and ...
    Here we show that disruption of the murine GSK-3beta gene results in embryonic lethality caused by severe liver degeneration during mid-gestation, a phenotype ...Missing: knockout | Show results with:knockout
  82. [82]
    The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 ...
    Our data show that (i) T58 and endogenous GSK-3 activity are required for Fbw7-mediated Myc degradation in vivo; (ii) T58 phosphorylation regulates the binding ...
  83. [83]
    GSK‐3β regulates the synaptic expression of NMDA receptors via ...
    May 28, 2020 · Our work has revealed that GSK-3β phosphorylation of this abundant lipid kinase stabilizes a pool of synaptic NMDA receptors in a subunit dependent manner.
  84. [84]
    GSK3 and tau: two convergence points in Alzheimer's disease
    GSK3 phosphorylates tau in most serine and threonine residues hyperphosphorylated in paired helical filaments, and GSK3 activity contributes both to amyloid-β ...
  85. [85]
    Tau phosphorylation by GSK-3beta promotes tangle-like filament ...
    Jun 28, 2007 · We have found that phosphorylation by GSK-3beta is sufficient to cause tau filaments to coalesce into tangle-like aggregates similar to those isolated from ...
  86. [86]
    GSK-3β, a pivotal kinase in Alzheimer disease - Frontiers
    GSK-3β also regulates Aβ production by interfering with APP cleavage at the γ-secretase complex step, since both APP and PS1 are also substrates of this ...Abstract · GSK-3β As a Molecular Link... · AD Therapies Involving GSK...
  87. [87]
    Selectively Silencing GSK-3 Isoforms Reduces Plaques and ...
    May 23, 2012 · Using these two models, we demonstrated that knocking down GSK-3α or GSK-3β reduces the accumulation of phosphorylated tau; however, a single ...
  88. [88]
    GSK-3β dysregulation contributes to parkinson's-like ... - Nature
    Nov 14, 2014 · Dopaminergic neuron loss in mice due to ... Impaired neurotransmission caused by overexpression of α-synuclein in nigral dopamine neurons.
  89. [89]
    GSK-3β Contributes to Parkinsonian Dopaminergic Neuron Death
    Jun 2, 2020 · Our data support the view that GSK-3β acts as a potential therapeutic target in PD and tideglusib would be a candidate drug for PD neuroprotective therapy.Missing: parkin | Show results with:parkin
  90. [90]
    Glycogen synthase kinase 3β gene polymorphisms may ... - PubMed
    Patients with genotype TT at rs334558 (P=0.044) had poorer response to lithium treatment. There was a trend that haplotype C-T increased the risk for bipolar I ...Missing: rs3755557 | Show results with:rs3755557
  91. [91]
    Glycogen synthase kinase 3 substrates in mood disorders and ...
    Jun 25, 2013 · Together, these observations strongly suggest that unusually high levels of GSK3 activity contribute to the pathogenesis of mood disorders.Function Of Gsk3 · Gsk3 Substrates That Are... · Neurotransmission
  92. [92]
    Glycogen Synthase Kinase-3 in the Etiology and Treatment of Mood ...
    Aug 8, 2011 · Evidence is reviewed that depression may be associated with impaired inhibitory control of GSK3, and mania by hyper-stimulation of GSK3.Missing: instability | Show results with:instability
  93. [93]
    Inhibition of GSK-3 ameliorates the pathogenesis of Huntington's ...
    Mar 19, 2021 · Our results provide strong evidence for a role for GSK-3 in the regulation of mHtt dynamics, and demonstrate the benefits of GSK-3 inhibition in ...Missing: Gli | Show results with:Gli
  94. [94]
    Schizophrenia as a GSK-3 dysregulation disorder - Cell Press
    Converging evidence suggests that the regulation of glycogen synthase kinase 3 (GSK-3) might be important in schizophrenia.
  95. [95]
    SERCA2 phosphorylation at serine 663 is a key regulator of ... - Nature
    Jun 8, 2023 · To determine a potential involvement of GSK3β on SERCA2 phosphorylation, we used three different strategies to assess the probability of the ...
  96. [96]
  97. [97]
  98. [98]
  99. [99]
    Chronic lithium treatment decreases mutant tau protein aggregation ...
    Here we report that chronic lithium treatment of a transgenic mouse strain expressing human tau with three missense mutations results in decreased glycogen ...Missing: reduces | Show results with:reduces
  100. [100]
    Lithium treatment arrests the development of neurofibrillary tangles ...
    Lithium treatment arrests the development of neurofibrillary tangles in mutant tau transgenic mice with advanced neurofibrillary pathology · Authors · Affiliation.
  101. [101]
    GSK-3β Contributes to Parkinsonian Dopaminergic Neuron Death
    Jun 3, 2020 · Our data support the view that GSK-3β acts as a potential therapeutic target in PD and tideglusib would be a candidate drug for PD ...
  102. [102]
    Glycogen Synthase Kinase-3 Inhibitors: Preclinical and Clinical ...
    ... Lys-85, a critical residue for ATP binding (Luo et al., 2016). Other examples include the Oxazole-carboxamide derivates such as PF-04802367 (PF-367) ...
  103. [103]
    Lithium: current state of the art and future directions
    Nov 28, 2024 · Lithium is effective both in treating acute mania, as an adjunctive antidepressant, and as a maintenance treatment in bipolar disorder.
  104. [104]
    Tideglusib reduces progression of brain atrophy in ... - PubMed
    Jan 31, 2014 · In patients with PSP, tideglusib reduced the progression of atrophy in the whole brain, particularly in the parietal and occipital lobes.Missing: 2012 revisited
  105. [105]
    Phase 2 study of elraglusib (9-ING-41), a glycogen synthase kinase ...
    May 31, 2023 · Elraglusib in combination with GnP demonstrates early evidence of clinical activity against advanced PDAC and an acceptable safety profile.
  106. [106]
    Machine Learning for Discovery of GSK3β Inhibitors | ACS Omega
    Single-point enzymatic reactions were performed with 1.2 ng/μL GSK3β, 200 ng/μL peptide substrate, 25 μM ATP, and 50 μM DTT with a final DMSO concentration of 1 ...
  107. [107]
  108. [108]
    GSK-3 inhibitor elraglusib (9-ING-41) to enhance tumor-infiltrating ...
    May 31, 2023 · Elraglusib promoted immune cell-mediated tumor cell killing, enhanced tumor cell pyroptosis, decreased tumor cell NF-κB-regulated survival protein expression.