1
|
Emmerich TD, Taylor-Chilton EJ, Caballero E, Hushcha I, Dickens K, Stasik I, Alder J, Saavedra-Castano S, Berenschot E, Tas NR, Susarrey-Arce A, Martinez-Gonzalez L, Oknianska A, Zwain T, Martinez A, Hayes JM. Structure-Based Discovery Targeting GSK-3α Reveals Potent Nanomolar Selective 4-Phenyl-1 H-benzofuro[3,2- b]pyrazolo[4,3- e]pyridine Inhibitor with Promising Glioblastoma and CNS-Active Potential in Cellular Models. J Med Chem 2025; 68:8679-8693. [PMID: 40198746 DOI: 10.1021/acs.jmedchem.5c00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Glycogen synthase kinase-3 (GSK-3) is linked with multiple CNS conditions, including glioblastoma (GBM). Compared to the GSK-3β isoform, structure-based inhibitor design targeting GSK-3α is limited. Virtual screening was employed to identify GSK-3α inhibitors with CNS-active potential. Using a GSK-3α homology model, an optimized protocol with three-dimensional (3D)-pharmacophore filtering and Glide-SP docking was used to screen the ZINC20 biogenic subset. From 14 compounds selected for binding assay validation, three novel hit compounds were identified, with 1 (4-phenyl-1H-benzofuro[3,2-b]pyrazolo[4,3-e]pyridine scaffold) exhibiting nanomolar activity against GSK-3α/β (IC50s ∼ 0.26 μM). Selectivity profiling (12 homologous kinases) revealed selectivity for GSK-3α/β and protein kinase A (PKA). Compound 1 was more potent against three GBM cell lines (cell viability IC50s = 3-6 μM at 72 h) compared to benchmark GSK-3 inhibitor, 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), and nontoxic to human astrocytes. It demonstrated CNS-active potential in an all-human in vitro blood-brain barrier GBM model, good in vitro metabolic stability, excellent predicted oral bioavailability and represents a promising lead compound for development.
Collapse
Affiliation(s)
- Thomas D Emmerich
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Eleanor J Taylor-Chilton
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Elena Caballero
- Centro de Investigaciones Biologicas, CSIC, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | - Iryna Hushcha
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Kathryn Dickens
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Izabela Stasik
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
- Biomedical Evidence-Based Transdisciplinary (BEST) Health Research Institute, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Jane Alder
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
- Biomedical Evidence-Based Transdisciplinary (BEST) Health Research Institute, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Santiago Saavedra-Castano
- Department of Chemical Engineering, MESA+ Institute and TechMed Centre, University of Twente, P.O. Box 217, Enschede 7500AE, The Netherlands
| | - Erwin Berenschot
- Department of Chemical Engineering, MESA+ Institute and TechMed Centre, University of Twente, P.O. Box 217, Enschede 7500AE, The Netherlands
| | - Niels R Tas
- Department of Chemical Engineering, MESA+ Institute and TechMed Centre, University of Twente, P.O. Box 217, Enschede 7500AE, The Netherlands
| | - Arturo Susarrey-Arce
- Department of Chemical Engineering, MESA+ Institute and TechMed Centre, University of Twente, P.O. Box 217, Enschede 7500AE, The Netherlands
| | - Loreto Martinez-Gonzalez
- Centro de Investigaciones Biologicas, CSIC, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | - Alina Oknianska
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
- Biomedical Evidence-Based Transdisciplinary (BEST) Health Research Institute, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Tamara Zwain
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
- Biomedical Evidence-Based Transdisciplinary (BEST) Health Research Institute, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Ana Martinez
- Centro de Investigaciones Biologicas, CSIC, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | - Joseph M Hayes
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
- Biomedical Evidence-Based Transdisciplinary (BEST) Health Research Institute, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| |
Collapse
|
2
|
Xiao H, Hou J, Wu Y, Zhao Y, Tu D, Ma X, Qiu R, Li L. The receptor-like cytoplasmic kinase OsSTRK1 regulates brassinosteroid signaling by phosphorylating OsGSK2. Cell Rep 2025; 44:115569. [PMID: 40220291 DOI: 10.1016/j.celrep.2025.115569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/16/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
In the BR signaling pathway, GSK3-like kinases are crucial negative regulators, and inactivation of GSK3-like kinases occurs through dephosphorylation by the phosphatase BSU1. However, the identity of the kinases that can phosphorylate GSK3-like kinases in BR signaling remains unknown. In this study, we identify that OsSTRK1 interacts with and phosphorylates OsGSK2 in rice to stabilize OsGSK2, preventing its interaction with the E3 ubiquitin ligase OsTUD1. Overexpression of OsSTRK1 leads to a BR-repressed phenotype and decreased sensitivity to BR, while RNAi of OsSTRK1 induces enhanced sensitivity to BR. We identify that Tyr-223 of OsGSK2 as a critical phosphorylation site that is essential for the role of OsSTRK1 in modulating BR signaling, thus influencing the growth and development of rice. Overall, our findings uncover the essential role of OsSTRK1 in the phosphorylation and activation of OsGSK2, thereby completing the assembly of the core components of the BR signaling pathway.
Collapse
Affiliation(s)
- Huangzhuo Xiao
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jiaqi Hou
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yequn Wu
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yating Zhao
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Daoyi Tu
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiaoci Ma
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ronghua Qiu
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lijia Li
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
3
|
Zhang F, Tang Y, Zhou H, Li K, West JA, Griffin JL, Lilley KS, Zhang N. The Yeast Gsk-3 Kinase Mck1 Is Necessary for Cell Wall Remodeling in Glucose-Starved and Cell Wall-Stressed Cells. Int J Mol Sci 2025; 26:3534. [PMID: 40332024 PMCID: PMC12027387 DOI: 10.3390/ijms26083534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
The cell wall integrity (CWI) pathway is responsible for transcriptional regulation of cell wall remodeling in response to cell wall stress. How cell wall remodeling mediated by the CWI pathway is effected by inputs from other signaling pathways is not well understood. Here, we demonstrate that the Mck1 kinase cooperates with Slt2, the MAP kinase of the CWI pathway, to promote cell wall thickening in glucose-starved cells. Integrative analyses of the transcriptome, proteome and metabolic profiling indicate that Mck1 is required for the accumulation of UDP-glucose (UDPG), the substrate for β-glucan synthesis, through the activation of two regulons: the Msn2/4-dependent stress response and the Cat8-/Adr1-mediated metabolic reprogram dependent on the SNF1 complex. Analysis of the phosphoproteome suggests that similar to mammalian Gsk-3 kinases, Mck1 is involved in the regulation of cytoskeleton-dependent cellular processes, metabolism, signaling and transcription. Specifically, Mck1 may be implicated in the Snf1-dependent metabolic reprogram through PKA inhibition and SAGA (Spt-Ada-Gcn5 acetyltransferase)-mediated transcription activation, a hypothesis further underscored by the significant overlap between the Mck1- and Gcn5-activated transcriptomes. Phenotypic analysis also supports the roles of Mck1 in actin cytoskeleton-mediated exocytosis to ensure plasma membrane homeostasis and cell wall remodeling in cell wall-stressed cells. Together, these findings not only reveal the novel functions of Mck1 in metabolic reprogramming and polarized growth but also provide valuable omics resources for future studies to uncover the underlying mechanisms of Mck1 and other Gsk-3 kinases in cell growth and stress response.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (F.Z.); (K.L.)
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yingzhi Tang
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (F.Z.); (K.L.)
| | - Houjiang Zhou
- MRC Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Kaiqiang Li
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (F.Z.); (K.L.)
| | - James A. West
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (F.Z.); (K.L.)
| | - Julian L. Griffin
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (F.Z.); (K.L.)
| | - Kathryn S. Lilley
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (F.Z.); (K.L.)
| | - Nianshu Zhang
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (F.Z.); (K.L.)
| |
Collapse
|
4
|
de la Serna-Soto M, Calleros L, Martos-Elvira M, Moreno-Piedra A, García-Villoria S, Griera M, Alcalde-Estévez E, Asenjo-Bueno A, Rodríguez-Puyol D, de Frutos S, Ruiz-Torres MP. Integrin-Linked Kinase (ILK) Promotes Mitochondrial Dysfunction by Decreasing CPT1A Expression in a Folic Acid-Based Model of Kidney Disease. Int J Mol Sci 2025; 26:1861. [PMID: 40076489 PMCID: PMC11899702 DOI: 10.3390/ijms26051861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Integrin-linked kinase (ILK) is a key scaffolding protein between extracellular matrix protein and the cytoskeleton and has been implicated previously in the pathogenesis of renal damage. However, its involvement in renal mitochondrial dysfunction remains to be elucidated. We studied the role of ILK and its downstream regulations in renal damage and mitochondria function both in vivo and vitro, using a folic acid (FA)-induced kidney disease model. Wild type (WT) and ILK conditional-knockdown (cKD-ILK) mice were injected with a single intraperitoneal dose of FA and studied after 15 days of chronic renal damage progression. Human Kidney tubular epithelial cells (HK2) were transfected with specific siRNAs targeting ILK, glycogen synthase kinase 3-β (GSK3β), or CCAAT/enhancer binding protein-β (C/EBPβ). The expressions and activities of renal ILK, GSK3β, C/EBPβ, mitochondrial oxidative phosphorylation enzymes, and mitochondrial membrane potential were assessed. Additionally, the expression of markers for fibrosis fibronectin (FN) and collagen 1 (COL1A1), for autophagy p62 and cytosolic light chain 3 (LC3B) isoforms II and I, and mitochondrial homeostasis marker carnitine palmitoyl-transferase 1A (CPT1A) were evaluated using immunoblotting, RT-qPCR, immunofluorescence, or colorimetric assays. FA upregulated ILK expression, leading to the decrease of GSK3β activity, increased tubular fibrosis, and produced mitochondrial dysfunction, both in vivo and vitro. These alterations were fully or partially reversed upon ILK depletion, mitigating FA-induced renal damage. The signaling axis composed by ILK, GSK3β, and C/EBPβ regulated CPT1A transcription as the limiting factor in the FA-based impaired mitochondrial activity. We highlight ILK as a potential therapeutical target for preserving mitochondrial function in kidney injury.
Collapse
Affiliation(s)
- Mariano de la Serna-Soto
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Laura Calleros
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - María Martos-Elvira
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Ariadna Moreno-Piedra
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Sergio García-Villoria
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Mercedes Griera
- Graphenano Medical Care S.L., Alcalá de Henares, 28871 Madrid, Spain;
| | - Elena Alcalde-Estévez
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Ana Asenjo-Bueno
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Diego Rodríguez-Puyol
- Department of Medicine, Universidad de Alcalá, Nephrology Service at Hospital Príncipe de Asturias, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain;
| | - Sergio de Frutos
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - María Piedad Ruiz-Torres
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| |
Collapse
|
5
|
Arioka M, Yi W, Igawa K, Ishikane S, Takahashi-Yanaga F. Differentiation-inducing factor-1 inhibits EMT by proteasomal degradation of TAZ and YAP in cervical and colon cancer cell lines. Eur J Pharmacol 2025; 987:177184. [PMID: 39645220 DOI: 10.1016/j.ejphar.2024.177184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/09/2024]
Abstract
We previously reported differentiation-inducing factor-1 (DIF-1) activated glycogen synthase kinase-3 (GSK-3) in various mammalian cells. GSK-3 has been proposed to regulate a number of signaling pathway including TAZ/YAP signaling pathway. To clarify the effect of DIF-1 on TAZ/YAP signaling pathway, we examined whether DIF-1 affect the expression levels of TAZ and YAP. We found that DIF-1-induced proteasomal- and GSK-3-dependent degradation of both TAZ and YAP in human cervical cancer cell line HeLa in a time- and dose-dependent manner. As TAZ/YAP signaling pathway is well known to accelerate the epithelial-mesenchymal transition (EMT) of the cancer cell, we examined the effect of TAZ/YAP signaling pathway on EMT-related proteins. Knockdown of TAZ and YAP proteins by siRNA significantly reduced the expression of fibronectin, vimentin, and Snail. We also found that DIF-1 suppressed the expression levels of TAZ/YAP target gene products and EMT-related protein. Further, overexpression of TAZ and YAP attenuated the inhibitory effects of DIF-1 on these protein expressions. Migration and trans-well invasion assays revealed that DIF-1 significantly inhibited HeLa cell migration and invasion. DIF-1-induced proteasomal- and GSK-3-dependent degradation of TAZ and YAP proteins and inhibition of cell migration and invasion were also observed in human colon cancer cell line HCT-116. These results suggest that DIF-1 inhibits the TAZ/YAP signaling pathway via GSK-3 activation. Further, it has been suggested that the inhibition of EMT induced by DIF-1 is involved with the suppression of TAZ/YAP signaling pathway.
Collapse
Affiliation(s)
- Masaki Arioka
- Department of Pharmacology, Faculty of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Wang Yi
- Department of Pharmacology, Faculty of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Kazunobu Igawa
- Department of Chemistry, Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Shin Ishikane
- Department of Pharmacology, Faculty of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Fumi Takahashi-Yanaga
- Department of Pharmacology, Faculty of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.
| |
Collapse
|
6
|
Jones T, Raman R, Puhl AC, Lane TR, Riabova O, Kazakova E, Makarov V, Ekins S. Discovery of Dual Targeting GSK-3β/HIV-1 Reverse Transcriptase Inhibitors as Neuroprotective Antiviral Agents. ACS Chem Neurosci 2025; 16:77-84. [PMID: 39663760 DOI: 10.1021/acschemneuro.4c00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
Abstract
Glycogen synthase kinase-3 beta (GSK-3β or GSK-3B) is a serine-threonine kinase involved in various pathways and cellular processes. Alteration in GSK-3β activity is associated with several neurological diseases including Alzheimer's disease (AD), bipolar disorder, and rare diseases like Rett syndrome. GSK-3β is also implicated in HIV-associated dementia (HAD), as it is upregulated in HIV-1-infected cells and plays a role in neuronal dysfunction. Therefore, a small molecule that can inhibit both GSK-3β and HIV-1 reverse transcriptase could offer neuroprotective therapy for patients suffering from HIV-1. Despite this, there are no known GSK-3β inhibitors currently approved, thus prompting us to screen our panel of various antiviral compounds against this kinase to better understand its structure-activity relationship. We show for the first time that the approved drugs, etravirine and rilpivirine, possess GSK-3β activity (IC50 619 nM and 502 nM, respectively). We have also identified 3 lead molecules exhibiting IC50 < 1 μM (11726169, 12326205, and 12326207), with compound 11726169 being the most potent in vitro GSK-3β inhibitor (IC50 = 12.1 nM). We also describe the generation of machine learning models for GSK-3β inhibition and their validation with our data as an external test set and propose their use for the future optimization of such inhibitors.
Collapse
Affiliation(s)
- Thane Jones
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Renuka Raman
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Olga Riabova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, Moscow 119071, Russia
| | - Elena Kazakova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, Moscow 119071, Russia
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, Moscow 119071, Russia
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
7
|
Ishikawa C, Mori N. A New Strategy for Adult T-Cell Leukemia Treatment Targeting Glycogen Synthase Kinase-3β. Eur J Haematol 2024; 113:852-862. [PMID: 39239903 DOI: 10.1111/ejh.14300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES The role of glycogen synthase kinase (GSK)-3β in adult T-cell leukemia (ATL) caused by human T-cell leukemia virus type 1 (HTLV-1) is paradoxical and enigmatic. Here, we investigated the role of GSK-3β and its potential as a therapeutic target for ATL. METHODS Cell proliferation/survival, cell cycle, apoptosis, and reactive oxygen species (ROS) generation were examined using the WST-8 assay, flow cytometry, and Hoechst 33342 staining, respectively. Expression of GSK-3β and cell cycle/death-related proteins, and survival signals was analyzed using RT-PCR, immunofluorescence staining, and immunoblotting. RESULTS HTLV-1-infected T-cell lines showed nuclear accumulation of GSK-3β. GSK-3β knockdown and its inhibition with 9-ING-41 and LY2090314 suppressed cell proliferation/survival. 9-ING-41 induced G2/M arrest by enhancing the expression of γH2AX, p53, p21, and p27, and suppressing the expression of CDK1, cyclin A/B, and c-Myc. It induced caspase-mediated apoptosis by decreasing the expression of Bcl-xL, Mcl-1, XIAP, c-IAP1/2, and survivin, and increasing the expression of Bak and Bax. 9-ING-41 also induced ferroptosis and necroptosis, promoted JNK phosphorylation, and suppressed IKKγ and JunB expression. It inhibited the phosphorylation of IκBα, Akt, and STAT3/5, induced ROS production, and reduced glycolysis-derived lactate levels. CONCLUSION GSK-3β functions as an oncogene in ATL and could be a potential therapeutic target.
Collapse
Affiliation(s)
- Chie Ishikawa
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Health Sciences, Transdisciplinary Research Organization for Subtropics and Island Studies, University of the Ryukyus, Nishihara, Japan
| | - Naoki Mori
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| |
Collapse
|
8
|
Rejnowicz E, Batchelor M, Leen E, Ahangar MS, Burgess SG, Richards MW, Kalverda AP, Bayliss R. Exploring the dynamics and interactions of the N-myc transactivation domain through solution nuclear magnetic resonance spectroscopy. Biochem J 2024; 481:1535-1556. [PMID: 39370942 PMCID: PMC11555651 DOI: 10.1042/bcj20240248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Myc proteins are transcription factors crucial for cell proliferation. They have a C-terminal domain that mediates Max and DNA binding, and an N-terminal disordered region culminating in the transactivation domain (TAD). The TAD participates in many protein-protein interactions, notably with kinases that promote stability (Aurora-A) or degradation (ERK1, GSK3) via the ubiquitin-proteasome system. We probed the structure, dynamics and interactions of N-myc TAD using nuclear magnetic resonance (NMR) spectroscopy following its complete backbone assignment. Chemical shift analysis revealed that N-myc has two regions with clear helical propensity: Trp77-Glu86 and Ala122-Glu132. These regions also have more restricted ps-ns motions than the rest of the TAD, and, along with the phosphodegron, have comparatively high transverse (R2) 15N relaxation rates, indicative of slower timescale dynamics and/or chemical exchange. Collectively these features suggest differential propensities for structure and interaction, either internal or with binding partners, across the TAD. Solution studies on the interaction between N-myc and Aurora-A revealed a previously uncharacterised binding site. The specificity and kinetics of sequential phosphorylation of N-myc by ERK1 and GSK3 were characterised using NMR and resulted in no significant structural changes outside the phosphodegron. When the phosphodegron was doubly phosphorylated, N-myc formed a robust interaction with the Fbxw7-Skp1 complex, but mapping the interaction by NMR suggests a more extensive interface. Our study provides foundational insights into N-myc TAD dynamics and a backbone assignment that will underpin future work on the structure, dynamics, interactions and regulatory post-translational modifications of this key oncoprotein.
Collapse
Affiliation(s)
- Ewa Rejnowicz
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Matthew Batchelor
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Eoin Leen
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Mohd Syed Ahangar
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Selena G. Burgess
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Mark W. Richards
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Arnout P. Kalverda
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Richard Bayliss
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| |
Collapse
|
9
|
Coats JT, Li S, Tanaka TU, Tauro S, Sutherland C, Saurin AT. Elraglusib Induces Cytotoxicity via Direct Microtubule Destabilization Independently of GSK3 Inhibition. CANCER RESEARCH COMMUNICATIONS 2024; 4:3013-3024. [PMID: 39470360 PMCID: PMC11586712 DOI: 10.1158/2767-9764.crc-24-0408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/01/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
SIGNIFICANCE Elraglusib was designed as a GSK3 inhibitor and is currently in clinical trials for several cancers. We show conclusively that the target of elraglusib that leads to cytotoxicity is MTs and not GSK3. This has significant implications for ongoing clinical trials of the compound and will help in understanding off-target side effects, inform future clinical trial design, and facilitate the development of biomarkers to predict response.
Collapse
Affiliation(s)
- Josh T. Coats
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Shuyu Li
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Tomoyuki U. Tanaka
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sudhir Tauro
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Calum Sutherland
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Adrian T. Saurin
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
10
|
Zhang Y, Zhong C, Shu X, Liu Q, Jiang Y. Estrogen Enhances FDFT1 Expression in Theca Cells of Chicken Hierarchical Ovarian Follicles by Increasing LSD1Ser54p Level Through GSK3β Phosphorylation at 216th Tyrosine. Biomolecules 2024; 14:1343. [PMID: 39595520 PMCID: PMC11591973 DOI: 10.3390/biom14111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024] Open
Abstract
The development of chicken ovarian follicles involves two key stages of primordial follicle recruitment and follicle selection that are tightly regulated by multiple reproductive hormones and cytokines. Our previous study revealed an estrogen-stimulated increase in the phosphorylation level of serine at position 54 of lysine demethylase 1A (LSD1Ser54p) in the theca cells of chicken hierarchical ovarian follicles (Post-TCs). In this study, we further found that the upregulation of LSD1Ser54p by estrogen was performed by glycogen synthase kinase 3 beta (GSK3β) and that GSK3β promoted LSD1Ser54p levels by directly binding to the SWIRM and AOL1 domains of LSD1. Upon estrogen stimulation, the phosphorylation level of tyrosine at position 216 of GSK3β (GSK3βTyr216p) increased, which enhanced the binding between LSD1 and GSK3β. The subsequent transcriptome sequencing on chicken Post-TCs treated with estrogen and CUT&RUN sequencing against the LSD1Ser54p protein revealed that the expression of the farnesyl-diphosphate farnesyltransferase 1 (FDFT1) gene was simultaneously upregulated by estrogen, GSK3β, and LSD1Ser54p. Moreover, the overexpression of FDFT1 further promoted cholesterol biosynthesis in chicken Post-TCs. In short, the findings of this study suggest that estrogen-induced tyrosine phosphorylation at position 216 of GSK3β can upregulate the level of LSD1Ser54p, leading to the activation of FDFT1 expression and subsequently promoting cholesterol biosynthesis in chicken Post-TCs, which may in turn enhance estrogen synthesis.
Collapse
Affiliation(s)
- Yanhong Zhang
- College of Life Science, Shandong Agricultural University, Tai’an 271018, China;
| | - Conghao Zhong
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Xinmei Shu
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an 271017, China;
| | - Qingxin Liu
- College of Life Science, Shandong Agricultural University, Tai’an 271018, China;
| | - Yunliang Jiang
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an 271017, China;
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Tai’an 271017, China
| |
Collapse
|
11
|
Enos MD, Gavagan M, Jameson N, Zalatan JG, Weis WI. Structural and functional effects of phosphopriming and scaffolding in the kinase GSK-3β. Sci Signal 2024; 17:eado0881. [PMID: 39226374 PMCID: PMC11461088 DOI: 10.1126/scisignal.ado0881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Glycogen synthase kinase 3β (GSK-3β) targets specific signaling pathways in response to distinct upstream signals. We used structural and functional studies to dissect how an upstream phosphorylation step primes the Wnt signaling component β-catenin for phosphorylation by GSK-3β and how scaffolding interactions contribute to this reaction. Our crystal structure of GSK-3β bound to a phosphoprimed β-catenin peptide confirmed the expected binding mode of the phosphoprimed residue adjacent to the catalytic site. An aspartate phosphomimic in the priming site of β-catenin adopted an indistinguishable structure but reacted approximately 1000-fold slower than the native phosphoprimed substrate. This result suggests that substrate positioning alone is not sufficient for catalysis and that native phosphopriming interactions are necessary. We also obtained a structure of GSK-3β with an extended peptide from the scaffold protein Axin that bound with greater affinity than that of previously crystallized Axin fragments. This structure neither revealed additional contacts that produce the higher affinity nor explained how substrate interactions in the GSK-3β active site are modulated by remote Axin binding. Together, our findings suggest that phosphopriming and scaffolding produce small conformational changes or allosteric effects, not captured in the crystal structures, that activate GSK-3β and facilitate β-catenin phosphorylation. These results highlight limitations in our ability to predict catalytic activity from structure and have potential implications for the role of natural phosphomimic mutations in kinase regulation and phosphosite evolution.
Collapse
Affiliation(s)
- Michael D. Enos
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94035, USA
- Department Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Maire Gavagan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Noel Jameson
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Jesse G. Zalatan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - William I. Weis
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94035, USA
- Department Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| |
Collapse
|
12
|
Alexander JM, Vazquez-Ramirez L, Lin C, Antonoudiou P, Maguire J, Wagner F, Jacob MH. Inhibition of GSK3α,β rescues cognitive phenotypes in a preclinical mouse model of CTNNB1 syndrome. EMBO Mol Med 2024; 16:2109-2131. [PMID: 39103699 PMCID: PMC11393422 DOI: 10.1038/s44321-024-00110-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024] Open
Abstract
CTNNB1 syndrome is a rare monogenetic disorder caused by CTNNB1 de novo pathogenic heterozygous loss-of-function variants that result in cognitive and motor disabilities. Treatment is currently lacking; our study addresses this critical need. CTNNB1 encodes β-catenin which is essential for normal brain function via its dual roles in cadherin-based synaptic adhesion complexes and canonical Wnt signal transduction. We have generated a Ctnnb1 germline heterozygous mouse line that displays cognitive and motor deficits, resembling key features of CTNNB1 syndrome in humans. Compared with wild-type littermates, Ctnnb1 heterozygous mice also exhibit decreases in brain β-catenin, β-catenin association with N-cadherin, Wnt target gene expression, and Na/K ATPases, key regulators of changes in ion gradients during high activity. Consistently, hippocampal neuron functional properties and excitability are altered. Most important, we identify a highly selective inhibitor of glycogen synthase kinase (GSK)3α,β that significantly normalizes the phenotypes to closely meet wild-type littermate levels. Our data provide new insights into brain molecular and functional changes, and the first evidence for an efficacious treatment with therapeutic potential for individuals with CTNNB1 syndrome.
Collapse
Affiliation(s)
- Jonathan M Alexander
- Tufts University School of Biomedical Sciences, Department of Neuroscience, Boston, MA, 02111, USA
| | - Leeanne Vazquez-Ramirez
- Tufts University School of Biomedical Sciences, Department of Neuroscience, Boston, MA, 02111, USA
| | - Crystal Lin
- Tufts University School of Biomedical Sciences, Department of Neuroscience, Boston, MA, 02111, USA
| | - Pantelis Antonoudiou
- Tufts University School of Biomedical Sciences, Department of Neuroscience, Boston, MA, 02111, USA
| | - Jamie Maguire
- Tufts University School of Biomedical Sciences, Department of Neuroscience, Boston, MA, 02111, USA
| | - Florence Wagner
- The Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA, 02142, USA
- Photys Therapeutics, Waltham, MA, USA
| | - Michele H Jacob
- Tufts University School of Biomedical Sciences, Department of Neuroscience, Boston, MA, 02111, USA.
| |
Collapse
|
13
|
Kühl F, Brand K, Lichtinghagen R, Huber R. GSK3-Driven Modulation of Inflammation and Tissue Integrity in the Animal Model. Int J Mol Sci 2024; 25:8263. [PMID: 39125833 PMCID: PMC11312333 DOI: 10.3390/ijms25158263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Nowadays, GSK3 is accepted as an enzyme strongly involved in the regulation of inflammation by balancing the pro- and anti-inflammatory responses of cells and organisms, thus influencing the initiation, progression, and resolution of inflammatory processes at multiple levels. Disturbances within its broad functional scope, either intrinsically or extrinsically induced, harbor the risk of profound disruptions to the regular course of the immune response, including the formation of severe inflammation-related diseases. Therefore, this review aims at summarizing and contextualizing the current knowledge derived from animal models to further shape our understanding of GSK3α and β and their roles in the inflammatory process and the occurrence of tissue/organ damage. Following a short recapitulation of structure, function, and regulation of GSK3, we will focus on the lessons learned from GSK3α/β knock-out and knock-in/overexpression models, both conventional and conditional, as well as a variety of (predominantly rodent) disease models reflecting defined pathologic conditions with a significant proportion of inflammation and inflammation-related tissue injury. In summary, the literature suggests that GSK3 acts as a crucial switch driving pro-inflammatory and destructive processes and thus contributes significantly to the pathogenesis of inflammation-associated diseases.
Collapse
Affiliation(s)
| | | | | | - René Huber
- Institute of Clinical Chemistry and Laboratory Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.K.); (K.B.); (R.L.)
| |
Collapse
|
14
|
El Hajjar L, Page A, Bridot C, Cantrelle FX, Landrieu I, Smet-Nocca C. Regulation of Glycogen Synthase Kinase-3β by Phosphorylation and O-β-Linked N-Acetylglucosaminylation: Implications on Tau Protein Phosphorylation. Biochemistry 2024; 63:1513-1533. [PMID: 38788673 DOI: 10.1021/acs.biochem.4c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Glycogen synthase kinase 3 (GSK3) plays a pivotal role in signaling pathways involved in insulin metabolism and the pathogenesis of neurodegenerative disorders. In particular, the GSK3β isoform is implicated in Alzheimer's disease (AD) as one of the key kinases involved in the hyperphosphorylation of tau protein, one of the neuropathological hallmarks of AD. As a constitutively active serine/threonine kinase, GSK3 is inactivated by Akt/PKB-mediated phosphorylation of Ser9 in the N-terminal disordered domain, and for most of its substrates, requires priming (prephosphorylation) by another kinase that targets the substrate to a phosphate-specific pocket near the active site. GSK3 has also been shown to be post-translationally modified by O-linked β-N-acetylglucosaminylation (O-GlcNAcylation), with still unknown functions. Here, we have found that binding of Akt inhibits GSK3β kinase activity on both primed and unprimed tau substrates. Akt-mediated Ser9 phosphorylation restores the GSK3β kinase activity only on primed tau, thereby selectively inactivating GSK3β toward unprimed tau protein. Additionally, we have shown that GSK3β is highly O-GlcNAcylated at multiple sites within the kinase domain and the disordered N- and C-terminal domains, including Ser9. In contrast to Akt-mediated regulation, neither the O-GlcNAc transferase nor O-GlcNAcylation significantly alters GSK3β kinase activity, but high O-GlcNAc levels reduce Ser9 phosphorylation by Akt. Reciprocally, Akt phosphorylation downregulates the overall O-GlcNAcylation of GSK3β, indicating a crosstalk between both post-translational modifications. Our results indicate that specific O-GlcNAc profiles may be involved in the phosphorylation-dependent Akt-mediated regulation of GSK3β kinase activity.
Collapse
Affiliation(s)
- Léa El Hajjar
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Adeline Page
- Protein Science Facility, SFR Biosciences Univ Lyon, ENS de Lyon, CNRS UAR3444, Inserm US8, Université Claude Bernard Lyon 1, 50 Avenue Tony Garnier, Lyon F-69007, France
| | - Clarisse Bridot
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - François-Xavier Cantrelle
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Isabelle Landrieu
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Caroline Smet-Nocca
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| |
Collapse
|
15
|
Patibandla C, van Aalten L, Dinkova-Kostova AT, Honda T, Cuadrado A, Fernández-Ginés R, McNeilly AD, Hayes JD, Cantley J, Sutherland C. Inhibition of glycogen synthase kinase-3 enhances NRF2 protein stability, nuclear localisation and target gene transcription in pancreatic beta cells. Redox Biol 2024; 71:103117. [PMID: 38479223 PMCID: PMC10950707 DOI: 10.1016/j.redox.2024.103117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/27/2024] [Accepted: 03/06/2024] [Indexed: 03/24/2024] Open
Abstract
Accumulation of reactive oxygen species (i.e., oxidative stress) is a leading cause of beta cell dysfunction and apoptosis in diabetes. NRF2 (NF-E2 p45-related factor-2) regulates the adaptation to oxidative stress, and its activity is negatively regulated by the redox-sensitive CUL3 (cullin-3) ubiquitin ligase substrate adaptor KEAP1 (Kelch-like ECH-associated protein-1). Additionally, NRF2 is repressed by the insulin-regulated Glycogen Synthase Kinase-3 (GSK3). We have demonstrated that phosphorylation of NRF2 by GSK3 enhances β-TrCP (beta-transducin repeat-containing protein) binding and ubiquitylation by CUL1 (cullin-1), resulting in increased proteasomal degradation of NRF2. Thus, we hypothesise that inhibition of GSK3 activity or β-TrCP binding upregulates NRF2 and so protects beta cells against oxidative stress. We have found that treating the pancreatic beta cell line INS-1 832/13 with the KEAP1 inhibitor TBE31 significantly enhanced NRF2 protein levels. The presence of the GSK3 inhibitor CT99021 or the β-TrCP-NRF2 protein-protein interaction inhibitor PHAR, along with TBE31, resulted in prolonged NRF2 stability and enhanced nuclear localisation (P < 0.05). TBE31-mediated induction of NRF2-target genes encoding NAD(P)H quinone oxidoreductase 1 (Nqo1), glutamate-cysteine ligase modifier (Gclm) subunit and heme oxygenase (Hmox1) was significantly enhanced by the presence of CT99021 or PHAR (P < 0.05) in both INS-1 832/13 and in isolated mouse islets. Identical results were obtained using structurally distinct GSK3 inhibitors and inhibition of KEAP1 with sulforaphane. In summary, we demonstrate that GSK3 and β-TrCP/CUL1 regulate the proteasomal degradation of NRF2, enhancing the impact of KEAP1 regulation, and so contributes to the redox status of pancreatic beta cells. Inhibition of GSK3, or β-TrCP/CUL1 binding to NRF2 may represent a strategy to protect beta cells from oxidative stress.
Collapse
Affiliation(s)
- Chinmai Patibandla
- Division of Cellular & Systems Medicine, James Arnott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, United Kingdom.
| | - Lidy van Aalten
- Division of Cellular & Systems Medicine, James Arnott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, United Kingdom
| | - Albena T Dinkova-Kostova
- Division of Cellular & Systems Medicine, James Arnott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, United Kingdom
| | - Tadashi Honda
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA; Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas Sols-Morreale UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Raquel Fernández-Ginés
- Instituto de Investigaciones Biomédicas Sols-Morreale UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Alison D McNeilly
- Division of Cellular & Systems Medicine, James Arnott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, United Kingdom
| | - John D Hayes
- Division of Cellular & Systems Medicine, James Arnott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, United Kingdom
| | - James Cantley
- Division of Cellular & Systems Medicine, James Arnott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, United Kingdom
| | - Calum Sutherland
- Division of Cellular & Systems Medicine, James Arnott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, United Kingdom
| |
Collapse
|
16
|
Ziak J, Dorskind JM, Trigg B, Sudarsanam S, Jin XO, Hand RA, Kolodkin AL. Microtubule-binding protein MAP1B regulates interstitial axon branching of cortical neurons via the tubulin tyrosination cycle. EMBO J 2024; 43:1214-1243. [PMID: 38388748 PMCID: PMC10987652 DOI: 10.1038/s44318-024-00050-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Regulation of directed axon guidance and branching during development is essential for the generation of neuronal networks. However, the molecular mechanisms that underlie interstitial (or collateral) axon branching in the mammalian brain remain unresolved. Here, we investigate interstitial axon branching in vivo using an approach for precise labeling of layer 2/3 callosal projection neurons (CPNs). This method allows for quantitative analysis of axonal morphology at high acuity and also manipulation of gene expression in well-defined temporal windows. We find that the GSK3β serine/threonine kinase promotes interstitial axon branching in layer 2/3 CPNs by releasing MAP1B-mediated inhibition of axon branching. Further, we find that the tubulin tyrosination cycle is a key downstream component of GSK3β/MAP1B signaling. These data suggest a cell-autonomous molecular regulation of cortical neuron axon morphology, in which GSK3β can release a MAP1B-mediated brake on interstitial axon branching upstream of the posttranslational tubulin code.
Collapse
Affiliation(s)
- Jakub Ziak
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Joelle M Dorskind
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
- Novartis Institutes for BioMedical Research, Boston, MA, USA
| | - Brian Trigg
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Sriram Sudarsanam
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Xinyu O Jin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Randal A Hand
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
- Prilenia Therapeutics, Boston, MA, USA
| | - Alex L Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA.
| |
Collapse
|
17
|
Zhan Y, Wen Y, Zheng F, Du LJ, Chen TY, Shen XL, Wu R, Tang XG. MiR-26b-3p Promotes Intestinal Motility Disorder by Targeting FZD10 to Inhibit GSK3β/β-Catenin Signaling and Induce Enteric Glial Cell Apoptosis. Mol Neurobiol 2024; 61:1543-1561. [PMID: 37728849 DOI: 10.1007/s12035-023-03600-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 08/20/2023] [Indexed: 09/21/2023]
Abstract
Enteric glial cells (EGCs) are the major component of the enteric nervous system and affect the pathophysiological process of intestinal motility dysfunction. MicroRNAs (miRNAs) play an important role in regulating gastrointestinal homeostasis. However, the mechanism of miRNA-mediated regulation of EGCs in intestinal dysmotility remains unclear. In this study, we investigated the effect of EGC apoptosis on intestinal dysmotility, and the effect of miR-26b-3p on EGC proliferation and apoptosis in vivo and in vitro. A loperamide hydrochloride (Lop)-induced constipated mouse model and an in vitro culture system of rat EGCs were established. The transcriptome was used to predict the differentially expressed gene miR-26b-3p and the target gene Frizzled 10 (FZD10), and their targeting binding relationship was verified by luciferase. EGCs were transfected with miR-26b-3p mimic or antagomir, and the FZD10 expression was down-regulated by siRNA. Immunofluorescence and flow cytometry were used to detect EGC apoptosis. MiR-26b-3p and FZD10 expressions were examined using quantitative real-time PCR (qRT-PCR). The CCK-8 assay was used to detect EGC proliferation. The protein levels were detected by Western blotting and enzyme-linked immunosorbent assay (ELISA). The results showed that miR-26b-3p was up-regulated in the Lop group, whereas FZD10 was down-regulated, and EGC apoptosis was increased in the colon of intestinal dysmotility mice. FZD10 down-regulation and miR-26b-3p mimic significantly increased glycogen synthase kinase-3β phosphorylation (p-GSK3β) levels, decreased β-catenin expression, and promoted EGC apoptosis. MiR-26b-3p antagomir alleviated intestinal dysmotility, promoted EGC increased activity of EGCs, and reduced EGC apoptosis in vivo. In conclusion, this study indicated that miR-26b-3p promotes intestinal motility disorders by targeting FZD10 to block GSK3β/β-catenin signaling and induces apoptosis in EGCs. Our results provide a new research target for the treatment and intervention of intestinal dysmotility.
Collapse
Affiliation(s)
- Yu Zhan
- Hospital of Chengdu University of TCM, Chengdu, China
- Anorectal Department, Affiliated Hospital of Integrative Chinese Medicine and Western Medicine of Chengdu University of TCM, Chengdu, China
- Chengdu First People's Hospital, Chengdu, China
| | - Yong Wen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fan Zheng
- Anorectal Department, People's Hospital of Deyang City, Deyang, China
- Universiti Sains Malaysia, George Town, Pulau Pinang, Malaysia
| | - Li-Juan Du
- The Third People's Hospital of Chengdu, Chengdu, China
- Southwest Jiaotong University College of Medicine, Chengdu, China
| | - Tai-Yu Chen
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xu-Long Shen
- Anorectal Department, Luzhou People's Hospital, Luzhou, China
| | - Rong Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xue-Gui Tang
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
18
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
19
|
Kaiser J, Nay K, Horne CR, McAloon LM, Fuller OK, Muller AG, Whyte DG, Means AR, Walder K, Berk M, Hannan AJ, Murphy JM, Febbraio MA, Gundlach AL, Scott JW. CaMKK2 as an emerging treatment target for bipolar disorder. Mol Psychiatry 2023; 28:4500-4511. [PMID: 37730845 PMCID: PMC10914626 DOI: 10.1038/s41380-023-02260-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/22/2023]
Abstract
Current pharmacological treatments for bipolar disorder are inadequate and based on serendipitously discovered drugs often with limited efficacy, burdensome side-effects, and unclear mechanisms of action. Advances in drug development for the treatment of bipolar disorder remain incremental and have come largely from repurposing drugs used for other psychiatric conditions, a strategy that has failed to find truly revolutionary therapies, as it does not target the mood instability that characterises the condition. The lack of therapeutic innovation in the bipolar disorder field is largely due to a poor understanding of the underlying disease mechanisms and the consequent absence of validated drug targets. A compelling new treatment target is the Ca2+-calmodulin dependent protein kinase kinase-2 (CaMKK2) enzyme. CaMKK2 is highly enriched in brain neurons and regulates energy metabolism and neuronal processes that underpin higher order functions such as long-term memory, mood, and other affective functions. Loss-of-function polymorphisms and a rare missense mutation in human CAMKK2 are associated with bipolar disorder, and genetic deletion of Camkk2 in mice causes bipolar-like behaviours similar to those in patients. Furthermore, these behaviours are ameliorated by lithium, which increases CaMKK2 activity. In this review, we discuss multiple convergent lines of evidence that support targeting of CaMKK2 as a new treatment strategy for bipolar disorder.
Collapse
Affiliation(s)
- Jacqueline Kaiser
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
- St Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
- School of Behavioural and Health Sciences, Australian Catholic University, Fitzroy, VIC, 3065, Australia
| | - Kevin Nay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Luke M McAloon
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
- St Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
- School of Behavioural and Health Sciences, Australian Catholic University, Fitzroy, VIC, 3065, Australia
| | - Oliver K Fuller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Abbey G Muller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Douglas G Whyte
- School of Behavioural and Health Sciences, Australian Catholic University, Fitzroy, VIC, 3065, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ken Walder
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, VIC, 3220, Australia
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, VIC, 3220, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, 3052, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Anthony J Hannan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - James M Murphy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Andrew L Gundlach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
- St Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia.
- St Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia.
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
20
|
Guo Z, Ma X, Chen X, Zhang RX, Yan H. Oxidative stress-induced temporal activation of ERK1/2 phosphorylates coreceptor of Wnt/β-catenin for myofibroblast formation in human lens epithelial cells. Mol Vis 2023; 29:206-216. [PMID: 38222447 PMCID: PMC10784218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 10/15/2023] [Indexed: 01/16/2024] Open
Abstract
Purpose Posterior capsular opacification (PCO) is the most common complication postcataract surgery, and its underlying mechanisms involve epithelial-mesenchymal transition (EMT) of remnant lens epithelial cells (LECs) in response to drastic changes in stimuli in the intraocular environment, such as oxidative stress and growth factors. Wnt/β-catenin signaling is a major pathway mediating oxidative stress-induced EMT in LECs, but its interplay with other transduction pathways remains little known in the development of PCO. ERK1/2 signaling is the downstream component of a phosphorelay pathway in response to extracellular stimuli (e.g., reactive oxygen species), and its activation regulates multiple cellular processes, including proliferation and EMT. Thus, this study aimed to investigate how ERK1/2 signaling and Wnt/β-catenin pathway crosstalk in oxidative stress-induced EMT in LECs. Methods Hydrogen peroxide (H2O2) at 50 μM treatment for 48 h was used to establish a moderate oxidative stress-induced EMT model in LECs. ERK1/2 signaling was inhibited using MEK1/2 inhibitor U0126 at 20 μM. Western blotting was used to quantify protein expression of various biomarkers of EMT and phosphorylated components in ERK1/2 and Wnt/β-catenin signaling. LEC proliferation was determined using an EdU staining assay and expression of proliferating cellular nuclear antigen (PCNA). Subcellular localization of biomarker proteins was visualized with immunofluorescent staining. Results Under the moderate level of H2O2-induced EMT in LECs, ERK1/2 signaling was activated, as evidenced by a marked increase in the ratio of phosphorylated ERK1/2 to total ERK1/2 at early (i.e., 5-15 min) and late time points (i.e., 12 h); the canonical Wnt/β-catenin pathway was activated by H2O2 at 48 h. LECs exposed to H2O2 exhibited hyperproliferation and EMT; however, these were restored by inhibition of ERK1/2 signaling demonstrated by reduced DNA synthesis and PCNA expression for cellular proliferation and altered expression of various EMT protein markers, including E-cadherin, α-SMA, and vimentin. More importantly, inhibition of ERK1/2 signaling reduced β-catenin accumulation in the activated Wnt/β-catenin signaling cascade. Specifically, there was significant downregulation in the phosphorylation level of LRP6 at Ser 1490 and GSK-3β at Ser 9, the key coreceptor of Wnt and regulator of β-catenin, respectively. Conclusions ERK1/2 signaling plays a crucial role in the moderate level of oxidative stress-induced EMT in LECs. Pharmacologically blocking ERK1/2 signaling significantly inhibited LEC proliferation and EMT. Mechanistically, ERK1/2 signaling regulated Wnt/β-catenin cascade by phosphorylating Wnt coreceptor LRP6 at Ser 1490 in the plasma membrane. These results shed light on a potential molecular switch of ERK1/2 and Wnt/β-catenin crosstalk underlying the development of PCO.
Collapse
Affiliation(s)
- Zaoxia Guo
- Shaanxi Eye Hospital, Xi'an People’s Hospital (Xi'an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi' an, Shaanxi, China
| | - Xiaopan Ma
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xi Chen
- Shaanxi Eye Hospital, Xi'an People’s Hospital (Xi'an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi' an, Shaanxi, China
| | - Rui Xue Zhang
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Hong Yan
- Shaanxi Eye Hospital, Xi'an People’s Hospital (Xi'an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi' an, Shaanxi, China
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| |
Collapse
|
21
|
Ziak J, Dorskind J, Trigg B, Sudarsanam S, Hand R, Kolodkin AL. MAP1B Regulates Cortical Neuron Interstitial Axon Branching Through the Tubulin Tyrosination Cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560024. [PMID: 37873083 PMCID: PMC10592918 DOI: 10.1101/2023.10.02.560024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Regulation of directed axon guidance and branching during development is essential for the generation of neuronal networks. However, the molecular mechanisms that underlie interstitial axon branching in the mammalian brain remain unresolved. Here, we investigate interstitial axon branching in vivo using an approach for precise labeling of layer 2/3 callosal projection neurons (CPNs), allowing for quantitative analysis of axonal morphology at high acuity and also manipulation of gene expression in well-defined temporal windows. We find that the GSK3β serine/threonine kinase promotes interstitial axon branching in layer 2/3 CPNs by releasing MAP1B-mediated inhibition of axon branching. Further, we find that the tubulin tyrosination cycle is a key downstream component of GSK3β/MAP1B signaling. We propose that MAP1B functions as a brake on axon branching that can be released by GSK3β activation, regulating the tubulin code and thereby playing an integral role in sculpting cortical neuron axon morphology.
Collapse
Affiliation(s)
- Jakub Ziak
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| | - Joelle Dorskind
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
- Novartis Institutes for BioMedical Research, Boston, MA
| | - Brian Trigg
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| | - Sriram Sudarsanam
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| | - Randal Hand
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
- Prilenia Therapeutics, Boston, MA
| | - Alex L. Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| |
Collapse
|
22
|
Gavagan M, Jameson N, Zalatan JG. The Axin scaffold protects the kinase GSK3β from cross-pathway inhibition. eLife 2023; 12:e85444. [PMID: 37548359 PMCID: PMC10442075 DOI: 10.7554/elife.85444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 08/04/2023] [Indexed: 08/08/2023] Open
Abstract
Multiple signaling pathways regulate the kinase GSK3β by inhibitory phosphorylation at Ser9, which then occupies the GSK3β priming pocket and blocks substrate binding. Since this mechanism should affect GSK3β activity toward all primed substrates, it is unclear why Ser9 phosphorylation does not affect other GSK3β-dependent pathways, such as Wnt signaling. We used biochemical reconstitution and cell culture assays to evaluate how Wnt-associated GSK3β is insulated from cross-activation by other signals. We found that the Wnt-specific scaffold protein Axin allosterically protects GSK3β from phosphorylation at Ser9 by upstream kinases, which prevents accumulation of pS9-GSK3β in the Axin•GSK3β complex. Scaffold proteins that protect bound proteins from alternative pathway reactions could provide a general mechanism to insulate signaling pathways from improper crosstalk.
Collapse
Affiliation(s)
- Maire Gavagan
- Department of Chemistry, University of WashingtonSeattleUnited States
| | - Noel Jameson
- Department of Chemistry, University of WashingtonSeattleUnited States
| | - Jesse G Zalatan
- Department of Chemistry, University of WashingtonSeattleUnited States
| |
Collapse
|
23
|
Livingstone JD, Gingras MJ, Leonenko Z, Beazely MA. Search for lithium isotope effects in neuronal HT22 cells. Biochem Biophys Rep 2023; 34:101461. [PMID: 37063814 PMCID: PMC10102007 DOI: 10.1016/j.bbrep.2023.101461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/11/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
Lithium has been used as a treatment for bipolar disorder for over half a century, but there has thus far been no clinical differentiation made between the two naturally occurring stable isotopes (6Li and 7Li). While the natural lithium salts commonly used in treatments are composed of a mixture of these two stable isotopes (approximately 7.59% 6Li and 92.41% 7Li), some preliminary research indicates the above two stable isotopes of lithium may have differential effects on rat behaviour and neurophysiology. Here, we evaluate whether lithium isotopes may have distinct effects on HT22 neuronal cell viability, GSK-3-β phosphorylation in HT22 cells, and GSK-3-β kinase activity. We report no significant difference in lithium isotope toxicity on HT22 cells, nor in GSK-3-β phosphorylation, nor in GSK-3-β kinase activity between the two isotopes of lithium.
Collapse
|
24
|
Coats JT, Tauro S, Sutherland C. Elraglusib (formerly 9-ING-41) possesses potent anti-lymphoma properties which cannot be attributed to GSK3 inhibition. Cell Commun Signal 2023; 21:131. [PMID: 37316860 DOI: 10.1186/s12964-023-01147-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
Elraglusib (formerly 9-ING-41) is an ATP-competitive inhibitor of glycogen synthase kinase-3β (GSK3β) undergoing clinical trials for the treatment of various cancers including non-Hodgkin lymphoma (NHL). The drug reduces proliferation of several NHL cell lines and has efficacy in xenograft models of the disease. To confirm the importance of its action on GSK3β, we treated 3 lymphoma cell lines with selective, structurally distinct GSK3 inhibitors: CT99021, SB216763, LY2090314, tideglusib, and elraglusib. Stabilization of β-catenin and reduced phosphorylation of CRMP2, two validated targets of GSK3, were used as functional read-outs for GSK3 inhibition. CT99021, SB216763, and LY2090314 failed to reduce proliferation or viability in any cell line at concentrations that stabilized β-catenin and reduced CRMP2 phosphorylation. There was partial reduction of CRMP2 phosphorylation but no significant effect on β-catenin at cytotoxic doses of elraglusib. There was no indication of GSK3 inhibition at doses of tideglusib that affected cell viability and apoptosis. Cell-free kinase screening confirmed several other targets of elraglusib, distinct from the GSK3 inhibitors with no anti-lymphoma actions, including PIM kinases and MST2. These data question GSK3 as the target of elraglusib in lymphoma, and hence the utility of GSK3 expression as a 'stand-alone', therapeutic biomarker in NHL. Video Abstract.
Collapse
Affiliation(s)
- Josh T Coats
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Sudhir Tauro
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Calum Sutherland
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.
| |
Collapse
|
25
|
Basheer N, Smolek T, Hassan I, Liu F, Iqbal K, Zilka N, Novak P. Does modulation of tau hyperphosphorylation represent a reasonable therapeutic strategy for Alzheimer's disease? From preclinical studies to the clinical trials. Mol Psychiatry 2023; 28:2197-2214. [PMID: 37264120 PMCID: PMC10611587 DOI: 10.1038/s41380-023-02113-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/28/2023] [Accepted: 05/05/2023] [Indexed: 06/03/2023]
Abstract
Protein kinases (PKs) have emerged as one of the most intensively investigated drug targets in current pharmacological research, with indications ranging from oncology to neurodegeneration. Tau protein hyperphosphorylation was the first pathological post-translational modification of tau protein described in Alzheimer's disease (AD), highlighting the role of PKs in neurodegeneration. The therapeutic potential of protein kinase inhibitors (PKIs)) and protein phosphatase 2 A (PP2A) activators in AD has recently been explored in several preclinical and clinical studies with variable outcomes. Where a number of preclinical studies demonstrate a visible reduction in the levels of phospho-tau in transgenic tauopathy models, no reduction in neurofibrillary lesions is observed. Amongst the few PKIs and PP2A activators that progressed to clinical trials, most failed on the efficacy front, with only a few still unconfirmed and potential positive trends. This suggests that robust preclinical and clinical data is needed to unequivocally evaluate their efficacy. To this end, we take a systematic look at the results of preclinical and clinical studies of PKIs and PP2A activators, and the evidence they provide regarding the utility of this approach to evaluate the potential of targeting tau hyperphosphorylation as a disease modifying therapy.
Collapse
Affiliation(s)
- Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia
| | - Tomáš Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY, 10314, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY, 10314, USA
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia.
- AXON Neuroscience R&D Services SE, Bratislava, 811 02, Slovakia.
| | - Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia.
- AXON Neuroscience CRM Services SE, Bratislava, 811 02, Slovakia.
| |
Collapse
|
26
|
Grygier P, Pustelny K, Nowak J, Golik P, Popowicz GM, Plettenburg O, Dubin G, Menezes F, Czarna A. Silmitasertib (CX-4945), a Clinically Used CK2-Kinase Inhibitor with Additional Effects on GSK3β and DYRK1A Kinases: A Structural Perspective. J Med Chem 2023; 66:4009-4024. [PMID: 36883902 PMCID: PMC10041529 DOI: 10.1021/acs.jmedchem.2c01887] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
A clinical casein kinase 2 inhibitor, CX-4945 (silmitasertib), shows significant affinity toward the DYRK1A and GSK3β kinases, involved in down syndrome phenotypes, Alzheimer's disease, circadian clock regulation, and diabetes. This off-target activity offers an opportunity for studying the effect of the DYRK1A/GSK3β kinase system in disease biology and possible line extension. Motivated by the dual inhibition of these kinases, we solved and analyzed the crystal structures of DYRK1A and GSK3β with CX-4945. We built a quantum-chemistry-based model to rationalize the compound affinity for CK2α, DYRK1A, and GSK3β kinases. Our calculations identified a key element for CK2α's subnanomolar affinity to CX-4945. The methodology is expandable to other kinase selectivity modeling. We show that the inhibitor limits DYRK1A- and GSK3β-mediated cyclin D1 phosphorylation and reduces kinase-mediated NFAT signaling in the cell. Given the CX-4945's clinical and pharmacological profile, this inhibitory activity makes it an interesting candidate with potential for application in additional disease areas.
Collapse
Affiliation(s)
- Przemyslaw Grygier
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | - Katarzyna Pustelny
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | - Jakub Nowak
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | | | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Zentrum Muenchen, Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Department Chemie, Technical University of Munich, Lichtenbergstrasse 4, Garching 85747, Germany
| | - Oliver Plettenburg
- Institute of Medicinal Chemistry, Helmholtz Munich, Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Institute of Organic Chemistry, Centre of Biomolecular Drug Research (BMWZ) and Laboratory of Nano and Quantum Engineering (LNQE), Leibniz University Hannover, Schneiderberg 1b, Hannover 30167, Germany
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Institute of Lung Health (ILH), Aulweg 130, Giessen 35392, Germany
| | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | - Filipe Menezes
- Institute of Structural Biology, Helmholtz Zentrum Muenchen, Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Department Chemie, Technical University of Munich, Lichtenbergstrasse 4, Garching 85747, Germany
| | - Anna Czarna
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| |
Collapse
|
27
|
Hartz RA, Ahuja VT, Sivaprakasam P, Xiao H, Krause CM, Clarke WJ, Kish K, Lewis H, Szapiel N, Ravirala R, Mutalik S, Nakmode D, Shah D, Burton CR, Macor JE, Dubowchik GM. Design, Structure-Activity Relationships, and In Vivo Evaluation of Potent and Brain-Penetrant Imidazo[1,2- b]pyridazines as Glycogen Synthase Kinase-3β (GSK-3β) Inhibitors. J Med Chem 2023; 66:4231-4252. [PMID: 36950863 DOI: 10.1021/acs.jmedchem.3c00133] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that regulates numerous cellular processes, including metabolism, proliferation, and cell survival. Due to its multifaceted role, GSK-3 has been implicated in a variety of diseases, including Alzheimer's disease, type 2 diabetes, cancer, and mood disorders. GSK-3β has been linked to the formation of the neurofibrillary tangles associated with Alzheimer's disease that arise from the hyperphosphorylation of tau protein. The design and synthesis of a series of imidazo[1,2-b]pyridazine derivatives that were evaluated as GSK-3β inhibitors are described herein. Structure-activity relationship studies led to the identification of potent GSK-3β inhibitors. In vivo studies with 47 in a triple-transgenic mouse Alzheimer's disease model showed that this compound is a brain-penetrant, orally bioavailable GSK-3β inhibitor that significantly lowered levels of phosphorylated tau.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ramu Ravirala
- Biocon-Bristol Myers Squibb Research and Development Center, Biocon Park, Plot No. 2 & 3, Bommasandra Phase IV, Jigani Link Road, Bangalore 560099, India
| | - Sayali Mutalik
- Biocon-Bristol Myers Squibb Research and Development Center, Biocon Park, Plot No. 2 & 3, Bommasandra Phase IV, Jigani Link Road, Bangalore 560099, India
| | - Deepa Nakmode
- Biocon-Bristol Myers Squibb Research and Development Center, Biocon Park, Plot No. 2 & 3, Bommasandra Phase IV, Jigani Link Road, Bangalore 560099, India
| | - Devang Shah
- Biocon-Bristol Myers Squibb Research and Development Center, Biocon Park, Plot No. 2 & 3, Bommasandra Phase IV, Jigani Link Road, Bangalore 560099, India
| | | | | | | |
Collapse
|
28
|
Role of Tau in Various Tauopathies, Treatment Approaches, and Emerging Role of Nanotechnology in Neurodegenerative Disorders. Mol Neurobiol 2023; 60:1690-1720. [PMID: 36562884 DOI: 10.1007/s12035-022-03164-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A few protein kinases and phosphatases regulate tau protein phosphorylation and an imbalance in their enzyme activity results in tau hyper-phosphorylation. Aberrant tau phosphorylation causes tau to dissociate from the microtubules and clump together in the cytosol to form neurofibrillary tangles (NFTs), which lead to the progression of neurodegenerative disorders including Alzheimer's disease (AD) and other tauopathies. Hence, targeting hyperphosphorylated tau protein is a restorative approach for treating neurodegenerative tauopathies. The cyclin-dependent kinase (Cdk5) and the glycogen synthase kinase (GSK3β) have both been implicated in aberrant tau hyperphosphorylation. The limited transport of drugs through the blood-brain barrier (BBB) for reaching the central nervous system (CNS) thus represents a significant problem in the development of drugs. Drug delivery systems based on nanocarriers help solve this problem. In this review, we discuss the tau protein, regulation of tau phosphorylation and abnormal hyperphosphorylation, drugs in use or under clinical trials, and treatment strategies for tauopathies based on the critical role of tau hyperphosphorylation in the pathogenesis of the disease. Pathology of neurodegenerative disease due to hyperphosphorylation and various therapeutic approaches including nanotechnology for its treatment.
Collapse
|
29
|
Liška K, Dočkal T, Houdek P, Sládek M, Lužná V, Semenovykh K, Drapšin M, Sumová A. Lithium affects the circadian clock in the choroid plexus - A new role for an old mechanism. Biomed Pharmacother 2023; 159:114292. [PMID: 36701987 DOI: 10.1016/j.biopha.2023.114292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
Lithium is an effective mood stabilizer, but the mechanism of its therapeutic action is not well understood. We investigated the effect of lithium on the circadian clock located in the ventricle barrier complex containing the choroid plexus (CP), a part of the glymphatic system that influences gross brain function via the production of cerebrospinal fluid. The mPer2Luc mice were injected with lithium chloride (LiCl) or vehicle, and their effects on the clock gene Nr1d1 in CP were detected by RT qPCR. CP organotypic explants were prepared to monitor bioluminescence rhythms in real time and examine the responses of the CP clock to LiCl and inhibitors of glycogen synthase kinase-3 (CHIR-99021) and protein kinase C (chelerythrine). LiCl affected Nr1d1 expression levels in CP in vivo and dose-dependently delayed the phase and prolonged the period of the CP clock in vitro. LiCl and CHIR-99021 had different effects on 1] CP clock parameters (amplitude, period, phase), 2] dexamethasone-induced phase shifts of the CP clock, and 3] dynamics of PER2 degradation and de novo accumulation. LiCl-induced phase delays were significantly reduced by chelerythrine, suggesting the involvement of PKC activity. The effects on the CP clock may be involved in the therapeutic effects of lithium and hypothetically improve brain function in psychiatric patients by aligning the function of the CP clock-related glymphatic system with the sleep-wake cycle. Importantly, our data argue for personalized timing of lithium treatment in BD patients.
Collapse
Affiliation(s)
- Karolína Liška
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tereza Dočkal
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Houdek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Sládek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vendula Lužná
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Kateryna Semenovykh
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Milica Drapšin
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Sumová
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
30
|
Chandel S, Singh R, Gautam A, Ravichandiran V. Screening of Azadirachta indica phytoconstituents as GSK-3β inhibitor and its implication in neuroblastoma: molecular docking, molecular dynamics, MM-PBSA binding energy, and in-vitro study. J Biomol Struct Dyn 2022; 40:12827-12840. [PMID: 34569452 DOI: 10.1080/07391102.2021.1977705] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3), a constitutively active serine/threonine kinase, primary regulator of various cellular activities varying from glycogen metabolism to cell proliferation and regulation. GSK-3β is associated with the pathogenesis of numerous human diseases, including cancer, metabolic disorder, and Alzheimer's disease. In this study, Azadirachta indica compounds were selected and further screened on the BOILED-Egg model. The compounds showing good GIT absorption were docked with the crystal structure of GSK-3β. The compounds with high docking score were submitted for the molecular dynamic simulation (MDS) and Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA). Based upon the MDS and MM-PBSA study, gedunin showed the highest binding energy throughout the MDS process. Gedunin was isolated from the Azadirachta indica, and its efficacy on GSK-3β inhibition was studied in the human neuroblastoma (SH-SY5Y) cells. Gedunin induced apoptosis and anti-proliferative activity by arresting G2/M phase, as evident by cell-cycle analysis. From immunoblot study, gedunin significantly enhanced the expression of an inhibitory form of GSK-3β (p-GSK-3β Ser9) in concentration-dependent manner. Our findings demonstrate that gedunin may act as an effective GSK-3β inhibitor suggesting that this compound may be used for the management of neuroblastoma. Further preclinical and clinical investigation is desirable.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shivani Chandel
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, India
| | - Rajveer Singh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, India
| | - Anupam Gautam
- Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany.,International Max Planck Research School "From Molecules to Organisms", Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, India
| |
Collapse
|
31
|
Chatterjee D, Beaulieu JM. Inhibition of glycogen synthase kinase 3 by lithium, a mechanism in search of specificity. Front Mol Neurosci 2022; 15:1028963. [PMID: 36504683 PMCID: PMC9731798 DOI: 10.3389/fnmol.2022.1028963] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/24/2022] [Indexed: 11/25/2022] Open
Abstract
Inhibition of Glycogen synthase kinase 3 (GSK3) is a popular explanation for the effects of lithium ions on mood regulation in bipolar disorder and other mental illnesses, including major depression, cyclothymia, and schizophrenia. Contribution of GSK3 is supported by evidence obtained from animal and patient derived model systems. However, the two GSK3 enzymes, GSK3α and GSK3β, have more than 100 validated substrates. They are thus central hubs for major biological functions, such as dopamine-glutamate neurotransmission, synaptic plasticity (Hebbian and homeostatic), inflammation, circadian regulation, protein synthesis, metabolism, inflammation, and mitochondrial functions. The intricate contributions of GSK3 to several biological processes make it difficult to identify specific mechanisms of mood stabilization for therapeutic development. Identification of GSK3 substrates involved in lithium therapeutic action is thus critical. We provide an overview of GSK3 biological functions and substrates for which there is evidence for a contribution to lithium effects. A particular focus is given to four of these: the transcription factor cAMP response element-binding protein (CREB), the RNA-binding protein FXR1, kinesin subunits, and the cytoskeletal regulator CRMP2. An overview of how co-regulation of these substrates may result in shared outcomes is also presented. Better understanding of how inhibition of GSK3 contributes to the therapeutic effects of lithium should allow for identification of more specific targets for future drug development. It may also provide a framework for the understanding of how lithium effects overlap with those of other drugs such as ketamine and antipsychotics, which also inhibit brain GSK3.
Collapse
Affiliation(s)
| | - Jean Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Dzaki N, Bu S, Lau SSY, Yong WL, Yu F. Drosophila GSK3β promotes microtubule disassembly and dendrite pruning in sensory neurons. Development 2022; 149:281771. [PMID: 36264221 DOI: 10.1242/dev.200844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022]
Abstract
The evolutionarily conserved Glycogen Synthase Kinase 3β (GSK3β), a negative regulator of microtubules, is crucial for neuronal polarization, growth and migration during animal development. However, it remains unknown whether GSK3β regulates neuronal pruning, which is a regressive process. Here, we report that the Drosophila GSK3β homologue Shaggy (Sgg) is cell-autonomously required for dendrite pruning of ddaC sensory neurons during metamorphosis. Sgg is necessary and sufficient to promote microtubule depolymerization, turnover and disassembly in the dendrites. Although Sgg is not required for the minus-end-out microtubule orientation in dendrites, hyperactivated Sgg can disturb the dendritic microtubule orientation. Moreover, our pharmacological and genetic data suggest that Sgg is required to promote dendrite pruning at least partly via microtubule disassembly. We show that Sgg and Par-1 kinases act synergistically to promote microtubule disassembly and dendrite pruning. Thus, Sgg and Par-1 might converge on and phosphorylate a common downstream microtubule-associated protein(s) to disassemble microtubules and thereby facilitate dendrite pruning.
Collapse
Affiliation(s)
- Najat Dzaki
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604
| | - Shufeng Bu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604.,Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Samuel Song Yuan Lau
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604
| | - Wei Lin Yong
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604.,Department of Biological Sciences, National University of Singapore, Singapore 117543
| |
Collapse
|
33
|
Wu H, Zhu N, Liu J, Ma J, Jiao R. Shaggy regulates tissue growth through Hippo pathway in Drosophila. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2131-2144. [PMID: 36057002 DOI: 10.1007/s11427-022-2156-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
The evolutionarily conserved Hippo pathway coordinates cell proliferation, differentiation and apoptosis to regulate organ growth and tumorigenesis. Hippo signaling activity is tightly controlled by various upstream signals including growth factors and cell polarity, but the full extent to which the pathway is regulated during development remains to be resolved. Here, we report the identification of Shaggy, the homolog of mammalian Gsk3β, as a novel regulator of the Hippo pathway in Drosophila. Our results show that Shaggy promotes the expression of Hippo target genes in a manner that is dependent on its kinase activity. Loss of Shaggy leads to Yorkie inhibition and downregulation of Hippo pathway target genes. Mechanistically, Shaggy acts upstream of the Hippo pathway and negatively regulates the abundance of the FERM domain containing adaptor protein Expanded. Our results reveal that Shaggy is functionally required for Crumbs/Slmb-mediated downregulation of Expanded in vivo, providing a potential molecular link between cellular architecture and the Hippo signaling pathway.
Collapse
Affiliation(s)
- Honggang Wu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Nannan Zhu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiyong Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jun Ma
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 311121, China
| | - Renjie Jiao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
34
|
Hottin C, Perron M, Roger JE. GSK3 Is a Central Player in Retinal Degenerative Diseases but a Challenging Therapeutic Target. Cells 2022; 11:cells11182898. [PMID: 36139472 PMCID: PMC9496697 DOI: 10.3390/cells11182898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. Although recent works strongly suggest that inhibiting GSK3 might also be a promising therapeutic approach for retinal degenerative diseases, its full potential is still under-evaluated. In this review, we summarize the literature on the role of GSK3 on the main cellular functions reported as deregulated during retinal degeneration, such as glucose homeostasis which is critical for photoreceptor survival, or oxidative stress, a major component of retinal degeneration. We also discuss the interest in targeting GSK3 for its beneficial effects on inflammation, for reducing neovascularization that occurs in some retinal dystrophies, or for cell-based therapy by enhancing Müller glia cell proliferation in diseased retina. Together, although GSK3 inhibitors hold promise as therapeutic agents, we highlight the complexity of targeting such a multitasked kinase and the need to increase our knowledge of the impact of reducing GSK3 activity on these multiple cellular pathways and biological processes.
Collapse
Affiliation(s)
- Catherine Hottin
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
35
|
Yousef MH, Salama M, El-Fawal HAN, Abdelnaser A. Selective GSK3β Inhibition Mediates an Nrf2-Independent Anti-inflammatory Microglial Response. Mol Neurobiol 2022; 59:5591-5611. [PMID: 35739410 PMCID: PMC9395457 DOI: 10.1007/s12035-022-02923-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022]
Abstract
Glycogen synthase kinase 3 (GSK3) is associated with the proinflammatory phenotype of microglia and has been shown to act in concert with nuclear factor kappa B (NF-κB). GSK3 is also a suppressor of nuclear factor erythroid 2-related factor 2 (Nrf2), the principal regulator of redox homeostasis. Agreeing with the oxidative paradigm of aging, Nrf2 is often deregulated in parainflammatory and neurodegenerative diseases. In this study, we aimed to explore a multimodal disease-modifying utility of GSK3 inhibition, beyond neuronal proteopathologies. Furthermore, we aimed to underscore the difference in therapeutic value between the two GSK3 paralogs by isoform-selective chemical inhibition. The anti-inflammatory effects of paralog-selective GSK3 inhibitors were evaluated as a function of the reductive capacity of each to mitigate LPS-induced activation of SIM-A9 microglia. The Griess method was employed to detect the nitrate-lowering capacity of selective GSK3 inhibition. Real-time PCR was used to assess post-treatment expression levels of pro-inflammatory markers and antioxidant genes; pro-inflammatory cytokines were assayed by ELISA. Nuclear lysates of treated cells were examined for Nrf2 and NF-κB accumulation by immunoblotting. Finally, to infer whether the counter-inflammatory activity of GSK3 inhibition was Nrf2-dependent, DsiRNA-mediated knockdown of Nrf2 was attempted. Results from our experiments reveal a superior anti-inflammatory and anti-oxidative efficacy for GSK3β-selective inhibition, compared to GSK3α-selective and non-selective pan-inhibition; hence, use of selective GSK3β inhibitors is likely to be more propitious than non-selective dual inhibitors administered at comparable doses. Moreover, our results suggest that the anti-inflammatory effects of GSK3 inhibition are not Nrf2 dependent.
Collapse
Affiliation(s)
- Mohamed H Yousef
- School of Sciences and Engineering, Biotechnology Graduate Program, The American University in Cairo, P.O. Box: 74, Cairo, Egypt
| | - Mohamed Salama
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, Cairo, Egypt
| | - Hassan A N El-Fawal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, Cairo, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, Cairo, Egypt.
| |
Collapse
|
36
|
Sukanya S, Choudhary BS, Mehta P, Filipek S, Malik R. Identification of CNS compatible small molecules as glycogen synthase kinase-3β (GSK-3β) inhibitors through structure-based virtual screening. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02912-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
37
|
Kim P, Park J, Lee DJ, Mizuno S, Shinohara M, Hong CP, Jeong Y, Yun R, Park H, Park S, Yang KM, Lee MJ, Jang SP, Kim HY, Lee SJ, Song SU, Park KS, Tanaka M, Ohshima H, Cho JW, Sugiyama F, Takahashi S, Jung HS, Kim SJ. Mast4 determines the cell fate of MSCs for bone and cartilage development. Nat Commun 2022; 13:3960. [PMID: 35803931 PMCID: PMC9270402 DOI: 10.1038/s41467-022-31697-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/28/2022] [Indexed: 11/26/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) differentiation into different lineages is precisely controlled by signaling pathways. Given that protein kinases play a crucial role in signal transduction, here we show that Microtubule Associated Serine/Threonine Kinase Family Member 4 (Mast4) serves as an important mediator of TGF-β and Wnt signal transduction in regulating chondro-osteogenic differentiation of MSCs. Suppression of Mast4 by TGF-β1 led to increased Sox9 stability by blocking Mast4-induced Sox9 serine 494 phosphorylation and subsequent proteasomal degradation, ultimately enhancing chondrogenesis of MSCs. On the other hand, Mast4 protein, which stability was enhanced by Wnt-mediated inhibition of GSK-3β and subsequent Smurf1 recruitment, promoted β-catenin nuclear localization and Runx2 activity, increasing osteogenesis of MSCs. Consistently, Mast4-/- mice demonstrated excessive cartilage synthesis, while exhibiting osteoporotic phenotype. Interestingly, Mast4 depletion in MSCs facilitated cartilage formation and regeneration in vivo. Altogether, our findings uncover essential roles of Mast4 in determining the fate of MSC development into cartilage or bone.
Collapse
Affiliation(s)
- Pyunggang Kim
- GILO Institute, GILO Foundation, Seoul, 06668, Korea
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam City, 463-400, Kyunggi-do, Korea
| | - Jinah Park
- GILO Institute, GILO Foundation, Seoul, 06668, Korea
- Amoris Bio Inc, Seoul, 06668, Korea
| | - Dong-Joon Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masahiro Shinohara
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, 359-8555, Japan
| | | | - Yealeen Jeong
- GILO Institute, GILO Foundation, Seoul, 06668, Korea
| | - Rebecca Yun
- GILO Institute, GILO Foundation, Seoul, 06668, Korea
| | - Hyeyeon Park
- GILO Institute, GILO Foundation, Seoul, 06668, Korea
| | - Sujin Park
- GILO Institute, GILO Foundation, Seoul, 06668, Korea
| | | | - Min-Jung Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | | | - Hyun-Yi Kim
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, 03722, Korea
- NGeneS Inc., Ansan-si, 15495, Korea
| | - Seung-Jun Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Sun U Song
- Research Institute, SCM Lifescience Inc., Incheon, Korea
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| | - Kyung-Soon Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam City, 463-400, Kyunggi-do, Korea
| | - Mikako Tanaka
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan
- Division of Dental Laboratory Technology, Meirin College, Niigata, 950-2086, Japan
| | - Hayato Ohshima
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan
| | - Jin Won Cho
- Department of Systems Biology and Glycosylation Network Research Center, Yonsei University, Seoul, Korea
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul, 06668, Korea.
- Medpacto Inc., Seoul, 06668, Korea.
- TheragenEtex Co., Gyeonggi-do, Korea.
| |
Collapse
|
38
|
Albeely AM, Williams OOF, Perreault ML. GSK-3β Disrupts Neuronal Oscillatory Function to Inhibit Learning and Memory in Male Rats. Cell Mol Neurobiol 2022; 42:1341-1353. [PMID: 33392916 PMCID: PMC11421759 DOI: 10.1007/s10571-020-01020-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/27/2020] [Indexed: 12/25/2022]
Abstract
Alterations in glycogen synthase kinase-3β (GSK-3β) activity have been implicated in disorders of cognitive impairment, including Alzheimer's disease and schizophrenia. Cognitive dysfunction is also characterized by the dysregulation of neuronal oscillatory activity, macroscopic electrical rhythms in brain that are critical to systems communication. A direct functional relationship between GSK-3β and neuronal oscillations has not been elucidated. Therefore, in the present study, using an adeno-associated viral vector containing a persistently active mutant form of GSK-3β, GSK-3β(S9A), the impact of elevated kinase activity in prefrontal cortex (PFC) or ventral hippocampus (vHIP) of rats on neuronal oscillatory activity was evaluated. GSK-3β(S9A)-induced changes in learning and memory were also assessed and the phosphorylation status of tau protein, a substrate of GSK-3β, examined. It was demonstrated that increasing GSK-3β(S9A) activity in either the PFC or vHIP had similar effects on neuronal oscillatory activity, enhancing theta and/or gamma spectral power in one or both regions. Increasing PFC GSK-3β(S9A) activity additionally suppressed high gamma PFC-vHIP coherence. These changes were accompanied by deficits in recognition memory, spatial learning, and/or reversal learning. Elevated pathogenic tau phosphorylation was also evident in regions where GSK-3β(S9A) activity was upregulated. The neurophysiological and learning and memory deficits induced by GSK-3β(S9A) suggest that aberrant GSK-3β signalling may not only play an early role in cognitive decline in Alzheimer's disease but may also have a more central involvement in disorders of cognitive dysfunction through the regulation of neurophysiological network function.
Collapse
Affiliation(s)
- Abdalla M Albeely
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E, Guelph, ON, N1G 2W1, Canada
- Collaborative Neuroscience Program, University of Guelph, 50 Stone Rd. E, Guelph, ON, Canada
| | - Olivia O F Williams
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E, Guelph, ON, N1G 2W1, Canada
| | - Melissa L Perreault
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E, Guelph, ON, N1G 2W1, Canada.
- Collaborative Neuroscience Program, University of Guelph, 50 Stone Rd. E, Guelph, ON, Canada.
| |
Collapse
|
39
|
Glycogen Synthase Kinase 3: Ion Channels, Plasticity, and Diseases. Int J Mol Sci 2022; 23:ijms23084413. [PMID: 35457230 PMCID: PMC9028019 DOI: 10.3390/ijms23084413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3) is a multifaceted serine/threonine (S/T) kinase expressed in all eukaryotic cells. GSK3β is highly enriched in neurons in the central nervous system where it acts as a central hub for intracellular signaling downstream of receptors critical for neuronal function. Unlike other kinases, GSK3β is constitutively active, and its modulation mainly involves inhibition via upstream regulatory pathways rather than increased activation. Through an intricate converging signaling system, a fine-tuned balance of active and inactive GSK3β acts as a central point for the phosphorylation of numerous primed and unprimed substrates. Although the full range of molecular targets is still unknown, recent results show that voltage-gated ion channels are among the downstream targets of GSK3β. Here, we discuss the direct and indirect mechanisms by which GSK3β phosphorylates voltage-gated Na+ channels (Nav1.2 and Nav1.6) and voltage-gated K+ channels (Kv4 and Kv7) and their physiological effects on intrinsic excitability, neuronal plasticity, and behavior. We also present evidence for how unbalanced GSK3β activity can lead to maladaptive plasticity that ultimately renders neuronal circuitry more vulnerable, increasing the risk for developing neuropsychiatric disorders. In conclusion, GSK3β-dependent modulation of voltage-gated ion channels may serve as an important pharmacological target for neurotherapeutic development.
Collapse
|
40
|
Balboni B, Tripathi SK, Veronesi M, Russo D, Penna I, Giabbai B, Bandiera T, Storici P, Girotto S, Cavalli A. Identification of Novel GSK-3β Hits Using Competitive Biophysical Assays. Int J Mol Sci 2022; 23:3856. [PMID: 35409221 PMCID: PMC8998611 DOI: 10.3390/ijms23073856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/05/2023] Open
Abstract
Glycogen synthase kinase 3 beta (GSK-3β) is an evolutionarily conserved serine-threonine kinase dysregulated in numerous pathologies, such as Alzheimer's disease and cancer. Even though GSK-3β is a validated pharmacological target most of its inhibitors have two main limitations: the lack of selectivity due to the high homology that characterizes the ATP binding site of most kinases, and the toxicity that emerges from GSK-3β complete inhibition which translates into the impairment of the plethora of pathways GSK-3β is involved in. Starting from a 1D 19F NMR fragment screening, we set up several biophysical assays for the identification of GSK-3β inhibitors capable of binding protein hotspots other than the ATP binding pocket or to the ATP binding pocket, but with an affinity able of competing with a reference binder. A phosphorylation activity assay on a panel of several kinases provided selectivity data that were further rationalized and corroborated by structural information on GSK-3β in complex with the hit compounds. In this study, we identified promising fragments, inhibitors of GSK-3β, while proposing an alternative screening workflow that allows facing the flaws that characterize the most common GSK-3β inhibitors through the identification of selective inhibitors and/or inhibitors able to modulate GSK-3β activity without leading to its complete inhibition.
Collapse
Affiliation(s)
- Beatrice Balboni
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (B.B.); (S.K.T.)
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Shailesh Kumar Tripathi
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (B.B.); (S.K.T.)
| | - Marina Veronesi
- D3 Pharmachemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (M.V.); (D.R.); (I.P.); (T.B.)
| | - Debora Russo
- D3 Pharmachemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (M.V.); (D.R.); (I.P.); (T.B.)
| | - Ilaria Penna
- D3 Pharmachemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (M.V.); (D.R.); (I.P.); (T.B.)
| | - Barbara Giabbai
- Structural Biology Laboratory, Elettra Sincrotrone Trieste S.C.p.A., Basovizza, 34149 Trieste, Italy; (B.G.); (P.S.)
| | - Tiziano Bandiera
- D3 Pharmachemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (M.V.); (D.R.); (I.P.); (T.B.)
| | - Paola Storici
- Structural Biology Laboratory, Elettra Sincrotrone Trieste S.C.p.A., Basovizza, 34149 Trieste, Italy; (B.G.); (P.S.)
| | - Stefania Girotto
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (B.B.); (S.K.T.)
| | - Andrea Cavalli
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy; (B.B.); (S.K.T.)
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
41
|
Mahmud F, Lai NS, How SE, Gansau JA, Mustaffa KMF, Leow CH, Osman H, Sidek HM, Embi N, Lee PC. Bioactivities and Mode of Actions of Dibutyl Phthalates and Nocardamine from Streptomyces sp. H11809. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072292. [PMID: 35408690 PMCID: PMC9000801 DOI: 10.3390/molecules27072292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/30/2022]
Abstract
Dibutyl phthalate (DBP) produced by Streptomyces sp. H11809 exerted inhibitory activity against human GSK-3β (Hs GSK-3β) and Plasmodiumfalciparum 3D7 (Pf 3D7) malaria parasites. The current study aimed to determine DBP’s plausible mode of action against Hs GSK-3β and Pf 3D7. Molecular docking analysis indicated that DBP has a higher binding affinity to the substrate-binding site (pocket 2; −6.9 kcal/mol) than the ATP-binding site (pocket 1; −6.1 kcal/mol) of Hs GSK-3β. It was suggested that the esters of DBP play a pivotal role in the inhibition of Hs GSK-3β through the formation of hydrogen bonds with Arg96/Glu97 amino acid residues in pocket 2. Subsequently, an in vitro Hs GSK-3β enzymatic assay revealed that DBP inhibits the activity of Hs GSK-3β via mixed inhibition inhibitory mechanisms, with a moderate IC50 of 2.0 µM. Furthermore, the decrease in Km value with an increasing DBP concentration suggested that DBP favors binding on free Hs GSK-3β over its substrate-bound state. However, the antimalarial mode of action of DBP remains unknown since the generation of a Pf 3D7 DBP-resistant clone was not successful. Thus, the molecular target of DBP might be indispensable for Pf survival. We also identified nocardamine as another active compound from Streptomyces sp. H11809 chloroform extract. It showed potent antimalarial activity with an IC50 of 1.5 μM, which is ~10-fold more potent than DBP, but with no effect on Hs GSK-3β. The addition of ≥12.5 µM ferric ions into the Pf culture reduced nocardamine antimalarial activity by 90% under in vitro settings. Hence, the iron-chelating ability of nocardamine was shown to starve the parasites from their iron source, eventually inhibiting their growth.
Collapse
Affiliation(s)
- Fauze Mahmud
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor 11800, Malaysia; (F.M.); (K.M.F.M.); (C.H.L.)
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia; (S.E.H.); (J.A.G.)
| | - Ngit Shin Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor 11800, Malaysia; (F.M.); (K.M.F.M.); (C.H.L.)
- Correspondence: (N.S.L.); (P.-C.L.); Tel.: +60-4653-4862 (N.S.L.); +60-8832-0000 (P.-C.L.); Fax: +60-4653-4803 (N.S.L.); +60-8843-2324 (P.-C.L.)
| | - Siew Eng How
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia; (S.E.H.); (J.A.G.)
| | - Jualang Azlan Gansau
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia; (S.E.H.); (J.A.G.)
| | - Khairul Mohd Fadzli Mustaffa
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor 11800, Malaysia; (F.M.); (K.M.F.M.); (C.H.L.)
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor 11800, Malaysia; (F.M.); (K.M.F.M.); (C.H.L.)
| | - Hasnah Osman
- School of Chemical Sciences, Universiti Sains Malaysia, Gelugor 11800, Malaysia;
| | - Hasidah Mohd Sidek
- Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (H.M.S.); (N.E.)
| | - Noor Embi
- Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (H.M.S.); (N.E.)
| | - Ping-Chin Lee
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia; (S.E.H.); (J.A.G.)
- Biotechnology Research Institute, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Correspondence: (N.S.L.); (P.-C.L.); Tel.: +60-4653-4862 (N.S.L.); +60-8832-0000 (P.-C.L.); Fax: +60-4653-4803 (N.S.L.); +60-8843-2324 (P.-C.L.)
| |
Collapse
|
42
|
Aourz N, Van Leuven F, Allaoui W, Van Eeckhaut A, De Bundel D, Smolders I. Unraveling the Effects of GSK-3β Isoform Modulation against Limbic Seizures and in the 6 Hz Electrical Kindling Model for Epileptogenesis. ACS Chem Neurosci 2022; 13:796-805. [PMID: 35253420 DOI: 10.1021/acschemneuro.1c00782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Two closely related glycogen synthase kinase-3 (GSK-3) isoforms have been identified in mammals: GSK-3α and GSK-3β. GSK-3β is the most prominent in the central nervous system and was previously shown to control neuronal excitability. We previously demonstrated that indirubin and its structural analogue and the nonselective GSK-3 inhibitor BIO-acetoxime exerted anticonvulsant effects in acute seizure models in zebrafish, mice, and rats. We here examined for the first time the anticonvulsant effect of TCS2002, a specific and potent inhibitor of GSK-3β, in two models for limbic seizures: the pilocarpine rat model for focal seizures and the acute 6 Hz corneal mouse model for refractory seizures. Next, we additionally used the 6 Hz kindling model to establish differences in seizure susceptibility and seizure progression in mice that either overexpress human GSK-3β (GSK-3β OE) or lack GSK-3β (GSK-3β-/-) in neurons. We demonstrate that TCS2002 exerts anticonvulsant actions against pilocarpine- and 6 Hz-evoked seizures. Compared to wild-type littermates, GSK-3β OE mice are less susceptible to seizures but are more rapidly kindled. Interestingly, compared to GSK-3β+/+ mice, neuronal GSK-3β-/- mice show increased susceptibility to 6 Hz-induced seizures. These contrasting observations suggest compensatory neurodevelopmental mechanisms that alter seizure susceptibility in GSK-3β OE and GSK-3β-/- mice. Although the pronounced anticonvulsant effects of selective and acute GSK-3β inhibition in the 6 Hz model identify GSK-3β as a potential drug target for pharmacoresistant seizures, our data on the sustained disruption of GSK-3β activity in the transgenic mice suggest a role for GSK-3 in kindling and warrants further research into the long-term effects of selective pharmacological GSK-3β inhibition.
Collapse
Affiliation(s)
- Najat Aourz
- Vrije Universiteit Brussel (VUB), Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information/Center for Neurosciences (C4N), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Fred Van Leuven
- Experimental Genetics Group (LEGTEGG), Department of Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - Wissal Allaoui
- Vrije Universiteit Brussel (VUB), Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information/Center for Neurosciences (C4N), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ann Van Eeckhaut
- Vrije Universiteit Brussel (VUB), Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information/Center for Neurosciences (C4N), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Dimitri De Bundel
- Vrije Universiteit Brussel (VUB), Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information/Center for Neurosciences (C4N), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ilse Smolders
- Vrije Universiteit Brussel (VUB), Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information/Center for Neurosciences (C4N), Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|
43
|
Zhou F, Yao L, Lu X, Li Y, Han X, Wang P. Therapeutic Targeting of GSK3β-Regulated Nrf2 and NFκB Signaling Pathways by Salvianolic Acid A Ameliorates Peritoneal Fibrosis. Front Med (Lausanne) 2022; 9:804899. [PMID: 35321474 PMCID: PMC8936188 DOI: 10.3389/fmed.2022.804899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/02/2022] [Indexed: 02/06/2023] Open
Abstract
Peritoneal fibrosis is a devastating complication in patients undergoing peritoneal dialysis, with no definite therapy yet available. Salvia miltiorrhiza and its major active component Salvianolic acid A (Sal A) have demonstrated a beneficial effect in myriad diseases. However, their effect on peritoneal fibrosis is unknown. In murine models of peritoneal dialysis, daily Sal A treatment substantially improved the peritoneal dialysis fluid (PDF) elicited peritoneal fibrosis, marked by thickening of the submesothelial compact zone, accumulation of extracellular matrix and increased expression of vimentin and PAI-1, concomitant with attenuation of GSK3β hyperactivity. This coincided with diminished nitrotyrosine in peritoneal tissues and increased Nrf2 nuclear translocation, entailing a lessened oxidative injury and reinforced Nrf2 antioxidant response. Meanwhile, inflammatory infiltration and maladaptive angiogenesis in peritoneal tissues provoked by PDF injury were also mitigated by Sal A, associated with a suppressed NFκB activation. Mechanistically, ectopic expression of the constitutively active GSK3β blunted the NFκB-suppressing and Nrf2-activating efficacy of Sal A in peritoneal mesothelial cells exposed to hypertonic dextrose, suggesting that GSK3β inhibition mediates the protective effect of Sal A. Collectively, our findings may open the avenue for developing a novel therapy based on Sal A for preventing peritoneal fibrosis in peritoneal dialysis.
Collapse
Affiliation(s)
- Fan Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Molecular Imaging, Zhengzhou, China
| | - Lan Yao
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoqing Lu
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yubao Li
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xingmin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Molecular Imaging, Zhengzhou, China
| | - Pei Wang
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Pei Wang
| |
Collapse
|
44
|
Bala A, Roy S, Das D, Marturi V, Mondal C, Patra S, Haldar PK, Samajdar G. Role of Glycogen Synthase Kinase-3 in the Etiology of Type 2 Diabetes Mellitus: A Review. Curr Diabetes Rev 2022; 18:e300721195147. [PMID: 34376135 DOI: 10.2174/1573399817666210730094225] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/15/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022]
Abstract
The risk of type 2 diabetes mellitus (T2DM) is increasing abundantly due to lifestyle-related obesity and associated cardiovascular problems. Presently, Glycogen synthase kinase-3 (GSK-3) has gained considerable attention from biomedical scientists to treat diabetes. Phosphorylation of GSK-3 permits a number of cellular activities like regulation of cell signaling, cellular metabolism, cell proliferation and cellular transport. Inhibiting GSK-3 activity by pharmacological intervention has become an important strategy for the management of T2DM. This review focuses on the schematic representation of fundamental GSK-3 enzymology and encompasses the GSK-3 inhibitors as a future therapeutic lead target for the management of T2DM that may significantly regulate insulin sensitivity to insulin receptor, glycogen synthesis and glucose metabolism. The various signaling mechanisms of inhibiting the GSK-3 by describing insulin signaling through Insulin Receptor Substrate (IRS-1), Phosphatidylinositol-3 Kinase (PI3K) and Protein Kinase B (PKB/ AKT) pathways that may hopefully facilitate the pharmacologist to design for antidiabetic drug evaluation model in near future have also been highlighted.
Collapse
Affiliation(s)
- Asis Bala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, EPIP Campus, Hajipur, Bihar 844102, India
- Division of Pharmacology and Toxicology, Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Road, Panihati, Sodepur, Kolkata-700114; India
| | - Susmita Roy
- Division of Pharmacology and Toxicology, Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Road, Panihati, Sodepur, Kolkata-700114; India
| | - Debanjana Das
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, EPIP Campus, Hajipur, Bihar 844102, India
| | - Venkatesh Marturi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, EPIP Campus, Hajipur, Bihar 844102, India
| | - Chaitali Mondal
- TCG Life Sciences (Chembiotek) Pvt. Ltd., Sector V, Salt Lake Electronics Complex, Kolkata, West Bengal 700091, India
| | - Susmita Patra
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Pallab Kanti Haldar
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Gourav Samajdar
- Division of Pharmacology and Toxicology, Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Road, Panihati, Sodepur, Kolkata-700114; India
| |
Collapse
|
45
|
Vainio L, Taponen S, Kinnunen SM, Halmetoja E, Szabo Z, Alakoski T, Ulvila J, Junttila J, Lakkisto P, Magga J, Kerkelä R. GSK3β Serine 389 Phosphorylation Modulates Cardiomyocyte Hypertrophy and Ischemic Injury. Int J Mol Sci 2021; 22:13586. [PMID: 34948382 PMCID: PMC8707850 DOI: 10.3390/ijms222413586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Prior studies show that glycogen synthase kinase 3β (GSK3β) contributes to cardiac ischemic injury and cardiac hypertrophy. GSK3β is constitutionally active and phosphorylation of GSK3β at serine 9 (S9) inactivates the kinase and promotes cellular growth. GSK3β is also phosphorylated at serine 389 (S389), but the significance of this phosphorylation in the heart is not known. We analyzed GSK3β S389 phosphorylation in diseased hearts and utilized overexpression of GSK3β carrying ser→ala mutations at S9 (S9A) and S389 (S389A) to study the biological function of constitutively active GSK3β in primary cardiomyocytes. We found that phosphorylation of GSK3β at S389 was increased in left ventricular samples from patients with dilated cardiomyopathy and ischemic cardiomyopathy, and in hearts of mice subjected to thoracic aortic constriction. Overexpression of either GSK3β S9A or S389A reduced the viability of cardiomyocytes subjected to hypoxia-reoxygenation. Overexpression of double GSK3β mutant (S9A/S389A) further reduced cardiomyocyte viability. Determination of protein synthesis showed that overexpression of GSK3β S389A or GSK3β S9A/S389A increased both basal and agonist-induced cardiomyocyte growth. Mechanistically, GSK3β S389A mutation was associated with activation of mTOR complex 1 signaling. In conclusion, our data suggest that phosphorylation of GSK3β at S389 enhances cardiomyocyte survival and protects from cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Laura Vainio
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Saija Taponen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Sini M. Kinnunen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Eveliina Halmetoja
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
| | - Zoltan Szabo
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
| | - Tarja Alakoski
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Johanna Ulvila
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
| | - Juhani Junttila
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
- Research Unit of Internal Medicine, Division of Cardiology, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
| | - Päivi Lakkisto
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Helsinki 00014, Finland;
- Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Johanna Magga
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Risto Kerkelä
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
| |
Collapse
|
46
|
You JS, Chen J. Aging Does Not Exacerbate Muscle Loss During Denervation and Lends Unique Muscle-Specific Atrophy Resistance With Akt Activation. Front Physiol 2021; 12:779547. [PMID: 34916960 PMCID: PMC8669767 DOI: 10.3389/fphys.2021.779547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/09/2021] [Indexed: 01/10/2023] Open
Abstract
Sarcopenia, or age-related skeletal muscle atrophy and weakness, imposes significant clinical and economic burdens on affected patients and societies. Neurological degeneration, such as motoneuron death, has been recognized as a key contributor to sarcopenia. However, little is known about how aged/sarcopenic muscle adapts to this denervation stress. Here, we show that mice at 27months of age exhibit clear signs of sarcopenia but no accelerated denervation-induced muscle atrophy when compared to 8-month-old mice. Surprisingly, aging lends unique atrophy resistance to tibialis anteria muscle, accompanied by an increase in the cascade of mammalian target of rapamycin complex 1 (mTORC1)-independent anabolic events involving Akt signaling, rRNA biogenesis, and protein synthesis during denervation. These results expand our understanding of age-dependent stress responses and may help develop better countermeasures to sarcopenia.
Collapse
Affiliation(s)
- Jae-Sung You
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | | |
Collapse
|
47
|
Graca FA, Sheffield N, Puppa M, Finkelstein D, Hunt LC, Demontis F. A large-scale transgenic RNAi screen identifies transcription factors that modulate myofiber size in Drosophila. PLoS Genet 2021; 17:e1009926. [PMID: 34780463 PMCID: PMC8629395 DOI: 10.1371/journal.pgen.1009926] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 11/29/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
Myofiber atrophy occurs with aging and in many diseases but the underlying mechanisms are incompletely understood. Here, we have used >1,100 muscle-targeted RNAi interventions to comprehensively assess the function of 447 transcription factors in the developmental growth of body wall skeletal muscles in Drosophila. This screen identifies new regulators of myofiber atrophy and hypertrophy, including the transcription factor Deaf1. Deaf1 RNAi increases myofiber size whereas Deaf1 overexpression induces atrophy. Consistent with its annotation as a Gsk3 phosphorylation substrate, Deaf1 and Gsk3 induce largely overlapping transcriptional changes that are opposed by Deaf1 RNAi. The top category of Deaf1-regulated genes consists of glycolytic enzymes, which are suppressed by Deaf1 and Gsk3 but are upregulated by Deaf1 RNAi. Similar to Deaf1 and Gsk3 overexpression, RNAi for glycolytic enzymes reduces myofiber growth. Altogether, this study defines the repertoire of transcription factors that regulate developmental myofiber growth and the role of Gsk3/Deaf1/glycolysis in this process.
Collapse
Affiliation(s)
- Flavia A. Graca
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Natalie Sheffield
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Melissa Puppa
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Liam C. Hunt
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
48
|
Sharma P, Tiufekchiev S, Lising V, Chung SW, Suk JS, Chung BM. Keratin 19 interacts with GSK3β to regulate its nuclear accumulation and degradation of cyclin D3. Mol Biol Cell 2021; 32:ar21. [PMID: 34406791 PMCID: PMC8693971 DOI: 10.1091/mbc.e21-05-0255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cyclin D3 regulates the G1/S transition and is frequently overexpressed in several cancer types including breast cancer, where it promotes tumor progression. Here we show that a cytoskeletal protein keratin 19 (K19) physically interacts with a serine/threonine kinase GSK3β and prevents GSK3β-dependent degradation of cyclin D3. The absence of K19 allowed active GSK3β to accumulate in the nucleus and degrade cyclin D3. Specifically, the head (H) domain of K19 was required to sustain inhibitory phosphorylation of GSK3β Ser9, prevent nuclear accumulation of GSK3β, and maintain cyclin D3 levels and cell proliferation. K19 was found to interact with GSK3β and K19–GSK3β interaction was mapped out to require Ser10 and Ser35 residues on the H domain of K19. Unlike wildtype K19, S10A and S35A mutants failed to maintain total and nuclear cyclin D3 levels and induce cell proliferation. Finally, we show that the K19–GSK3β-cyclin D3 pathway affected sensitivity of cells toward inhibitors to cyclin-dependent kinase 4 and 6 (CDK4/6). Overall, these findings establish a role for K19 in the regulation of GSK3β-cyclin D3 pathway and demonstrate a potential strategy for overcoming resistance to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Pooja Sharma
- Department of Biology, The Catholic University of America, Washington, DC 20064
| | - Sarah Tiufekchiev
- Department of Biology, The Catholic University of America, Washington, DC 20064
| | - Victoria Lising
- Department of Biology, The Catholic University of America, Washington, DC 20064
| | - Seung Woo Chung
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231
| | - Jung Soo Suk
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231
| | - Byung Min Chung
- Department of Biology, The Catholic University of America, Washington, DC 20064
| |
Collapse
|
49
|
Targeting the coronavirus nucleocapsid protein through GSK-3 inhibition. Proc Natl Acad Sci U S A 2021; 118:2113401118. [PMID: 34593624 PMCID: PMC8594528 DOI: 10.1073/pnas.2113401118] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2021] [Indexed: 12/20/2022] Open
Abstract
COVID-19 is taking a major toll on personal health, healthcare systems, and the global economy. With three betacoronavirus epidemics in less than 20 y, there is an urgent need for therapies to combat new and existing coronavirus outbreaks. Our analysis of clinical data from over 300,000 patients in three major health systems demonstrates a 50% reduced risk of COVID-19 in patients taking lithium, a direct inhibitor of glycogen synthase kinase-3 (GSK-3). We further show that GSK-3 is essential for phosphorylation of the SARS-CoV-2 nucleocapsid protein and that GSK-3 inhibition blocks SARS-CoV-2 infection in human lung epithelial cells. These findings suggest an antiviral strategy for COVID-19 and new coronaviruses that may arise in the future. 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.
Collapse
|
50
|
Combined intermittent fasting and ERK inhibition enhance the anti-tumor effects of chemotherapy via the GSK3β-SIRT7 axis. Nat Commun 2021; 12:5058. [PMID: 34433808 PMCID: PMC8387475 DOI: 10.1038/s41467-021-25274-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 07/29/2021] [Indexed: 12/28/2022] Open
Abstract
Dietary interventions such as intermittent fasting (IF) have emerged as an attractive strategy for cancer therapies; therefore, understanding the underlying molecular mechanisms is pivotal. Here, we find SIRT7 decline markedly attenuates the anti-tumor effect of IF. Mechanistically, AMP-activated protein kinase (AMPK) phosphorylating SIRT7 at T263 triggers further phosphorylation at T255/S259 by glycogen synthase kinase 3β (GSK3β), which stabilizes SIRT7 by decoupling E3 ligase UBR5. SIRT7 hyperphosphorylation achieves anti-tumor activity by disrupting the SKP2-SCF E3 ligase, thus preventing SKP2-mediated K63-linked AKT polyubiquitination and subsequent activation. In contrast, GSK3β-SIRT7 axis is inhibited by EGF/ERK2 signaling, with ERK2 inactivating GSK3β, thus accelerating SIRT7 degradation. Unfavorably, glucose deprivation or chemotherapy hijacks the GSK3β-SIRT7 axis via ERK2, thus activating AKT and ensuring survival. Notably, Trametinib, an FDA-approved MEK inhibitor, enhances the efficacy of combination therapy with doxorubicin and IF. Overall, we have revealed the GSK3β-SIRT7 axis that must be fine-tuned in the face of the energetic and oncogenic stresses in malignancy. The combination of intermittent fasting and chemotherapy can improve the response to treatment. Here, the authors show that SIRT7 activation is required to inactivate Akt during intermittent fasting and that the combination of intermittent fasting and inhibitors that block the Erk pathway can improve efficacy of treatment.
Collapse
|