Chronic kidney disease (CKD) is a serious medical condition that poses substantial burdens on patients, families, healthcare systems, and society as a whole. It is characterized by progressive kidney damage and loss of function in the kidney, often compounded by underlying conditions such as diabetes, hypertension, and autoimmune diseases. Glycogen synthase kinase 3 beta (GSK3β), a highly conserved serine/threonine kinase originally implicated in insulin signaling, has emerged as a convergent point of multiple pathways implicated in the pathogenesis and progression of CKD. In the kidney, GSK3β regulates cell fate across diverse cells, including podocytes, mesangial cells, and renal tubular cells, through its interactions with key signaling pathways such as Wnt/β-catenin, NF-κB, Nrf2, PI3K/Akt, and cytoskeleton remodeling pathways. Evidence suggests that dysregulation of GSK3β is closely associated with pathological changes in the kidney, including podocyte injury, mesangial expansion, interstitial fibrosis, and tubular atrophy, which collectively drive chronic kidney destruction. In CKD, GSK3β is overexpressed and thus hyperactive in kidney cells. This sustained hyperactivity perpetuates oxidative stress and profibrotic signaling, particularly in renal tubular cells, thus accelerating the transition from acute kidney injury to CKD. Pharmacological targeting of GSK3β with selective inhibitors has shown promise in preclinical models, by reducing kidney injury, attenuating renal fibrosis, and promoting renal recovery, positioning GSK3β as a potential therapeutic target for CKD. This review highlights recent advances in understanding the molecular and cellular mechanisms through which GSK3β contributes to CKD and underscores its potential as a therapeutic target for various chronic renal diseases.

Chronic kidney disease (CKD) is characterized by the gradual but persistent decline of renal function, ultimately resulting in end-stage renal disease that necessitates dialysis or kidney transplantation [1,2]. CKD represents a significant global health challenge [3-5], with its prevalence steadily increasing due to aging populations, rising rates of comorbidities like diabetes, hypertension, and other risk factors. According to the Global Burden of Disease study, it is estimated that over 850 million people worldwide are affected by some form of kidney disease. CKD commonly arises from underlying conditions such as diabetes mellitus, hypertension, and autoimmune diseases, with these etiologic factors driving disease progression through common pathomechanisms such as chronic inflammation, oxidative stress, and fibrosis [6,7]. Current treatments aim to manage these underlying causes, control blood pressure, and slow kidney damage, primarily through the use of renin–angiotensin–aldosterone system inhibitors, sodium-glucose cotransporter 2 inhibitors, and other renoprotective agents [8-12]. While these therapies can delay disease progression, they often fail to halt or reverse kidney damage in many patients. Moreover, patients with advanced CKD face a higher risk of cardiovascular complications and mortality, further emphasizing the limitations of existing treatment options [13,14]. In light of these challenges, there is an urgent need for novel therapeutic strategies that more effectively address the complex and multifactorial pathophysiology of CKD.

The insulin signaling pathway plays a crucial role in kidney health and diseases, including CKD [15,16]. Within this pathway, glycogen synthase kinase 3 (GSK3) has garnered attention for its potential involvement in kidney injury and repair. Originally characterized in the early 1980 s as a key transducer of insulin signaling responsible for regulating glycogen synthesis, GSK3 is a highly evolutionarily conserved intracellular serine/threonine kinase. It functions by phosphorylating and inhibiting glycogen synthase, thereby playing a central role in glucose metabolism [17]. Beyond its metabolic function, GSK3 has been recognized as a key regulator of various cellular processes, such as proliferation, differentiation, apoptosis, and inflammation. Dysregulation of GSK3 activity has been implicated in a number of diseases, such as neurodegenerative disorders, cancer, diabetes, and inflammatory conditions, positioning it as a promising therapeutic target [18].

Mammalian GSK3 exists in two isoforms: GSK3α (51 kDa) and GSK3β (47 kDa), each encoded by distinct genes located on chromosomes 19 and 3, respectively. Although sharing 97% homology in their catalytic domains and 84% homology in overall structure, GSK3α and GSK3β are not functionally identical and redundant [17,19-21]. The fact that GSK3α knockout (KO) mice display increased insulin sensitivity and lower lipid levels, while embryos lacking GSK3β are not viable, underscores that the α and β isoforms of GSK3 are not functionally interchangeable in some critical celluar processes, with neither able to compensate for the loss of the other [22]. The two isoforms are differentially expressed in a tissue-dependent pattern. In the kidney, both GSK3 isoforms are expressed in renal parenchyma, but the β isoform rather than the α isoform of GSK3 is predominantly expressed and particularly enriched in podocytes and renal tubular cells [23]. Given these findings, most research studies focus on the role of GSK3β in the progression of kidney diseases. In contrast to the findings in global KO mice, podocyte-specific KO of only GSK3 β or GSK3α in mice either at the embryonic stage or during young adulthood resulted in no discernible phenotype, whereas combined KO of both GSK3α and GSK3β specifically in glomerular podocytes in embryonic or adult mice caused severe podocyte injury, glomerulosclerosis, heavy proteinuria, and significant impairment of kidney function [24].

Burgeoning evidence suggests that GSK3β plays a key role in the pathogenesis of CKD [25-28]. GSK3β dysregulation has been associated with various CKD, including glomerular disease, diabetic kidney disease (DKD), and chronic renal tubulointerstitial disease, in both human patients and preclinical models. In cell signaling networks, GSK3β resides at the nexus of multiple pathways involved in CKD, including mitochondria permeability transition (MPT), cytoskeletal organization, development control, insulin signaling pathway, canonical wingless (Wnt) pathway, NF-κB pathway, and the NF-E2-related factor 2 (Nrf2) antioxidant response signaling. GSK3β has emerged as the integration point of multiple kidney injury pathways and has become one of the most attractive therapeutic targets for CKD, which involves multiple pathogenic signaling mechanisms. Indeed, targeting GSK3β via genetic KO or pharmacological inhibitors can mitigate kidney injury in CKD by reducing cell death, inflammation, and fibrosis, thereby improving outcomes in preclinical models [25-27].

In light of the central role of GSK3β in renal injury and its therapeutic potential, this review consolidates current research on the involvement of GSK3β in CKD, with an aim to delineate the molecular mechanisms through which GSK3β influences CKD progression and highlight the therapeutic implications of targeting this kinase. By synthesizing insights from recent studies, this article seeks to provide a comprehensive understanding of GSK3β as a critical factor in CKD development, paving the way for innovative therapeutic strategies to enhance kidney health and improve patient outcomes.

Molecular structure of GSK3β

GSK3β, encoded by the GSK3B gene, is a protein composed of 420 amino acids, structured with 11 exons. It features an N-terminal catalytic domain and a C-terminal regulatory domain (Figure 1) [29]. GSK3β is prominently expressed in several tissues, such as skeletal muscle [30], liver [31], adipose [32], brain [33], cardiac tissue [34], and kidney (Figure 2) [35]. Key structural elements of GSK3β include the glycine-rich loop for ATP stabilization, the catalytic loop essential for phosphoryl-transfer, and the activation loop that regulates access to the catalytic site. The catalytic domain includes the ATP-binding pocket, which is vital for its kinase activity, allowing ATP to bind and facilitate the transfer of phosphate groups to substrate proteins [36,37]. Comparative sequence analysis of GSK3β across different species, such as humans (UniProt ID: P49841), mice (UniProt ID: Q9WV60), and rats (UniProt ID: P18266), reveals a high degree of conservation in key functional regions (Table 1 and Figure 3). The wide distribution and sequence conservation of GSK3β suggest that this protein performs functions of fundamental biological importance.

Protein three-dimensional structure modeling of GSK3β.

Figure 1:
Protein three-dimensional structure modeling of GSK3β.

The full-length three-dimensional protein structure of GSK3β was based on and SWISS-MODLE (https://swissmodel.expasy.org/interactive). Views of the three- dimensional model of human GSK3β.

Figure 1:
Protein three-dimensional structure modeling of GSK3β.

The full-length three-dimensional protein structure of GSK3β was based on and SWISS-MODLE (https://swissmodel.expasy.org/interactive). Views of the three- dimensional model of human GSK3β.

Close modal

Single-cell database analysis reveals widespread expression of GSK3β across kidney cell populations.

Figure 2:
Single-cell database analysis reveals widespread expression of GSK3β across kidney cell populations.

GSK3β is predominantly expressed in various kidney cell types, including podocytes, mesangial cells, and renal tubular epithelial cells. A post hoc analysis was performed on the single nucleus RNA sequencing (snRNAseq) transcriptome of human kidneys based on the Wu and Uchimura et al. Healthy Adult Kidney Dataset (RBK RID: 144KPM) that is publicly available from Kidney Interactive Transcriptomics (https://humphreyslab.com/SingleCell/) reveals the mRNA expression of GSK3B in various kidney cells (A, B) . CNT, connecting tubule; DCT, distal convoluted tubule; EC, endothelial cell; IC-A, alpha intercalated cell; IC-B, beta intercalated cell; LH (AL), loop of henle (ascending limb); LH (DL), loop of henle (descending limb); PC, collecting duct principal cell; PT (S1), proximal tubular (Segment 1); PT (S2), proximal tubular (Segment 2); PT (S3), proximal tubular (Segment 3)

Figure 2:
Single-cell database analysis reveals widespread expression of GSK3β across kidney cell populations.

GSK3β is predominantly expressed in various kidney cell types, including podocytes, mesangial cells, and renal tubular epithelial cells. A post hoc analysis was performed on the single nucleus RNA sequencing (snRNAseq) transcriptome of human kidneys based on the Wu and Uchimura et al. Healthy Adult Kidney Dataset (RBK RID: 144KPM) that is publicly available from Kidney Interactive Transcriptomics (https://humphreyslab.com/SingleCell/) reveals the mRNA expression of GSK3B in various kidney cells (A, B) . CNT, connecting tubule; DCT, distal convoluted tubule; EC, endothelial cell; IC-A, alpha intercalated cell; IC-B, beta intercalated cell; LH (AL), loop of henle (ascending limb); LH (DL), loop of henle (descending limb); PC, collecting duct principal cell; PT (S1), proximal tubular (Segment 1); PT (S2), proximal tubular (Segment 2); PT (S3), proximal tubular (Segment 3)

Close modal
Table 1:
Protein sequence and homology analysis by BLAST for GSK3β in various organisms.
SpeciesAccessionGMQECoverageSeq identity
Homo sapiens NP_001139628.1 0.88 100% 100% 
Mus musculus NP_062801.1 0.88 100% 99.05% 
Rattus norvegicus NP_114469.1 0.88 100% 99.05% 
SpeciesAccessionGMQECoverageSeq identity
Homo sapiens NP_001139628.1 0.88 100% 100% 
Mus musculus NP_062801.1 0.88 100% 99.05% 
Rattus norvegicus NP_114469.1 0.88 100% 99.05% 

GMQE, global model quality estimate.

GSK3β is highly conserved in various organisms.

Figure 3:
GSK3β is highly conserved in various organisms.

Amino acid sequence alignment analysis of GSK3β in various organisms. Multiple sequence alignments were analyzed using the DNAMAN analysis software package (DNAMAN version 6.0).

Figure 3:
GSK3β is highly conserved in various organisms.

Amino acid sequence alignment analysis of GSK3β in various organisms. Multiple sequence alignments were analyzed using the DNAMAN analysis software package (DNAMAN version 6.0).

Close modal

Schematic of GSK3β signaling pathways.

Figure 4:
Schematic of GSK3β signaling pathways.

Following activation, GSK3β triggers a variety of signaling pathways, including Wnt/β-catenin pathway, NRF2 pathway, NF-κB pathway, and TGF-β/Smad pathway, which are likely implicated in the regulation of cellular function and homeostasis by GSK3β. Beyond these pathways, GSK3β plays an important role in cytoskeletal remodeling and mitochondrial regulation, impacting cellular motility, structural integrity and viability. AKI, acute kidney injury; CBP, CREB-binding proteins; CK1, casein kinase 1; CREB, cAMP response element-binding protein; CRMP2, collapsin response mediator protein 2; MPT, mitochondria permeability transition; NRF2, NF-E2-related factor 2; P, phosphorylation; Ub, ubiquitin; VDAC, voltage-dependent anion channel. The diagram was drawn by Figdraw.

Figure 4:
Schematic of GSK3β signaling pathways.

Following activation, GSK3β triggers a variety of signaling pathways, including Wnt/β-catenin pathway, NRF2 pathway, NF-κB pathway, and TGF-β/Smad pathway, which are likely implicated in the regulation of cellular function and homeostasis by GSK3β. Beyond these pathways, GSK3β plays an important role in cytoskeletal remodeling and mitochondrial regulation, impacting cellular motility, structural integrity and viability. AKI, acute kidney injury; CBP, CREB-binding proteins; CK1, casein kinase 1; CREB, cAMP response element-binding protein; CRMP2, collapsin response mediator protein 2; MPT, mitochondria permeability transition; NRF2, NF-E2-related factor 2; P, phosphorylation; Ub, ubiquitin; VDAC, voltage-dependent anion channel. The diagram was drawn by Figdraw.

Close modal

While GSK3β was initially identified as a cytoplasmic protein, recent studies have demonstrated its presence in other cellular compartments, including the nucleus and mitochondria [38]. Mitochondrial GSK3β plays a crucial role in regulating endoplasm-mitochondria calcium crosstalk [39], oxidative stress [40], and mitochondrial electron transport chain [41,42]. Even in the cytoplasm, GSK3β is not uniformly distributed but is compartmentalized in subcellular structures, such as growth cones and multivesicular bodies, suggesting that specific pools of GSK3β are involved in distinct cellular processes or signaling events. Indeed, inhibition of GSK3β through the insulin signaling pathway has a negligible effect on GSK3β-regulated Wnt/β-catenin signaling.

Biological function of GSK3β

The activity of GSK3β is tightly regulated by specific post-translational modifications. In general, the functions of GSK3β are dependent on tyrosine residue phosphorylation (Tyr216) as an active form. Conversely, serine residue phosphorylation (Ser9) is associated with an inhibitory status [43,44]. Ubiquitination at Lysine 183 leads to a reduction in GSK3β levels by promoting its degradation through the ubiquitin-proteasome pathway, while acetylation at Lysine 15 stabilizes GSK3β by modulating its functional activity [45-49].

As a kinase, GSK3β plays a crucial role in regulating numerous biological processes [18], either by catalyzing the phosphorylation of signaling proteins that serve as GSK3β substrates or by mediating the degradation of target proteins. To date, over 100 proteins, ranging from signaling transducers and transcription factors to cytoskeleton regulators, have been identified as substrates of GSK3β, undergoing phosphorylation and regulation by its activity. Dysregulation of GSK3β-mediated signaling and cellular processes contributes to the onset and progression of numerous diseases. In the kidney, GSK3β is highly expressed in renal parenchymal cells [23], and its deficiency in the mature kidney is associated with alterations in CKD [25-28]. These observations underscore the vital role of GSK3β in maintaining kidney integrity and overall physiological stability.

GSK3β acts as a central convergence point for multiple key cellular signaling pathways, orchestrating a range of critical processes that regulate cellular function and homeostasis. It plays a pivotal role in the Wnt/β-catenin pathway, which is crucial for cell proliferation, differentiation, development, as well as tissue injury, repair, and regeneration. GSK3β is also a pivotal modulator of the NRF2 pathway, which controls oxidative stress responses and cellular defense mechanisms. Moreover, GSK3β is integral to the TGF-β (the transforming growth factor-beta)/Smad3 signaling pathway, where it influences cellular differentiation and fibrogenesis. In addition, GSK3β interacts with the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, a key mediator of insulin signaling and cellular survival. GSK3β is involved in the NF-κB pathway, which is central to the inflammatory reaction and immune response. Beyond these pathways, GSK3β plays an important role in cytoskeletal remodeling and mitochondrial regulation, impacting cellular motility, structural integrity and viability. Through its broad involvement in these essential cellular processes, GSK3β emerges as a critical regulator of both normal cellular processes and pathogenesis of various diseases, including CKD (Figure 4).

GSK3β and Wnt/β-catenin signaling pathway

GSK3β functions as a negative regulator in the classical Wnt/β-catenin pathway by promoting the degradation of β-catenin [29]. In the absence of Wnt activation signals, the Wnt-specific scaffold protein Axin forms a complex with GSK3β, β-catenin, and casein kinase I. This complex facilitates the phosphorylation of β-catenin at Ser45 by casein kinase I, followed by phosphorylation at Thr41, Ser33, and Ser37 by GSK3β, leading to β-catenin ubiquitination and subsequent degradation. In contrast, when Wnt activation signals are present, Wnt activates Dvl, which, in conjunction with the GSK3β-binding protein Frat, facilitates the dissociation of the Axin complex [29,50-53]. This dissociation prevents β-catenin phosphorylation, allowing its stabilization and activation. Cross-talk between GSK3β and the Wnt/β-catenin pathway has been implicated in the development of renal fibrosis [54,55]. The role of GSK3β in the Wnt/β-catenin signaling pathway during kidney injury, however, remains controversial. Moderate activation of the Wnt/β-catenin pathway supports kidney cell survival and regeneration, promoting recovery and minimizing initial damage. Conversely, excessive Wnt/β-catenin activation can drive fibrosis by increasing extracellular matrix production, activating fibroblasts, exacerbating inflammation through up-regulation of cytokines and chemokines, resulting in podocyte injury along with interstitial fibrosis and tubular atrophy. Some studies have suggested that the ubiquitination and subsequent degradation of GSK3β may activate Wnt/β-catenin signaling, which in turn contributes to the progression of acute kidney injury (AKI) [56]. However, conditional ablation of GSK3β in podocytes had minimal effect on the β-catenin pathway and did not result in renal phenotypes in young mice, as demonstrated in our study [28] and supported by findings from Hurcombe et al. [24]. This lack of impact is likely due to compensation by GSK3α, which seems to function redundantly with GSK3β in mediating Wnt/β-catenin signaling [57].

GSK3β and NF-κB signaling pathway

The activation of NF-κB is a tightly regulated process initiated by upstream signaling cascades that activate the IκB kinase complex. Under basal conditions, NF-κB dimers, predominantly the p65/p50 heterodimer, are retained in the cytoplasm through interaction with the inhibitory protein IκB [22,58]. Upon stimulation, IκB kinase complex phosphorylates IκB, triggering its ubiquitination and subsequent proteasomal degradation. This process exposes a nuclear localization sequence on the NF-κB dimer, facilitating its rapid translocation into the nucleus, where it binds to specific DNA sequences and initiates the transcription of genes involved in inflammation [59]. GSK3β has been shown to enhance NF-κB transcriptional activity by phosphorylating key components, such as the p65 (RelA) subunit. More specifically, the C-terminal region of RelA/p65 contains a highly conserved domain that shares homology with the consensus docking sequence for GSK3β, which is essential for the efficient phosphorylation of Ser-468 and facilitating the physical interaction between RelA/p65 and GSK3β [60]. Additionally, GSK3β regulates NF-κB activation and RelB degradation through site-specific phosphorylation of BCL10, and it also influences histone modifications, which further modulate the expression of inflammatory genes by facilitating the binding of NF-κB and the p65/p50 complex to promoter regions [61-63]. GSK3β also interacts with PP2Ac to regulate NF-κB activity, and the novel GSK3β inhibitor 5n has been found to protect against AKI through PP2Ac-dependent mechanisms [64]. Furthermore, GSK3β inhibitors reduce oxidative stress and production of inflammatory mediators by inhibiting NF-κB p65 and IκB phosphorylation via the TLR/MyD88 pathway [65]. Mirabegron, through modulation of the GSK3β/NF-κB p65 signaling pathway, exerts protective effects on renal tissue and preserves renal function in the context of renal ischemia reperfusion [66]. Thus, GSK3β plays a crucial role in regulating inflammation via the NF-κB pathway, significantly contributing to renal inflammation and chronic kidney disease [67,68]. Targeting GSK3β therefore represents a promising therapeutic strategy for treating inflammation in CKD.

GSK3β and NRF2 signaling pathway

As the master regulator of antioxidant responses, Nrf2 is subjected to a complex and meticulously regulated pathophysiological process that is governed by various signaling pathways [26,69]. Under conditions of low oxidative stress, Nrf2 is retained in the cytoplasm, where it is bound to the inhibitory protein Kelch-like enoyl-CoA hydratase-associated protein 1 (Keap1). However, upon oxidative stress, Nrf2 is activated, following its dissociation from Keap1 and subsequent translocation into the nucleus. In the nucleus, Nrf2 binds to a conserved antioxidant response element (ARE), initiating the transcription of cytoprotective genes [70]. Following severe and persistent kidney injury, the sustained hyperactivity of GSK3β impairs the nuclear accumulation of Nrf2 through a Keap1-independent mechanism, exacerbating oxidative stress and playing a pivotal role in the progression from AKI to CKD [26]. Inhibition of GSK3β, for example by low-dose lithium, may exert protective effects through direct regulation of the Nrf2 antioxidant response [71]. In typical circumstances, short-term and mild kidney injury would activate the reperfusion injury salvage kinase (RISK) pathway, which is a major prosurvival signaling cascade that embraces the PI3K/Akt pathway and its downstream target, GSK3β [72]. Activation of the RISK pathway inhibits GSK3β, thereby preventing the degradation of NRF2, allowing NRF2 to accumulate, translocate to the nucleus, and activate the expression of antioxidant genes. This underscores the pivotal role of GSK3β in regulating NRF2 stability and function in CKD [73].

GSK3β and TGF-β/Smad signaling pathway

The TGF-β signaling pathway is a crucial regulator of various cellular processes, including proliferation, differentiation, apoptosis, and fibrogenesis [74-76]. The pathway is initiated when TGF-β ligands, such as TGFβ1, TGFβ2, and TGFβ3 (77-79), bind to TGF-β receptors [80,81], leading to the phosphorylation of receptor-regulated Smad proteins, mainly Smad2 and Smad3 [82,83]. These phosphorylated Smads then form a complex with Smad4, which translocates to the nucleus to regulate the transcription of specific target genes [84,85]. GSK3β is a key regulator of the TGF-β1 pathway [78]. In the heart, diminished GSK3β activity enhances TGF-β1-induced phosphorylation of Smad3 at the C-terminal Ser423/425 site (which leads to activation), indicating that GSK3β typically inhibits this phosphorylation [86]. Additionally, GSK3β controls Smad3 stability by phosphorylating it at the Thr66 site, leading to its ubiquitination and degradation [87].

The Smad linker region, situated between the MH1 and MH2 domains, plays a distinct role in Smad function compared to the C-terminal domain [88]. While phosphorylation of the C-terminal domain activates Smad, phosphorylation within the linker region inhibits its transcriptional activity [89]. It is proposed that GSK3β-mediated phosphorylation at Ser204 in the linker region provides an additional mechanism to suppress Smad3 activity [90]. Furthermore, GSK3β inhibition enhances the activity of cAMP response element-binding protein (CREB), which competes for CREB-binding protein (CBP) — a transcriptional coactivator essential for TGF-β1/Smad signaling to drive the profibrogenic plasticity in tubular epithelial cells (TECs) [91]. GSK3β also indirectly influences TGF-β/Smad signaling through cross-regulation with other signaling pathways, including Wnt/β-catenin signaling [29,92-94].

GSK3β and PI3K/Akt signaling pathway

The PI3K/Akt signaling is the core of the prosurvival RISK pathway that is triggered by insulin or a variety of growth factors. It represents a critical transduction cascade that is essential for orchestrating a range of essential cellular processes, including proliferation, survival, metabolism, migration, and apoptosis [95,96]. Activation of PI3K results in the production of phosphatidylinositol [3,4,5]-triphosphate, which activates Akt. Subsequently, Akt phosphorylates a diverse range of target proteins, thereby regulating a multitude of cellular functions [95,96]. A key target of the PI3K/Akt pathway is GSK3β [97]. Under basal conditions, GSK3β is active, regulating glycogen biosynthesis, cell cycle progression, and apoptosis. However, upon Akt activation, GSK3β is phosphorylated at the Ser9 residue, resulting in its inactivation [98]. This inactivation is particularly significant in processes such as TGF-β1-induced epithelial-mesenchymal transition (EMT) [98]. Akt phosphorylates and inactivates GSK3β, leading to the stabilization of proteins such as cyclin D1, indicating the role of the PI3K/Akt/GSK3β signaling pathway in regulating cell survival and proliferation [99]. Notably, while GSK3β is generally inactivated by the PI3K/Akt pathway upon insulin treatment, exposure of kidney cells to a type 2 diabetic milieu containing high ambient insulin and glucose as well as microinflammation causes a reduction in Ser9 phosphorylation, leading to GSK3β hyperactivity. This overactivated GSK3β can then phosphorylate insulin receptor substrate 1 (IRS1) at the Ser332 site, impairing insulin signaling and contributing to insulin resistance, a condition often associated with diabetic nephropathy [100].

GSK3β and cytoskeleton remodeling

The cytoskeleton is essential for maintaining cellular shape, internal organization, and polarity in various cell types, including kidney cells. GSK3β is a central regulator of cytoskeletal dynamics, playing a crucial role in maintaining kidney cell function and influencing cellular response to injury [101-103]. It modulates microtubule-associated proteins (MAPs) crucial for microtubule stability and organization [104] and influences the actin cytoskeleton by regulating Rho family GTPases, which are vital for cell migration and adhesion [105,106]. As highly specialized epithelial cells that cover the exterior of the glomerular capillary and regulate glomerular permeability, podocytes possess an enriched actin and microtubule cytoskeletal network. Proper balance in cytoskeletal dynamics is critically required for podocytes to maintain the integrity of the glomerular filtration barrier. Recent research has highlighted the role of GSK3β in the dynamic remodeling of the cytoskeleton in podocytes [102]. GSK3β regulates the activity of MAPs, such as Tau and collapsin response mediator protein 2 (CRMP2), which are vital for microtubule polymerization and stability [107,108]. Upon podocyte injury induced by adriamycin, GSK3β is overactivated, and this results in the hyperphosphorylation of Tau and CRMP2, leading to microtubule destabilization, retraction of podocyte processes, and cellular dysfunction. Conversely, inhibition of GSK3β, as observed with lithium treatment, counteracts these effects by stabilizing microtubules and promoting extension and repair of podocyte processes, which are essential for glomerular adaptation to podocyte loss [102]. Dysregulation of GSK3β has been associated with CKD such as diabetic nephropathy and glomerulosclerosis, where changes in cytoskeletal dynamics exacerbate disease progression [109]. Thus, GSK3β emerges as a key integrator of cellular function and structure in kidney cells, making it a promising therapeutic target.

CKD is a heterogeneous condition caused by a wide range of etiologies, each with distinct underlying pathomechanisms, clinical presentations, and progression patterns, all of which led to chronic impairment of kidney function and structure. CKD is characterized by progressive decline in kidney function and gradual, persistent renal destruction, accompanied by complex pathological features, including glomerulosclerosis, interstitial fibrosis, tubular atrophy, and inflammatory infiltration, all of which contribute to disease progression [110-112]. Accumulating evidence indicates that GSK3β is aberrantly activated in injured renal parenchymal cells, where it exacerbates CKD by promoting fibrogenesis, glomerulosclerosis, and tubulointerstitial damage [25,26,91,113-115]. Mechanistically, GSK3β participates in multiple pathogenic pathways, including the Wnt/β-catenin pathway, TGF-β/Smad pathways, inflammation cascades, and apoptosis regulation. Its hyperactivity is particularly prominent in DKD, correlating with glomerular injury, tubular damage, and proteinuria [16,109,113]. In addition, inhibiting GSK3β may effectively slow the progression from AKI to CKD by mitigating renal fibrosis [25,26]. Beyond parenchymal renal cells, GSK3β also modulates immune responses across multiple immune cell populations, which also significantly influence kidney inflammation and CKD pathogenesis. In macrophages, dendritic cells, B lymphocytes, and T lymphocytes, GSK3β governs cytokine production, metabolic reprogramming, antigen presentation, and immune polarization—processes that may underpin both acute and chronic immune-mediated renal inflammation [116-118]. Based on these findings, GSK3β is considered a crucial therapeutic target in CKD.

GSK3β in polycystic kidney disease

Polycystic kidney disease (PKD) is an inherited disorder characterized by the gradual formation and expansion of fluid-filled cysts in the kidneys. These cysts exert pressure on healthy renal tissue, and this can eventually lead to impaired kidney function [119-122]. The pathogenesis of PKD is intricate, involving dysregulated cell proliferation, abnormal fluid secretion, and aberrant extracellular matrix remodeling within the kidneys. Recent studies have highlighted the critical involvement of GSK3β in PKD pathogenesis, particularly in regulating epithelial cell differentiation, polarity, and proliferation[123]—processes that are disrupted in PKD. GSK3β expression is significantly elevated in collecting ducts in PKD, where it mediates responses to AVP, leading to increased proliferation and fluid secretion [124-126]. Collecting ducts are central to cyst formation, and GSK3β inhibition has been shown to reduce cyst formation and renal cyst hyperplasia [126,127]. Elevated renal cAMP levels are a hallmark of PKD [128,129]. Notably, GSK3β modulates AVP-sensitive adenylate cyclase activity and cAMP production. Targeted GSK3β inhibition or renal-specific GSK3β KO can markedly diminish adenylate cyclase activity and cAMP accumulation [126,130,131]. GSK3β operates within a feed-forward mechanism involving cAMP- and CREB-mediated signaling pathways. In this loop, elevated cAMP levels enhance the activity of the transcription factor CREB, which binds to the GSK3β promoter region, thereby upregulating the expression of GSK3β [127]. In turn, GSK3β positively regulates the production of cAMP. The inactivation or deletion of GSK3β suppresses adenylate cyclase activity and reduces cAMP synthesis [131]. In PKD, FoxM1 promotes cyst growth in PKD1 mutant ADPKD [132]. Conversely, in AKI, GSK3β acts as an upstream suppressor of FoxM1, with GSK3β inhibition or gene deletion leading to increased FoxM1 expression and improved tubular repair [27]. Reasons for the above conflicting results are unclear but highlight the dual role of FoxM1, warranting further investigation to elucidate the underlying mechanisms. While GSK3β’s role in later-stage PKD, particularly in cyst expansion and fibrosis, is well-documented, its involvement in the early stages of the disease remains less clear [126,131,133]. In juvenile rats, LiCl exposure induced microcysts in collecting ducts, whereas in adult rats, prolonged LiCl treatment increased cell proliferation without cyst formation [134,135]. Kjaersgaard et al. found an inverse association between renal GSK3β activity and postnatal kidney development. Specifically, GSK3β facilitates cell proliferation at early stages, whereas its elevated abundance or activity sustains an anti-proliferative effect in mature kidneys. Lithium, by acting on the distal nephron—particularly early collecting duct epithelial cells—attenuates GSK3β activity, leading to dysregulated cell proliferation associated with dilatations and microcysts and subsequent structural changes [135]. Previous research has associated chronic lithium treatment with renal microcyst formation [136], and even rarely with concomitant occurrence of PKD [137]. Autopsy findings from long-term lithium users revealed microcysts and tubular dilation in both renal cortex and medulla [138], with structural alterations most prominent in the collecting ducts and distal tubules [139]. These lithium-induced microcysts are GSK3-positive with enhanced inhibitory phosphorylation of GSK3β at serine 9, and the cyst epithelium is proliferative [140]. In human biopsy samples from lithium users, renal microcysts show AQP2 positivity, implicating the collecting duct as their likely origin [135]. β-catenin is a key target of GSK3 signaling, and overexpression of β-catenin in murine renal epithelial cells has been shown to cause severe cystic changes shortly after birth [141]. As a classic GSK3 inhibitor, lithium at high doses activates the Wnt/β-catenin pathway [142]. Thus, it is plausible that renal microcyst formation following chronic lithium treatment could, at least in part, be attributed to the overactivation of the β-catenin pathway. However, studies by Doble et al. [57] demonstrate that inactivation of at least three of the four alleles of GSK3α and GSK3β is necessary to induce significant changes in β-catenin activity. This implies that there may be a potential therapeutic window during which pharmacologic GSK3β inhibition could be achieved without altering β-catenin activities in peripheral organs such as the kidney. While long-term high-dose lithium use for treating psychiatric disorders is associated with an increased risk of renal toxicity, recent evidence indicates that short-term low-dose lithium may suppress GSK3β activity without activating β-catenin, paradoxically offering renoprotective effects.

GSK3β in DKD

DKD is one of the most common complications of diabetes mellitus and is a principal cause of chronic kidney disease. Its prevalence is on the rise, correlating with the escalating incidence of diabetes mellitus [143,144]. As a key transducer of insulin signaling, GSK3β plays a central role in regulating glucose homeostasis by phosphorylating glycogen synthase, thereby affecting glycogen synthesis, insulin signaling, and glucose metabolism [31,145]. Dysregulation of GSK3β has been associated with insulin resistance and the development of DKD [16,109,113,146,147].

As a diagnostic criterion of DKD, albuminuria is an indicator of early impairment of the glomerular filtration barrier by diabetes. Podocytes are a crucial constituent of the glomerular filtration barrier. In the progression of DKD, they can undergo apoptosis, detachment, or structural changes like foot process effacement, cytoskeletal remodeling, and glomerular basement membrane thickening, all of which contribute to the disruption of the filtration barrier [102,148,149]. Recent experimental and clinical studies have highlighted the important role of GSK3β in the pathogenesis of DKD and podocyte injury. For example, Paeng et al. [149] demonstrated that GSK3β is overactive in podocytes exposed to high ambient glucose, contributing to podocyte apoptosis. Additionally, GSK3β hyperactivity has been observed in glomerular podocytes under type 2 diabetic conditions, associated with IRS1 hyperphosphorylation, impaired Nrf2 antioxidant response, and premature senescence, underscoring its potential as a therapeutic target in type 2 diabetic glomerular injury [43]. Sarsasapogenin has been shown to ameliorate experimental DKD by targeting the GSK3β signaling pathway. This beneficial effect is associated with restoration of podocyte autophagy [150]. Similarly, hepatocyte growth factor has been found to prevent high glucose (HG)-induced podocyte injury through an autophagy-promoting mechanism that is dependent on GSK3β inhibition [151], further implicating the PI3K/Akt-GSK3β axis in lysosomal function improvement and DKD protection [152]. TDAG51 deficiency has been identified as a protective factor against HG-induced podocyte damage, primarily through the activation of Nrf2 by modulating the AKT-GSK3β pathway, further illustrating the multifaceted role of GSK3β in diabetic podocyte injury [153]. These findings collectively underscore the potential of GSK3β as a central therapeutic target for mitigating podocyte injury and delaying the progression of DKD. A number of pharmacological agents have GSK3β inhibitory activity. For instance, β-hydroxybutyrate is able to directly target the ATP-binding pocket of GSK3β and thereby block its kinase activity, as evidenced by molecular modeling and docking analysis. Through inhibiting GSK3β, β-hydroxybutyrate reinforces Nrf2 antioxidant response in glomerular podocytes, resulting in reduced senescence and glomerular injury in mouse models of type 1 DKD [35]. It is conceivable that therapeutic targeting of GSK3β for treating DKD is feasible by using existing or novel medications with GSK3β inhibitory activities.

In addition to glomerular podocytes, glomerular mesangial cells and renal TECs are injured in DKD and are involved in the pathogenesis of DKD. A growing body of evidence suggests that GSK3β also plays a role in diabetic mesangial and tubular injuries [154,155]. DKD may present with mesangial cell proliferation and expansion of mesangial matrix, leading to the formation of typical Kimmelstiel–Wilson nodules. Lin et al. [156] demonstrated that HG activates GSK3β and caspase-3, leading to apoptosis in glomerular mesangial cells. Pharmacological inhibition of GSK3β with specific inhibitors like bromoindirubin-3'-oxime or LiCl counteracts these effects by enhancing Akt phosphorylation and β-catenin translocation. Furthermore, knockdown of GSK3β reduces proliferation of mesangial cells in the DKD [157]. Diabetic kidney injury is accompanied by renal tubulointerstitial lesions, which are known to be closely associated with the decline in renal function [158,159]. GSK3β is overactivated in renal tubular cells subjected to hyperglycemic stimulation. Inhibiting the GSK3β/β-TrCP axis enhances NRF2-ARE binding and stabilizes NRF2, which in turn alleviates renal tubular injury [160]. Phosphorylated GSK3β plays a role in the elevation of SREBP-1 expression and lipid deposition in renal tubular cells following HG treatment [161]. Consistent with these, the results of renal biopsy specimens from patients at different stages of diabetic nephropathy demonstrated a consistent increase in the expression of total and activated GSK3β in glomeruli and tubules, correlated with the severity of diabetic nephropathy [162]. Contrary to the above data suggesting the pathogenic role of GSK3β hyperactivity in DKD, Mariappan et al. found that activation of GSK3β has a beneficial effect on diabetes-induced kidney damage [163]. They administered sodium nitroprusside, a nitric oxide donor that is able to activate GSK3β, to mice with streptozotocin-induced type 1 diabetes and observed improvements in renal hypertrophy, matrix protein accumulation, and albuminuria, independent of any correction of hyperglycemia. Although nitroprusside activates GSK3β, it is not a GSK3β-selective agonist. Additionally, nitroprusside is known to exert hypotensive effects, which may confound the interpretation of the improved albuminuria and diabetic kidney injury in their study. Collectively, while most studies suggest a detrimental role of GSK3β activation in DKD, to conclusively define the role of GSK3β in DKD, it is necessary to employ DN models with kidney-specific GSK3β hyperactivity or KO, or to use highly selective and rigorous inhibitors.

GSK3β in glomerular diseases

Glomerular diseases represent one of the leading causes of CKD [4]. The progression of glomerular disease is dependent on the damage to glomerular cells, such as glomerular podocytes. Injury to glomerular cells often results in the hallmark clinical features, including proteinuria and a decline in renal function [164]. While the exact pathomechanisms of glomerular injury are obscure, converging evidence recently points to a role for GSK3β.

In cultured glomerular podocytes, doxorubicin induced cell death and disruption of the actin cytoskeleton, accompanied by marked overactivation of GSK3β and minimal activation of Nrf2. Ectopic expression of a kinase-dead mutant of GSK3β in cultured podocytes enhanced doxorubicin-induced Nrf2 activation and mitigated podocyte injury. In contrast, expression of the constitutively active mutant of GSK3β attenuated the Nrf2 response to doxorubicin and exacerbated podocyte damage [73]. In mouse models of doxorubicin nephropathy and nephrotoxic serum nephritis, podocyte-specific KO of GSK3β or pharmacological inhibition with SB216763 (a highly selective inhibitor of GSK3) significantly ameliorated podocyte injury and reduced albuminuria. This effect was primarily dependent on the restoration of NRF2 antioxidant response [73,165]. GSK3β inhibitor (such as TDZD-8), or targeted GSK3β KO in podocytes, has been demonstrated to preserve actin cytoskeleton integrity following adriamycin insult [28,40]. These interventions decreased the phosphorylation and activation of paxillin, a focal adhesion-associated adaptor protein, and reduced podocyte hypermotility [166]. Notably, a discrete pool of GSK3β located within podocyte mitochondria interacts with and phosphorylates cyclophilin F, a critical component of the MPT. Treatment with TDZD-8 effectively inhibits this phosphorylation event, thereby preventing the onset of oxidative stress-induced mitochondrial dysfunction and offering substantial protection against podocyte death [40]. Recent studies have extended the pathogenic scope of GSK3β beyond lipopolysaccharide (LPS)- or adriamycin-induced models to other glomerular diseases relevant to human pathology. In kidney biopsies from patients with primary focal segmental glomerulosclerosis (FSGS), increased expression of phosphorylated GSK3β in podocytes was observed [167]. Moreover, glomerular expression of GSK3β was elevated by 2.6-fold in FSGS and 3.1-fold in lupus nephritis in human kidney biopsies based on computerized morphometric analysis [28]. Elevated GSK3β levels were also detected in peripheral blood mononuclear cells from patients with FSGS and membranous nephropathy [168]. Furthermore, GSK3β contributes to lupus nephritis development in lupus-prone mice, in part through activation of the NLRP3/IL-1β axis. Notably, the selective GSK3β inhibitor TDZD-8 effectively blocked NLRP3/IL-1β activation in peripheral blood mononuclear cells from patients with systemic lupus erythematosus [117].

In addition to the regulation of NRF2-mediated antioxidant response, podocyte cytoskeleton integrity and MPT, GSK3β plays a pivotal role in the canonical Wnt/β-catenin signaling pathway which has been recently implicated in primary glomerular disorders [67,68,115,142]. Balanced Wnt/β-catenin signaling in podocytes is known to be critical for glomerular filtration barrier maintenance [169]. In consistency, Dai et al. [142] triggered Wnt/β-catenin signaling in mice with high-dose LiCl, resulting in podocyte damage and transient proteinuria. Although these findings may align with clinical observations of lithium nephrotoxicity [170-172], it is important to highlight that lithium nephrotoxicity is uncommon in clinical practice, and its severity is influenced by both the dose and duration of lithium administration [173]. The current FDA-approved dose of lithium is high to ensure that lithium effectively crosses the blood-brain barrier for the treatment of psychiatric disorders [174,175]. In contrast, our recent study demonstrates that low-dose lithium can effectively inhibit GSK3β activity and confer protection on peripheral organs, including the kidneys, without causing adverse side effects [35,176,177]. Additionally, podocyte-specific KO of GSK3β showed minimal impact on the β-catenin pathway, and this is likely due to the functional redundancy between GSK3β and GSK3α [24,28,57]. Therefore, the use of highly selective GSK3β inhibitors or lithium at low doses presents a promising therapeutic approach for the treatment of glomerular diseases.

Progressive CKD is associated with de novo acquisition of pro-inflammatory phenotypes by kidney epithelial cells, like glomerular podocytes, contributing to renal inflammation and dysfunction. NF-κB signaling plays a central role in this pro-inflammatory phenotypic shift and can be modulated by GSK3β. NF-κB acts as a double-edged sword in the pathogenesis of glomerular diseases [67,68,178-180]. On one hand, NF-κB directs the transcription of a multitude of injurious mediators involved in podocyte injury, most of which are related to inflammatory response and immune reaction, including proinflammatory cytokines (e.g. MCP-1) and immune regulatory cathepsin L and B7-1, which also have been implicated in podocyte cytoskeleton disorganization [181]. On the other hand, NF-κB is a survival factor essential for cellular response to stress or injury, functioning for self-protection. The actual pathobiological effects of NF-κB are dictated by its post-translational modifications, such as phosphorylations, whose patterns may influence the recruitment of different transcriptional cofactors, direct distinct profiles of gene expression, and result in varying and even opposing actions. Broad-range inhibition of NF-κB unselectively suppresses the expression of a broad range of NF-κB target genes, mitigates podocyte acquisition of pro-inflammatory phenotypes, such as expression of MCP-1, cathepsin L, and B7-1, but unfortunately sensitizes podocytes to death. In contrast, GSK3β phosphorylates NF-κB specifically at serine 467, which specifies the expression of selective NF-κB target molecules, including podocytopathic mediators like MCP-1, cathepsin L, and B7-1, but not the pro-survival Bcl-xL [182]. Inhibition of GSK3β by lithium or TDZD-8 mitigated the expression of podocytopathic mediators, ameliorated podocyte injury, but barely affected Bcl-xL expression or sensitized apoptosis. In vivo, in mice with LPS or adriamycin-induced podocytopathy, GSK3β inhibitors attenuated RelA/p65 phosphorylation specifically at serine 467 and improved proteinuria and podocyte injury. In contrast, TPCK, a broad-range inhibitor of NF-κB, improved albuminuria and podocyte injury by a much lesser extent but aggravated glomerular cell apoptosis. Collectively, GSK3β-directed fine-tuning of NF-κB might serve as a novel therapeutic target for glomerular disease.

GSK3β in renal tubulointerstitial fibrosis

Renal fibrosis is a hallmark of progressive CKD, characterized by tubular atrophy and the excessive deposition of extracellular matrix (ECM) within the tubulointerstitium. Regardless of the original etiology of kidney disease—whether ischemia-reperfusion, nephrotoxicity, or renal obstruction—the renal tubule possesses an intrinsic capacity for self-repair. However, when the extent of injury surpasses the adaptive ability of the kidney for self-repair, a cascade of maladaptive kidney repair may be triggered. These include G2/M cell cycle arrest, cellular dedifferentiation, premature senescence, and the production of numerous fibrotic and pro-inflammatory mediators [183,184].

In mouse models of acute tubular injury, the tubular activity of GSK3β was significantly increased. Inhibition of GSK3β ameliorated renal injury by ameliorating tubular cell apoptosis and injury, indicating the potential value of GSK3β in blocking the progression of AKI and the subsequent transformation to CKD [185]. Analysis of the renal transcriptome database showed that patients with progressive CKD exhibited GSK3β overexpression in the renal tubulointerstitium, where the pre-defined hallmark gene sets involved in fibrogenesis were remarkably enriched [91]. Our study demonstrated that targeted inhibition of GSK3β in renal tubules—either through genetic KO or inhibitor treatment—potentiates Nrf2 antioxidant response via a Keap1-independent mechanism to ameliorate renal oxidative stress and attenuate AKI to CKD transition [26]. Additionally, GSK3β inhibition promotes CREB activity in renal tubules in FA-elicited progressive CKD [91]. CREB competes for CBP, a transcriptional coactivator essential for the TGF-β1/Smad signaling pathway to drive the profibrogenic plasticity of renal TECs. This resulted in downregulation of the transcriptional activity of the TGFβ1/Smad pathway, leading to an amelioration of renal fibrosis. Mechanistically, it is postulated that CREB serine 129 lies in the consensus sequence motif of GSK3β, and phosphorylation by GSK3β at serine 129 may act as a suppressive signal for CREB activity [186-188]. Consistently in mouse models of progression of AKI caused by ischemia-reperfusion injury to CKD, Singh et al. [189] demonstrated that pharmacological inhibition of GSK3β using TDZD-8 significantly attenuated the development of renal fibrosis. This protective effect was primarily mediated through the suppression of the TGFβ1/Smad3 signaling pathway, which in turn reduced the deposition of extracellular matrix components such as collagen-1 and fibronectin. Moreover, constitutive activation of GSK3β was found to directly enhance α-SMA expression, further implicating GSK3β in the fibrotic process. Additional recent evidence demonstrates that inhibition of GSK3β enhances transcription factor EB -mediated lysosomal synthesis and facilitates ECM degradation, thereby improving renal fibrosis [190].

A variety of medications with GSK3β inhibitory activities have been used to treat a range of diseases, and several of these have already undergone clinical trials (Table 2, https://clinicaltrials.gov/). However, there remains a significant lack of clinical studies evaluating GSK3β inhibitors for renal diseases [191]. In fact, the majority of research on GSK3β inhibitors in the context of kidney disease is still in the preclinical phase, relying on animal models and in vitro experiments (Table 3). The potential of specific GSK3β inhibitors in treating CKD is increasingly supported by preclinical findings, particularly those involving GSK3β-regulated pathways implicated in renal inflammation, fibrosis, and dysfunction. Among the many tested compounds, lithium, TDZD-8, Tideglusib, and SB216763 are notable for their demonstrated efficacy in reducing kidney injury and fibrosis, making them promising candidates for CKD therapy.

Table 2:
List of selected GSK3β inhibitors in clinical trials.
InhibitorsIndicationsClinical trialsStudy phaseTarget or pathway
Lithium Cardiac surgery associated AKI NCT03056248 Phase 4 PI3K/Akt, JAK/STAT3 
 Parkinson’s Disease NCT04273932 Phase 1 Wnt/β-catenin 
 Osteosarcoma NCT01669369 Phase 4 MAPK 
 Stroke NCT01112813 Phase 3 AKT/GSK3β/β-catenin and AKT/FoxO3a/β-catenin 
 Amyotrophic lateral sclerosis NCT00790582 Phase 2 Autophagy 
 Low-Grade neuroendocrine tumors NCT00501540 Phase 2 GSK3β 
 Long COVID NCT06108297 Phase 1 Inflammation 
9‐ING‐41 Myelofibrosis NCT04218071 Phase 2 GSK3β 
 Pancreatic cancer NCT05077800 Phase 2 GSK3β, β-catenin, chemotherapy resistance 
 Salivary gland carcinoma NCT05010629 Phase 2 GSK3β 
 Advanced cancers NCT03678883 Phase 2 GSK3β, PD-1, TIGIT, LAG-3, CXCR3 
LY2090314 Metastatic pancreatic cancer NCT01632306 Phase 2 Cell proliferation 
 Acute leukemia NCT01214603 Phase 2 GSK3α, GSK3β 
 Advanced or metastatic cancer NCT01287520 Phase 1 GSK3α, GSK3β 
Tideglusib Alzheimer´S disease NCT01350362 Phase 2 Tau 
 Alzheimer´S disease NCT00948259 Phase 2 Tau 
InhibitorsIndicationsClinical trialsStudy phaseTarget or pathway
Lithium Cardiac surgery associated AKI NCT03056248 Phase 4 PI3K/Akt, JAK/STAT3 
 Parkinson’s Disease NCT04273932 Phase 1 Wnt/β-catenin 
 Osteosarcoma NCT01669369 Phase 4 MAPK 
 Stroke NCT01112813 Phase 3 AKT/GSK3β/β-catenin and AKT/FoxO3a/β-catenin 
 Amyotrophic lateral sclerosis NCT00790582 Phase 2 Autophagy 
 Low-Grade neuroendocrine tumors NCT00501540 Phase 2 GSK3β 
 Long COVID NCT06108297 Phase 1 Inflammation 
9‐ING‐41 Myelofibrosis NCT04218071 Phase 2 GSK3β 
 Pancreatic cancer NCT05077800 Phase 2 GSK3β, β-catenin, chemotherapy resistance 
 Salivary gland carcinoma NCT05010629 Phase 2 GSK3β 
 Advanced cancers NCT03678883 Phase 2 GSK3β, PD-1, TIGIT, LAG-3, CXCR3 
LY2090314 Metastatic pancreatic cancer NCT01632306 Phase 2 Cell proliferation 
 Acute leukemia NCT01214603 Phase 2 GSK3α, GSK3β 
 Advanced or metastatic cancer NCT01287520 Phase 1 GSK3α, GSK3β 
Tideglusib Alzheimer´S disease NCT01350362 Phase 2 Tau 
 Alzheimer´S disease NCT00948259 Phase 2 Tau 

AKI, acute kidney injury. COVID, corona virus disease.

Table 3:
GSK3β inhibitors in animal models.
InhibitorsSpeciesModelConclusionReference
Lithium C57BL/6 mice STZ injection Protects islet β-cells via Nrf2 activation [192
 C57BL/6 mice Ageing Anti-senescence [35
 C57BL/6 mice Cisplatin or IRI AKI Promotes recovery from AKI [176
 C57BL/6 mice Cisplatin AKI Improve autophagy [177
 C57BL/6 mice FA injury Anti-fibrosis [91
TDZD8 C57BL/6 mice LPS Anti-inflammation [67
 C57BL/6 mice Doxorubicin injection Desensitizing MPT [40
SB216763 C57BL/6 mice Doxorubicin injection Reinforces the Nrf2 antioxidant [73
Tideglusib db/db mice Protects podocytes via Nrf2 activation [43
InhibitorsSpeciesModelConclusionReference
Lithium C57BL/6 mice STZ injection Protects islet β-cells via Nrf2 activation [192
 C57BL/6 mice Ageing Anti-senescence [35
 C57BL/6 mice Cisplatin or IRI AKI Promotes recovery from AKI [176
 C57BL/6 mice Cisplatin AKI Improve autophagy [177
 C57BL/6 mice FA injury Anti-fibrosis [91
TDZD8 C57BL/6 mice LPS Anti-inflammation [67
 C57BL/6 mice Doxorubicin injection Desensitizing MPT [40
SB216763 C57BL/6 mice Doxorubicin injection Reinforces the Nrf2 antioxidant [73
Tideglusib db/db mice Protects podocytes via Nrf2 activation [43

AKI , acute kidney injury. FA, folic acid. IRI, ischemia-reperfusion injury. LPS, lipopolysaccharide. MPT, mitochondria permeability transition. NRF2, NF-E2-related factor 2. STZ, streptozotocin. .

Lithium, a competitive inhibitor of GSK3 with respect to magnesium (Mg2+), has been clinically used for over 50 years as an FDA approved first-line treatment for bipolar affective disorders (Figure 5A), [193]. It is one of the earliest GSK3β inhibitors identified and has been extensively studied for its kidney effects. In kidney disease models, low-dose lithium has shown effectiveness in reducing oxidative stress and fibrosis by stabilizing the Nrf2 antioxidant response and mitigating pro-inflammatory signaling. Interestingly, studies indicate that delayed administration of lithium can also improve outcomes in AKI, reducing the risk of transition to CKD. The renoprotective dose of lithium is much lower than that used for psychiatric diseases, emphasizing the need for precise dosing strategies in CKD to avoid adverse effects while maximizing therapeutic benefit. A randomized, double-blinded, placebo-controlled pilot trial has been conducted on patients undergoing cardiac surgery with cardiopulmonary bypass to assess the effectiveness of short-term low-dose lithium therapy in averting cardiac surgery-associated AKI [191].

2D chemical structure of the GSK3β inhibitor.

Figure 5:
2D chemical structure of the GSK3β inhibitor.

(A) 2D chemical structure of lithium chloride. (B) 2D chemical structure of TDZD-8. (C) 2D chemical structure of SB216763. (D) 2D chemical structure of Tideglusib. The 2D chemical structure were download from Pubchem (https://pubchem.ncbi.nlm.nih.gov/).

Figure 5:
2D chemical structure of the GSK3β inhibitor.

(A) 2D chemical structure of lithium chloride. (B) 2D chemical structure of TDZD-8. (C) 2D chemical structure of SB216763. (D) 2D chemical structure of Tideglusib. The 2D chemical structure were download from Pubchem (https://pubchem.ncbi.nlm.nih.gov/).

Close modal

TDZD-8, a highly selective non-ATP competitive GSK3β inhibitor, has shown strong efficacy in attenuating fibrosis and inflammation in CKD models (Figure 5B) [40]. By modulating the TGF-β/Smad3 pathway, TDZD-8 reduces fibrotic signaling, making it particularly effective in models of ischemia-reperfusion injury and obstructive nephropathy, where fibrosis is a key pathological feature.

SB216763, a selective ATP-competitive inhibitor of GSK3β, has demonstrated efficacy in reducing podocyte injury and tubular apoptosis in CKD models (Figure 5C). Its ability to activate the Nrf2 antioxidant pathway and reduce NF-κB-driven inflammation makes SB216763 a promising candidate for therapeutic intervention in CKD, supporting both podocyte and tubular cell survival.

Tideglusib, a non-ATP competitive inhibitor of GSK3β, originally developed for neurodegenerative diseases, exhibits renoprotective effects in DKD models by reducing podocyte damage and oxidative stress through GSK3β modulation (Figure 5D) [43]. Tideglusib’s efficacy in protecting glomerular cells suggests it may benefit patients with glomerular forms of CKD, especially those with diabetes-related renal complications.

While these GSK3β inhibitors offer significant promise for CKD treatment, challenges remain, including optimizing dose regimens, enhancing renal selectivity, and evaluating long-term safety. Nonetheless, these inhibitors highlight the potential of GSK3β-targeted therapies to slow CKD progression by addressing core molecular drivers of kidney injury, underscoring the need for continued research into precise and effective therapeutic strategies for CKD.

The central role of GSK3β in the progression of CKD is now widely acknowledged. CKD, often stemming from primary conditions such as diabetes and glomerular diseases, progresses through intricate mechanisms driven by chronic inflammation, apoptosis, and fibrosis. GSK3β acts as a key regulatory kinase within multiple signaling pathways, including Wnt/β-catenin, NF-κB, Nrf2, TGFβ-Smad, which together orchestrate critical aspects of kidney damage in CKD. A growing body of evidence provides comprehensive data on GSK3β’s functions across various kidney diseases, such as DKD, PKD, and glomerular diseases, with a focus on its impact on inflammatory responses, fibrotic transformation, and oxidative stress. Evidence from preclinical models indicates that GSK3β inhibition may serve as a therapeutic approach, potentially reducing renal injury and fibrosis while promoting cellular repair. Nonetheless, critical questions remain regarding the long-term effectiveness, specificity, and safety of GSK3β-targeted therapies, with particular concern for potential off-target effects and variability in response across renal cell types. Additionally, debate persists about GSK3β’s mechanistic role in diverse renal pathologies, suggesting further complexity in its function and regulation.

Despite these uncertainties, GSK3β remains a promising therapeutic target with broad implications for CKD management. This review highlights the need for continued research to elucidate the precise functions of GSK3β within renal pathophysiology and to develop effective, targeted therapies that may improve outcomes for patients suffering from CKD and related renal diseases.

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

This work was supported in part by the University of Toledo incentive funds and by grant R01DK133203 and U01AG086161 from the US National Institutes of Health. The funders had no role in the design and conduct of this study, collection and interpretation of the data, or preparation and approval of the manuscript.

M.Z.: Writing—original draft. M.T.: Writing—review & editing. R.G.: Writing—review & editing, Conceptualization, Funding acquisition.

All authors thank Figdraw (www.figdraw.com) for providing the illustration tool, and Dr. Ashaq Hussain for proofreading the manuscript.

AKI

acute kidney injury

ARE

antioxidant response element

CKD

chronic kidney disease

CREB

cAMP response element-binding protein

CRMP2

collapsin response mediator protein 2

DKD

diabetic kidney disease

GSK3β

glycogen synthase kinase 3 beta

HG

high glucose

KEAP1

Kelch-like enoyl-CoA hydratase-associated protein 1

LPS

lipopolysaccharide

MAPS

microtubule-associated proteins

MPT

mitochondria permeability transition

NRF2

NF-E2-related factor 2

PKD

Polycystic kidney disease

RISK

reperfusion injury salvage kinase

TECs

tubular epithelial cells

1
Cockwell
,
P.
and
Fisher
,
L.A
. (
2020
)
The global burden of chronic kidney disease
.
The Lancet
395
,
662
664
https://doi.org/10.1016/S0140-6736(19)32977-0
2
Kalantar-Zadeh
,
K.
,
Jafar
,
T.H.
,
Nitsch
,
D.
,
Neuen
,
B.L.
and
Perkovic
,
V
. (
2021
)
Chronic kidney disease
.
The Lancet
398
,
786
802
https://doi.org/10.1016/S0140-6736(21)00519-5
3
Yang
,
C.
,
Wang
,
H.
,
Zhao
,
X.
,
Matsushita
,
K.
,
Coresh
,
J.
,
Zhang
,
L.
et al.
(
2020
)
CKD in China: evolving spectrum and public health implications
.
Am. J. Kidney Dis.
76
,
258
264
https://doi.org/10.1053/j.ajkd.2019.05.032
4
Hu
,
J.
,
Ke
,
R.
,
Teixeira
,
W.
,
Dong
,
Y.
,
Ding
,
R.
,
Yang
,
J.
et al.
(
2023
)
Global, regional, and national burden of ckd due to glomerulonephritis from 1990 to 2019: a systematic analysis from the global burden of disease study 2019
.
Clin. J. Am. Soc. Nephrol.
18
,
60
71
https://doi.org/10.2215/CJN.0000000000000017
5
Zhao
,
W.M.
,
Li
,
X.L.
,
Shi
,
R.
,
Zhu
,
Y.
,
Wang
,
Z.J.
,
Wang
,
X.R.
et al.
(
2024
)
Global, regional and national burden of CKD in children and adolescents from 1990 to 2019
.
Nephrol. Dial. Transplant.
39
,
1268
1278
https://doi.org/10.1093/ndt/gfad269
6
Fogo
,
A.B
. (
2007
)
Mechanisms of progression of chronic kidney disease
.
Pediatr. Nephrol.
22
,
2011
2022
https://doi.org/10.1007/s00467-007-0524-0
7
Swartling
,
O.
,
Rydell
,
H.
,
Stendahl
,
M.
,
Segelmark
,
M.
,
Trolle Lagerros
,
Y.
and
Evans
,
M
. (
2021
)
CKD Progression and mortality among men and women: a nationwide study in Sweden
.
Am. J. Kidney Dis.
78
,
190
199
https://doi.org/10.1053/j.ajkd.2020.11.026
8
Siragy
,
H.M.
and
Carey
,
R.M
. (
2010
)
Role of the intrarenal renin-angiotensin-aldosterone system in chronic kidney disease
.
Am. J. Nephrol.
31
,
541
550
https://doi.org/10.1159/000313363
9
Chung
,
E.Y.
,
Ruospo
,
M.
,
Natale
,
P.
,
Bolignano
,
D.
,
Navaneethan
,
S.D.
,
Palmer
,
S.C.
et al.
(
2020
)
Aldosterone antagonists in addition to renin angiotensin system antagonists for preventing the progression of chronic kidney disease
.
Cochrane Database Syst. Rev.
10
, CD007004 https://doi.org/10.1002/14651858.CD007004.pub4
10
Fu
,
E.L.
,
Evans
,
M.
,
Clase
,
C.M.
,
Tomlinson
,
L.A.
,
Dekker
,
F.W
, et al.
(
2021
)
Stopping renin-angiotensin system inhibitors in patients with advanced CKD and risk of adverse outcomes: a nationwide study
.
J. Am. Soc. Nephrol.
32
,
424
435
https://doi.org/10.1681/ASN.2020050682
11
Wheeler
,
D.C.
,
Stefánsson
,
B.V.
,
Jongs
,
N.
,
Chertow
,
G.M.
,
Greene
,
T.
,
Hou
,
F.F.
et al.
(
2021
)
Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial
.
Lancet Diabetes Endocrinol.
9
,
22
31
https://doi.org/10.1016/S2213-8587(20)30369-7
12
Bakris
,
G.L.
,
Agarwal
,
R.
,
Anker
,
S.D.
,
Pitt
,
B.
,
Ruilope
,
L.M.
,
Rossing
,
P.
et al.
(
2020
)
Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes
.
N. Engl. J. Med.
383
,
2219
2229
https://doi.org/10.1056/NEJMoa2025845
13
Matsushita
,
K.
,
Ballew
,
S.H.
,
Wang
,
A.Y.M.
,
Kalyesubula
,
R.
,
Schaeffner
,
E.
and
Agarwal
,
R
. (
2022
)
Epidemiology and risk of cardiovascular disease in populations with chronic kidney disease
.
Nat. Rev. Nephrol.
18
,
696
707
https://doi.org/10.1038/s41581-022-00616-6
14
Jankowski
,
J.
,
Floege
,
J.
,
Fliser
,
D.
,
Böhm
,
M.
and
Marx
,
N
. (
2021
)
Cardiovascular disease in chronic kidney disease: pathophysiological insights and therapeutic options
.
Circulation
143
,
1157
1172
https://doi.org/10.1161/CIRCULATIONAHA.120.050686
15
Bailey
,
J.L.
,
Zheng
,
B.
,
Hu
,
Z.
,
Price
,
S.R.
and
Mitch
,
W.E
. (
2006
)
Chronic kidney disease causes defects in signaling through the insulin receptor substrate/phosphatidylinositol 3-kinase/akt pathway
.
J. Am. Soc. Nephrol.
17
,
1388
1394
https://doi.org/10.1681/ASN.2004100842
16
Liang
,
L.L.
,
He
,
M.F.
,
Zhou
,
P.P.
,
Pan
,
S.K.
,
Liu
,
D.W.
and
Liu
,
Z.S
. (
2024
)
GSK3β: a ray of hope for the treatment of diabetic kidney disease
.
FASEB J.
38
, e23458 https://doi.org/10.1096/fj.202302160R
17
Embi
,
N.
,
Rylatt
,
D.B.
and
Cohen
,
P
. (
1980
)
Glycogen synthase kinase-3 from rabbit skeletal muscle. Separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase
.
Eur. J. Biochem.
107
,
519
527
18
Wang
,
L.
,
Li
,
J.
and
Di
,
L
. (
2022
)
Glycogen synthesis and beyond, a comprehensive review of GSK3 as a key regulator of metabolic pathways and a therapeutic target for treating metabolic diseases
.
Med. Res. Rev.
42
,
946
982
https://doi.org/10.1002/med.21867
19
Azoulay-Alfaguter
,
I.
,
Yaffe
,
Y.
,
Licht-Murava
,
A.
,
Urbanska
,
M.
,
Jaworski
,
J.
,
Pietrokovski
,
S.
et al.
(
2011
)
Distinct molecular regulation of glycogen synthase kinase-3alpha isozyme controlled by its N-terminal region: functional role in calcium/calpain signaling
.
J. Biol. Chem.
286
,
13470
13480
https://doi.org/10.1074/jbc.M110.127969
20
Woodgett
,
J.R
. (
1990
)
Molecular cloning and expression of glycogen synthase kinase-3/factor A
.
EMBO J.
9
,
2431
2438
https://doi.org/10.1002/j.1460-2075.1990.tb07419.x
21
MacAulay
,
K.
,
Doble
,
B.W.
,
Patel
,
S.
,
Hansotia
,
T.
,
Sinclair
,
E.M.
,
Drucker
,
D.J.
et al.
(
2007
)
Glycogen synthase kinase 3alpha-specific regulation of murine hepatic glycogen metabolism
.
Cell Metab.
6
,
329
337
https://doi.org/10.1016/j.cmet.2007.08.013
22
Hoeflich
,
K.P.
,
Luo
,
J.
,
Rubie
,
E.A.
,
Tsao
,
M.-S.
,
Jin
,
O.
and
Woodgett
,
J.R
. (
2000
)
Requirement for glycogen synthase kinase-3β in cell survival and NF-κB activation
.
Nature
406
,
86
90
https://doi.org/10.1038/35017574
23
Chen
,
L.
,
Chou
,
C.-L.
and
Knepper
,
M.A
. (
2021
)
A Comprehensive Map of mRNAs and Their Isoforms across All 14 Renal Tubule Segments of Mouse
.
J. Am. Soc. Nephrol.
32
,
897
912
https://doi.org/10.1681/ASN.2020101406
24
Hurcombe
,
J.A.
,
Hartley
,
P.
,
Lay
,
A.C.
,
Ni
,
L.
,
Bedford
,
J.J.
,
Leader
,
J.P.
et al.
(
2019
)
Podocyte GSK3 is an evolutionarily conserved critical regulator of kidney function
.
Nat. Commun.
10
,
403
. https://doi.org/10.1038/s41467-018-08235-1
25
Jiang
,
C.
,
Zhu
,
W.
,
Yan
,
X.
,
Shao
,
Q.
,
Xu
,
B.
,
Zhang
,
M.
et al.
(
2016
)
Rescue therapy with Tanshinone IIA hinders transition of acute kidney injury to chronic kidney disease via targeting GSK3β
.
Sci. Rep.
6
,
36698
. https://doi.org/10.1038/srep36698
26
Lu
,
M.
,
Wang
,
P.
,
Qiao
,
Y.
,
Jiang
,
C.
,
Ge
,
Y.
,
Flickinger
,
B.
et al.
(
2019
)
GSK3β-mediated Keap1-independent regulation of Nrf2 antioxidant response: A molecular rheostat of acute kidney injury to chronic kidney disease transition
.
Redox Biol.
26
, 101275 https://doi.org/10.1016/j.redox.2019.101275
27
Sinha
,
S.
,
Dwivedi
,
N.
,
Woodgett
,
J.
,
Tao
,
S.
,
Howard
,
C.
,
Fields
,
T.A.
et al.
(
2020
)
Glycogen synthase kinase-3β inhibits tubular regeneration in acute kidney injury by a FoxM1-dependent mechanism
.
FASEB J.
34
,
13597
13608
https://doi.org/10.1096/fj.202000526RR
28
Li
,
C.
,
Ge
,
Y.
,
Dworkin
,
L.
,
Peng
,
A.
and
Gong
,
R
. (
2016
)
The β isoform of GSK3 mediates podocyte autonomous injury in proteinuric glomerulopathy
.
J. Pathol.
239
,
23
35
https://doi.org/10.1002/path.4692
29
Doble
,
B.W.
and
Woodgett
,
J.R
. (
2003
)
GSK-3: tricks of the trade for a multi-tasking kinase
.
J. Cell. Sci.
116
,
1175
1186
https://doi.org/10.1242/jcs.00384
30
McManus
,
E.J.
,
Sakamoto
,
K.
,
Armit
,
L.J.
,
Ronaldson
,
L.
,
Shpiro
,
N.
,
Marquez
,
R.
et al.
(
2005
)
Role that phosphorylation of GSK3 plays in insulin and Wnt signalling defined by knockin analysis
.
EMBO J.
24
,
1571
1583
https://doi.org/10.1038/sj.emboj.7600633
31
Patel
,
S.
,
Doble
,
B.W.
,
MacAulay
,
K.
,
Sinclair
,
E.M.
,
Drucker
,
D.J.
and
Woodgett
,
J.R
. (
2008
)
Tissue-specific role of glycogen synthase kinase 3beta in glucose homeostasis and insulin action
.
Mol. Cell. Biol.
28
,
6314
6328
https://doi.org/10.1128/MCB.00763-08
32
Song
,
W.J.
,
Song
,
E.A.C.
,
Jung
,
M.S.
,
Choi
,
S.H.
,
Baik
,
H.H.
,
Jin
,
B.K.
et al.
(
2015
)
Phosphorylation and inactivation of glycogen synthase kinase 3β (GSK3β) by dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A)
.
J. Biol. Chem.
290
,
2321
2333
https://doi.org/10.1074/jbc.M114.594952
33
Yao
,
H.B.
,
Shaw
,
P.C.
,
Wong
,
C.C.
and
Wan
,
D.C.C
. (
2002
)
Expression of glycogen synthase kinase-3 isoforms in mouse tissues and their transcription in the brain
.
J. Chem. Neuroanat.
23
,
291
297
https://doi.org/10.1016/S0891-0618(02)00014-5
34
Haq
,
S.
,
Choukroun
,
G.
,
Kang
,
Z.B.
,
Ranu
,
H.
,
Matsui
,
T.
,
Rosenzweig
,
A.
et al.
(
2000
)
Glycogen synthase kinase-3beta is a negative regulator of cardiomyocyte hypertrophy
.
J. Cell Biol.
151
,
117
130
https://doi.org/10.1083/jcb.151.1.117
35
Fang
,
Y.
,
Chen
,
B.
,
Liu
,
Z.
,
Gong
,
A.Y.
,
Gunning
,
W.T.
,
Ge
,
Y.
et al.
(
2022
)
Age-related GSK3β overexpression drives podocyte senescence and glomerular aging
.
J. Clin. Invest.
132
, e141848 https://doi.org/10.1172/JCI141848
36
Stamos
,
J.L.
,
Chu
,
M.L.H.
,
Enos
,
M.D.
,
Shah
,
N.
and
Weis
,
W.I
. (
2014
)
Structural basis of GSK-3 inhibition by N-terminal phosphorylation and by the Wnt receptor LRP6
.
Elife
3
, e01998 https://doi.org/10.7554/eLife.01998
37
Mangangcha
,
I.R.
,
Brojen Singh
,
R.K.
,
Lebeche
,
D.
and
Ali
,
S
. (
2022
)
Xanthone glucoside 2-β-D-glucopyranosyl-1,3,6,7-tetrahydroxy-9H-xanthen-9-one binds to the ATP-binding pocket of glycogen synthase kinase 3β and inhibits its activity: implications in prostate cancer and associated cardiovascular disease risk
.
Journal of Biomolecular Structure and Dynamics
40
,
7868
7884
https://doi.org/10.1080/07391102.2021.1902857
38
Bijur
,
G.N.
and
Jope
,
R.S
. (
2003
)
Glycogen synthase kinase-3 beta is highly activated in nuclei and mitochondria
.
Neuroreport
14
,
2415
2419
39
Gomez
,
L.
,
Thiebaut
,
P.-A.
,
Paillard
,
M.
,
Ducreux
,
S.
,
Abrial
,
M.
,
Crola Da Silva
,
C.
et al.
(
2016
)
The SR/ER-mitochondria calcium crosstalk is regulated by GSK3β during reperfusion injury
.
Cell Death Differ.
23
,
313
322
https://doi.org/10.1038/cdd.2015.101
40
Wang
,
Z.
,
Bao
,
H.
,
Ge
,
Y.
,
Zhuang
,
S.
,
Peng
,
A.
and
Gong
,
R
. (
2015
)
Pharmacological targeting of GSK3B confers protection against podocytopathy and proteinuria by desensitizing mitochondrial permeability transition
.
Br. J. Pharmacol.
172
,
895
909
https://doi.org/10.1111/bph.12952
41
King
,
T.D.
,
Clodfelder-Miller
,
B.
,
Barksdale
,
K.A.
and
Bijur
,
G.N
. (
2008
)
Unregulated mitochondrial GSK3β activity results in NADH:Ubiquinone oxidoreductase deficiency
.
Neurotox. Res.
14
,
367
382
https://doi.org/10.1007/BF03033861
42
Chiara
,
F.
and
Rasola
,
A
. (
2013
)
GSK-3 and mitochondria in cancer cells
.
Front. Oncol.
3
, 16 https://doi.org/10.3389/fonc.2013.00016
43
Chen
,
M.
,
Fang
,
Y.
,
Ge
,
Y.
,
Qiu
,
S.
,
Dworkin
,
L.
and
Gong
,
R
. (
2024
)
The redox-sensitive GSK3β is a key regulator of glomerular podocyte injury in type 2 diabetic kidney disease
.
Redox Biol.
72
, 103127 https://doi.org/10.1016/j.redox.2024.103127
44
Beurel
,
E.
,
Grieco
,
S.F.
and
Jope
,
R.S
. (
2015
)
Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases
.
Pharmacol. Ther.
148
,
114
131
https://doi.org/10.1016/j.pharmthera.2014.11.016
45
Sundaresan
,
N.R.
,
Bindu
,
S.
,
Pillai
,
V.B.
,
Samant
,
S.
,
Pan
,
Y.
,
Huang
,
J.-Y.
et al.
(
2015
)
SIRT3 Blocks Aging-Associated Tissue Fibrosis in Mice by Deacetylating and Activating Glycogen Synthase Kinase 3β
.
Mol. Cell. Biol.
36
,
678
692
https://doi.org/10.1128/MCB.00586-15
46
Ko
,
R.
,
Park
,
J.H.
,
Ha
,
H.
,
Choi
,
Y.
and
Lee
,
S.Y
. (
2015
)
Glycogen synthase kinase 3β ubiquitination by TRAF6 regulates TLR3-mediated pro-inflammatory cytokine production
.
Nat. Commun.
6
,
6765
, 6765 https://doi.org/10.1038/ncomms7765
47
Jin
,
K.
,
Li
,
Y.
,
Wei
,
R.
,
Liu
,
Y.
,
Wang
,
S.
and
Tian
,
H
. (
2024
)
BZW2 promotes malignant progression in lung adenocarcinoma through enhancing the ubiquitination and degradation of GSK3β
.
Cell Death Discov.
10
, 105 https://doi.org/10.1038/s41420-024-01879-7
48
Zhou
,
Q.
,
Li
,
S.
,
Li
,
M.
,
Ke
,
D.
,
Wang
,
Q.
,
Yang
,
Y.
et al.
(
2022
)
Human tau accumulation promotes glycogen synthase kinase-3β acetylation and thus upregulates the kinase: A vicious cycle in Alzheimer neurodegeneration
.
EBioMedicine
78
,
103970
, 103970 https://doi.org/10.1016/j.ebiom.2022.103970
49
Suber
,
T.
,
Wei
,
J.
,
Jacko
,
A.M.
,
Nikolli
,
I.
,
Zhao
,
Y.
,
Zhao
,
J
, et al.
(
2017
)
SCFFBXO17 E3 ligase modulates inflammation by regulating proteasomal degradation of glycogen synthase kinase-3β in lung epithelia
.
J. Biol. Chem.
292
,
7452
7461
https://doi.org/10.1074/jbc.M116.771667
50
Van Amerongen
,
R.
,
Nawijn
,
M.
,
Franca-Koh
,
J.
,
Zevenhoven
,
J.
,
Van der Gulden
,
H.
,
Jonkers
,
J.
et al.
(
2005
)
Frat is dispensable for canonical Wnt signaling in mammals
.
Genes Dev.
19
,
425
430
https://doi.org/10.1101/gad.326705
51
Wang
,
W.
,
Li
,
X.
,
Lee
,
M.
,
Jun
,
S.
,
Aziz
,
K.E.
,
Feng
,
L.
et al.
(
2015
)
FOXKs Promote Wnt/β-Catenin Signaling by Translocating DVL into the Nucleus
.
Dev. Cell
32
,
707
718
https://doi.org/10.1016/j.devcel.2015.01.031
52
Yang
,
D.
,
Li
,
S.
,
Duan
,
X.
,
Ren
,
J.
,
Liang
,
S.
,
Yakoumatos
,
L.
et al.
(
2020
)
TLR4 induced Wnt3a-Dvl3 restrains the intensity of inflammation and protects against endotoxin-driven organ failure through GSK3β/β-catenin signaling
.
Mol. Immunol.
118
,
153
164
https://doi.org/10.1016/j.molimm.2019.12.013
53
Gavagan
,
M.
,
Jameson
,
N.
and
Zalatan
,
J.G
. (
2023
)
The Axin scaffold protects the kinase GSK3β from cross-pathway inhibition
.
Elife
12
https://doi.org/10.7554/eLife.85444
54
Ho
,
C.
,
Lee
,
P.-H.
,
Hsu
,
Y.-C.
,
Huang
,
Y.-T.
,
Wang
,
F.-S.
and
Lin
,
C.-L
. (
2012
)
Sustained wnt/β-catenin signaling rescues high glucose induction of transforming growth factor-β1-mediated renal fibrosis
.
Am. J. Med. Sci.
344
,
374
382
https://doi.org/10.1097/MAJ.0b013e31824369c5
55
Liu
,
Y.
,
Feng
,
Q.
,
Miao
,
J.
,
Wu
,
Q.
,
Zhou
,
S.
,
Shen
,
W.
et al.
(
2020
)
C‐X‐C motif chemokine receptor 4 aggravates renal fibrosis through activating JAK/STAT/GSK3β/β‐catenin pathway
.
J. Cell. Mol. Med.
24
,
3837
3855
https://doi.org/10.1111/jcmm.14973
56
Sun
,
Y.
,
Fan
,
Y.
,
Wang
,
Z.
,
Li
,
M.
,
Su
,
D.
,
Liu
,
Y.
et al.
(
2022
)
S100A16 promotes acute kidney injury by activating HRD1-induced ubiquitination and degradation of GSK3β and CK1α
.
Cell. Mol. Life Sci.
79
,
184
, 184 https://doi.org/10.1007/s00018-022-04213-5
57
Doble
,
B.W.
,
Patel
,
S.
,
Wood
,
G.A.
,
Kockeritz
,
L.K.
and
Woodgett
,
J.R
. (
2007
)
Functional Redundancy of GSK-3α and GSK-3β in Wnt/β-Catenin Signaling Shown by Using an Allelic Series of Embryonic Stem Cell Lines
.
Dev. Cell
12
,
957
971
https://doi.org/10.1016/j.devcel.2007.04.001
58
Cildir
,
G.
,
Low
,
K.C.
and
Tergaonkar
,
V
. (
2016
)
Noncanonical NF-κB Signaling in Health and Disease
.
Trends Mol. Med.
22
,
414
429
https://doi.org/10.1016/j.molmed.2016.03.002
59
Oeckinghaus
,
A.
,
Hayden
,
M.S.
and
Ghosh
,
S
. (
2011
)
Crosstalk in NF-κB signaling pathways
.
Nat. Immunol.
12
,
695
708
https://doi.org/10.1038/ni.2065
60
Gong
,
R.
,
Rifai
,
A.
,
Ge
,
Y.
,
Chen
,
S.
and
Dworkin
,
L.D
. (
2008
)
Hepatocyte growth factor suppresses proinflammatory NFkappaB activation through GSK3beta inactivation in renal tubular epithelial cells
.
J. Biol. Chem.
283
,
7401
7410
https://doi.org/10.1074/jbc.M710396200
61
Ougolkov
,
A.V.
,
Bone
,
N.D.
,
Fernandez-Zapico
,
M.E.
,
Kay
,
N.E.
and
Billadeau
,
D.D
. (
2007
)
Inhibition of glycogen synthase kinase-3 activity leads to epigenetic silencing of nuclear factor kappaB target genes and induction of apoptosis in chronic lymphocytic leukemia B cells
.
Blood
110
,
735
742
https://doi.org/10.1182/blood-2006-12-060947
62
Abd-Ellah
,
A.
,
Voogdt
,
C.
,
Krappmann
,
D.
,
Möller
,
P.
and
Marienfeld
,
R.B
. (
2018
)
GSK3β modulates NF-κB activation and RelB degradation through site-specific phosphorylation of BCL10
.
Sci. Rep.
8
, 1352 https://doi.org/10.1038/s41598-018-19822-z
63
Grayck
,
M.R.
,
McCarthy
,
W.C.
,
Solar
,
M.
,
Golden
,
E.
,
Balasubramaniyan
,
N.
,
Zheng
,
L
, et al.
(
2024
)
GSK3beta/NF-kappaB -dependent transcriptional regulation of homeostatic hepatocyte Tnf production
.
Am. J. Physiol. Gastrointest. Liver Physiol.
326
,
G374
G384
https://doi.org/10.1152/ajpgi.00229.2023
64
Cai
,
Y.T.
,
Li
,
Z.
,
Wang
,
Y.Y.
,
Li
,
C.
and
Ma
,
Q.Y
. (
2024
)
A novel GSK3β inhibitor 5n attenuates acute kidney injury
.
Heliyon
10
, e29159 https://doi.org/10.1016/j.heliyon.2024.e29159
65
Su
,
S.
,
Zhang
,
P.
,
Zhang
,
Q.
and
Yin
,
Z
. (
2019
)
GSK-3β Inhibitor Induces Expression of the TLR4/MyD88/NF-κB Signaling Pathway to Protect Against Renal Ischemia-Reperfusion Injury During Rat Kidney Transplantation
.
Inflammation
42
,
2105
2118
https://doi.org/10.1007/s10753-019-01074-2
66
Ahmed
,
M.A.
,
Kamel
,
E.O.
and
Abd-Eldayem
,
A.M
. (
2024
)
Role of cAMP/pCREB and GSK-3β/NF-κB p65 signaling pathways in the renoprotective effect of mirabegron against renal ischemia-reperfusion injury in rats
.
Eur. J. Pharmacol.
974
,
176617
https://doi.org/10.1016/j.ejphar.2024.176617
67
Bao
,
H.
,
Ge
,
Y.
,
Peng
,
A.
and
Gong
,
R
. (
2015
)
Fine-tuning of NFκB by glycogen synthase kinase 3β directs the fate of glomerular podocytes upon injury
.
Kidney Int.
87
,
1176
1190
https://doi.org/10.1038/ki.2014.428
68
Wang
,
P.
,
Zhou
,
S.
,
Ge
,
Y.
,
Lu
,
M.
,
Liu
,
Z.
and
Gong
,
R
. (
2017
)
Valproate hampers podocyte acquisition of immune phenotypes via intercepting the GSK3β facilitated NFkB activation
.
Oncotarget
8
,
88332
88344
https://doi.org/10.18632/oncotarget.19917
69
Silva-Palacios
,
A.
,
Ostolga-Chavarría
,
M.
,
Zazueta
,
C.
and
Königsberg
,
M
. (
2018
)
Nrf2: Molecular and epigenetic regulation during aging
.
Ageing Res. Rev.
47
,
31
40
https://doi.org/10.1016/j.arr.2018.06.003
70
Ahmed
,
S.M.U.
,
Luo
,
L.
,
Namani
,
A.
,
Wang
,
X.J.
and
Tang
,
X
. (
2017
)
Nrf2 signaling pathway: Pivotal roles in inflammation
.
Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease
1863
,
585
597
https://doi.org/10.1016/j.bbadis.2016.11.005
71
Jiang
,
Y.
,
Bao
,
H.
,
Ge
,
Y.
,
Tang
,
W.
,
Cheng
,
D.
,
Luo
,
K.
et al.
(
2015
)
Therapeutic targeting of GSK3β enhances the Nrf2 antioxidant response and confers hepatic cytoprotection in hepatitis C
.
Gut
64
,
168
179
https://doi.org/10.1136/gutjnl-2013-306043
72
Hausenloy
,
D
. (
2004
)
New directions for protecting the heart against ischaemia–reperfusion injury: targeting the Reperfusion Injury Salvage Kinase (RISK)-pathway
.
Cardiovasc. Res.
61
,
448
460
https://doi.org/10.1016/j.cardiores.2003.09.024
73
Zhou
,
S.
,
Wang
,
P.
,
Qiao
,
Y.
,
Ge
,
Y.
,
Wang
,
Y.
,
Quan
,
S.
et al.
(
2016
)
Genetic and Pharmacologic Targeting of Glycogen Synthase Kinase 3β Reinforces the Nrf2 Antioxidant Defense against Podocytopathy
.
J. Am. Soc. Nephrol.
27
,
2289
2308
https://doi.org/10.1681/ASN.2015050565
74
Moustakas
,
A.
,
Pardali
,
K.
,
Gaal
,
A.
and
Heldin
,
C.-H
. (
2002
)
Mechanisms of TGF-β signaling in regulation of cell growth and differentiation
.
Immunol. Lett.
82
,
85
91
https://doi.org/10.1016/S0165-2478(02)00023-8
75
Doi
,
S.
,
Zou
,
Y.
,
Togao
,
O.
,
Pastor
,
J.V.
,
John
,
G.B.
,
Wang
,
L
, et al.
(
2011
)
Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice
.
J. Biol. Chem.
286
,
8655
8665
https://doi.org/10.1074/jbc.M110.174037
76
Liu
,
F.
,
Cao
,
Y.
,
Zhang
,
C.
and
Su
,
H
. (
2023
)
Decreased DANCR contributes to high glucose-induced extracellular matrix accumulation in human renal mesangial cell via regulating the TGF-β/Smad signaling
.
FASEB J.
37
, e22926 https://doi.org/10.1096/fj.202300146R
77
Akhurst
,
R.J.
and
Hata
,
A
. (
2012
)
Targeting the TGFβ signalling pathway in disease
.
Nat. Rev. Drug Discov.
11
,
790
811
https://doi.org/10.1038/nrd3810
78
Massague
,
J
. (
2012
)
TGFbeta signalling in context
.
Nat. Rev. Mol. Cell Biol.
13
,
616
630
79
Moustakas
,
A.
and
Heldin
,
C.H
. (
2009
)
The regulation of TGFβ signal transduction
.
Development (Rome)
136
,
3699
3714
https://doi.org/10.1242/dev.030338
80
Pan
,
R.
,
Zhang
,
Y.
,
Zheng
,
M.
,
Zang
,
B.
and
Jin
,
M
. (
2017
)
Hydroxysafflor yellow A suppresses MRC-5 Cell activation induced by TGF-β1 by blocking TGF-β1 Binding to TβRII
.
Front. Pharmacol.
8
, 264 https://doi.org/10.3389/fphar.2017.00264
81
Choi
,
H.I.
,
Park
,
J.S.
,
Kim
,
D.H.
,
Kim
,
C.S.
,
Bae
,
E.H.
,
Ma
,
S.K.
et al.
(
2019
)
PGC-1α Suppresses the activation of TGF-β/Smad signaling via targeting TGFβRI downregulation by let-7b/c upregulation
.
Int. J. Mol. Sci.
20
, 5084 https://doi.org/10.3390/ijms20205084
82
You
,
Y.K.
,
Luo
,
Q.
,
Wu
,
W.F.
,
Zhang
,
J.J.
,
Zhu
,
H.J.
,
Lao
,
L.
et al.
(
2019
)
Petchiether A attenuates obstructive nephropathy by suppressing TGF-β/Smad3 and NF-κB signalling
.
J. Cell. Mol. Med.
23
,
5576
5587
https://doi.org/10.1111/jcmm.14454
83
Su
,
Q.
,
Chen
,
X.
,
Ling
,
X.
,
Li
,
D.
,
Ren
,
X.
,
Zhao
,
Y.
et al.
(
2023
)
SUMOylation of Smad2 mediates TGF-β-regulated endothelial-mesenchymal transition
.
J. Biol. Chem.
299
,
105244
, 105244 https://doi.org/10.1016/j.jbc.2023.105244
84
Schmierer
,
B.
and
Hill
,
C.S
. (
2007
)
TGFβ–SMAD signal transduction: molecular specificity and functional flexibility
.
Nat. Rev. Mol. Cell Biol.
8
,
970
982
https://doi.org/10.1038/nrm2297
85
Heldin
,
C.-H.
and
Moustakas
,
A
. (
2016
)
Signaling receptors for TGF-β family members
.
Cold Spring Harb. Perspect. Biol.
8
, a022053 https://doi.org/10.1101/cshperspect.a022053
86
Lal
,
H.
,
Ahmad
,
F.
,
Zhou
,
J.
,
Yu
,
J.E.
,
Vagnozzi
,
R.J.
,
Guo
,
Y.
et al.
(
2014
)
Cardiac fibroblast glycogen synthase kinase-3β regulates ventricular remodeling and dysfunction in ischemic heart
.
Circulation
130
,
419
430
https://doi.org/10.1161/CIRCULATIONAHA.113.008364
87
Guo
,
X.
,
Ramirez
,
A.
,
Waddell
,
D.S.
,
Li
,
Z.
,
Liu
,
X.
and
Wang
,
X.-F
. (
2008
)
Axin and GSK3-β control Smad3 protein stability and modulate TGF-β signaling
.
Genes Dev.
22
,
106
120
https://doi.org/10.1101/gad.1590908
88
Kamato
,
D.
,
Burch
,
M.L.
,
Piva
,
T.J.
,
Rezaei
,
H.B.
,
Rostam
,
M.A.
,
Xu
,
S.
et al.
(
2013
)
Transforming growth factor-β signalling: role and consequences of Smad linker region phosphorylation
.
Cell. Signal.
25
,
2017
2024
https://doi.org/10.1016/j.cellsig.2013.06.001
89
Wang
,
G.
,
Matsuura
,
I.
,
He
,
D.
and
Liu
,
F
. (
2009
)
Transforming growth factor-β-inducible Phosphorylation of Smad3
.
Journal of Biological Chemistry
284
,
9663
9673
https://doi.org/10.1074/jbc.M809281200
90
Millet
,
C.
,
Yamashita
,
M.
,
Heller
,
M.
,
Yu
,
L.-R.
,
Veenstra
,
T.D.
and
Zhang
,
Y.E
. (
2009
)
A negative feedback control of transforming growth factor-β signaling by glycogen synthase kinase 3-mediated smad3 linker phosphorylation at ser-204
.
Journal of Biological Chemistry
284
,
19808
19816
https://doi.org/10.1074/jbc.M109.016667
91
Chen
,
B.
,
Wang
,
P.
,
Liang
,
X.
,
Jiang
,
C.
,
Ge
,
Y.
,
Dworkin
,
L.D.
et al.
(
2021
)
Permissive effect of GSK3β on profibrogenic plasticity of renal tubular cells in progressive chronic kidney disease
.
Cell Death Dis.
12
, 432 https://doi.org/10.1038/s41419-021-03709-5
92
Guo
,
Y.
,
Gupte
,
M.
,
Umbarkar
,
P.
,
Singh
,
A.P.
,
Sui
,
J.Y.
,
Force
,
T.
et al.
(
2017
)
Entanglement of GSK-3β, β-catenin and TGF-β1 signaling network to regulate myocardial fibrosis
.
J. Mol. Cell. Cardiol.
110
,
109
120
https://doi.org/10.1016/j.yjmcc.2017.07.011
93
Guo
,
X.
and
Wang
,
X.-F
. (
2009
)
Signaling cross-talk between TGF-β/BMP and other pathways
.
Cell Res.
19
,
71
88
https://doi.org/10.1038/cr.2008.302
94
Furuhashi
,
M.
,
Yagi
,
K.
,
Yamamoto
,
H.
,
Furukawa
,
Y.
,
Shimada
,
S.
,
Nakamura
,
Y.
et al.
(
2001
)
Axin facilitates Smad3 activation in the transforming growth factor beta signaling pathway
.
Mol. Cell. Biol.
21
,
5132
5141
https://doi.org/10.1128/MCB.21.15.5132-5141.2001
95
Burke
,
J.E.
,
Triscott
,
J.
,
Emerling
,
B.M.
and
Hammond
,
G.R.V
. (
2023
)
Beyond PI3Ks: targeting phosphoinositide kinases in disease
.
Nat. Rev. Drug Discov.
22
,
357
386
https://doi.org/10.1038/s41573-022-00582-5
96
Fruman
,
D.A.
,
Chiu
,
H.
,
Hopkins
,
B.D.
,
Bagrodia
,
S.
,
Cantley
,
L.C.
and
Abraham
,
R.T
. (
2017
)
The PI3K pathway in human disease
.
Cell
170
,
605
635
https://doi.org/10.1016/j.cell.2017.07.029
97
Gong
,
R.
,
Rifai
,
A.
and
Dworkin
,
L.D
. (
2005
)
Activation of PI3K–Akt–GSK3β pathway mediates hepatocyte growth factor inhibition of RANTES expression in renal tubular epithelial cells
.
Biochem. Biophys. Res. Commun.
330
,
27
33
https://doi.org/10.1016/j.bbrc.2005.02.122
98
Lan
,
A.
,
Qi
,
Y.
and
Du
,
J
. (
2014
)
Akt2 Mediates TGF-1-Induced epithelial to mesenchymal transition by deactivating GSK3/Snail signaling pathway in renal tubular epithelial cells
.
Cell. Physiol. Biochem.
34
,
368
382
https://doi.org/10.1159/000363006
99
Lai
,
S.S.
,
Zhao
,
D.D.
,
Cao
,
P.
,
Lu
,
K.
,
Luo
,
O.Y.
,
Chen
,
W.B.
et al.
(
2016
)
PP2Acα positively regulates the termination of liver regeneration in mice through the AKT/GSK3β/Cyclin D1 pathway
.
J. Hepatol.
64
,
352
360
https://doi.org/10.1016/j.jhep.2015.09.025
100
Leng
,
S.
,
Zhang
,
W.
,
Zheng
,
Y.
,
Liberman
,
Z.
,
Rhodes
,
C.J.
,
Eldar-Finkelman
,
H.
et al.
(
2010
)
Glycogen synthase kinase 3 beta mediates high glucose-induced ubiquitination and proteasome degradation of insulin receptor substrate 1
.
J. Endocrinol.
206
,
171
181
https://doi.org/10.1677/JOE-09-0456
101
Liang
,
X.
,
Chen
,
B.
,
Wang
,
P.
,
Ge
,
Y.
,
Malhotra
,
D.K.
,
Dworkin
,
L.D.
et al.
(
2020
)
Triptolide potentiates the cytoskeleton-stabilizing activity of cyclosporine A in glomerular podocytes via a GSK3β dependent mechanism
.
Am. J. Transl. Res.
12
,
800
812
102
Xu
,
W.
,
Ge
,
Y.
,
Liu
,
Z.
and
Gong
,
R
. (
2015
)
Glycogen synthase kinase 3β orchestrates microtubule remodeling in compensatory glomerular adaptation to podocyte depletion
.
J. Biol. Chem.
290
,
1348
1363
https://doi.org/10.1074/jbc.M114.593830
103
Yucel
,
G.
and
Oro
,
A.E
. (
2011
)
Cell Migration: GSK3β steers the cytoskeleton’s tip
.
Cell
144
,
319
321
https://doi.org/10.1016/j.cell.2011.01.023
104
Leibinger
,
M.
,
Andreadaki
,
A.
,
Golla
,
R.
,
Levin
,
E.
,
Hilla
,
A.M.
,
Diekmann
,
H.
et al.
(
2017
)
Boosting CNS axon regeneration by harnessing antagonistic effects of GSK3 activity
.
Proc. Natl. Acad. Sci. U.S.A.
114
,
E5454
E5463
https://doi.org/10.1073/pnas.1621225114
105
Yokota
,
Y.
,
Eom
,
T.-Y.
,
Stanco
,
A.
,
Kim
,
W.-Y.
,
Rao
,
S.
,
Snider
,
W.D.
et al.
(
2010
)
Cdc42 and Gsk3 modulate the dynamics of radial glial growth, inter-radial glial interactions and polarity in the developing cerebral cortex
.
Development
137
,
4101
4110
https://doi.org/10.1242/dev.048637
106
Chikano
,
Y.
,
Domoto
,
T.
,
Furuta
,
T.
,
Sabit
,
H.
,
Kitano-Tamura
,
A.
,
Pyko
,
I.V.
et al.
(
2015
)
Glycogen synthase kinase 3β sustains invasion of glioblastoma via the focal adhesion kinase, Rac1, and c-Jun N-terminal kinase-mediated pathway
.
Mol. Cancer Ther.
14
,
564
574
https://doi.org/10.1158/1535-7163.MCT-14-0479
107
Fukata
,
Y.
,
Itoh
,
T.J.
,
Kimura
,
T.
,
Ménager
,
C.
,
Nishimura
,
T.
,
Shiromizu
,
T.
et al.
(
2002
)
CRMP-2 binds to tubulin heterodimers to promote microtubule assembly
.
Nat. Cell Biol.
4
,
583
591
https://doi.org/10.1038/ncb825
108
Bunker
,
J.M.
,
Wilson
,
L.
,
Jordan
,
M.A.
and
Feinstein
,
S.C
. (
2004
)
Modulation of microtubule dynamics by tau in living cells: implications for development and neurodegeneration
.
Mol. Biol. Cell
15
,
2720
2728
https://doi.org/10.1091/mbc.e04-01-0062
109
Fang
,
Y.
,
Chen
,
B.
,
Gong
,
A.Y.
,
Malhotra
,
D.K.
,
Gupta
,
R.
,
Dworkin
,
L.D.
et al.
(
2021
)
The ketone body β-hydroxybutyrate mitigates the senescence response of glomerular podocytes to diabetic insults
.
Kidney Int.
100
,
1037
1053
https://doi.org/10.1016/j.kint.2021.06.031
110
Chen
,
T.K.
,
Knicely
,
D.H.
and
Grams
,
M.E
. (
2019
)
Chronic Kidney Disease Diagnosis and Management
.
JAMA
322
,
1294
https://doi.org/10.1001/jama.2019.14745
111
Leaf
,
I.A.
and
Duffield
,
J.S
. (
2017
)
What can target kidney fibrosis?
Nephrol. Dial. Transplant.
32
,
i89
i97
https://doi.org/10.1093/ndt/gfw388
112
Hodgkins
,
K.S.
and
Schnaper
,
H.W
. (
2012
)
Tubulointerstitial injury and the progression of chronic kidney disease
.
Pediatr. Nephrol.
27
,
901
909
https://doi.org/10.1007/s00467-011-1992-9
113
Zeng
,
L.
,
Ng
,
J.K.-C.
,
Fung
,
W.W.-S.
,
Chan
,
G.C.-K.
,
Chow
,
K.-M.
and
Szeto
,
C.-C
. (
2023
)
Intrarenal and urinary glycogen synthase kinase-3 beta levels in diabetic and nondiabetic chronic kidney disease
.
Kidney Blood Press. Res.
48
,
241
248
https://doi.org/10.1159/000530210
114
Gong
,
R.
,
Ge
,
Y.
,
Chen
,
S.
,
Liang
,
E.
,
Esparza
,
A.
,
Sabo
,
E.
et al.
(
2008
)
Glycogen synthase kinase 3beta: a novel marker and modulator of inflammatory injury in chronic renal allograft disease
.
Am. J. Transplant.
8
,
1852
1863
https://doi.org/10.1111/j.1600-6143.2008.02319.x
115
Waters
,
A.
and
Koziell
,
A
. (
2009
)
Activation of Canonical Wnt Signaling Meets with Podocytopathy
.
J. Am. Soc. Nephrol.
20
,
1864
1866
https://doi.org/10.1681/ASN.2009070762
116
Beurel
,
E.
,
Michalek
,
S.M.
and
Jope
,
R.S
. (
2010
)
Innate and adaptive immune responses regulated by glycogen synthase kinase-3 (GSK3)
.
Trends Immunol.
31
,
24
31
https://doi.org/10.1016/j.it.2009.09.007
117
Zhao
,
J.
,
Wang
,
H.
,
Huang
,
Y.
,
Zhang
,
H.
,
Wang
,
S.
,
Gaskin
,
F.
et al.
(
2015
)
Lupus nephritis: glycogen synthase kinase 3β promotion of renal damage through activation of the NLRP3 inflammasome in lupus-prone mice
.
Arthritis Rheumatol.
67
,
1036
1044
https://doi.org/10.1002/art.38993
118
Jellusova
,
J.
,
Cato
,
M.H.
,
Apgar
,
J.R.
,
Ramezani-Rad
,
P.
,
Leung
,
C.R.
,
Chen
,
C.
et al.
(
2017
)
Gsk3 is a metabolic checkpoint regulator in B cells
.
Nat. Immunol.
18
,
303
312
https://doi.org/10.1038/ni.3664
119
Du
,
Z.
,
Tian
,
X.
,
Ma
,
M.
,
Somlo
,
S.
,
Weinstein
,
A.M.
and
Wang
,
T
. (
2021
)
Restoration of proximal tubule flow–activated transport prevents cyst growth in polycystic kidney disease
.
JCI Insight
6
https://doi.org/10.1172/jci.insight.146041
120
Li
,
X
. (
2011
)
Phosphorylation, protein kinases and ADPKD
.
Biochim. Biophys. Acta
1812
,
1219
1224
https://doi.org/10.1016/j.bbadis.2011.03.001
121
Dwivedi
,
N.
,
Jamadar
,
A.
,
Mathew
,
S.
,
Fields
,
T.A.
and
Rao
,
R
. (
2023
)
Myofibroblast depletion reduces kidney cyst growth and fibrosis in autosomal dominant polycystic kidney disease
.
Kidney Int.
103
,
144
155
https://doi.org/10.1016/j.kint.2022.08.036
122
Jamadar
,
A.
,
Suma
,
S.M.
,
Mathew
,
S.
,
Fields
,
T.A.
,
Wallace
,
D.P.
,
Calvet
,
J.P.
et al.
(
2021
)
The tyrosine-kinase inhibitor Nintedanib ameliorates autosomal-dominant polycystic kidney disease
.
Cell Death Dis.
12
,
947
, 947 https://doi.org/10.1038/s41419-021-04248-9
123
Force
,
T.
and
Woodgett
,
J.R
. (
2009
)
Unique and overlapping functions of GSK-3 Isoforms in cell differentiation and proliferation and cardiovascular development
.
Journal of Biological Chemistry
284
,
9643
9647
https://doi.org/10.1074/jbc.R800077200
124
Kaiser
,
M.
and
Edemir
,
B
. (
2020
)
Lithium Chloride and GSK3 Inhibition Reduce Aquaporin-2 Expression in Primary Cultured Inner Medullary Collecting Duct Cells Due to Independent Mechanisms
.
Cells
9
,
1060
https://doi.org/10.3390/cells9041060
125
Rao
,
R
. (
2012
)
Glycogen synthase kinase-3 regulation of urinary concentrating ability
.
Curr. Opin. Nephrol. Hypertens.
21
,
541
546
https://doi.org/10.1097/MNH.0b013e32835571d4
126
Tao
,
S.
,
Kakade
,
V.R.
,
Woodgett
,
J.R.
,
Pandey
,
P.
,
Suderman
,
E.D.
,
Rajagopal
,
M.
et al.
(
2015
)
Glycogen synthase kinase-3β promotes cyst expansion in polycystic kidney disease
.
Kidney Int.
87
,
1164
1175
https://doi.org/10.1038/ki.2014.427
127
Kakade
,
V.R.
,
Tao
,
S.
,
Rajagopal
,
M.
,
Zhou
,
X.
,
Li
,
X.
,
Yu
,
A.S.L.
et al.
(
2016
)
A cAMP and CREB-mediated feed-forward mechanism regulates GSK3β in polycystic kidney disease
.
J. Mol. Cell Biol.
8
,
464
476
https://doi.org/10.1093/jmcb/mjw022
128
Torres
,
V.E.
and
Harris
,
P.C
. (
2014
)
Strategies Targeting cAMP Signaling in the Treatment of Polycystic Kidney Disease
.
J. Am. Soc. Nephrol.
25
,
18
32
https://doi.org/10.1681/ASN.2013040398
129
Wallace
,
D.P
. (
2011
)
Cyclic AMP-mediated cyst expansion
.
Biochim. Biophys. Acta
1812
,
1291
1300
https://doi.org/10.1016/j.bbadis.2010.11.005
130
Nørregaard
,
R.
,
Tao
,
S.
,
Nilsson
,
L.
,
Woodgett
,
J.R.
,
Kakade
,
V.
,
Yu
,
A.S.L.
et al.
(
2015
)
Glycogen synthase kinase 3α regulates urine concentrating mechanism in mice
.
Am. J. Physiol. Renal Physiol.
308
,
F650
60
https://doi.org/10.1152/ajprenal.00516.2014
131
Rao
,
R.
,
Patel
,
S.
,
Hao
,
C.
,
Woodgett
,
J.
and
Harris
,
R
. (
2010
)
GSK3β Mediates Renal Response to Vasopressin by Modulating Adenylate Cyclase Activity
.
J. Am. Soc. Nephrol.
21
,
428
437
https://doi.org/10.1681/ASN.2009060672
132
Yu
,
W.
,
Wang
,
G.
,
Li
,
L.X.
,
Zhang
,
H.
,
Gui
,
X.
,
Zhou
,
J.X.
et al.
(
2023
)
Transcription factor FoxM1 promotes cyst growth in PKD1 mutant ADPKD
.
Hum. Mol. Genet.
32
,
1114
1126
https://doi.org/10.1093/hmg/ddac273
133
Mun
,
H.
and
Park
,
J.H
. (
2016
)
Inflammation and Fibrosis in ADPKD
.
Adv. Exp. Med. Biol.
933
,
35
44
https://doi.org/10.1007/978-981-10-2041-4_4
134
Christensen
,
B.M.
,
Kim
,
Y.-H.
,
Kwon
,
T.-H.
and
Nielsen
,
S
. (
2006
)
Lithium treatment induces a marked proliferation of primarily principal cells in rat kidney inner medullary collecting duct
.
American Journal of Physiology-Renal Physiology
291
,
F39
F48
https://doi.org/10.1152/ajprenal.00383.2005
135
Kjaersgaard
,
G.
,
Madsen
,
K.
,
Marcussen
,
N.
,
Christensen
,
S.
,
Walter
,
S.
and
Jensen
,
B.L
. (
2012
)
Tissue injury after lithium treatment in human and rat postnatal kidney involves glycogen synthase kinase-3β-positive epithelium
.
American Journal of Physiology-Renal Physiology
302
,
F455
F465
https://doi.org/10.1152/ajprenal.00144.2011
136
Alsady
,
M.
,
Baumgarten
,
R.
and
Deen
,
P.M.T
. (
2016
)
Lithium in the kidney: Friend and foe?
J. Am. Soc. Nephrol.
27
,
1587
1595
https://doi.org/10.1681/ASN.2015080907
137
Atagun
,
M.I.
,
Oral
,
E.T.
and
Sevinc
,
C
. (
2013
)
Polycystic kidney disease in a patient using lithium chronically
.
Turk Psikiyatri Derg.
24
,
213
216
138
Davis
,
J.
,
Desmond
,
M.
and
Berk
,
M
. (
2018
)
Lithium and nephrotoxicity: Unravelling the complex pathophysiological threads of the lightest metal
.
Nephrology (Carlton).
23
,
897
903
https://doi.org/10.1111/nep.13263
139
Hestbech
,
J.
,
Hansen
,
H.E.
,
Amdisen
,
A.
and
Olsen
,
S
. (
1977
)
Chronic renal lesions following long-term treatment with lithium
.
Kidney Int.
12
,
205
213
https://doi.org/10.1038/ki.1977.102
140
Kjaersgaard
,
G.
,
Madsen
,
K.
,
Marcussen
,
N.
and
Jensen
,
B.L
. (
2014
)
Lithium induces microcysts and polyuria in adolescent rat kidney independent of cyclooxygenase-2
.
Physiol. Rep.
2
, e00202 https://doi.org/10.1002/phy2.202
141
Saadi-Kheddouci
,
S.
,
Berrebi
,
D.
,
Romagnolo
,
B.
,
Cluzeaud
,
F.
,
Peuchmaur
,
M.
,
Kahn
,
A.
et al.
(
2001
)
Early development of polycystic kidney disease in transgenic mice expressing an activated mutant of the beta-catenin gene
.
Oncogene
20
,
5972
5981
https://doi.org/10.1038/sj.onc.1204825
142
Dai
,
C.
,
Stolz
,
D.B.
,
Kiss
,
L.P.
,
Monga
,
S.P.
,
Holzman
,
L.B.
and
Liu
,
Y
. (
2009
)
Wnt/β-Catenin Signaling Promotes Podocyte Dysfunction and Albuminuria
.
J. Am. Soc. Nephrol.
20
,
1997
2008
https://doi.org/10.1681/ASN.2009010019
143
Chen
,
X.
,
Xiao
,
L.
,
Yu
,
S.
,
Ren
,
Z.
,
Wang
,
W.
,
Jia
,
Y.
et al.
(
2023
)
GYY4137, a H2S donor, ameliorates kidney injuries in diabetic mice by modifying renal ROS-associated enzymes
.
Biomedicine & Pharmacotherapy
162
,
114694
https://doi.org/10.1016/j.biopha.2023.114694
144
Umanath
,
K.
and
Lewis
,
J.B
. (
2018
)
Update on Diabetic Nephropathy: Core Curriculum 2018
.
Am. J. Kidney Dis.
71
,
884
895
https://doi.org/10.1053/j.ajkd.2017.10.026
145
Pearce
,
N.J.
,
Arch
,
J.R.S.
,
Clapham
,
J.C.
,
Coghlan
,
M.P.
,
Corcoran
,
S.L.
,
Lister
,
C.A.
et al.
(
2004
)
Development of glucose intolerance in male transgenic mice overexpressing human glycogen synthase kinase-3beta on a muscle-specific promoter
.
Metab. Clin. Exp.
53
,
1322
1330
https://doi.org/10.1016/j.metabol.2004.05.008
146
Liu
,
Y.
,
Tanabe
,
K.
,
Baronnier
,
D.
,
Patel
,
S.
,
Woodgett
,
J.
,
Cras-Méneur
,
C.
et al.
(
2010
)
Conditional ablation of Gsk-3β in islet beta cells results in expanded mass and resistance to fat feeding-induced diabetes in mice
.
Diabetologia
53
,
2600
2610
https://doi.org/10.1007/s00125-010-1882-x
147
Tanabe
,
K.
,
Liu
,
Z.
,
Patel
,
S.
,
Doble
,
B.W.
,
Li
,
L.
,
Cras-Méneur
,
C.
et al.
(
2008
)
Genetic deficiency of glycogen synthase kinase-3beta corrects diabetes in mouse models of insulin resistance
.
PLoS Biol.
6
, e37, e37 https://doi.org/10.1371/journal.pbio.0060037
148
Zolnierkiewicz
,
O.
and
Rogacka
,
D
. (
2024
)
Hyperglycemia - A culprit of podocyte pathology in the context of glycogen metabolism
.
Arch. Biochem. Biophys.
753
,
109927
149
Paeng
,
J.
,
Chang
,
J.H.
,
Lee
,
S.H.
,
Nam
,
B.Y.
,
Kang
,
H.-Y.
,
Kim
,
S.
et al.
(
2014
)
Enhanced glycogen synthase kinase-3β activity mediates podocyte apoptosis under diabetic conditions
.
Apoptosis
19
,
1678
1690
https://doi.org/10.1007/s10495-014-1037-5
150
Li
,
X.Z.
,
Jiang
,
H.
,
Xu
,
L.
,
Liu
,
Y.Q.
,
Tang
,
J.W.
,
Shi
,
J.S
, et al.
(
2021
)
Sarsasapogenin restores podocyte autophagy in diabetic nephropathy by targeting GSK3beta signaling pathway
.
Biochem. Pharmacol.
192
,
114675
.
151
Zhang
,
C.
,
Hou
,
B.
,
Yu
,
S.
,
Chen
,
Q.
,
Zhang
,
N.
and
Li
,
H
. (
2016
)
HGF alleviates high glucose-induced injury in podocytes by GSK3β inhibition and autophagy restoration
.
Biochimica et Biophysica Acta (BBA) - Molecular Cell Research
1863
,
2690
2699
https://doi.org/10.1016/j.bbamcr.2016.08.004
152
Hou
,
B.
,
Li
,
Y.
,
Li
,
X.
,
Zhang
,
C.
,
Zhao
,
Z.
,
Chen
,
Q.
et al.
(
2020
)
HGF protected against diabetic nephropathy via autophagy-lysosome pathway in podocyte by modulating PI3K/Akt-GSK3β-TFEB axis
.
Cell. Signal.
75
,
109744
https://doi.org/10.1016/j.cellsig.2020.109744
153
Liu
,
C.
,
Li
,
Y.
and
Wang
,
X
. (
2022
)
TDAG51-Deficiency podocytes are protected from high-glucose-induced damage through Nrf2 activation via the AKT-GSK-3β pathway
.
Inflammation
45
,
1520
1533
https://doi.org/10.1007/s10753-022-01638-9
154
Shen
,
S.
,
Ji
,
C.
and
Wei
,
K
. (
2022
)
Cellular senescence and regulated cell death of tubular epithelial cells in diabetic kidney disease
.
Front. Endocrinol. (Lausanne)
13
, 924299 https://doi.org/10.3389/fendo.2022.924299
155
Jiang
,
S.
and
Su
,
H
. (
2023
)
Cellular crosstalk of mesangial cells and tubular epithelial cells in diabetic kidney disease
.
Cell Commun. Signal
21
,
288
, 288 https://doi.org/10.1186/s12964-023-01323-w
156
Lin
,
C.-L.
,
Wang
,
J.-Y.
,
Huang
,
Y.-T.
,
Kuo
,
Y.-H.
,
Surendran
,
K.
and
Wang
,
F.-S
. (
2006
)
Wnt/β-Catenin signaling modulates survival of high glucose–stressed mesangial cells
.
J. Am. Soc. Nephrol.
17
,
2812
2820
https://doi.org/10.1681/ASN.2005121355
157
Yang
,
Z.
,
Sun
,
X.
,
Deng
,
X.
,
Jiang
,
W.
,
Liu
,
H
, et al.
(
2024
)
A novel lncRNA TCONS_00071187 upregulated by activated GSK3β promotes high glucose-induced mesangial cell proliferation in the diabetic nephropathy
.
Cell. Mol. Biol. (Noisy-le-grand)
70
,
219
225
https://doi.org/10.14715/cmb/2024.70.1.30
158
Gilbert
,
R.E
. (
2017
)
Proximal tubulopathy: Prime mover and key therapeutic target in diabetic kidney disease
.
Diabetes
66
,
791
800
https://doi.org/10.2337/db16-0796
159
Slyne
,
J.
,
Slattery
,
C.
,
McMorrow
,
T.
and
Ryan
,
M.P
. (
2015
)
New developments concerning the proximal tubule in diabetic nephropathy: in vitro models and mechanisms
.
Nephrol. Dial. Transplant.
30
,
iv60
iv67
https://doi.org/10.1093/ndt/gfv264
160
Mathur
,
A.
,
Pandey
,
V.K.
and
Kakkar
,
P
. (
2018
)
Activation of GSK3β/β-TrCP axis via PHLPP1 exacerbates Nrf2 degradation leading to impairment in cell survival pathway during diabetic nephropathy
.
Free Radical Biology and Medicine
120
,
414
424
https://doi.org/10.1016/j.freeradbiomed.2018.04.550
161
Liu
,
W.
,
Hao
,
J.
,
Zhu
,
L.
,
Li
,
F.
,
Liu
,
Q.
,
Liu
,
S.
et al.
(
2013
)
Phospho-GSK-3β is involved in the high-glucose-mediated lipid deposition in renal tubular cells in diabetes
.
Int. J. Biochem. Cell Biol.
45
,
2066
2075
https://doi.org/10.1016/j.biocel.2013.07.007
162
Liang
,
X.
,
Wang
,
P.
,
Chen
,
B.
,
Ge
,
Y.
,
Gong
,
A.Y.
,
Flickinger
,
B.
et al.
(
2020
)
Glycogen synthase kinase 3β hyperactivity in urinary exfoliated cells predicts progression of diabetic kidney disease
.
Kidney Int.
97
,
175
192
https://doi.org/10.1016/j.kint.2019.08.036
163
Mariappan
,
M.M.
,
Prasad
,
S.
,
D’Silva
,
K.
,
Cedillo
,
E.
,
Sataranatarajan
,
K.
,
Barnes
,
J.L.
et al.
(
2014
)
Activation of Glycogen Synthase Kinase 3β Ameliorates Diabetes-induced Kidney Injury
.
Journal of Biological Chemistry
289
,
35363
35375
https://doi.org/10.1074/jbc.M114.587840
164
Wiggins
,
R.C
. (
2007
)
The spectrum of podocytopathies: a unifying view of glomerular diseases
.
Kidney Int.
71
,
1205
1214
https://doi.org/10.1038/sj.ki.5002222
165
Li
,
C.
,
Ge
,
Y.
,
Peng
,
A.
and
Gong
,
R
. (
2015
)
The redox sensitive glycogen synthase kinase 3β suppresses the self-protective antioxidant response in podocytes upon oxidative glomerular injury
.
Oncotarget
6
,
39493
39506
https://doi.org/10.18632/oncotarget.6303
166
Xu
,
W.
,
Ge
,
Y.
,
Liu
,
Z.
and
Gong
,
R
. (
2014
)
Glycogen synthase kinase 3β dictates podocyte motility and focal adhesion turnover by modulating paxillin activity: implications for the protective effect of low-dose lithium in podocytopathy
.
Am. J. Pathol.
184
,
2742
2756
https://doi.org/10.1016/j.ajpath.2014.06.027
167
Naves
,
M.A.
,
Requião-Moura
,
L.R.
,
Soares
,
M.F.
,
Silva-Júnior
,
J.A.
,
Mastroianni-Kirsztajn
,
G.
and
Teixeira
,
V.P.C
. (
2012
)
Podocyte Wnt/ß-catenin pathway is activated by integrin-linked kinase in clinical and experimental focal segmental glomerulosclerosis
.
J. Nephrol.
25
,
401
409
https://doi.org/10.5301/jn.5000017
168
Ardalan
,
M.
,
Hejazian
,
S.M.
,
Sharabiyani
,
H.F.
,
Farnood
,
F.
,
Ghafari Aghdam
,
A.
,
Bastami
,
M.
et al.
(
2020
)
Dysregulated levels of glycogen synthase kinase-3β (GSK-3β) and miR-135 in peripheral blood samples of cases with nephrotic syndrome
.
PeerJ
8
, e10377 https://doi.org/10.7717/peerj.10377
169
Kato
,
H.
,
Gruenwald
,
A.
,
Suh
,
J.H.
,
Miner
,
J.H.
,
Barisoni-Thomas
,
L.
,
Taketo
,
M.M.
et al.
(
2011
)
Wnt/β-catenin pathway in podocytes integrates cell adhesion, differentiation, and survival
.
J. Biol. Chem.
286
,
26003
26015
https://doi.org/10.1074/jbc.M111.223164
170
Markowitz
,
G.S.
,
Radhakrishnan
,
J.
,
Kambham
,
N.
,
Valeri
,
A.M.
,
Hines
,
W.H.
and
D’Agati
,
V.D
. (
2000
)
Lithium nephrotoxicity: a progressive combined glomerular and tubulointerstitial nephropathy
.
J. Am. Soc. Nephrol.
11
,
1439
1448
https://doi.org/10.1681/ASN.V1181439
171
Petersen
,
C.E.
,
Amaral
,
S.
and
Frosch
,
E
. (
2008
)
Lithium-Induced Nephrotic Syndrome in a Prepubertal Boy
.
J. Child Adolesc. Psychopharmacol.
18
,
210
213
https://doi.org/10.1089/cap.2007.0118
172
Grünfeld
,
J.-P.
and
Rossier
,
B.C
. (
2009
)
Lithium nephrotoxicity revisited
.
Nat. Rev. Nephrol.
5
,
270
276
https://doi.org/10.1038/nrneph.2009.43
173
Presne
,
C.
,
Fakhouri
,
F.
,
Noël
,
L.-H.
,
Stengel
,
B.
,
Even
,
C.
,
Kreis
,
H.
et al.
(
2003
)
Lithium-induced nephropathy: Rate of progression and prognostic factors
.
Kidney Int.
64
,
585
592
https://doi.org/10.1046/j.1523-1755.2003.00096.x
174
Forlenza
,
O.V.
,
De-Paula
,
V.J.R.
and
Diniz
,
B.S.O
. (
2014
)
Neuroprotective effects of lithium: implications for the treatment of alzheimer’s disease and related neurodegenerative disorders
.
ACS Chem. Neurosci.
5
,
443
450
https://doi.org/10.1021/cn5000309
175
Gong
,
R.
,
Wang
,
P.
and
Dworkin
,
L
. (
2016
)
What we need to know about the effect of lithium on the kidney
.
American Journal of Physiology-Renal Physiology
311
,
F1168
F1171
https://doi.org/10.1152/ajprenal.00145.2016
176
Bao
,
H.
,
Ge
,
Y.
,
Wang
,
Z.
,
Zhuang
,
S.
,
Dworkin
,
L.
,
Peng
,
A.
et al.
(
2014
)
Delayed administration of a single dose of lithium promotes recovery from AKI
.
J. Am. Soc. Nephrol.
25
,
488
500
https://doi.org/10.1681/ASN.2013040350
177
Bao
,
H.
,
Zhang
,
Q.
,
Liu
,
X.
,
Song
,
Y.
,
Li
,
X.
,
Wang
,
Z.
et al.
(
2019
)
Lithium targeting of AMPK protects against cisplatin-induced acute kidney injury by enhancing autophagy in renal proximal tubular epithelial cells
.
FASEB J.
33
,
14370
14381
https://doi.org/10.1096/fj.201901712R
178
Reiser
,
J.
,
Loos
,
M.
,
Oh
,
J.
,
Asanuma
,
K.
,
Giardino
,
L
, et al.
(
2004
)
Induction of B7-1 in podocytes is associated with nephrotic syndrome
.
J. Clin. Invest.
113
,
1390
1397
https://doi.org/10.1172/JCI20402
179
Liu
,
Z.
,
Hsu
,
H.
,
Goeddel
,
D.V.
and
Karin
,
M
. (
1996
)
Dissection of TNF receptor 1 effector Functions: JNK activation is not linked to apoptosis while NF-κB activation prevents cell death
.
Cell
87
,
565
576
https://doi.org/10.1016/S0092-8674(00)81375-6
180
Van Antwerp
,
D.J.
,
Martin
,
S.J.
,
Kafri
,
T.
,
Green
,
D.R.
and
Verma
,
I.M
. (
1996
)
Suppression of TNF-α-induced apoptosis by NF-κB
.
Science
274
,
787
789
https://doi.org/10.1126/science.274.5288.787
181
Brähler
,
S.
,
Ising
,
C.
,
Hagmann
,
H.
,
Rasmus
,
M.
,
Hoehne
,
M.
,
Kurschat
,
C.
et al.
(
2012
)
Intrinsic proinflammatory signaling in podocytes contributes to podocyte damage and prolonged proteinuria
.
Am. J. Physiol. Renal Physiol.
303
,
F1473
85
https://doi.org/10.1152/ajprenal.00031.2012
182
Huang
,
B.
,
Yang
,
X.-D.
,
Lamb
,
A.
and
Chen
,
L.-F
. (
2010
)
Posttranslational modifications of NF-kappaB: another layer of regulation for NF-kappaB signaling pathway
.
Cell. Signal.
22
,
1282
1290
https://doi.org/10.1016/j.cellsig.2010.03.017
183
Nørregaard
,
R.
,
Mutsaers
,
H.A.M.
,
Frøkiær
,
J.
and
Kwon
,
T.H
. (
2023
)
Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis
.
Physiol. Rev.
103
,
2827
2872
https://doi.org/10.1152/physrev.00027.2022
184
Li
,
L.
,
Fu
,
H.
and
Liu
,
Y
. (
2022
)
The fibrogenic niche in kidney fibrosis: components and mechanisms
.
Nat. Rev. Nephrol.
18
,
545
557
https://doi.org/10.1038/s41581-022-00590-z
185
Jamadar
,
A.
and
Rao
,
R
. (
2020
)
Glycogen Synthase Kinase-3 Signaling in Acute Kidney Injury
.
Nephron
144
,
609
612
https://doi.org/10.1159/000509354
186
Fiol
,
C.J.
,
Williams
,
J.S.
,
Chou
,
C.H.
,
Wang
,
Q.M.
,
Roach
,
P.J.
and
Andrisani
,
O.M
. (
1994
)
A secondary phosphorylation of CREB341 at Ser129 is required for the cAMP-mediated control of gene expression. A role for glycogen synthase kinase-3 in the control of gene expression
.
Journal of Biological Chemistry
269
,
32187
32193
https://doi.org/10.1016/S0021-9258(18)31619-3
187
Bullock
,
B.P.
and
Habener
,
J.F
. (
1998
)
Phosphorylation of the camp response element binding protein CREB by cAMP-Dependent Protein Kinase A and Glycogen Synthase Kinase-3 Alters DNA-binding affinity, conformation, and increases net charge
.
Biochemistry
37
,
3795
3809
https://doi.org/10.1021/bi970982t
188
Grimes
,
C.A.
and
Jope
,
R.S
. (
2001
)
CREB DNA binding activity is inhibited by glycogen synthase kinase-3 beta and facilitated by lithium
.
J. Neurochem.
78
,
1219
1232
https://doi.org/10.1046/j.1471-4159.2001.00495.x
189
Singh
,
S.P.
,
Tao
,
S.
,
Fields
,
T.A.
,
Webb
,
S.
,
Harris
,
R.C.
and
Rao
,
R
. (
2015
)
Glycogen synthase kinase-3 inhibition attenuates fibroblast activation and development of fibrosis following renal ischemia-reperfusion in mice
.
Dis. Model. Mech.
8
,
931
940
https://doi.org/10.1242/dmm.020511
190
Ren
,
J.
,
Wei
,
H.
,
Sun
,
J.
,
Feng
,
X.
,
Zhang
,
Y.
,
Yuan
,
H.
et al.
(
2022
)
GSK3β-dependent lysosome biogenesis: An effective pathway to mitigate renal fibrosis with LM49
.
Front. Pharmacol.
13
, 925489 https://doi.org/10.3389/fphar.2022.925489
191
Sharif
,
S.
,
Chen
,
B.
,
Brewster
,
P.
,
Chen
,
T.
,
Dworkin
,
L.
and
Gong
,
R
. (
2021
)
Rationale and design of assessing the effectiveness of short-term low-dose lithium therapy in averting cardiac surgery-associated acute kidney injury: a randomized, double blinded, placebo controlled pilot trial
.
Front. Med. (Lausanne)
8
, 639402 https://doi.org/10.3389/fmed.2021.639402
192
Zhang
,
J.
,
Anshul
,
F.
,
Malhotra
,
D.K.
,
Jaume
,
J.
,
Dworkin
,
L.D.
and
Gong
,
R
. (
2021
)
Microdose Lithium Protects against Pancreatic Islet Destruction and Renal Impairment in Streptozotocin-Elicited Diabetes
.
Antioxidants (Basel).
10
,
138
https://doi.org/10.3390/antiox10010138
193
Ryves
,
W.J.
and
Harwood
,
A.J
. (
2001
)
Lithium inhibits glycogen synthase kinase-3 by Competition for magnesium
.
Biochem. Biophys. Res. Commun.
280
,
720
725
https://doi.org/10.1006/bbrc.2000.4169
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).