27,181 research outputs found

    GSK3 inhibition reduces inflammatory responses of microglia and upregulates Il-10 production

    Get PDF
    Introduction: Neurodegeneration resulting from pathogen invasion or tissue damage has been associated with activation of microglia, and exacerbated by the release of neurotoxic mediators such as pro-inflammatory cytokines, chemokines and reactive oxygen species. Activation of microglia stimulated by lipopolysaccharide is mediated in part by GSK-3 signaling molecule. Induced IL-10 expression via GSK-3 inhibition is noteworthy since IL-10 has been remarkably shown to suppress inflammation. Objectives: We aimed to inactivate microglia through inhibition of GSK-3 signaling and to determine its effects on the production of pro- and anti-inflammatory mediators. Methods: LPS-stimulated BV-2 cells were treated with a GSK-3 inhibitor (LiCl, NP12, SB216763 or CHIR99021). Inhibition of GSK-3 was determined by the phosphorylation status of GSK-3β. The effects of GSK-3 inhibition on microglial inflammatory response were investigated by examining various mediators and CD200R marker. Production of nitric oxide (NO), glutamate and pro- and anti-inflammatory cytokines were measured using flow cytometry, Griess assay, glutamate assay and Cytometric Bead Array (CBA) respectively. Results: GSK-3β signaling in LPS-stimulated microglia was blocked by GSK-3 inhibitor through increased phosphorylation at Serine 9 residue. GSK-3 inhibitors had also led to reducing in microglia activity via increased expression of CD200R. Inhibition of GSK-3 also diminished inflammatory mediators such as nitric oxide (NO), glutamate, pro-inflammatory cytokines (TNF-α and IL-6) and chemokine, MCP-1. Reduction of pro-inflammatory mediators by GSK-3 inhibitor was coincided with increased IL-10 production. Conclusions: Suppression of microglia-mediated inflammatory response was facilitated by GSK-3 inhibition with associated increased in IL-10 production

    GSK-3 in Neurodegenerative Diseases

    Get PDF
    Glycogen synthase kinase-3 (GSK-3) regulates multiple cellular processes, and its dysregulation is implicated in the pathogenesis of diverse diseases. In this paper we will focus on the dysfunction of GSK-3 in Alzheimer's disease and Parkinson's disease. Specifically, GSK-3 is known to interact with tau, β-amyloid (Aβ), and α-synuclein, and as such may be crucially involved in both diseases. Aβ production, for example, is regulated by GSK-3, and its toxicity is mediated by GSK-induced tau phosphorylation and degeneration. α-synuclein is a substrate for GSK-3 and GSK-3 inhibition protects against Parkinsonian toxins. Lithium, a GSK-3 inhibitor, has also been shown to affect tau, Aβ, and α-synuclein in cell culture, and transgenic animal models. Thus, understanding the role of GSK-3 in neurodegenerative diseases will enhance our understanding of the basic mechanisms underlying the pathogenesis of these disorders and also facilitate the identification of new therapeutic avenues

    Acute glycogen synthase kinase-3 inhibition modulates human cardiac conduction

    Get PDF
    Glycogen synthase kinase 3 (GSK-3) inhibition has emerged as a potential therapeutic target for several diseases, including cancer. However, the role for GSK-3 regulation of human cardiac electrophysiology remains ill-defined. We demonstrate that SB216763, a GSK-3 inhibitor, can acutely reduce conduction velocity in human cardiac slices. Combined computational modeling and experimental approaches provided mechanistic insight into GSK-3 inhibition-mediated changes, revealing that decreased sodium-channel conductance and tissue conductivity may underlie the observed phenotypes. Our study demonstrates that GSK-3 inhibition in human myocardium alters electrophysiology and may predispose to an arrhythmogenic substrate; therefore, monitoring for adverse arrhythmogenic events could be considered

    Differential effects of glycogen synthase kinase 3 inhibitors on migration in glioblastoma cell lines

    Full text link
    Glioblastoma multiforme (GBM) represents the deadliest brain cancer pathology. Current therapeutic treatment focuses on surgical resection of tumors, followed by adjuvant radiation and/or chemotherapy. The invasive nature of GBM however limits the effectiveness of such therapy. Drug development for GBM has centered on attempts to manage this infiltrative capacity. One avenue of such development focuses on aberrant cellular signaling pathways characteristic of malignancies. This study sought to understand the mechanism by which glycogen synthase kinase 3 (GSK-3) inhibitors differentially regulate GBM migration. Measurement of in vitro cellular migration has traditionally utilized Boyden chamber (transwell) assays and collagen migration assays. Administration of known GSK-3 inhibitors demonstrate a differential effect, whereby migration is inhibited in transwell assays, but maintained in the collagen assay. Cell aggregates from 4 different GBM cell lines were utilized in comparison of GSK-3 inhibitor effects in both the transwell and collagen assays. Contrary to previous findings, GSK-3 inhibitors demonstrated limited effect in reducing migration in the transwell assay, while demonstrating modest effects in the collagen assay. A mechanistic understanding of GSK-3 inhibitor function remains elusive. Previous studies have demonstrated GSK-3 effects on various cytoskeletal proteins, including the WAVE-2 complex, vital for cytoskeletal stability. Administration of GSK-3 inhibitors had previously led to a downregulation of WAVE2 levels. After administration of known GSK-3 inhibitors, this study failed to reproduce previous differences in total WAVE2 protein levels compared to control untreated samples. This evidence indicates a different mode of cytoskeletal regulation must be driving GSK-3 inhibition’s effect on GBM migration. GSK-3 inhibitors represent one promising class of chemotherapeutic agents for treatment of GBM. This study corroborated previous findings about the effect of classical GSK-3 inhibitors (BIO, CHI99021) in reducing GBM migration. Further investigation is necessary to isolate the specific cellular mechanisms responsible for GSK-3’s effects. With rapid development of other therapeutic strategies, GSK-3 inhibitors present one class of drugs available to clinicians for effective therapeutic management of GBM

    Identification of AIP as a GSK-3 binding protein

    Get PDF
    GSK-3, a well-known serine/threonine kinase is one of the key players controlling numerous cellular and physiological processes such as protein synthesis, cell poliferation, cellular differentiation, apoptosis and microtubule dynamics. Therefore, GSK-3 phosphorylates and regulates the functions of a diverse group of substrates including many transcription factors, components regulating the cell cycles and signaling proteins. However, the mechanisms by which GSK-3 regulates the functions of many substrates specifically and selectively are not known. In order to understand the molecular basis of GSK-3 regulation and specificity, we attempt to search for novel GSK-3 binding proteins using yeast two-hybrid screening. We have identified AIP (Aurora-A Kinase Interacting Protein) as a protein that interacts with GSK-3. AIP has been reported to be a novel negative regulator of Aurora-A kinase where it might down-regulates Aurora-A kinase through proteasome dependent degradation. Our study showed that AIP is able to bind both the homologous forms of GSK-3, GSK-3a and GSK-3b in intact cells. This binding is not affected by SB216763, a specific GSK-3 inhibitor, indicating that the kinase activity of GSK-3 is not required for the interaction. AIP has the consensus motif –S-X-X-X-S- for substrate phosphorylation by GSK-3b and i sphosphorylated by GSK-3b in vitro. Our results suggest that AIPis a novel binding partner of GSK-3

    Involvement of the Glycogen Synthase Kinase-3 Signaling Pathway in TBI Pathology and Neurocognitive Outcome

    Get PDF
    BACKGROUND: Traumatic brain injury (TBI) sets in motion cascades of biochemical changes that result in delayed cell death and altered neuronal architecture. Studies have demonstrated that inhibition of glycogen synthase kinase-3 (GSK-3) effectively reduces apoptosis following a number of stimuli. The Wnt family of proteins, and growth factors are two major factors that regulate GSK-3 activity. In the absence of stimuli, GSK-3 is constitutively active and is complexed with Axin, adenomatous polyposis coli (APC), and casein kinase Iα (CK1α) and phosphorylates ß-Catenin leading to its degradation. Binding of Wnt to Frizzled receptors causes the translocation of GSK-3 to the plasma membrane, where it phosphorylates and inactivates the Frizzled co-receptor lipoprotein-related protein 6 (LRP6). Furthermore, the translocation of GSK-3 reduces ß-Catenin phosphorylation and degradation, leading to ß-Catenin accumulation and gene expression. Growth factors activate Akt, which in turn inhibits GSK-3 activity by direct phosphorylation, leading to a reduction in apoptosis. METHODOLOGY/PRINCIPAL FINDINGS: Using a rodent model, we found that TBI caused a rapid, but transient, increase in LRP6 phosphorylation that is followed by a modest decrease in ß-Catenin phosphorylation. Phospho-GSK-3β immunoreactivity was found to increase three days post injury, a time point at which increased Akt activity following TBI has been observed. Lithium influences several neurochemical cascades, including inhibiting GSK-3. When the efficacy of daily lithium was assessed, reduced hippocampal neuronal cell loss and learning and memory improvements were observed. These influences were partially mimicked by administration of the GSK-3-selective inhibitor SB-216763, as this drug resulted in improved motor function, but only a modest improvement in memory retention and no overt neuroprotection. CONCLUSION/SIGNIFICANCE: Taken together, our findings suggest that selective inhibition of GSK-3 may offer partial cognitive improvement. As a broad spectrum inhibitor of GSK-3, lithium offers neuroprotection and robust cognitive improvement, supporting its clinical testing as a treatment for TBI

    AT7519, a Novel Small Molecule Multi-Cyclin Dependent Kinase Inhibitor, Induces Apoptosis in Multiple Myeloma VIA GSK3 beta

    Get PDF
    Dysregulated cell cycling is a universal hallmark of cancer and is often mediated by abnormal activation of cyclin-dependent kinases (CDKs) and their cyclin partners. Overexpression of individual complexes are reported in multiple myeloma (MM), making them attractive therapeutic targets. In this study, we investigate the preclinical activity of a novel small-molecule multi-CDK inhibitor, AT7519, in MM. We show the anti-MM activity of AT7519 displaying potent cytotoxicity and apoptosis; associated with in vivo tumor growth inhibition and prolonged survival. At the molecular level, AT7519 inhibited RNA polymerase II (RNA pol II) phosphorylation, a CDK9, 7 substrate, associated with decreased RNA synthesis confirmed by [(3)H] Uridine incorporation. In addition, AT7519 inhibited glycogen synthase kinase 3beta (GSK-3beta) phosphorylation; conversely pretreatment with a selective GSK-3 inhibitor and shRNA GSK-3beta knockdown restored MM survival, suggesting the involvement of GSK-3beta in AT7519-induced apoptosis. GSK-3beta activation was independent of RNA pol II dephosphorylation confirmed by alpha-amanitin, a specific RNA pol II inihibitor, showing potent inhibition of RNA pol II phosphorylation without corresponding effects on GSK-3beta phosphorylation. These results offer new insights into the crucial, yet controversial role of GSK-3beta in MM and show significant anti-MM activity of AT7519, providing the rationale for its clinical evaluation in MM

    A screen for transcription factor targets of glycogen synthase kinase-3 highlights an inverse correlation of NFκB and androgen receptor signaling in prostate cancer

    Get PDF
    Expression of Glycogen Synthase Kinase-3 (GSK-3) is elevated in prostate cancer and its inhibition reduces prostate cancer cell proliferation, in part by reducing androgen receptor (AR) signaling. However, GSK-3 inhibition can also activate signals that promote cell proliferation and survival, which may preclude the use of GSK-3 inhibitors in the clinic. To identify such signals in prostate cancer, we screened for changes in transcription factor target DNA binding activity in GSK-3-silenced cells. Among the alterations was a reduction in AR DNA target binding, as predicted from previous studies, and an increase in NFκB DNA target binding. Consistent with the latter, gene silencing of GSK-3 or inhibition using the GSK-3 inhibitor CHIR99021 increased basal NFκB transcriptional activity. Activation of NFκB was accompanied by an increase in the level of the NFκB family member RelB. Conversely, silencing RelB reduced activation of NFκB by CHIR99021. Furthermore, the reduction of prostate cancer cell proliferation by CHIR99021 was potentiated by inhibition of NFκB signaling using the IKK inhibitor PS1145. Finally, stratification of human prostate tumor gene expression data for GSK3 revealed an inverse correlation between NFκB-dependent and androgen-dependent gene expression, consistent with the results from the transcription factor target DNA binding screen. In addition, there was a correlation between expression of androgen-repressed NFκB target genes and reduced survival of patients with metastatic prostate cancer. These findings highlight an association between GSK-3/AR and NFκB signaling and its potential clinical importance in metastatic prostate cancer

    IN SILICO STUDIES ON NEW INDAZOLE DERIVATIVES AS GSK-3β INHIBITORS

    Get PDF
    Objective: In silico studies were conducted on newly proposed Indazole derivatives as GSK-3β inhibitors to select the best possible drug candidates based on drug properties and bioactivity score of the compounds. Methods: 31 Indazole derivatives and active GSK-3β Indazole inhibitor 3-(5-chloro-1-methyl-indol-3-yl)-4-[1-[3-(triazol-1-yl)propyl]indazol-3-yl]pyrrole-2,5-dione(IC50 of 0.003 μM) were subjected to predict the mutagenic, tumorigenic, irritant, reproductive risks, and drug-relevant properties using OSIRIS Property Explorer. Further bioactivity scores were determined using Molinspiration online tools. Results: The results of new GSK-3β inhibitors were compared with potent GSK-3β Indazole inhibitor to examine the prospective of the optimized compounds. The best possible drug candidates were reported after comprehensive analysis on predicted cLogP, solubility, molecular weight, topological molecular polar surface area (TPSA), drug- likeness, drug score properties and bioactivity score for different human targets like GPCR, ion channel, kinase, nuclear receptor, protease and enzymes. Conclusion: Five compounds 282, 141, 161, 108 and 456 were reported as the best drug like candidates for GSK-3β regulation

    Herpes Simplex Virus type-1 infection induces synaptic dysfunction in cultured cortical neurons via GSK-3 activation and intraneuronal amyloid-β protein accumulation

    Get PDF
    Increasing evidence suggests that recurrent Herpes Simplex Virus type 1 (HSV-1) infection spreading to the CNS is a risk factor for Alzheimer's Disease (AD) but the underlying mechanisms have not been fully elucidated yet. Here we demonstrate that in cultured mouse cortical neurons HSV-1 induced Ca 2+ -dependent activation of glycogen synthase kinase (GSK)-3. This event was critical for the HSV-1-dependent phosphorylation of amyloid precursor protein (APP) at Thr668 and the following intraneuronal accumulation of amyloid-β protein (Aβ). HSV-1-infected neurons also exhibited: i) significantly reduced expression of the presynaptic proteins synapsin-1 and synaptophysin; ii) depressed synaptic transmission. These effects depended on GSK-3 activation and intraneuronal accumulation of Aβ. In fact, either the selective GSK-3 inhibitor, SB216763, or a specific antibody recognizing Aβ (4G8) significantly counteracted the effects induced by HSV-1 at the synaptic level. Moreover, in neurons derived from APP KO mice and infected with HSV-1 Aβ accumulation was not found and synaptic protein expression was only slightly reduced when compared to wild-type infected neurons. These data further support our contention that HSV-1 infections spreading to the CNS may contribute to AD phenotype
    corecore