Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.034
Filter
1.
Pharmacol Rev ; 76(3): 323-357, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697859

ABSTRACT

Over the last six decades, lithium has been considered the gold standard treatment for the long-term management of bipolar disorder due to its efficacy in preventing both manic and depressive episodes as well as suicidal behaviors. Nevertheless, despite numerous observed effects on various cellular pathways and biologic systems, the precise mechanism through which lithium stabilizes mood remains elusive. Furthermore, there is recent support for the therapeutic potential of lithium in other brain diseases. This review offers a comprehensive examination of contemporary understanding and predominant theories concerning the diverse mechanisms underlying lithium's effects. These findings are based on investigations utilizing cellular and animal models of neurodegenerative and psychiatric disorders. Recent studies have provided additional support for the significance of glycogen synthase kinase-3 (GSK3) inhibition as a crucial mechanism. Furthermore, research has shed more light on the interconnections between GSK3-mediated neuroprotective, antioxidant, and neuroplasticity processes. Moreover, recent advancements in animal and human models have provided valuable insights into how lithium-induced modifications at the homeostatic synaptic plasticity level may play a pivotal role in its clinical effectiveness. We focused on findings from translational studies suggesting that lithium may interface with microRNA expression. Finally, we are exploring the repurposing potential of lithium beyond bipolar disorder. These recent findings on the therapeutic mechanisms of lithium have provided important clues toward developing predictive models of response to lithium treatment and identifying new biologic targets. SIGNIFICANCE STATEMENT: Lithium is the drug of choice for the treatment of bipolar disorder, but its mechanism of action in stabilizing mood remains elusive. This review presents the latest evidence on lithium's various mechanisms of action. Recent evidence has strengthened glycogen synthase kinase-3 (GSK3) inhibition, changes at the level of homeostatic synaptic plasticity, and regulation of microRNA expression as key mechanisms, providing an intriguing perspective that may help bridge the mechanistic gap between molecular functions and its clinical efficacy as a mood stabilizer.


Subject(s)
Lithium Compounds , Humans , Animals , Lithium Compounds/pharmacology , Lithium Compounds/therapeutic use , Antimanic Agents/pharmacology , Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Neuronal Plasticity/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors
2.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667283

ABSTRACT

Astrocytes and ependymal cells have been reported to be able to switch from a mature cell identity towards that of a neural stem/progenitor cell. Astrocytes are widely scattered in the brain where they exert multiple functions and are routinely targeted for in vitro and in vivo reprogramming. Ependymal cells serve more specialized functions, lining the ventricles and the central canal, and are multiciliated, epithelial-like cells that, in the spinal cord, act as bi-potent progenitors in response to injury. Here, we isolate or generate ependymal cells and post-mitotic astrocytes, respectively, from the lateral ventricles of the mouse brain and we investigate their capacity to reverse towards a progenitor-like identity in culture. Inhibition of the GSK3 and TGFß pathways facilitates the switch of mature astrocytes to Sox2-expressing, mitotic cells that generate oligodendrocytes. Although this medium allows for the expansion of quiescent NSCs, isolated from live rats by "milking of the brain", it does not fully reverse astrocytes towards the bona fide NSC identity; this is a failure correlated with a concomitant lack of neurogenic activity. Ependymal cells could be induced to enter mitosis either via exposure to neuraminidase-dependent stress or by culturing them in the presence of FGF2 and EGF. Overall, our data confirm that astrocytes and ependymal cells retain a high capacity to reverse to a progenitor identity and set up a simple and highly controlled platform for the elucidation of the molecular mechanisms that regulate this reversal.


Subject(s)
Astrocytes , Ependyma , Phenotype , Animals , Astrocytes/metabolism , Astrocytes/cytology , Ependyma/cytology , Ependyma/metabolism , Mice , Cells, Cultured , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Cell Differentiation , Brain/cytology , Brain/metabolism , Rats , SOXB1 Transcription Factors/metabolism , Mice, Inbred C57BL , Mitosis , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Animals, Newborn
3.
Curr Med Chem ; 30(13): 1502-1528, 2023.
Article in English | MEDLINE | ID: mdl-35078392

ABSTRACT

All cells and intracellular components are remodeled and recycled in order to replace the old and damaged cells. Autophagy is a process by which damaged, and unwanted cells are degraded in the lysosomes. There are three different types of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. Autophagy has an effect on adaptive and innate immunity, suppression of any tumour, and the elimination of various microbial pathogens. The process of autophagy has both positive and negative effects, and this pertains to any specific disease or its stage of progression. Autophagy involves various processes which are controlled by various signaling pathways, such as Jun N-terminal kinase, GSK3, ERK1, Leucine-rich repeat kinase 2, and PTEN-induced putative kinase 1 and parkin RBR E3. Protein kinases are also important for the regulation of autophagy as they regulate the process of autophagy either by activation or inhibition. The present review discusses the kinase catalyzed phosphorylated reactions, the kinase inhibitors, types of protein kinase inhibitors and their binding properties to protein kinase domains, the structures of active and inactive kinases, and the hydrophobic spine structures in active and inactive protein kinase domains. The intervention of autophagy by targeting specific kinases may form the mainstay of treatment of many diseases and lead the road to future drug discovery.


Subject(s)
Autophagy , Protein Kinase Inhibitors , Humans , Autophagy/drug effects , Autophagy/immunology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Immunity, Innate , Signal Transduction , Protein Kinase Inhibitors/pharmacology
4.
Med Oncol ; 40(1): 44, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36481875

ABSTRACT

Glycogen Synthase Kinase-3 (GSK-3) was recently implicated in the dysregulated biology of acute myeloid leukemia (AML). Low concentrations of GSK-3 inhibitors, SB216763 and BIO, suppressed the proliferation of AML cells with FLT3-ITD as early as 24 h after treatment. BIO was used in subsequent assays since it exhibited higher inhibitory effects than SB216763. BIO-induced G1 cell cycle arrest by regulating the expression of cyclin D2 and p21 in MV4-11 cells, and promoted apoptosis by regulating the cleaved-caspase3 signaling pathways. In vivo assays demonstrated that BIO suppressed tumor growth, while metabolomics assay showed that BIO reduced the levels of ATP and pyruvate in MV4-11 cells suggesting that it inhibited glycolysis. BIO markedly suppressed cell growth and induced apoptosis of AML cells with FLT3-ITD by partially inhibiting glycolysis, suggesting that BIO may be a promising therapeutic candidate for AML.


Subject(s)
Glycogen Synthase Kinase 3 , Leukemia , Humans , Cell Proliferation , fms-Like Tyrosine Kinase 3/genetics , Glycogen Synthase Kinase 3/antagonists & inhibitors , Metabolomics , Cell Line, Tumor , Cell Cycle Checkpoints
5.
Thorac Cancer ; 13(16): 2318-2330, 2022 08.
Article in English | MEDLINE | ID: mdl-35790883

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma (MPM) is one of the most aggressive tumors with few effective treatments worldwide. It has been suggested that alternative splicing at the transcriptome level plays an indispensable role in MPM. METHODS: We analyzed the splicing profile of 84 MPM patients from the TCGA cohort by using seven typical splicing types. We classified MPM patients based on their splicing status and conducted a comprehensive analysis of the correlation between the splicing classification and clinical characteristics, genetic variation, pathway changes, immune heterogeneity, and potential therapeutic targets. RESULTS: The expression of the alternative splicing regulator SRPK1 is significantly higher in MPM tissues than in normal tissues, and correlates with poor survival. SRPK1 deficiency promotes MPM cell apoptosis and inhibits cell migration in vitro. We divided the MPM patients into four clusters based on their splicing profile and identified two clusters associated with the shortest (cluster 3) and longest (cluster 4) survival time. We present the different gene signatures of each cluster that are related to survival and splicing. Comprehensive analysis of data from the GDSC and TCGA databases revealed that cluster 3 MPM patients could respond well to the small-molecule inhibitor CHIR-99021, a small-molecule inhibitor of GSK-3. CONCLUSION: We performed unsupervised clustering of alternative splicing data from 84 MPM patients from the TCGA database and identified a cluster associated with the worst prognosis that was sensitive to a GSK-3 inhibitor.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Alternative Splicing , Cell Line, Tumor , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Lung Neoplasms/pathology , Mesothelioma/drug therapy , Mesothelioma/genetics , Mesothelioma/pathology , Pleural Neoplasms/drug therapy , Pleural Neoplasms/genetics , Pleural Neoplasms/pathology , Protein Serine-Threonine Kinases
6.
Emerg Microbes Infect ; 11(1): 391-405, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34985411

ABSTRACT

ABSTRACTHIV-1 latency posts a major obstacle for HIV-1 eradication. Currently, no desirable latency reversing agents (LRAs) have been implicated in the "Shock and Kill" strategy to mobilize the latently infected cells to be susceptible for clearance by immune responses. Identification of key cellular pathways that modulate HIV-1 latency helps to develop efficient LRAs. In this study, we demonstrate that the Wnt downstream ß-catenin/TCF1 pathway is a crucial modulator for HIV-1 latency. The pharmacological activation of the ß-catenin/TCF1 pathway with glycogen synthase kinase-3 (GSK3) inhibitors promoted transcription of HIV-1 proviral DNA and reactivated latency in CD4+ T cells; the GSK3 kinase inhibitor 6-bromoindirubin-3'-oxime (6-BIO)-induced HIV-1 reactivation was subsequently confirmed in resting CD4+ T cells from cART-suppressed patients and SIV-infected rhesus macaques. These findings advance our understanding of the mechanisms responsible for viral latency, and provide the potent LRA that can be further used in conjunction of immunotherapies to eradicate viral reservoirs.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Glycogen Synthase Kinase 3/antagonists & inhibitors , HIV-1/growth & development , Indoles/pharmacology , Oximes/pharmacology , Virus Activation/drug effects , Virus Latency/drug effects , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , HIV-1/drug effects , HIV-1/genetics , HeLa Cells , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Macaca mulatta , Transcription, Genetic/drug effects , U937 Cells , Virus Activation/genetics , Virus Latency/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism
7.
Cancer Lett ; 524: 259-267, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34715251

ABSTRACT

Merkel cell carcinoma is an aggressive skin cancer frequently caused by the Merkel cell polyomavirus (MCPyV). Since proliferation of MCPyV-positive MCC tumor cells strictly depends on expression of the virus-encoded T antigens (TA), these proteins theoretically represent ideal targets for different kinds of therapeutic approaches. Here we developed a cell-based assay to identify compounds which specifically inhibit growth of MCC cells by repressing TA expression. Applying this technique we screened a kinase inhibitor library and identified six compounds targeting glycogen synthase kinase 3 (GSK3) such as CHIR99021 as suppressors of TA transcription in MCC cells. Involvement of GSK3α and -ß in the regulation of TA-expression was confirmed by combining GSK3A knockout with inducible GSK3B shRNA knockdown since double knockouts could not be generated. Finally, we demonstrate that CHIR99021 exhibits in vivo antitumor activity in an MCC xenograft mouse model suggesting GSK3 inhibitors as potential therapeutics for the treatment of MCC in the future.


Subject(s)
Antigens, Viral, Tumor/genetics , Carcinoma, Merkel Cell/drug therapy , Glycogen Synthase Kinase 3/genetics , Skin Neoplasms/drug therapy , Animals , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/pathology , Carcinoma, Merkel Cell/virology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Merkel cell polyomavirus/drug effects , Merkel cell polyomavirus/pathogenicity , Mice , Pyridines/pharmacology , Pyrimidines/pharmacology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
8.
J Med Chem ; 65(2): 1342-1351, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34464131

ABSTRACT

Herein we present the evaluation of 11C-labeled-maleimides as radiotracers for positron emission tomography imaging of GSK-3 associated with Alzheimer's disease (AD). 3-Acetyl-4-(1-[11C]-methyl-1H-indol-3-yl)[1H]pyrrole-2,5-dione ([11C]-2) was obtained by direct methylation using [11C]-CH3I and Cs2CO3 in DMF with a 31 ± 4% radiochemical yield and a radiochemical purity of 97.7 ± 0.8%. [11C]-2 was stable both in its final formulation and in human plasma for 120 min and had a plasma protein binding of 70 ± 1% and a LogD7.4 value of 1.84 ± 0.04. [11C]-2 ex vivo biodistributions in healthy animals demonstrated significant brain uptake and retention, showing its ability to penetrate the intact blood-brain barrier. In vivo PET imaging in mice bearing AD showed, with respect to normal animals, significant differences in uptake in the hypothalamus, the striatum, and the amygdala and a significant increase in amygdala uptake in later stages of the pathology. These results are very promising, and further studies are being performed for a complete validation of this compound as novel tracer for AD.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Carbon Radioisotopes/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Maleimides/chemistry , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/pharmacology , Alzheimer Disease/diagnostic imaging , Animals , Brain/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proof of Concept Study , Radiochemistry , Radiopharmaceuticals/pharmacology
9.
J Dent Res ; 101(1): 46-53, 2022 01.
Article in English | MEDLINE | ID: mdl-34152872

ABSTRACT

Small-molecule drugs targeting glycogen synthase kinase 3 (GSK3) as inhibitors of the protein kinase activity are able to stimulate reparative dentine formation. To develop this approach into a viable clinical treatment for exposed pulp lesions, we synthesized a novel, small-molecule noncompetitive adenosine triphosphate (ATP) drug that can be incorporated into a biodegradable hydrogel for placement by syringe into the tooth. This new drug, named NP928, belongs to the thiadiazolidinone (TDZD) family and has equivalent activity to similar drugs of this family such as tideglusib. However, NP928 is more water soluble than other TDZD drugs, making it more suitable for direct delivery into pulp lesions. We have previously reported that biodegradable marine collagen sponges can successfully deliver TDZD drugs to pulp lesions, but this involves in-theater preparation of the material, which is not ideal in a clinical context. To improve surgical handling and delivery, here we incorporated NP928 into a specifically tailored hydrogel that can be placed by syringe into a damaged tooth. This hydrogel is based on biodegradable hyaluronic acid and can be gelled in situ upon dental blue light exposure, similarly to other common dental materials. NP928 released from hyaluronic acid-based hydrogels upregulated Wnt/ß-catenin activity in pulp stem cells and fostered reparative dentine formation compared to marine collagen sponges delivering equivalent concentrations of NP928. This drug-hydrogel combination has the potential to be rapidly developed into a therapeutic procedure that is amenable to general dental practice.


Subject(s)
Dentin, Secondary , Dentinogenesis , Glycogen Synthase Kinase 3/antagonists & inhibitors , Thiadiazoles/pharmacology , Dental Pulp , Dentinogenesis/drug effects , Humans , Hydrogels
10.
Malays J Pathol ; 43(3): 413-424, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34958063

ABSTRACT

INTRODUCTION: GSK-3 is an immune regulator that plays a role in the modulation of cytokine-producing effector T cells associated with inflammation and demyelination of the CNS in EAE. OBJECTIVE: This study aimed to evaluate the treatment paradigm of a single dose of GSK-3 inhibitor administration at various time courses for the protection of the CNS from EAE. MATERIALS AND METHODS: Effects of GSK-3 inhibition on intracellular cytokine levels were evaluated from in vitro naïve CD4+ T cell cultures. Immunized C57BL/6 female mice with MOG35-55 in conjunction with CFA and Ptx were used as a chronic inflammatory EAE disease model. Tideglusib (NP12), a Thiadiazolidinone class, selective, and non-ATP competitive GSK-3 inhibitor, was injected intraperitoneally at pre-EAE, same-day of immunization or disease onset. After 30 days post-immunization, brain, and spinal cord tissues were collected for inflammation and demyelination analysis by H&E and luxol fast blue staining, respectively, whereas cytokine profiles of the serum were assessed by cytokine beads array. RESULTS: The inhibition of GSK-3 in CD4+ T cells increased IL-10 production. The administration of Tideglusib during pre-EAE and same-day, but not during disease onset, significantly reduced clinical symptoms and delayed disease onset. Histopathological analysis of spinal cord tissues showed a significant decline in the number of inflammatory cell infiltration with a concomitant reduction in demyelination through the blocking of GSK-3, especially during pre-EAE and sameday. Upregulation of IL-10 via GSK-3 inhibition coincided with the downregulation of cytokineassociated effector T cells, including IFN-γ, IL-9, IL-17A, IL-17F, IL-21, and IL-23. Increased IL-4 production, however, was only significant in the pre-EAE group. CONCLUSION: The neuroprotective effects of Tideglusib against EAE are time-dependent. Downregulation of Th1 and Th17 hallmark cytokines by Tideglusib in EAE may be associated with IL-10 production.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Glycogen Synthase Kinase 3/antagonists & inhibitors , Animals , CD4-Positive T-Lymphocytes/pathology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glycogen Synthase Kinase 3 beta , Mice , Mice, Inbred C57BL , Neuroprotective Agents , Th17 Cells/pathology , Thiadiazoles/pharmacology , Time Factors
11.
Eur J Med Chem ; 226: 113889, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34649182

ABSTRACT

GSK3 is a promising target for the treatment of Alzheimer's disease. Here, we describe the design and synthesize of a series of GSK3 degraders based on a click chemistry platform. A series of highly potent GSK3 degraders were obtained. Among them, PT-65 exhibited most potent degradation potency against GSK3α (DC50 = 28.3 nM) and GSK3ß (DC50 = 34.2 nM) in SH-SY5Y cells. SPR assay confirmed that PT-65 binds to GSK3ß with high affinity (KD = 12.41 nM). The proteomic study indicated that PT-65 could selectively induced GSK3 degradation. Moreover, PT-65 could effectively suppress GSK3ß and Aß mediated tau hyperphosphorylation in a dose-dependent manner and protect SH-SY5Y cells from Aß caused cell damage. We also confirmed that PT-65 could suppress OA induced tau hyperphosphorylation and ameliorate learning and memory impairments in vivo model of AD. In summary, PT-65 might be a promising candidate for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Drug Discovery , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Alzheimer Disease/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glycogen Synthase Kinase 3/metabolism , Humans , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Proteolysis/drug effects , Structure-Activity Relationship
12.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Article in English | MEDLINE | ID: mdl-34593624

ABSTRACT

The coronaviruses responsible for severe acute respiratory syndrome (SARS-CoV), COVID-19 (SARS-CoV-2), Middle East respiratory syndrome-CoV, and other coronavirus infections express a nucleocapsid protein (N) that is essential for viral replication, transcription, and virion assembly. Phosphorylation of N from SARS-CoV by glycogen synthase kinase 3 (GSK-3) is required for its function and inhibition of GSK-3 with lithium impairs N phosphorylation, viral transcription, and replication. Here we report that the SARS-CoV-2 N protein contains GSK-3 consensus sequences and that this motif is conserved in diverse coronaviruses, raising the possibility that SARS-CoV-2 may be sensitive to GSK-3 inhibitors, including lithium. We conducted a retrospective analysis of lithium use in patients from three major health systems who were PCR-tested for SARS-CoV-2. We found that patients taking lithium have a significantly reduced risk of COVID-19 (odds ratio = 0.51 [0.35-0.74], P = 0.005). We also show that the SARS-CoV-2 N protein is phosphorylated by GSK-3. Knockout of GSK3A and GSK3B demonstrates that GSK-3 is essential for N phosphorylation. Alternative GSK-3 inhibitors block N phosphorylation and impair replication in SARS-CoV-2 infected lung epithelial cells in a cell-type-dependent manner. Targeting GSK-3 may therefore provide an approach to treat COVID-19 and future coronavirus outbreaks.


Subject(s)
COVID-19/prevention & control , Coronavirus Nucleocapsid Proteins/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Lithium Compounds/therapeutic use , Adult , Aged , Female , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , Humans , Lithium Compounds/pharmacology , Male , Middle Aged , Molecular Targeted Therapy , Phosphoproteins/metabolism , Phosphorylation/drug effects , Retrospective Studies
13.
Biomolecules ; 11(7)2021 07 16.
Article in English | MEDLINE | ID: mdl-34356660

ABSTRACT

Leishmaniasis is a public health disease that requires the development of more effective treatments and the identification of novel molecular targets. Since blocking the PI3K/AKT pathway has been successfully studied as an effective anticancer strategy for decades, we examined whether the same approach would also be feasible in Leishmania due to their high amount and diverse set of annotated proteins. Here, we used a best reciprocal hits protocol to identify potential protein kinase homologues in an annotated human PI3K/AKT pathway. We calculated their ligandibility based on available bioactivity data of the reported homologues and modelled their 3D structures to estimate the druggability of their binding pockets. The models were used to run a virtual screening method with molecular docking. We found and studied five protein kinases in five different Leishmania species, which are AKT, CDK, AMPK, mTOR and GSK3 homologues from the studied pathways. The compounds found for different enzymes and species were analysed and suggested as starting point scaffolds for the design of inhibitors. We studied the kinases' participation in protein-protein interaction networks, and the potential deleterious effects, if inhibited, were supported with the literature. In the case of Leishmania GSK3, an inhibitor of its human counterpart, prioritized by our method, was validated in vitro to test its anti-Leishmania activity and indirectly infer the presence of the enzyme in the parasite. The analysis contributes to improving the knowledge about the presence of similar signalling pathways in Leishmania, as well as the discovery of compounds acting against any of these kinases as potential molecular targets in the parasite.


Subject(s)
Leishmania/drug effects , Leishmania/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protozoan Proteins/metabolism , Binding Sites , Drug Evaluation, Preclinical , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Maps , Protein Kinases/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry
14.
Molecules ; 26(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34361750

ABSTRACT

The purpose of this work is to investigate the protein kinase inhibitory activity of constituents from Acacia auriculiformis stem bark. Column chromatography and NMR spectroscopy were used to purify and characterize betulin from an ethyl acetate soluble fraction of acacia bark. Betulin, a known inducer of apoptosis, was screened against a panel of 16 disease-related protein kinases. Betulin was shown to inhibit Abelson murine leukemia viral oncogene homolog 1 (ABL1) kinase, casein kinase 1ε (CK1ε), glycogen synthase kinase 3α/ß (GSK-3 α/ß), Janus kinase 3 (JAK3), NIMA Related Kinase 6 (NEK6), and vascular endothelial growth factor receptor 2 kinase (VEGFR2) with activities in the micromolar range for each. The effect of betulin on the cell viability of doxorubicin-resistant K562R chronic myelogenous leukemia cells was then verified to investigate its putative use as an anti-cancer compound. Betulin was shown to modulate the mitogen-activated protein (MAP) kinase pathway, with activity similar to that of imatinib mesylate, a known ABL1 kinase inhibitor. The interaction of betulin and ABL1 was studied by molecular docking, revealing an interaction of the inhibitor with the ABL1 ATP binding pocket. Together, these data demonstrate that betulin is a multi-target inhibitor of protein kinases, an activity that can contribute to the anticancer properties of the natural compound and to potential treatments for leukemia.


Subject(s)
Acacia/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Triterpenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Apoptosis/genetics , Binding Sites , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase 1 epsilon/genetics , Casein Kinase 1 epsilon/metabolism , Cell Proliferation/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , K562 Cells , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , NIMA-Related Kinases/antagonists & inhibitors , NIMA-Related Kinases/genetics , NIMA-Related Kinases/metabolism , Plant Bark/chemistry , Plant Extracts/chemistry , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Triterpenes/chemistry , Triterpenes/isolation & purification , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Bioorg Med Chem ; 40: 116179, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33991821

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) has been implicated in numerous pathologies making GSK-3 an attractive therapeutic target. Our group has identified a compound termed COB-187 that is a potent and selective inhibitor of GSK-3. In this study, we probed the mechanism by which COB-187 inhibits GSK-3ß. Progress curves, generated via real-time monitoring of kinase activity, indicated that COB-187 inhibition of GSK-3ß is time-dependent and subsequent jump dilution assays revealed that COB-187 binding to GSK-3ß is reversible. Further, a plot of the kinetic constant (kobs) versus COB-187 concentration suggested that, within the range of concentrations studied, COB-187 binds to GSK-3ß via an induced-fit mechanism. There is a critical cysteine residue at the entry to the active site of GSK-3ß (Cys-199). We generated a mutant version of GSK-3ß wherein Cys-199 was substituted with an alanine. This mutation caused a dramatic decrease in the activity of COB-187; specifically, an IC50 in the nM range for wild type versus >100 µM for the mutant. A screen of COB-187 against 34 kinases that contain a conserved cysteine in their active site revealed that COB-187 is highly selective for GSK-3 indicating that COB-187's inhibition of GSK-3ß via Cys-199 is specific. Combined, these findings suggest that COB-187 inhibits GSK-3ß via a specific, reversible, time and Cys-199-dependent mechanism.


Subject(s)
Cystine/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Binding Sites/drug effects , Cystine/metabolism , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Humans , Molecular Structure , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Time Factors
16.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-33848269

ABSTRACT

Transitions between cell fates commonly occur in development and disease. However, reversing an unwanted cell transition in order to treat disease remains an unexplored area. Here, we report a successful process of guiding ill-fated transitions toward normalization in vascular calcification. Vascular calcification is a severe complication that increases the all-cause mortality of cardiovascular disease but lacks medical therapy. The vascular endothelium is a contributor of osteoprogenitor cells to vascular calcification through endothelial-mesenchymal transitions, in which endothelial cells (ECs) gain plasticity and the ability to differentiate into osteoblast-like cells. We created a high-throughput screening and identified SB216763, an inhibitor of glycogen synthase kinase 3 (GSK3), as an inducer of osteoblastic-endothelial transition. We demonstrated that SB216763 limited osteogenic differentiation in ECs at an early stage of vascular calcification. Lineage tracing showed that SB216763 redirected osteoblast-like cells to the endothelial lineage and reduced late-stage calcification. We also found that deletion of GSK3ß in osteoblasts recapitulated osteoblastic-endothelial transition and reduced vascular calcification. Overall, inhibition of GSK3ß promoted the transition of cells with osteoblastic characteristics to endothelial differentiation, thereby ameliorating vascular calcification.


Subject(s)
Cell Differentiation/drug effects , Osteogenesis/drug effects , Vascular Calcification/metabolism , Animals , Cell Line , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Mice , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology
17.
Bioorg Chem ; 112: 104839, 2021 07.
Article in English | MEDLINE | ID: mdl-33813310

ABSTRACT

Plasmodium falciparum glycogen synthase kinase-3 (PfGSK-3) has been identified as a potential target for the development of novel drugs against multi-drug resistant malaria. A series of benzofuran-based compounds was synthesised and evaluated as inhibitors of recombinantly expressed and purified PfGSK-3 and human glycogen synthase kinase-3 beta (HsGSK-3ß). Of this series, five compounds (5k, 5m, 5p, 5r, 5s) preferentially inhibited PfGSK-3, with four of these compounds exhibiting IC50 values in the sub-micromolar range (0.00048-0.440 µM). Evaluation of the structure-activity relationships required for PfGSK-3 selective inhibition indicated that a C6-OCH3 substitution on ring A is preferred, while the effect of the ring B substituent on activity, in decreasing order is: C4'-CN > C4'-F > C3'-OCH3 > C3',4'-diCl. To date, development of PfGSK-3 inhibitors has been limited to the 4-phenylthieno[2,3-b]pyridine class. Chalcone-based scaffolds, such as the benzofurans described herein, are promising new hits which can be explored for future design of PfGSK-3 selective inhibitors.


Subject(s)
Benzofurans/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Plasmodium falciparum/enzymology , Protein Kinases/pharmacology , Benzofurans/chemical synthesis , Benzofurans/chemistry , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Humans , Molecular Structure , Protein Kinases/chemical synthesis , Protein Kinases/chemistry , Structure-Activity Relationship
18.
Nat Commun ; 12(1): 1863, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767186

ABSTRACT

Embryonic stem cells (ESCs) can be maintained in the naïve state through inhibition of Mek1/2 and Gsk3 (2i). A relevant effect of 2i is the inhibition of Cdk8/19, which are negative regulators of the Mediator complex, responsible for the activity of enhancers. Inhibition of Cdk8/19 (Cdk8/19i) stimulates enhancers and, similar to 2i, stabilizes ESCs in the naïve state. Here, we use mass spectrometry to describe the molecular events (phosphoproteome, proteome, and metabolome) triggered by 2i and Cdk8/19i on ESCs. Our data reveal widespread commonalities between these two treatments, suggesting overlapping processes. We find that post-transcriptional de-repression by both 2i and Cdk8/19i might support the mitochondrial capacity of naive cells. However, proteome reprogramming in each treatment is achieved by different mechanisms. Cdk8/19i acts directly on the transcriptional machinery, activating key identity genes to promote the naïve program. In contrast, 2i stabilizes the naïve circuitry through, in part, de-phosphorylation of downstream transcriptional effectors.


Subject(s)
Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Glycogen Synthase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Benzamides/pharmacology , Cell Line , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Phosphorylation/physiology , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
19.
Neurobiol Dis ; 154: 105336, 2021 07.
Article in English | MEDLINE | ID: mdl-33753290

ABSTRACT

In Huntington's disease (HD), the mutant huntingtin (mHtt) accumulates as toxic aggregates in the striatum tissue, with deleterious effects on motor-coordination and cognitive functions. Reducing the levels of mHtt is therefore a promising therapeutic strategy. We have previously reported that GSK-3 is a negative regulator of the autophagy/lysosome pathway, which is responsible for intracellular degradation, and is critically important for maintaining neuronal vitality. Thus, we hypothesized that inhibition of GSK-3 may trigger mHtt clearance thereby reducing mHtt cytotoxicity and improving HD symptoms. Here, we demonstrate that depletion or suppression of autophagy results in a massive accumulation of mHtt aggregates. Accordingly, mHtt aggregates were localized in lysosomes, but, mostly mislocalized from lysosomes in the absence of functional autophagy. Overexpression of GSK-3, particularly the α isozyme, increased the number of mHtt aggregates, while silencing GSK-3α/ß, or treatment with a selective GSK-3 inhibitor, L807mts, previously described by us, reduced the amounts of mHtt aggregates. This effect was mediated by increased autophagic and lysosomal activity. Treating R6/2 mouse model of HD with L807mts, reduced striatal mHtt aggregates and elevated autophagic and lysosomal markers. The L807mts treatment also reduced hyperglycemia and improved motor-coordination functions in these mice. In addition, L807mts restored the expression levels of Sirt1, a critical neuroprotective factor in the HD striatum, along with its targets BDNF, DRPP-32, and active Akt, all provide neurotrophic/pro-survival support and typically decline in the HD brain. Our results provide strong evidence for a role for GSK-3 in the regulation of mHtt dynamics, and demonstrate the benefits of GSK-3 inhibition in reducing mHtt toxicity, providing neuroprotective support, and improving HD symptoms.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , Animals , Cell Line, Tumor , Glycogen Synthase Kinase 3/genetics , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Lysosomes/metabolism , Male , Mice , Mice, Inbred CBA , Mice, Transgenic
20.
Cell Rep ; 34(10): 108834, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691117

ABSTRACT

At implantation, the embryo establishes contacts with the maternal endometrium. This stage is associated with a high incidence of preclinical pregnancy losses. While the maternal factors underlying uterine receptivity have been investigated, the signals required by the embryo for successful peri-implantation development remain elusive. To explore these, we studied integrin ß1 signaling, as embryos deficient for this receptor degenerate at implantation. We demonstrate that the coordinated action of pro-survival signals and localized actomyosin suppression via integrin ß1 permits the development of the embryo beyond implantation. Failure of either process leads to developmental arrest and apoptosis. Pharmacological stimulation through fibroblast growth factor 2 (FGF2) and insulin-like growth factor 1 (IGF1), coupled with ROCK-mediated actomyosin inhibition, rescues the deficiency of integrin ß1, promoting progression to post-implantation stages. Mutual exclusion between integrin ß1 and actomyosin seems to be conserved in the human embryo, suggesting the possibility that these mechanisms could also underlie the transition of the human epiblast from pre- to post-implantation.


Subject(s)
Integrin beta1/metabolism , Morphogenesis , Actomyosin/metabolism , Amides/pharmacology , Animals , Embryo Implantation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development , Female , Fibroblast Growth Factor 2/pharmacology , Germ Layers/growth & development , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Insulin-Like Growth Factor I/pharmacology , Integrin beta1/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Morphogenesis/drug effects , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pyridines/pharmacology , Signal Transduction , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...