101
|
Martelli AM, Evangelisti C, Paganelli F, Chiarini F, McCubrey JA. GSK-3: a multifaceted player in acute leukemias. Leukemia 2021; 35:1829-1842. [PMID: 33811246 DOI: 10.1038/s41375-021-01243-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023]
Abstract
Glycogen synthase kinase 3 (GSK-3) consists of two isoforms (α and β) that were originally linked to glucose metabolism regulation. However, GSK-3 is also involved in several signaling pathways controlling many different key functions in healthy cells. GSK-3 is a unique kinase in that its isoforms are constitutively active, while they are inactivated mainly through phosphorylation at Ser residues by a variety of upstream kinases. In the early 1990s, GSK-3 emerged as a key player in cancer cell pathophysiology. Since active GSK-3 promotes destruction of multiple oncogenic proteins (e.g., β-catenin, c-Myc, Mcl-1) it was considered to be a tumor suppressor. Accordingly, GSK-3 is frequently inactivated in human cancer via aberrant regulation of upstream signaling pathways. More recently, however, it has emerged that GSK-3 isoforms display also oncogenic properties, as they up-regulate pathways critical for neoplastic cell proliferation, survival, and drug-resistance. The regulatory roles of GSK-3 isoforms in cell cycle, apoptosis, DNA repair, tumor metabolism, invasion, and metastasis reflect the therapeutic relevance of these kinases and provide the rationale for combining GSK-3 inhibitors with other targeted drugs. Here, we discuss the multiple and often conflicting roles of GSK-3 isoforms in acute leukemias. We also review the current status of GSK-3 inhibitor development for innovative leukemia therapy.
Collapse
Affiliation(s)
- Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Camilla Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Francesca Paganelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" Unit of Bologna, Bologna, Italy.,IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Chiarini
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" Unit of Bologna, Bologna, Italy.,IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
102
|
Wagstaff EL, Heredero Berzal A, Boon CJF, Quinn PMJ, ten Asbroek ALMA, Bergen AA. The Role of Small Molecules and Their Effect on the Molecular Mechanisms of Early Retinal Organoid Development. Int J Mol Sci 2021; 22:7081. [PMID: 34209272 PMCID: PMC8268497 DOI: 10.3390/ijms22137081] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Early in vivo embryonic retinal development is a well-documented and evolutionary conserved process. The specification towards eye development is temporally controlled by consecutive activation or inhibition of multiple key signaling pathways, such as the Wnt and hedgehog signaling pathways. Recently, with the use of retinal organoids, researchers aim to manipulate these pathways to achieve better human representative models for retinal development and disease. To achieve this, a plethora of different small molecules and signaling factors have been used at various time points and concentrations in retinal organoid differentiations, with varying success. Additions differ from protocol to protocol, but their usefulness or efficiency has not yet been systematically reviewed. Interestingly, many of these small molecules affect the same and/or multiple pathways, leading to reduced reproducibility and high variability between studies. In this review, we make an inventory of the key signaling pathways involved in early retinogenesis and their effect on the development of the early retina in vitro. Further, we provide a comprehensive overview of the small molecules and signaling factors that are added to retinal organoid differentiation protocols, documenting the molecular and functional effects of these additions. Lastly, we comparatively evaluate several of these factors using our established retinal organoid methodology.
Collapse
Affiliation(s)
- Ellie L. Wagstaff
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Peter M. J. Quinn
- Jonas Children’s Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center—New York-Presbyterian Hospital, New York, NY 10032, USA;
| | | | - Arthur A. Bergen
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Netherlands Institute for Neuroscience (NIN-KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
103
|
Abstract
Circadian clocks are biochemical time-keeping machines that synchronize animal behavior and physiology with planetary rhythms. In Drosophila, the core components of the clock comprise a transcription/translation feedback loop and are expressed in seven neuronal clusters in the brain. Although it is increasingly evident that the clocks in each of the neuronal clusters are regulated differently, how these clocks communicate with each other across the circadian neuronal network is less clear. Here, we review the latest evidence that describes the physical connectivity of the circadian neuronal network . Using small ventral lateral neurons as a starting point, we summarize how one clock may communicate with another, highlighting the signaling pathways that are both upstream and downstream of these clocks. We propose that additional efforts are required to understand how temporal information generated in each circadian neuron is integrated across a neuronal circuit to regulate rhythmic behavior.
Collapse
Affiliation(s)
- Myra Ahmad
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Wanhe Li
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Deniz Top
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
104
|
Sambon M, Wins P, Bettendorff L. Neuroprotective Effects of Thiamine and Precursors with Higher Bioavailability: Focus on Benfotiamine and Dibenzoylthiamine. Int J Mol Sci 2021; 22:ijms22115418. [PMID: 34063830 PMCID: PMC8196556 DOI: 10.3390/ijms22115418] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 11/25/2022] Open
Abstract
Thiamine (vitamin B1) is essential for brain function because of the coenzyme role of thiamine diphosphate (ThDP) in glucose and energy metabolism. In order to compensate thiamine deficiency, several thiamine precursors with higher bioavailability were developed since the 1950s. Among these, the thioester benfotiamine (BFT) has been extensively studied and has beneficial effects both in rodent models of neurodegeneration and in human clinical studies. BFT has antioxidant and anti-inflammatory properties that seem to be mediated by a mechanism independent of the coenzyme function of ThDP. BFT has no adverse effects and improves cognitive outcome in patients with mild Alzheimer’s disease (AD). Recent in vitro studies show that another thiamine thioester, dibenzoylthiamine (DBT) is even more efficient that BFT, especially with respect to its anti-inflammatory potency. Thiamine thioesters have pleiotropic properties linked to an increase in circulating thiamine concentrations and possibly in hitherto unidentified metabolites in particular open thiazole ring derivatives. The identification of the active neuroprotective derivatives and the clarification of their mechanism of action open extremely promising perspectives in the field of neurodegenerative, neurodevelopmental and psychiatric conditions.
Collapse
|
105
|
Papadopoli D, Pollak M, Topisirovic I. The role of GSK3 in metabolic pathway perturbations in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119059. [PMID: 33989699 DOI: 10.1016/j.bbamcr.2021.119059] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 01/11/2023]
Abstract
Malignant transformation and tumor progression are accompanied by significant perturbations in metabolic programs. As such, cancer cells support high ATP turnover to construct the building blocks needed to fuel neoplastic growth. The coordination of metabolic networks in malignant cells is dependent on the collaboration with cellular signaling pathways. Glycogen synthase kinase 3 (GSK3) lies at the convergence of several signaling axes, including the PI3K/AKT/mTOR, AMPK, and Wnt pathways, which influence cancer initiation, progression and therapeutic responses. Accordingly, GSK3 modulates metabolic processes, including protein and lipid synthesis, glucose and mitochondrial metabolism, as well as autophagy. In this review, we highlight current knowledge of the role of GSK3 in metabolic perturbations in cancer.
Collapse
Affiliation(s)
- David Papadopoli
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, 5100 Maisonneuve Blvd West, Montréal, QC H4A 3T2, Canada.
| | - Michael Pollak
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, 5100 Maisonneuve Blvd West, Montréal, QC H4A 3T2, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Décarie Blvd, Montréal, QC H4A 3J1, Canada
| | - Ivan Topisirovic
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, 5100 Maisonneuve Blvd West, Montréal, QC H4A 3T2, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Décarie Blvd, Montréal, QC H4A 3J1, Canada; Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC H3G 1Y6, Canada
| |
Collapse
|
106
|
Welz B, Bikker R, Hoffmeister L, Diekmann M, Christmann M, Brand K, Huber R. Activation of GSK3 Prevents Termination of TNF-Induced Signaling. J Inflamm Res 2021; 14:1717-1730. [PMID: 33986607 PMCID: PMC8111165 DOI: 10.2147/jir.s300806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Background Termination of TNF-induced signaling plays a key role in the resolution of inflammation with dysregulations leading to severe pathophysiological conditions (sepsis, chronic inflammatory disease, cancer). Since a recent phospho-proteome analysis in human monocytes suggested GSK3 as a relevant kinase during signal termination, we aimed at further elucidating its role in this context. Materials and Methods For the analyses, THP-1 monocytic cells and primary human monocytes were used. Staurosporine (Stauro) was applied to activate GSK3 by inhibiting kinases that mediate inhibitory GSK3α/β-Ser21/9 phosphorylation (eg, PKC). For GSK3 inhibition, Kenpaulone (Ken) was used. GSK3- and PKC-siRNAs were applied for knockdown experiments. Protein expression and phosphorylation were assessed by Western blot or ELISA and mRNA expression by qPCR. NF-κB activation was addressed using reporter gene assays. Results Constitutive GSK3β and PKCβ expression and GSK3α/β-Ser21/9 and PKCα/βII-Thr638/641 phosphorylation were not altered during TNF long-term incubation. Stauro-induced GSK3 activation (demonstrated by Bcl3 reduction) prevented termination of TNF-induced signaling as reflected by strongly elevated IL-8 expression (used as an indicator) following TNF long-term incubation. A similar increase was observed in TNF short-term-exposed cells, and this effect was inhibited by Ken. PKCα/β-knockdown modestly increased, whereas GSK3α/β-knockdown inhibited TNF-induced IL-8 expression. TNF-dependent activation of two NF-κB-dependent indicator plasmids was enhanced by Stauro, demonstrating transcriptional effects. A TNF-induced increase in p65-Ser536 phosphorylation was further enhanced by Stauro, whereas IκBα proteolysis and IKKα/β-Ser176/180 phosphorylation were not affected. Moreover, PKCβ-knockdown reduced levels of Bcl3. A20 and IκBα mRNA, both coding for signaling inhibitors, were dramatically less affected under our conditions when compared to IL-8, suggesting differential transcriptional effects. Conclusion Our results suggest that GSK3 activation is involved in preventing the termination of TNF-induced signaling. Our data demonstrate that activation of GSK3 – either pathophysiologically or pharmacologically induced – may destroy the finely balanced condition necessary for the termination of inflammation-associated signaling.
Collapse
Affiliation(s)
- Bastian Welz
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Rolf Bikker
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Leonie Hoffmeister
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Mareike Diekmann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Martin Christmann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Korbinian Brand
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - René Huber
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| |
Collapse
|
107
|
Lee EJ, Neppl RL. Influence of Age on Skeletal Muscle Hypertrophy and Atrophy Signaling: Established Paradigms and Unexpected Links. Genes (Basel) 2021; 12:genes12050688. [PMID: 34063658 PMCID: PMC8147613 DOI: 10.3390/genes12050688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle atrophy in an inevitable occurrence with advancing age, and a consequence of disease including cancer. Muscle atrophy in the elderly is managed by a regimen of resistance exercise and increased protein intake. Understanding the signaling that regulates muscle mass may identify potential therapeutic targets for the prevention and reversal of muscle atrophy in metabolic and neuromuscular diseases. This review covers the major anabolic and catabolic pathways that regulate skeletal muscle mass, with a focus on recent progress and potential new players.
Collapse
|
108
|
Wisessaowapak C, Visitnonthachai D, Watcharasit P, Satayavivad J. Prolonged arsenic exposure increases tau phosphorylation in differentiated SH-SY5Y cells: The contribution of GSK3 and ERK1/2. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 84:103626. [PMID: 33621689 DOI: 10.1016/j.etap.2021.103626] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/19/2021] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
Arsenic is a metalloid that has been hypothesized to be an environmental risk factor for Alzheimer's disease (AD), a disease having hyperphosphorylated tau aggregate as a marker. The present study demonstrated that prolonged exposure to sodium arsenite at low micromolar range (1-10 μM) reduced Tau 1 (recognizing dephosphorylated tau at residues 189-207) and elevated pS202 tau in differentiated human neuroblastoma SH-SY5Y cells indicating that arsenic increases tau phosphorylation in neurons. Sodium arsenite elevated GSK3β kinase activity, while GSK3 inhibitors, BIO, SB216763, and lithium, reversed the Tau 1 reduction by sodium arsenite. Additionally, sodium arsenite increased levels of active phosphorylation of ERK1/2, and inhibition of ERK1/2 by U0126 partially improved the Tau1 reduction. These results suggest that arsenic may cause tau hyperphosphorylation in neurons through the activation of GSK3 and ERK1/2. Furthermore, sodium arsenite augmented tau phosphorylation in the membrane and cytosolic fractions. Inductions of GSK3 activity by sodium arsenite treatment were observed in the membrane fraction, as evidenced by a reduction of β-catenin, a protein signaled for degradation following phosphorylation by GSK3. An enhancement of ERK1/2 phosphorylation by sodium arsenite was also witnessed in the cytosol. Additionally, sodium arsenite increased insoluble tau aggregation. These results suggest that arsenic induces tau hyperphosphorylation in the membrane fraction which may lead to its redistribution from the membrane fraction to the cytosol, where it promotes neurofibrillary formation. Collectively, we demonstrate that prolonged arsenic exposure increases tau phosphorylation, partly through GSK3 and ERK1/2 activation, and insoluble tau aggregates, hence possibly contributing to the development of sporadic AD.
Collapse
Affiliation(s)
- Churaibhon Wisessaowapak
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Environmental Toxicology Program, Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok, 10210, Thailand
| | | | - Piyajit Watcharasit
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Environmental Toxicology Program, Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok, 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Ministry of Higher Education Science Research and Innovation, Thailand.
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Environmental Toxicology Program, Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok, 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Ministry of Higher Education Science Research and Innovation, Thailand
| |
Collapse
|
109
|
Kobar K, Collett K, Prykhozhij SV, Berman JN. Zebrafish Cancer Predisposition Models. Front Cell Dev Biol 2021; 9:660069. [PMID: 33987182 PMCID: PMC8112447 DOI: 10.3389/fcell.2021.660069] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer predisposition syndromes are rare, typically monogenic disorders that result from germline mutations that increase the likelihood of developing cancer. Although these disorders are individually rare, resulting cancers collectively represent 5-10% of all malignancies. In addition to a greater incidence of cancer, affected individuals have an earlier tumor onset and are frequently subjected to long-term multi-modal cancer screening protocols for earlier detection and initiation of treatment. In vivo models are needed to better understand tumor-driving mechanisms, tailor patient screening approaches and develop targeted therapies to improve patient care and disease prognosis. The zebrafish (Danio rerio) has emerged as a robust model for cancer research due to its high fecundity, time- and cost-efficient genetic manipulation and real-time high-resolution imaging. Tumors developing in zebrafish cancer models are histologically and molecularly similar to their human counterparts, confirming the validity of these models. The zebrafish platform supports both large-scale random mutagenesis screens to identify potential candidate/modifier genes and recently optimized genome editing strategies. These techniques have greatly increased our ability to investigate the impact of certain mutations and how these lesions impact tumorigenesis and disease phenotype. These unique characteristics position the zebrafish as a powerful in vivo tool to model cancer predisposition syndromes and as such, several have already been created, including those recapitulating Li-Fraumeni syndrome, familial adenomatous polyposis, RASopathies, inherited bone marrow failure syndromes, and several other pathogenic mutations in cancer predisposition genes. In addition, the zebrafish platform supports medium- to high-throughput preclinical drug screening to identify compounds that may represent novel treatment paradigms or even prevent cancer evolution. This review will highlight and synthesize the findings from zebrafish cancer predisposition models created to date. We will discuss emerging trends in how these zebrafish cancer models can improve our understanding of the genetic mechanisms driving cancer predisposition and their potential to discover therapeutic and/or preventative compounds that change the natural history of disease for these vulnerable children, youth and adults.
Collapse
Affiliation(s)
- Kim Kobar
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Keon Collett
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | | | - Jason N. Berman
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
110
|
Xu T, Wang S, Li X, Li X, Qu K, Tong H, Zhang R, Bai S, Fan J. Lithium chloride represses abdominal aortic aneurysm via regulating GSK3β/SIRT1/NF-κB signaling pathway. Free Radic Biol Med 2021; 166:1-10. [PMID: 33588051 DOI: 10.1016/j.freeradbiomed.2021.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/14/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
Lithium chloride (LiCl), a pharmacological compound, was effective in reducing inflammation, but whether it can protect against abdominal aortic aneurysm (AAA) is largely unknown. This study is designed to investigate therapeutic effects of LiCl on AAA and the potential mechanism. Rat AAA models were induced by periaortic application of CaCl2. AAA rats were treated by daily intraperitoneal injection of LiCl or vehicle alone to study the protection effects of LiCl in vivo. Rat primary vascular smooth muscle cells (VSMCs) stimulated with tumor necrosis factor (TNF)-α served as an in vitro model. LiCl treatment prevented the development of AAA through inhibiting the inflammatory cells infiltration and inflammatory cytokines overproduction, as well as attenuating superoxide production and elastin degradation in aorta of AAA rats. Additionally, the downregulation of p-GSK3β(Ser9) and SIRT1, upregulation of NF-κB(p-65), MMP-2 and MMP-9 in AAA were abolished by LiCl treatment. In vitro by upregulating p-GSK3β(Ser9), LiCl significantly induced SIRT1 expression, along with inhibition of the NF-κB activation and decreased elastin level elicited in VSMCs by TNF-α stimulation. SIRT1 activator SRT1720 achieved similar repressive effects as LiCl on TNF-α-induced NF-κB activation and decreased elastin in VSMCs. Moreover, administration of LiCl also caused regression of established rats AAA. This study provided the first evidence that LiCl prevented the development of AAA through inhibiting inflammation, MMPs, and superoxide production, and facilitating the biosynthesis of elastin. The beneficial effect of LiCl may be mediated by regulation GSK3β/SIRT1/NF-κB cascade.
Collapse
Affiliation(s)
- Tong Xu
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Shoushuai Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xiang Li
- Department of Cell Biology, Key Laboratory of Cell Biology of National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology of Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xiuquan Li
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Kaiyun Qu
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hao Tong
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Ruijie Zhang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Shuling Bai
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Jun Fan
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China.
| |
Collapse
|
111
|
The PKA-p38MAPK-NFAT5-Organic Osmolytes Pathway in Duchenne Muscular Dystrophy: From Essential Player in Osmotic Homeostasis, Inflammation and Skeletal Muscle Regeneration to Therapeutic Target. Biomedicines 2021; 9:biomedicines9040350. [PMID: 33808305 PMCID: PMC8066813 DOI: 10.3390/biomedicines9040350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 11/30/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), the absence of dystrophin from the dystrophin-associated protein complex (DAPC) causes muscle membrane instability, which leads to myofiber necrosis, hampered regeneration, and chronic inflammation. The resulting disabled DAPC-associated cellular pathways have been described both at the molecular and the therapeutical level, with the Toll-like receptor nuclear factor kappa-light-chain-enhancer of activated B cells pathway (NF-ƘB), Janus kinase/signal transducer and activator of transcription proteins, and the transforming growth factor-β pathways receiving the most attention. In this review, we specifically focus on the protein kinase A/ mitogen-activated protein kinase/nuclear factor of activated T-cells 5/organic osmolytes (PKA-p38MAPK-NFAT5-organic osmolytes) pathway. This pathway plays an important role in osmotic homeostasis essential to normal cell physiology via its regulation of the influx/efflux of organic osmolytes. Besides, NFAT5 plays an essential role in cell survival under hyperosmolar conditions, in skeletal muscle regeneration, and in tissue inflammation, closely interacting with the master regulator of inflammation NF-ƘB. We describe the involvement of the PKA-p38MAPK-NFAT5-organic osmolytes pathway in DMD pathophysiology and provide a clear overview of which therapeutic molecules could be of potential benefit to DMD patients. We conclude that modulation of the PKA-p38MAPK-NFAT5-organic osmolytes pathway could be developed as supportive treatment for DMD in conjunction with genetic therapy.
Collapse
|
112
|
Sayas CL, Ávila J. GSK-3 and Tau: A Key Duet in Alzheimer's Disease. Cells 2021; 10:721. [PMID: 33804962 PMCID: PMC8063930 DOI: 10.3390/cells10040721] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 02/07/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a ubiquitously expressed serine/threonine kinase with a plethora of substrates. As a modulator of several cellular processes, GSK-3 has a central position in cell metabolism and signaling, with important roles both in physiological and pathological conditions. GSK-3 has been associated with a number of human disorders, such as neurodegenerative diseases including Alzheimer's disease (AD). GSK-3 contributes to the hyperphosphorylation of tau protein, the main component of neurofibrillary tangles (NFTs), one of the hallmarks of AD. GSK-3 is further involved in the regulation of different neuronal processes that are dysregulated during AD pathogenesis, such as the generation of amyloid-β (Aβ) peptide or Aβ-induced cell death, axonal transport, cholinergic function, and adult neurogenesis or synaptic function. In this review, we will summarize recent data about GSK-3 involvement in these processes contributing to AD pathology, mostly focusing on the crucial interplay between GSK-3 and tau protein. We further discuss the current development of potential AD therapies targeting GSK-3 or GSK-3-phosphorylated tau.
Collapse
Affiliation(s)
- Carmen Laura Sayas
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), 38200 Tenerife, Spain
| | - Jesús Ávila
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC) y la Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, 28031 Madrid, Spain
| |
Collapse
|
113
|
Rizk M, Saker Z, Harati H, Fares Y, Bahmad HF, Nabha S. Deciphering the roles of glycogen synthase kinase 3 (GSK3) in the treatment of autism spectrum disorder and related syndromes. Mol Biol Rep 2021; 48:2669-2686. [PMID: 33650079 DOI: 10.1007/s11033-021-06237-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Autism spectrum disorder (ASD) is a complex and multifactorial neurodevelopmental disorder characterized by the presence of restricted interests and repetitive behaviors besides deficits in social communication. Syndromic ASD is a subset of ASD caused by underlying genetic disorders, most commonly Fragile X Syndrome (FXS) and Rett Syndrome (RTT). Various mutations and consequent malfunctions in core signaling pathways have been identified in ASD, including glycogen synthase kinase 3 (GSK3). A growing body of evidence suggests a key role of GSK3 dysregulation in the pathogenesis of ASD and its related disorders. Here, we provide a synopsis of the implication of GSK3 in ASD, FXS, and RTT as a promising therapeutic target for the treatment of ASD.
Collapse
Affiliation(s)
- Mahdi Rizk
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.,Department of Neurosurgery, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA
| | - Sanaa Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
114
|
Xiong Y, Ju L, Yuan L, Chen L, Wang G, Xu H, Peng T, Luo Y, Xiao Y, Wang X. KNSTRN promotes tumorigenesis and gemcitabine resistance by activating AKT in bladder cancer. Oncogene 2021; 40:1595-1608. [PMID: 33452459 DOI: 10.1038/s41388-020-01634-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 01/29/2023]
Abstract
KNSTRN is a component of the mitotic spindle, which was rarely investigated in tumorigenesis. AKT plays an essential role in tumorigenesis by modulating the phosphorylation of various substrates. The activation of AKT is regulated by PTEN and PIP3. Here, we prove KNSTRN is positively correlated with malignancy of bladder cancer and KNSTRN activates AKT phosphorylation at Thr308 and Ser473. More importantly, our study reveals that both KNSTRN and PTEN interact with PH domain of AKT at cell membrane. The amount of KNSTRN interacted with AKT is negatively related to PTEN. Furthermore, PIP3 pull-down assay proves that KNSTRN promoted AKT movement to PIP3. These data suggest KNSTRN may activate AKT phosphorylation by promoting AKT movement to PIP3 and alleviating PTEN suppression. Based on the activation of AKT phosphorylation, our study demonstrates that KNSTRN promotes bladder cancer metastasis and gemcitabine resistance in vitro and in vivo. Meanwhile, the effect of KNSTRN on tumorigenesis and gemcitabine resistance could be restored by AKT specific inhibitor MK2206 or AKT overexpression. In conclusion, we identify an oncogene KNSTRN that promotes tumorigenesis and gemcitabine resistance by activating AKT phosphorylation and may serve as a therapeutic target in bladder cancer.
Collapse
Affiliation(s)
- Yaoyi Xiong
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetic Resource Preservation Center of Hubei Province, Wuhan, China.,Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lushun Yuan
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetic Resource Preservation Center of Hubei Province, Wuhan, China.,Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Huimin Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianchen Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yongwen Luo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China. .,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China. .,Human Genetic Resource Preservation Center of Hubei Province, Wuhan, China. .,Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China. .,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China. .,Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China. .,Medical Research Institute, Wuhan University, Wuhan, China.
| |
Collapse
|
115
|
Patel S, Werstuck GH. Macrophage Function and the Role of GSK3. Int J Mol Sci 2021; 22:ijms22042206. [PMID: 33672232 PMCID: PMC7926541 DOI: 10.3390/ijms22042206] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
Macrophages are present in nearly all vertebrate tissues, where they respond to a complex variety of regulatory signals to coordinate immune functions involved in tissue development, metabolism, homeostasis, and repair. Glycogen synthase kinase 3 (GSK3) is a ubiquitously expressed protein kinase that plays important roles in multiple pathways involved in cell metabolism. Dysregulation of GSK3 has been implicated in several prevalent metabolic disorders, and recent findings have highlighted the importance of GSK3 activity in the regulation of macrophages, especially with respect to the initiation of specific pathologies. This makes GSK3 a potential therapeutic target for the development of novel drugs to modulate immunometabolic responses. Here, we summarize recent findings that have contributed to our understanding of how GSK3 regulates macrophage function, and we discuss the role of GSK3 in the development of metabolic disorders and diseases.
Collapse
Affiliation(s)
- Sarvatit Patel
- Thrombosis and Atherosclerosis Research Institute, 237 Barton Street E, Hamilton, ON L9L 2X2, Canada;
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Geoff H. Werstuck
- Thrombosis and Atherosclerosis Research Institute, 237 Barton Street E, Hamilton, ON L9L 2X2, Canada;
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
- Correspondence: ; Tel.: +1-905-521-2100 (ext. 40747)
| |
Collapse
|
116
|
McKenna M, Balasuriya N, Zhong S, Li SSC, O'Donoghue P. Phospho-Form Specific Substrates of Protein Kinase B (AKT1). Front Bioeng Biotechnol 2021; 8:619252. [PMID: 33614606 PMCID: PMC7886700 DOI: 10.3389/fbioe.2020.619252] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/29/2020] [Indexed: 11/17/2022] Open
Abstract
Protein kinase B (AKT1) is hyper-activated in diverse human tumors. AKT1 is activated by phosphorylation at two key regulatory sites, Thr308 and Ser473. Active AKT1 phosphorylates many, perhaps hundreds, of downstream cellular targets in the cytosol and nucleus. AKT1 is well-known for phosphorylating proteins that regulate cell survival and apoptosis, however, the full catalog of AKT1 substrates remains unknown. Using peptide arrays, we recently discovered that each phosphorylated form of AKT1 (pAKT1S473, pAKT1T308, and ppAKT1S473,T308) has a distinct substrate specificity, and these data were used to predict potential new AKT1 substrates. To test the high-confidence predictions, we synthesized target peptides representing putative AKT1 substrates. Peptides substrates were synthesized by solid phase synthesis and their purity was confirmed by mass spectrometry. Most of the predicted peptides showed phosphate accepting activity similar to or greater than that observed with a peptide derived from a well-established AKT1 substrate, glycogen synthase kinase 3β (GSK-3β). Among the novel substrates, AKT1 was most active with peptides representing PIP3-binding protein Rab11 family-interacting protein 2 and cysteinyl leukotriene receptor 1, indicating their potential role in AKT1-dependent cellular signaling. The ppAKT1S473,T308 enzyme was highly selective for peptides containing a patch of basic residues at −5, −4, −3 and aromatic residues (Phe/Tyr) at +1 positions from the phosphorylation site. The pAKT1S473 variant preferred more acidic peptides, Ser or Pro at +4, and was agnostic to the residue at −5. The data further support our hypothesis that Ser473 phosphorylation plays a key role in modulating AKT1 substrate selectivity.
Collapse
Affiliation(s)
- McShane McKenna
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Nileeka Balasuriya
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Shanshan Zhong
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Shawn Shun-Cheng Li
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Patrick O'Donoghue
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada.,Department of Chemistry, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
117
|
Zhang N, Tian YN, Zhou LN, Li MZ, Chen HD, Song SS, Huan XJ, Bao XB, Zhang A, Miao ZH, He JX. Glycogen synthase kinase 3β inhibition synergizes with PARP inhibitors through the induction of homologous recombination deficiency in colorectal cancer. Cell Death Dis 2021; 12:183. [PMID: 33589588 PMCID: PMC7884722 DOI: 10.1038/s41419-021-03475-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/17/2022]
Abstract
Monotherapy with poly ADP-ribose polymerase (PARP) inhibitors results in a limited objective response rate (≤60% in most cases) in patients with homologous recombination repair (HRR)-deficient cancer, which suggests a high rate of resistance in this subset of patients to PARP inhibitors (PARPi). To overcome resistance to PARPi and to broaden their clinical use, we performed high-throughput screening of 99 anticancer drugs in combination with PARPi to identify potential therapeutic combinations. Here, we found that GSK3 inhibitors (GSK3i) exhibited a strong synergistic effect with PARPi in a panel of colorectal cancer (CRC) cell lines with diverse genetic backgrounds. The combination of GSK3β and PARP inhibition causes replication stress and DNA double-strand breaks, resulting in increased anaphase bridges and abnormal spindles. Mechanistically, inhibition or genetic depletion of GSK3β was found to impair the HRR of DNA and reduce the mRNA and protein level of BRCA1. Finally, we demonstrated that inhibition or depletion of GSK3β could enhance the in vivo sensitivity to simmiparib without toxicity. Our results provide a mechanistic understanding of the combination of PARP and GSK3 inhibition, and support the clinical development of this combination therapy for CRC patients.
Collapse
Affiliation(s)
- Ning Zhang
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Yu-Nan Tian
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Li-Na Zhou
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Meng-Zhu Li
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Hua-Dong Chen
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Shan-Shan Song
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Xia-Juan Huan
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Xu-Bin Bao
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Ao Zhang
- Department of Medicinal Chemistry, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ze-Hong Miao
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| | - Jin-Xue He
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
118
|
Snitow ME, Bhansali RS, Klein PS. Lithium and Therapeutic Targeting of GSK-3. Cells 2021; 10:255. [PMID: 33525562 PMCID: PMC7910927 DOI: 10.3390/cells10020255] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Lithium salts have been in the therapeutic toolbox for better or worse since the 19th century, with purported benefit in gout, hangover, insomnia, and early suggestions that lithium improved psychiatric disorders. However, the remarkable effects of lithium reported by John Cade and subsequently by Mogens Schou revolutionized the treatment of bipolar disorder. The known molecular targets of lithium are surprisingly few and include the signaling kinase glycogen synthase kinase-3 (GSK-3), a group of structurally related phosphomonoesterases that includes inositol monophosphatases, and phosphoglucomutase. Here we present a brief history of the therapeutic uses of lithium and then focus on GSK-3 as a therapeutic target in diverse diseases, including bipolar disorder, cancer, and coronavirus infections.
Collapse
Affiliation(s)
| | | | - Peter S. Klein
- Department of Medicine, Perelman School of Medicine,
University of Pennsylvania, 3400 Spruce St., Philadelphia, PA 19104, USA; (M.E.S.); (R.S.B.)
| |
Collapse
|
119
|
Zhong Y, Yang S, Cui J, Wang J, Li L, Chen Y, Chen J, Feng P, Huang S, Li H, Han Y, Tang G, Hu K. Novel 18F-Labeled Isonicotinamide-Based Radioligands for Positron Emission Tomography Imaging of Glycogen Synthase Kinase-3β. Mol Pharm 2021; 18:1277-1284. [PMID: 33492962 DOI: 10.1021/acs.molpharmaceut.0c01133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Glycogen synthase kinase-3β (GSK-3β), a cytoplasmic serine/threonine protein kinase, is involved in several human pathologies including Alzheimer's disease, bipolar disorder, diabetes, and cancer. Positron emission tomography (PET) imaging of GSK-3β could aid in investigating GSK-3β levels under normal and pathological conditions. In this study, we designed and synthesized fluorinated PET radioligands starting with recently identified isonicotinamide derivatives that showed potent affinity to GSK-3β. After extensive in vitro inhibitory activity assays and analyzing U87 cell uptake, we identified [18F]10a-d as potential tracers with good specificity and high affinity. They were then subjected to further in vivo evaluation in rodent brain comprising PET imaging and metabolism studies. The radioligands [18F]10b-d penetrated the blood-brain barrier and accumulated in GSK-3β-rich regions, including amygdala, cerebellum, and hippocampus. Also, it could be specifically blocked using the corresponding standard compounds. With these results, this work sets the basis for further development of novel 18F-labeled GSK-3β PET probes.
Collapse
Affiliation(s)
- Yuhua Zhong
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Shaoxi Yang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Jianyu Cui
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Jie Wang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Lin Li
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Yilin Chen
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Junjie Chen
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Pengju Feng
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Shun Huang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Hongsheng Li
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Yanjian Han
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Ganghua Tang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Kongzhen Hu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| |
Collapse
|
120
|
Draffin JE, Sánchez‐Castillo C, Fernández‐Rodrigo A, Sánchez‐Sáez X, Ávila J, Wagner FF, Esteban JA. GSK3α, not GSK3β, drives hippocampal NMDAR-dependent LTD via tau-mediated spine anchoring. EMBO J 2021; 40:e105513. [PMID: 33197065 PMCID: PMC7809792 DOI: 10.15252/embj.2020105513] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 11/09/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK3) is an important signalling protein in the brain and modulates different forms of synaptic plasticity. Neuronal functions of GSK3 are typically attributed to one of its two isoforms, GSK3β, simply because of its prevalent expression in the brain. Consequently, the importance of isoform-specific functions of GSK3 in synaptic plasticity has not been fully explored. We now directly address this question for NMDA receptor-dependent long-term depression (LTD) in the hippocampus. Here, we specifically target the GSK3 isoforms with shRNA knock-down in mouse hippocampus and with novel isoform-selective drugs to dissect their roles in LTD. Using electrophysiological and live imaging approaches, we find that GSK3α, but not GSK3β, is required for LTD. The specific engagement of GSK3α occurs via its transient anchoring in dendritic spines during LTD induction. We find that the major GSK3 substrate, the microtubule-binding protein tau, is required for this spine anchoring of GSK3α and mediates GSK3α-induced LTD. These results link GSK3α and tau in a common mechanism for synaptic depression and rule out a major role for GSK3β in this process.
Collapse
Affiliation(s)
- Jonathan E Draffin
- Centro de Biología Molecular Severo OchoaCSIC‐Universidad Autónoma de MadridMadridSpain
| | | | | | - Xavier Sánchez‐Sáez
- Centro de Biología Molecular Severo OchoaCSIC‐Universidad Autónoma de MadridMadridSpain
- Present address:
Department of Physiology, Genetics and MicrobiologyUniversity of AlicanteAlicanteSpain
| | - Jesús Ávila
- Centro de Biología Molecular Severo OchoaCSIC‐Universidad Autónoma de MadridMadridSpain
| | - Florence F Wagner
- Stanley Center for Psychiatric ResearchBroad Institute of Massachusetts Institute of Technology and Harvard UniversityCambridgeMAUSA
| | - José A Esteban
- Centro de Biología Molecular Severo OchoaCSIC‐Universidad Autónoma de MadridMadridSpain
| |
Collapse
|
121
|
Chen L, Zuo Y, Pan R, Ye Z, Wei K, Xia S, Li W, Tan J, Xia X. GSK-3β Regulates the Expression of P21 to Promote the Progression of Chordoma. Cancer Manag Res 2021; 13:201-214. [PMID: 33469364 PMCID: PMC7810826 DOI: 10.2147/cmar.s289883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/19/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Chordoma is a rare malignant bone tumor transformed from the remnants of notochord. It is characterized as highly aggressive and locally invasive, difficult to be completely removed by surgery, and has a poor clinical prognosis. Glycogen synthase kinase 3 beta (GSK-3β) is involved in many cellular processes. GSK-3β overexpression has been shown to promote the development of many cancers, according to previous studies. However, the role of GSK-3β in chordoma remains unclear. Methods Immunohistochemistry (IHC) and Western blotting (WB) were performed on clinical specimens to measure GSK-3β expression in chordoma, and immunofluorescence and quantitative real-time polymerase chain reaction (QRT-PCR) were performed to examine the expression of GSK-3β and P21 in cell lines. Cell proliferation was detected by the CCK-8 assay and colony formation analysis, cell migration and invasion checked by Transwell experiments, and cell apoptosis was determined by Annexin V/propidium iodide staining. P21 was predicted as a downstream target gene of GSK-3β using STRING and UNIHI databases. Moreover, we used immunoprecipitation to confirm that GSK-3β and P21 interacted with each other. The double luciferase reporter gene assay showed that GSK-3β could regulate the promoter activity of P21. Finally, the role of the GSK-3β -P21 pathway in chordoma tumorigenesis was analyzed in vivo in nude mice. Results Our study showed that GSK-3β was significantly higher in chordoma tissues than in paracancer tissues, and siRNA knockdown of GSK-3β inhibited chordoma cell proliferation and promoted cell apoptosis. Additionally, our research found that GSK-3β bound and downregulated the expression of the P21 gene, and the expression of silencing P21 partially reversed the inhibitory effect of knockdown GSK-3β on chordoma. Furthermore, xenografts showed that knockdown GSK-3β inhibited the formation of chordomas in vivo. Conclusion Our results indicated that the GSK-3β-P21 axis may be an important signaling pathway for the occurrence and development of chordoma, providing a new therapeutic target for the clinical treatment of this disorder.
Collapse
Affiliation(s)
- Li Chen
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi 541004, People's Republic of China
| | - Yi Zuo
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi 541004, People's Republic of China
| | - Ru Pan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi 541004, People's Republic of China
| | - Zhen Ye
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi 541004, People's Republic of China
| | - Kailun Wei
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi 541004, People's Republic of China
| | - Shaohuai Xia
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Wencai Li
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi 541004, People's Republic of China
| | - Xuewei Xia
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin, Guangxi 541004, People's Republic of China
| |
Collapse
|
122
|
|
123
|
Guerrero-Hue M, Rayego-Mateos S, Vázquez-Carballo C, Palomino-Antolín A, García-Caballero C, Opazo-Rios L, Morgado-Pascual JL, Herencia C, Mas S, Ortiz A, Rubio-Navarro A, Egea J, Villalba JM, Egido J, Moreno JA. Protective Role of Nrf2 in Renal Disease. Antioxidants (Basel) 2020; 10:antiox10010039. [PMID: 33396350 PMCID: PMC7824104 DOI: 10.3390/antiox10010039] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the fastest-growing causes of death and is predicted to become by 2040 the fifth global cause of death. CKD is characterized by increased oxidative stress and chronic inflammation. However, therapies to slow or prevent CKD progression remain an unmet need. Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor that plays a key role in protection against oxidative stress and regulation of the inflammatory response. Consequently, the use of compounds targeting Nrf2 has generated growing interest for nephrologists. Pre-clinical and clinical studies have demonstrated that Nrf2-inducing strategies prevent CKD progression and protect from acute kidney injury (AKI). In this article, we review current knowledge on the protective mechanisms mediated by Nrf2 against kidney injury, novel therapeutic strategies to induce Nrf2 activation, and the status of ongoing clinical trials targeting Nrf2 in renal diseases.
Collapse
Affiliation(s)
- Melania Guerrero-Hue
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Sandra Rayego-Mateos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Cristina Vázquez-Carballo
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
| | - Alejandra Palomino-Antolín
- Research Unit, Hospital Universitario Santa Cristina, IIS-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (A.P.-A.); (J.E.)
- Departament of Pharmacology and Therapeutics, Medicine Faculty, Instituto Teófilo Hernando, Autónoma University, 28029 Madrid, Spain
| | - Cristina García-Caballero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Lucas Opazo-Rios
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - José Luis Morgado-Pascual
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Carmen Herencia
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
| | - Sebastián Mas
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Red Nacional Investigaciones Nefrológicas (REDINREN), 28040 Madrid, Spain
| | - Alfonso Rubio-Navarro
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Javier Egea
- Research Unit, Hospital Universitario Santa Cristina, IIS-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (A.P.-A.); (J.E.)
- Departament of Pharmacology and Therapeutics, Medicine Faculty, Instituto Teófilo Hernando, Autónoma University, 28029 Madrid, Spain
| | - José Manuel Villalba
- Department of Cell Biology, Physiology, and Immunology, Agrifood Campus of International Excellence (ceiA3), University of Cordoba, 14014 Cordoba, Spain;
| | - Jesús Egido
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
- Department of Cell Biology, Physiology, and Immunology, Agrifood Campus of International Excellence (ceiA3), University of Cordoba, 14014 Cordoba, Spain;
- Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
- Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-957-218-039
| |
Collapse
|
124
|
Viana GSB, Vale EMD, Araujo ARAD, Coelho NC, Andrade SM, Costa ROD, Aquino PEAD, Sousa CNSD, Medeiros ISD, Vasconcelos SMMD, Neves KRT. Rapid and long-lasting antidepressant-like effects of ketamine and their relationship with the expression of brain enzymes, BDNF, and astrocytes. ACTA ACUST UNITED AC 2020; 54:e10107. [PMID: 33331415 PMCID: PMC7747878 DOI: 10.1590/1414-431x202010107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/28/2020] [Indexed: 12/28/2022]
Abstract
Ketamine (KET) is an N-methyl-D-aspartate (NMDA) antagonist with rapid and long-lasting antidepressant effects, but how the drug shows its sustained effects is still a matter of controversy. The objectives were to evaluate the mechanisms for KET rapid (30 min) and long-lasting (15 and 30 days after) antidepressant effects in mice. A single dose of KET (2, 5, or 10 mg/kg, po) was administered to male Swiss mice and the forced swim test (FST) was performed 30 min, 15, or 30 days later. Imipramine (IMI, 30 mg/kg, ip), a tricyclic antidepressant drug, was used as reference. The mice were euthanized, separated into two time-point groups (D1, first day after KET injection; D30, 30 days later), and brain sections were processed for glycogen synthase kinase-3 (GSK-3), histone deacetylase (HDAC), brain-derived neurotrophic factor (BDNF), and glial fibrillary acidic protein (GFAP) immunohistochemical assays. KET (5 and 10 mg/kg) presented rapid and long-lasting antidepressant-like effects. As expected, the immunoreactivities for brain GSK-3 and HDAC decreased compared to control groups in all areas (striatum, DG, CA1, CA3, and mainly pre-frontal cortex, PFC) after KET injection. Increases in BDNF immunostaining were demonstrated in the PFC, DG, CA1, and CA3 areas at D1 and D30 time-points. GFAP immunoreactivity was also increased in the PFC and striatum at both time-points. In conclusion, KET changed brain BDNF and GFAP expressions 30 days after a single administration. Although neuroplasticity could be involved in the observed effects of KET, more studies are needed to explain the mechanisms for the drug’s sustained antidepressant-like effects.
Collapse
Affiliation(s)
- G S B Viana
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - E M do Vale
- Departamento de Biofisiologia, Faculdade de Medicina Estácio de Juazeiro do Norte, Juazeiro do Norte, CE, Brasil
| | - A R A de Araujo
- Departamento de Biofisiologia, Faculdade de Medicina Estácio de Juazeiro do Norte, Juazeiro do Norte, CE, Brasil
| | - N C Coelho
- Departamento de Biofisiologia, Faculdade de Medicina Estácio de Juazeiro do Norte, Juazeiro do Norte, CE, Brasil
| | - S M Andrade
- Departamento de Biofisiologia, Faculdade de Medicina Estácio de Juazeiro do Norte, Juazeiro do Norte, CE, Brasil
| | - R O da Costa
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P E A de Aquino
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - C N S de Sousa
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - I S de Medeiros
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - S M M de Vasconcelos
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - K R T Neves
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
125
|
Ishikane S, Ikushima E, Igawa K, Tomooka K, Takahashi-Yanaga F. Differentiation-inducing factor-1 potentiates adipogenic differentiation and attenuates the osteogenic differentiation of bone marrow-derived mesenchymal stem cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118909. [PMID: 33189784 DOI: 10.1016/j.bbamcr.2020.118909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cells (MSCs) are an attractive cell source for tissue regeneration and repair. However, their low differentiation efficacy currently impedes the development of MSC therapy. Therefore, in this study, we investigated the effects of differentiation-inducing factor-1 (DIF-1) on the differentiation efficacy of bone marrow-derived MSCs (BM-MSCs) into adipogenic or osteogenic lineages. BM-MSCs, which were obtained from Sprague-Dawley rats, were positive for the MSC markers (CD29, CD73, and CD90). DIF-1 alone neither affected cell surface antigen expression nor induced adipogenic or osteogenic differentiation. However, DIF-1 significantly enhanced the effects of adipogenic differentiation stimuli, which were evaluated as the number of oil red-O positive cells and the expression of adipocyte differentiation markers (peroxisome proliferator-activated receptor gamma, adipocyte fatty acid-binding protein, and adiponectin). In contrast, DIF-1 significantly attenuated the effects of osteogenic differentiation stimuli, which were evaluated as alizarin red-S positive calcium deposition, and the expression of osteoblast differentiation markers alkaline phosphatase, runt-related transcription factor 2, and osteopontin. We further investigated the mechanism by which DIF-1 affects MSC differentiation efficacy and found that glycogen synthase kinase-3 was the main factor mediating the action of DIF-1 on the adipogenic differentiation of BM-MSCs, whereas it was only partially involved in osteogenic differentiation. These results suggest that DIF-1 supports MSC differentiation toward the desired cell fate by enhancing the differentiation efficacy.
Collapse
Affiliation(s)
- Shin Ishikane
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyusyu, Fukuoka 807-8555, Japan.
| | - Eigo Ikushima
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyusyu, Fukuoka 807-8555, Japan
| | - Kazunobu Igawa
- Department of Molecular and Material Science, Institute for Materials Chemistry and Engineering, Kyushu University, Chikushi Campus 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| | - Katsuhiko Tomooka
- Department of Molecular and Material Science, Institute for Materials Chemistry and Engineering, Kyushu University, Chikushi Campus 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| | - Fumi Takahashi-Yanaga
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyusyu, Fukuoka 807-8555, Japan
| |
Collapse
|
126
|
Esteban-Villarrubia J, Soto-Castillo JJ, Pozas J, San Román-Gil M, Orejana-Martín I, Torres-Jiménez J, Carrato A, Alonso-Gordoa T, Molina-Cerrillo J. Tyrosine Kinase Receptors in Oncology. Int J Mol Sci 2020; 21:E8529. [PMID: 33198314 PMCID: PMC7696731 DOI: 10.3390/ijms21228529] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Tyrosine kinase receptors (TKR) comprise more than 60 molecules that play an essential role in the molecular pathways, leading to cell survival and differentiation. Consequently, genetic alterations of TKRs may lead to tumorigenesis and, therefore, cancer development. The discovery and improvement of tyrosine kinase inhibitors (TKI) against TKRs have entailed an important step in the knowledge-expansion of tumor physiopathology as well as an improvement in the cancer treatment based on molecular alterations over many tumor types. The purpose of this review is to provide a comprehensive review of the different families of TKRs and their role in the expansion of tumor cells and how TKIs can stop these pathways to tumorigenesis, in combination or not with other therapies. The increasing growth of this landscape is driving us to strengthen the development of precision oncology with clinical trials based on molecular-based therapy over a histology-based one, with promising preliminary results.
Collapse
Affiliation(s)
- Jorge Esteban-Villarrubia
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Juan José Soto-Castillo
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Javier Pozas
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - María San Román-Gil
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Inmaculada Orejana-Martín
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Javier Torres-Jiménez
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Alfredo Carrato
- Medical Oncology Department, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (A.C.); (J.M.-C.)
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (A.C.); (J.M.-C.)
| | - Javier Molina-Cerrillo
- Medical Oncology Department, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (A.C.); (J.M.-C.)
| |
Collapse
|
127
|
Liu WL, Chiang FT, Kao JTW, Chiou SH, Lin HL. GSK3 modulation in acute lung injury, myocarditis and polycystic kidney disease-related aneurysm. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118798. [PMID: 32693109 PMCID: PMC7368652 DOI: 10.1016/j.bbamcr.2020.118798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/17/2022]
Abstract
GSK3 are involved in different physical and pathological conditions and inflammatory regulated by macrophages contribute to significant mechanism. Infection stimuli may modulate GSK3 activity and influence host cell adaption, immune cells infiltration or cytokine expressions. To further address the role of GSK3 modulation in macrophages, the signal transduction of three major organs challenged by endotoxin, virus and genetic inherited factors are briefly introduced (lung injury, myocarditis and autosomal dominant polycystic kidney disease). As a result of pro-inflammatory and anti-inflammatory functions of GSK3 in different microenvironments and stages of macrophages (M1/M2), the rational resolution should be considered by adequately GSK3.
Collapse
Affiliation(s)
- Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan,Division of Critical Care Medicine, Department of Emergency and Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan,Center For Innovation, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Fu-Tien Chiang
- Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan,Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Juliana Tze-Wah Kao
- Division of Nephrology, Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan,Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan,Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Heng-Liang Lin
- Center For Innovation, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan; Division of Fund Managing, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
128
|
Tung HYL, Limtung P. Mutations in the phosphorylation sites of SARS-CoV-2 encoded nucleocapsid protein and structure model of sequestration by protein 14-3-3. Biochem Biophys Res Commun 2020; 532:134-138. [PMID: 32829876 PMCID: PMC7428706 DOI: 10.1016/j.bbrc.2020.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/11/2020] [Indexed: 02/09/2023]
Abstract
SARS-CoV-2 is the etiologic agent of COVID-19. There is currently no effective means of preventing infections by SARS-CoV-2, except through restriction of population movement and contact. An understanding of the origin, evolution and biochemistry (molecular biology) of SARS-CoV-2 is a prerequisite to its control. Mutations in the phosphorylation sites of SARS-CoV-2 encoded nucleocapsid protein isolated from various populations and locations, are described. Mutations occurred in the phosphorylation sites, all located within a stretch which forms a phosphorylation dependent interaction site, including C-TAK1 phosphorylation sites for 14-3-3. The consequences of these mutations are discussed and a structure-based model for the role of protein 14-3-3 in the sequestration and inhibition of SARS-CoV-2 nucleocapsid protein's function is presented. It is proposed that the phosphorylation of SARS-CoV-2 nucleocapsid protein and its sequestration by Protein 14-3-3 is a cellular response mechanism for the control and inhibition of the replication, transcription and packaging of the SARS-CoV-2 genome.
Collapse
Affiliation(s)
- H Y Lim Tung
- Peptide and Protein Chemistry Research Laboratory, Nacbraht Biomedical Research Institute, 3164 21st Street Suite 122, Astoria (NYC), NY, 11106, USA.
| | - Pierre Limtung
- Peptide and Protein Chemistry Research Laboratory, Nacbraht Biomedical Research Institute, 3164 21st Street Suite 122, Astoria (NYC), NY, 11106, USA
| |
Collapse
|
129
|
Andreev S, Pantsar T, El-Gokha A, Ansideri F, Kudolo M, Anton DB, Sita G, Romasco J, Geibel C, Lämmerhofer M, Goettert MI, Tarozzi A, Laufer SA, Koch P. Discovery and Evaluation of Enantiopure 9 H-pyrimido[4,5- b]indoles as Nanomolar GSK-3β Inhibitors with Improved Metabolic Stability. Int J Mol Sci 2020; 21:ijms21217823. [PMID: 33105671 PMCID: PMC7659979 DOI: 10.3390/ijms21217823] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 12/25/2022] Open
Abstract
Glycogen synthase kinase-3β (GSK-3β) is a potential target in the field of Alzheimer's disease drug discovery. We recently reported a new class of 9H-pyrimido[4,5-b]indole-based GSK-3β inhibitors, of which 3-(3-((7-chloro-9H-pyrimido[4,5-b]indol-4-yl)(methyl)amino)piperidin-1-yl)propanenitrile (1) demonstrated promising inhibitory potency. However, this compound underwent rapid degradation by human liver microsomes. Starting from 1, we prepared a series of amide-based derivatives and studied their structure-activity relationships against GSK-3β supported by 1 µs molecular dynamics simulations. The biological potency of this series was substantially enhanced by identifying the eutomer configuration at the stereocenter. Moreover, the introduction of an amide bond proved to be an effective strategy to eliminate the metabolic hotspot. The most potent compounds, (R)-3-(3-((7-chloro-9H-pyrimido[4,5-b]indol-4-yl)(methyl)amino)piperidin-1-yl)-3-oxopropanenitrile ((R)-2) and (R)-1-(3-((7-bromo-9Hpyrimido[4,5-b]indol-4-yl)(methyl)amino)piperidin-1-yl)propan-1-one ((R)-28), exhibited IC50 values of 480 nM and 360 nM, respectively, and displayed improved metabolic stability. Their favorable biological profile is complemented by minimal cytotoxicity and neuroprotective properties.
Collapse
Affiliation(s)
- Stanislav Andreev
- Institute of Pharmaceutical Sciences, Department of Medicinal and Pharmaceutical Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (S.A.); (T.P.); (A.E.-G.); (F.A.); (M.K.); (S.A.L.)
| | - Tatu Pantsar
- Institute of Pharmaceutical Sciences, Department of Medicinal and Pharmaceutical Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (S.A.); (T.P.); (A.E.-G.); (F.A.); (M.K.); (S.A.L.)
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Ahmed El-Gokha
- Institute of Pharmaceutical Sciences, Department of Medicinal and Pharmaceutical Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (S.A.); (T.P.); (A.E.-G.); (F.A.); (M.K.); (S.A.L.)
- Chemistry Department, Faculty of Science, Menoufia University, Gamal Abdel-Nasser Street, Shebin El-Kom 32511, Egypt
| | - Francesco Ansideri
- Institute of Pharmaceutical Sciences, Department of Medicinal and Pharmaceutical Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (S.A.); (T.P.); (A.E.-G.); (F.A.); (M.K.); (S.A.L.)
| | - Mark Kudolo
- Institute of Pharmaceutical Sciences, Department of Medicinal and Pharmaceutical Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (S.A.); (T.P.); (A.E.-G.); (F.A.); (M.K.); (S.A.L.)
| | - Débora Bublitz Anton
- Cell Culture Laboratory, Postgraduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado 95914-014, Brazil; (D.B.A.); (M.I.G.)
| | - Giulia Sita
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy;
| | - Jenny Romasco
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, Corso D’Augusto, 237, 47921 Rimini, Italy; (J.R.); (A.T.)
| | - Christian Geibel
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (C.G.); (M.L.)
| | - Michael Lämmerhofer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (C.G.); (M.L.)
| | - Márcia Ines Goettert
- Cell Culture Laboratory, Postgraduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado 95914-014, Brazil; (D.B.A.); (M.I.G.)
| | - Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, Corso D’Augusto, 237, 47921 Rimini, Italy; (J.R.); (A.T.)
| | - Stefan A. Laufer
- Institute of Pharmaceutical Sciences, Department of Medicinal and Pharmaceutical Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (S.A.); (T.P.); (A.E.-G.); (F.A.); (M.K.); (S.A.L.)
| | - Pierre Koch
- Institute of Pharmaceutical Sciences, Department of Medicinal and Pharmaceutical Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; (S.A.); (T.P.); (A.E.-G.); (F.A.); (M.K.); (S.A.L.)
- Department of Pharmaceutical/Medicinal Chemistry II, Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-(941)-943-2847
| |
Collapse
|
130
|
Li W, Lu P, Lu Y, Wei H, Niu X, Xu J, Wang K, Zhang H, Li R, Qiu Z, Wang N, Jia P, Zhang Y, Zhang S, Lu H, Chen X, Liu Y, Zhang P. 17β-Estradiol Protects Neural Stem/Progenitor Cells Against Ketamine-Induced Injury Through Estrogen Receptor β Pathway. Front Neurosci 2020; 14:576813. [PMID: 33100963 PMCID: PMC7556164 DOI: 10.3389/fnins.2020.576813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Ketamine inhibits neural stem/progenitor cell (NSPC) proliferation and disrupts normal neurogenesis in the developing brain. 17β-Estradiol alleviates neurogenesis damage and enhances behavioral performance after ketamine administration. However, the receptor pathway of 17β-estradiol that protects NSPCs from ketamine-induced injury remains unknown. In the present study, we investigated the role of estrogen receptor α (ER-α) and estrogen receptor β (ER-β) in 17β-estradiol’s protection against ketamine-exposed NSPCs and explored its potential mechanism. The primary cultured NSPCs were identified by immunofluorescence and then treated with ketamine and varying doses of ER-α agonist 4,4′,4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT) or ER-β agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) for 24 h. NSPC proliferation was analyzed by 5-bromo-2-deoxyuridine incorporation test. The expression of phosphorylated glycogen synthase kinase-3β (p-GSK-3β) was quantified by western blotting. It was found that treatment with different concentrations of PPT did not alter the inhibition of ketamine on NSPC proliferation. However, treatment with DPN attenuated the inhibition of ketamine on NSPC proliferation at 24 h after their exposure (P < 0.05). Furthermore, treatment with DPN increased p-GSK-3β expression in NSPCs exposed to ketamine. These findings indicated that ER-β mediates probably the protective effects of 17β-estradiol on ketamine-damaged NSPC proliferation and GSK-3β is involved in this process
Collapse
Affiliation(s)
- Weisong Li
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Niu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jing Xu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhengguo Qiu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ning Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pengyu Jia
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shuyue Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haixia Lu
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
131
|
Hou Y, Hu Z, Gong X, Yang B. HSPB8 overexpression prevents disruption of blood-brain barrier after intracerebral hemorrhage in rats through Akt/GSK3β/β-catenin signaling pathway. Aging (Albany NY) 2020; 12:17568-17581. [PMID: 32889520 PMCID: PMC7521513 DOI: 10.18632/aging.103773] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Blood brain barrier (BBB) disruption is a crucial factor contributing to secondary brain injury after intracerebral hemorrhage (ICH). Heat shock protein B8 (HSPB8) has been recently reported to confer neuroprotection against against ischaemic stroke through maintaining BBB integrity. However, the role of HSPB8 in ICH is still elusive. In this study, we found that HSPB8 was upregulated by ICH and extensively expressed in neurovascular structure including endothelial cells and astrocytes. lentivirus intracerebroventricular (i.c.v) injection achieved a widespread and persistent HSPB8 overexpression in brain tissues. HSPB8 overexpression significantly ameliorated neurobehavioral deficits and brain edema at 24 and 72h following ICH. Moreover, HSPB8 overexpression remarkedly inhibited BBB disruption and significantly increase the level of p-Akt, p-GSKβ and intranuclear β-catenin 24h post-ICH. This effect was obviously reversed by Akt specific inhibitor, MK2206. Based on these findings, HSPB8 exerted its protective effect on BBB, at least partly, via Akt/ p-GSKβ/β-catenin pathways.
Collapse
Affiliation(s)
- Ying Hou
- Department of Neurology, 2nd Xiangya Hospital, Central South University Changsha, Hunan Province, China
| | - Zhiping Hu
- Department of Neurology, 2nd Xiangya Hospital, Central South University Changsha, Hunan Province, China
| | - Xiyu Gong
- Department of Neurology, 2nd Xiangya Hospital, Central South University Changsha, Hunan Province, China
| | - Binbin Yang
- Department of Neurology, 2nd Xiangya Hospital, Central South University Changsha, Hunan Province, China
| |
Collapse
|
132
|
Zhang L, Li X, Zhang N, Yang X, Hou T, Fu W, Yuan F, Wang L, Wen H, Tian Y, Zhang H, Lu X, Zhu WG. WDFY2 Potentiates Hepatic Insulin Sensitivity and Controls Endosomal Localization of the Insulin Receptor and IRS1/2. Diabetes 2020; 69:1887-1902. [PMID: 32641353 DOI: 10.2337/db19-0699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 06/04/2020] [Indexed: 11/13/2022]
Abstract
Endosomes help activate the hepatic insulin-evoked Akt signaling pathway, but the underlying regulatory mechanisms are unclear. Previous studies have suggested that the endosome-located protein WD repeat and FYVE domain-containing 2 (WDFY2) might be involved in metabolic disorders, such as diabetes. Here, we generated Wdfy2 knockout (KO) mice and assessed the metabolic consequences. These KO mice exhibited systemic insulin resistance, with increased gluconeogenesis and suppressed glycogen accumulation in the liver. Mechanistically, we found that the insulin-stimulated activation of Akt2 and its substrates FoxO1 and GSK-3β is attenuated in the Wdfy2 KO liver and H2.35 hepatocytes, suggesting that WDFY2 acts as an important regulator of hepatic Akt2 signaling. We further found that WDFY2 interacts with the insulin receptor (INSR) via its WD1-4 domain and localizes the INSR to endosomes after insulin stimulation. This process ensures that the downstream insulin receptor substrates 1 and 2 (IRS1/2) can be recruited to the endosomal INSR. IRS1/2-INSR binding promotes IRS1/2 phosphorylation and subsequent activation, initiating downstream Akt2 signaling in the liver. Interestingly, adeno-associated viral WDFY2 delivery ameliorated metabolic defects in db/db mice. These findings demonstrate that WDFY2 activates insulin-evoked Akt2 signaling by controlling endosomal localization of the INSR and IRS1/2 in hepatocytes. This pathway might constitute a new potential target for diabetes prevention or treatment.
Collapse
Affiliation(s)
- Luyao Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xue Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Nan Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Tianyun Hou
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| | - Wan Fu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Fengjie Yuan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lina Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - He Wen
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan Tian
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Hongquan Zhang
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xifeng Lu
- Department of Physiology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei-Guo Zhu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
133
|
Korona D, Nightingale D, Fabre B, Nelson M, Fischer B, Johnson G, Lees J, Hubbard S, Lilley K, Russell S. Characterisation of protein isoforms encoded by the Drosophila Glycogen Synthase Kinase 3 gene shaggy. PLoS One 2020; 15:e0236679. [PMID: 32760087 PMCID: PMC7410302 DOI: 10.1371/journal.pone.0236679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/09/2020] [Indexed: 12/15/2022] Open
Abstract
The Drosophila shaggy gene (sgg, GSK-3) encodes multiple protein isoforms with serine/threonine kinase activity and is a key player in diverse developmental signalling pathways. Currently it is unclear whether different Sgg proteoforms are similarly involved in signalling or if different proteoforms have distinct functions. We used CRISPR/Cas9 genome engineering to tag eight different Sgg proteoform classes and determined their localization during embryonic development. We performed proteomic analysis of the two major proteoform classes and generated mutant lines for both of these for transcriptomic and phenotypic analysis. We uncovered distinct tissue-specific localization patterns for all of the tagged proteoforms we examined, most of which have not previously been characterised directly at the protein level, including one proteoform initiating with a non-standard codon. Collectively, this suggests complex developmentally regulated splicing of the sgg primary transcript. Further, affinity purification followed by mass spectrometric analyses indicate a different repertoire of interacting proteins for the two major proteoforms we examined, one with ubiquitous expression (Sgg-PB) and one with nervous system specific expression (Sgg-PA). Specific mutation of these proteoforms shows that Sgg-PB performs the well characterised maternal and zygotic segmentations functions of the sgg locus, while Sgg-PA mutants show adult lifespan and locomotor defects consistent with its nervous system localisation. Our findings provide new insights into the role of GSK-3 proteoforms and intriguing links with the GSK-3α and GSK-3β proteins encoded by independent vertebrate genes. Our analysis suggests that different proteoforms generated by alternative splicing are likely to perform distinct functions.
Collapse
Affiliation(s)
- Dagmara Korona
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Nightingale
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, United Kingdom
| | - Bertrand Fabre
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, United Kingdom
| | - Michael Nelson
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre Manchester, University of Manchester, Manchester, United Kingdom
| | - Bettina Fischer
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Glynnis Johnson
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan Lees
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, United Kingdom
| | - Simon Hubbard
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre Manchester, University of Manchester, Manchester, United Kingdom
| | - Kathryn Lilley
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, United Kingdom
| | - Steven Russell
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
134
|
Ni M, Zhou H, Zhang J, Jin D, Lu T, Busuttil RW, Kupiec-Weglinski JW, Wang X, Zhai Y. Isoform- and Cell Type-Specific Roles of Glycogen Synthase Kinase 3 N-Terminal Serine Phosphorylation in Liver Ischemia Reperfusion Injury. THE JOURNAL OF IMMUNOLOGY 2020; 205:1147-1156. [PMID: 32680958 DOI: 10.4049/jimmunol.2000397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/21/2020] [Indexed: 12/31/2022]
Abstract
Glycogen synthase kinase 3 (Gsk3) α and β are both constitutively active and inhibited upon stimulation by N-terminal serine phosphorylation. Although roles of active Gsk3 in liver ischemia reperfusion injury (IRI) have been well appreciated, whether Gsk3 N-terminal serine phosphorylation has any functional significance in the disease process remains unclear. In a murine liver partial warm ischemia model, we studied Gsk3 N-terminal serine mutant knock-in (KI) mice and showed that liver IRI was decreased in Gsk3αS21A but increased in Gsk3βS9A mutant KI mice. Bone marrow chimeric experiments revealed that the Gsk3α, but not β, mutation in liver parenchyma protected from IRI, and both mutations in bone marrow-derived cells exacerbated liver injuries. Mechanistically, mutant Gsk3α protected hepatocytes from inflammatory (TNF-α) cell death by the activation of HIV-1 TAT-interactive protein 60 (TIP60)-mediated autophagy pathway. The pharmacological inhibition of TIP60 or autophagy diminished the protection of the Gsk3α mutant hepatocytes from inflammatory cell death in vitro and the Gsk3α mutant KI mice from liver IRI in vivo. Thus, Gsk3 N-terminal serine phosphorylation inhibits liver innate immune activation but suppresses hepatocyte autophagy in response to inflammation. Gsk3 αS21, but not βS9, mutation is sufficient to sustain Gsk4 activities in hepatocytes and protect livers from IRI via TIP60 activation.
Collapse
Affiliation(s)
- Ming Ni
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Haoming Zhou
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Jing Zhang
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Dan Jin
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Department of Obstetrics and Gynecology, Shanghai Jiaotong University, Shanghai 200025, China; and
| | - Tianfei Lu
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Liver Surgery, Renji Hospital, Shanghai Jiaotong University, Shanghai 200025, China
| | - Ronald W Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Jerzy W Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Xuehao Wang
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China;
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095;
| |
Collapse
|
135
|
Aceto G, Re A, Mattera A, Leone L, Colussi C, Rinaudo M, Scala F, Gironi K, Barbati SA, Fusco S, Green T, Laezza F, D'Ascenzo M, Grassi C. GSK3β Modulates Timing-Dependent Long-Term Depression Through Direct Phosphorylation of Kv4.2 Channels. Cereb Cortex 2020; 29:1851-1865. [PMID: 29790931 DOI: 10.1093/cercor/bhy042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/15/2018] [Accepted: 02/07/2018] [Indexed: 12/31/2022] Open
Abstract
Spike timing-dependent plasticity (STDP) is a form of activity-dependent remodeling of synaptic strength that underlies memory formation. Despite its key role in dictating learning rules in the brain circuits, the molecular mechanisms mediating STDP are still poorly understood. Here, we show that spike timing-dependent long-term depression (tLTD) and A-type K+ currents are modulated by pharmacological agents affecting the levels of active glycogen-synthase kinase 3 (GSK3) and by GSK3β knockdown in layer 2/3 of the mouse somatosensory cortex. Moreover, the blockade of A-type K+ currents mimics the effects of GSK3 up-regulation on tLTD and occludes further changes in synaptic strength. Pharmacological, immunohistochemical and biochemical experiments revealed that GSK3β influence over tLTD induction is mediated by direct phosphorylation at Ser-616 of the Kv4.2 subunit, a molecular determinant of A-type K+ currents. Collectively, these results identify the functional interaction between GSK3β and Kv4.2 channel as a novel mechanism for tLTD modulation providing exciting insight into the understanding of GSK3β role in synaptic plasticity.
Collapse
Affiliation(s)
- Giuseppe Aceto
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agnese Re
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Andrea Mattera
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lucia Leone
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| | - Claudia Colussi
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Marco Rinaudo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federico Scala
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Katia Gironi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Salvatore Fusco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| | - Thomas Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Marcello D'Ascenzo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| |
Collapse
|
136
|
Wang X, Chen N, Du Z, Ling Z, Zhang P, Yang J, Khaleel M, Khoury AN, Li J, Li S, Huang H, Zhou X, Han Y, Wei F. Bioinformatics analysis integrating metabolomics of m 6A RNA microarray in intervertebral disc degeneration. Epigenomics 2020; 12:1419-1441. [PMID: 32627576 DOI: 10.2217/epi-2020-0101] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aim: To explore the potential functions and mechanism of N6.methyladenosine (m6A) abnormality of RNAs in nucleus pulposus from the intervertebral disc degeneration (IDD). Materials & methods: We performed rat model, m6A epitranscriptomic microarray, bioinformatics analysis and metabolomics. Results: In IDD, most of the differentially methylated RNAs showed a significant demethylation situation. The competing endogenous RNA network LOC102555094/miR-431/GSK-3β combining downstream Wnt pathway were identified in bioinformatics analysis. For metabolomics, activation of Wnt pathway led to reprogramming of glucose metabolism and enzyme activation of PKM2. Finally, quantitative real-time PCR and methylated RNA immunoprecipitation coupled with quantitative real-time PCR revealed the positive correlation between demethylation of LOC102555094 and expression of both FTO and ZFP217. Conclusion: LOC102555094 might be demethylated by ZFP217, activating FTO and LOC102555094/miR-431/GSK-3β/Wnt played a crucial role in IDD.
Collapse
Affiliation(s)
- Xiaoshuai Wang
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Rd, Shenzhen, 518107, China
| | - Ningning Chen
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Rd, Shenzhen, 518107, China
| | - Zefeng Du
- Department of Clinical Medicine, Zhongshan Medical College of Sun Yat-sen University, No. 74, Zhongshan Er Rd, Guangzhou, 510030, China
| | - Zemin Ling
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510030, China
| | - Penghui Zhang
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Rd, Shenzhen, 518107, China
| | - Jiaming Yang
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Rd, Shenzhen, 518107, China
| | - Mohammed Khaleel
- Department of Orthopaedic Surgery, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Anthony N Khoury
- Hip Preservation Center, Baylor University Medical Center at Dallas, TX 75390, USA
| | - Jianwen Li
- Affiliated Dongguan People's Hospital of Southern Medical University, Dongguan, 523000, China
| | - Songbo Li
- Affiliated Dongguan People's Hospital of Southern Medical University, Dongguan, 523000, China
| | - Haoyang Huang
- Department of Clinical Medicine, Zhongshan Medical College of Sun Yat-sen University, No. 74, Zhongshan Er Rd, Guangzhou, 510030, China
| | - Xinwei Zhou
- Department of Clinical Medicine, Zhongshan Medical College of Sun Yat-sen University, No. 74, Zhongshan Er Rd, Guangzhou, 510030, China
| | - Yueyin Han
- Department of Clinical Medicine, Zhongshan Medical College of Sun Yat-sen University, No. 74, Zhongshan Er Rd, Guangzhou, 510030, China
| | - Fuxin Wei
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Rd, Shenzhen, 518107, China
| |
Collapse
|
137
|
Trostnikov MV, Veselkina ER, Krementsova AV, Boldyrev SV, Roshina NV, Pasyukova EG. Modulated Expression of the Protein Kinase GSK3 in Motor and Dopaminergic Neurons Increases Female Lifespan in Drosophila melanogaster. Front Genet 2020; 11:668. [PMID: 32695143 PMCID: PMC7339944 DOI: 10.3389/fgene.2020.00668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
Most eukaryotic genes express multiple transcripts and proteins, and a sophisticated gene expression strategy plays a crucial role in ensuring the cell-specificity of genetic information and the correctness of phenotypes. The Drosophila melanogaster gene shaggy encodes several isoforms of the conserved glycogen synthase kinase 3 (GSK3), which is vitally important for multiple biological processes. To characterize the phenotypic effects of differential shaggy expression, we explored how the multidirectional modulation of the expression of the main GSK3 isoform, Shaggy-PB, in different tissues and cells affects lifespan. To this end, we used lines with transgenic constructs that encode mutant variants of the protein. The effect of shaggy misexpression on lifespan depended on the direction of the presumed change in GSK3 activity and the type of tissue/cell. The modulation of GSK3 activity in motor and dopaminergic neurons improved female lifespan but caused seemingly negative changes in the structural (mitochondrial depletion; neuronal loss) and functional (perturbed locomotion) properties of the nervous system, indicating the importance of analyzing the relationship between lifespan and healthspan in invertebrate models. Our findings provide new insights into the molecular and cellular bases of lifespan extension, demonstrating that the fine-tuning of transcript-specific shaggy expression in individual groups of neurons is sufficient to provide a sex-specific increase in survival and slow aging.
Collapse
Affiliation(s)
- Mikhail V Trostnikov
- Laboratory of Genome Variation, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina R Veselkina
- Laboratory of Genome Variation, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Anna V Krementsova
- Laboratory of Genome Variation, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia.,Laboratory of Kinetics and Mechanisms of Enzymatic and Catalytic Reactions, N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Stepan V Boldyrev
- Laboratory of Genome Variation, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia.,Laboratory of Genetic Basis of Biodiversity, N. I. Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Natalia V Roshina
- Laboratory of Genome Variation, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia.,Laboratory of Genetic Basis of Biodiversity, N. I. Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Elena G Pasyukova
- Laboratory of Genome Variation, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
138
|
Gavagan M, Fagnan E, Speltz EB, Zalatan JG. The Scaffold Protein Axin Promotes Signaling Specificity within the Wnt Pathway by Suppressing Competing Kinase Reactions. Cell Syst 2020; 10:515-525.e5. [PMID: 32553184 DOI: 10.1016/j.cels.2020.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/02/2020] [Accepted: 05/15/2020] [Indexed: 10/24/2022]
Abstract
Scaffold proteins are thought to promote signaling specificity by accelerating reactions between bound kinase and substrate proteins. To test the long-standing hypothesis that the scaffold protein Axin accelerates glycogen synthase kinase 3β (GSK3β)-mediated phosphorylation of β-catenin in the Wnt signaling network, we measured GSK3β reaction rates with multiple substrates in a minimal, biochemically reconstituted system. We observed an unexpectedly small, ∼2-fold Axin-mediated rate increase for the β-catenin reaction when measured in isolation. In contrast, when both β-catenin and non-Wnt pathway substrates are present, Axin accelerates the β-catenin reaction by preventing competition with alternative substrates. At high competitor concentrations, Axin produces >10-fold rate effects. Thus, while Axin alone does not markedly accelerate the β-catenin reaction, in physiological settings where multiple GSK3β substrates are present, Axin may promote signaling specificity by suppressing interactions with competing, non-Wnt pathway targets. This mechanism for scaffold-mediated control of competition enables a shared kinase to perform distinct functions in multiple signaling networks.
Collapse
Affiliation(s)
- Maire Gavagan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Erin Fagnan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth B Speltz
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Jesse G Zalatan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
139
|
Li J, Ma S, Chen J, Hu K, Li Y, Zhang Z, Su Z, Woodgett JR, Li M, Huang Q. GSK-3β Contributes to Parkinsonian Dopaminergic Neuron Death: Evidence From Conditional Knockout Mice and Tideglusib. Front Mol Neurosci 2020; 13:81. [PMID: 32581704 PMCID: PMC7283909 DOI: 10.3389/fnmol.2020.00081] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) dysregulation has been implicated in nigral dopaminergic neurodegeneration, one of the main pathological features of Parkinson’s disease (PD). The two isoforms, GSK-3α and GSK-3β, have both been suggested to play a detrimental role in neuronal death. To date, several studies have focused on the role of GSK-3β on PD pathogenesis, while the role of GSK-3α has been largely overlooked. Here, we report in situ observations that both GSK-3α and GSK-3β are dephosphorylated at a negatively acting regulatory serine, indicating kinase activation, selectively in nigral dopaminergic neurons following exposure of mice to 1-methyl-4-pheny-1,2,3,6-tetrahydropyridine (MPTP). To identify whether GSK-3α and GSK-3β display functional redundancy in regulating parkinsonian dopaminergic cell death, we analysed dopaminergic neuron-specific Gsk3a null (Gsk3aΔDat) and Gsk3b null (Gsk3bΔDat) mice, respectively. We found that Gsk3bΔDat, but not Gsk3aΔDat, showed significant resistance to MPTP insult, revealing non-redundancy of GSK-3α and GSK-3β in PD pathogenesis. In addition, we tested the neuroprotective effect of tideglusib, the most clinically advanced inhibitor of GSK-3, in the MPTP model of PD. Administration of higher doses (200 mg/kg and 500 mg/kg) of tideglusib exhibited significant neuroprotection, whereas 50 mg/kg tideglusib failed to prevent dopaminergic neurodegeneration from MPTP toxicity. Administration of 200 mg/kg tideglusib improved motor symptoms of MPTP-treated mice. Together, these data demonstrate GSK-3β and not GSK-3α is critical for parkinsonian neurodegeneration. Our data support the view that GSK-3β acts as a potential therapeutic target in PD and tideglusib would be a candidate drug for PD neuroprotective therapy.
Collapse
Affiliation(s)
- Junyu Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | - Kunhua Hu
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yongyi Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zeyu Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zixiang Su
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiaoying Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
140
|
Amici M, Lee Y, Pope RJP, Bradley CA, Cole A, Collingridge GL. GSK-3β regulates the synaptic expression of NMDA receptors via phosphorylation of phosphatidylinositol 4 kinase type IIα. Eur J Neurosci 2020; 54:6815-6825. [PMID: 32463939 PMCID: PMC8554790 DOI: 10.1111/ejn.14841] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 05/01/2020] [Accepted: 05/20/2020] [Indexed: 12/03/2022]
Abstract
Deregulation of GSK‐3β is strongly implicated in a variety of serious brain conditions, such as Alzheimer disease, bipolar disorder and schizophrenia. To understand how GSK‐3β becomes dysregulated in these conditions, it is important to understand its physiological functions in the central nervous system. In this context, GSK‐3β plays a role in the induction of NMDA receptor‐dependent long‐term depression (LTD) and several substrates for GSK‐3β have been identified in this form of synaptic plasticity, including KLC‐2, PSD‐95 and tau. Stabilization of NMDA receptors at synapses has also been shown to involve GSK‐3β, but the substrates involved are currently unknown. Recent work has identified phosphatidylinositol 4 kinase type IIα (PI4KIIα) as a neuronal GSK‐3β substrate that can potentially regulate the surface expression of AMPA receptors. In the present study, we investigated the synaptic role of PI4KIIα in organotypic rat hippocampal slices. We found that knockdown of PI4KIIα has no effect on synaptic AMPA receptor‐mediated synaptic transmission but substantially reduces NMDA receptor‐mediated synaptic transmission. Furthermore, the ability of the selective GSK‐3 inhibitor, CT99021, to reduce the amplitude of NMDA receptor‐mediated currents was occluded in shRNA‐PI4KIIα transfected neurons. The effects of knocking down PI4KIIα were fully rescued by a shRNA‐resistant wild‐type construct, but not by a mutant construct that cannot be phosphorylated by GSK‐3β. These data suggest that GSK‐3β phosphorylates PI4KIIα to stabilize NMDA receptors at the synapse.
Collapse
Affiliation(s)
- Mascia Amici
- Glutamate Receptor Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Yeseul Lee
- Glutamate Receptor Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Robert J P Pope
- Glutamate Receptor Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Clarrisa A Bradley
- Neurosciences and Mental Health, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Adam Cole
- Neurosignalling and Mood Disorders Group, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Graham L Collingridge
- Glutamate Receptor Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Disease, Department of Physiology, The University of Toronto, Toronto, ON, Canada
| |
Collapse
|
141
|
Teves ME, Roldan ERS, Krapf D, Strauss III JF, Bhagat V, Sapao P. Sperm Differentiation: The Role of Trafficking of Proteins. Int J Mol Sci 2020; 21:E3702. [PMID: 32456358 PMCID: PMC7279445 DOI: 10.3390/ijms21103702] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/10/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
Sperm differentiation encompasses a complex sequence of morphological changes that takes place in the seminiferous epithelium. In this process, haploid round spermatids undergo substantial structural and functional alterations, resulting in highly polarized sperm. Hallmark changes during the differentiation process include the formation of new organelles, chromatin condensation and nuclear shaping, elimination of residual cytoplasm, and assembly of the sperm flagella. To achieve these transformations, spermatids have unique mechanisms for protein trafficking that operate in a coordinated fashion. Microtubules and filaments of actin are the main tracks used to facilitate the transport mechanisms, assisted by motor and non-motor proteins, for delivery of vesicular and non-vesicular cargos to specific sites. This review integrates recent findings regarding the role of protein trafficking in sperm differentiation. Although a complete characterization of the interactome of proteins involved in these temporal and spatial processes is not yet known, we propose a model based on the current literature as a framework for future investigations.
Collapse
Affiliation(s)
- Maria E. Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond VA 23298, USA;
| | - Eduardo R. S. Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales (CSIC), 28006-Madrid, Spain
| | - Diego Krapf
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Jerome F. Strauss III
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond VA 23298, USA;
| | - Virali Bhagat
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond VA 23298, USA;
| | - Paulene Sapao
- Department of Chemistry, Virginia Commonwealth University, Richmond VA, 23298, USA;
| |
Collapse
|
142
|
Jaworski T. Control of neuronal excitability by GSK-3beta: Epilepsy and beyond. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118745. [PMID: 32450268 DOI: 10.1016/j.bbamcr.2020.118745] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 12/22/2022]
Abstract
Glycogen synthase kinase 3beta (GSK-3β) is an enzyme with a variety of cellular functions in addition to the regulation of glycogen metabolism. In the central nervous system, different intracellular signaling pathways converge on GSK-3β through a cascade of phosphorylation events that ultimately control a broad range of neuronal functions in the development and adulthood. In mice, genetically removing or increasing GSK-3β cause distinct functional and structural neuronal phenotypes and consequently affect cognition. Precise control of GSK-3β activity is important for such processes as neuronal migration, development of neuronal morphology, synaptic plasticity, excitability, and gene expression. Altered GSK-3β activity contributes to aberrant plasticity within neuronal circuits leading to neurological, psychiatric disorders, and neurodegenerative diseases. Therapeutically targeting GSK-3β can restore the aberrant plasticity of neuronal networks at least in animal models of these diseases. Although the complete repertoire of GSK-3β neuronal substrates has not been defined, emerging evidence shows that different ion channels and their accessory proteins controlling excitability, neurotransmitter release, and synaptic transmission are regulated by GSK-3β, thereby supporting mechanisms of synaptic plasticity in cognition. Dysregulation of ion channel function by defective GSK-3β activity sustains abnormal excitability in the development of epilepsy and other GSK-3β-linked human diseases.
Collapse
Affiliation(s)
- Tomasz Jaworski
- Laboratory of Animal Models, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
143
|
Freitas MJ, Silva JV, Brothag C, Regadas-Correia B, Fardilha M, Vijayaraghavan S. Isoform-specific GSK3A activity is negatively correlated with human sperm motility. Mol Hum Reprod 2020; 25:171-183. [PMID: 30824926 DOI: 10.1093/molehr/gaz009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/29/2019] [Accepted: 02/19/2019] [Indexed: 01/04/2023] Open
Abstract
In mouse and bovine sperm, GSK3 activity is inversely proportional to motility. Targeted disruption of the GSK3A gene in testis results in normal spermatogenesis, but mature sperm present a reduced motility, rendering male mice infertile. On the other hand, GSK3B testis-specific KO is fertile. Yet in human sperm, an isoform-specific correlation between GSK3A and sperm motility was never established. In order to analyze GSK3 function in human sperm motility, normospermic and asthenozoospermic samples from adult males were used to correlate GSK3 expression and activity levels with human sperm motility profiles. Moreover, testicular and sperm GSK3 interactomes were identified using a yeast two-hybrid screen and coimmunoprecipitation, respectively. An extensive in-silico analysis of the GSK3 interactome was performed. The results proved that inhibited GSK3A (serine phosphorylated) presents a significant strong positive correlation (r = 0.822, P = 0.023) with the percentage of progressive human sperm, whereas inhibited GSK3B is not significantly correlated with sperm motility (r = 0.577, P = 0.175). The importance of GSK3 in human sperm motility was further reinforced by in-silico analysis of the GSK3 interactome, which revealed a high level of involvement of GSK3 interactors in sperm motility-related functions. The limitation of techniques used for GSK3 interactome identification can be a drawback, since none completely mimics the physiological environment. Our findings prove that human sperm motility relies on isoform-specific functions of GSK3A within this cell. Given the reported relevance of GSK3 protein-protein interactions in sperm motility, we hypothesized that they stand as potential targets for male contraceptive strategies based on sperm motility modulation.
Collapse
Affiliation(s)
- M J Freitas
- Signal Transduction Laboratory, Institute for Research in Biomedicine-iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - J V Silva
- Signal Transduction Laboratory, Institute for Research in Biomedicine-iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal.,Reproductive Genetics & Embryo-fetal Development Group, Institute for Innovation and Health Research (I3S), University of Porto, Porto, Portugal.,Department of Microscopy, Laboratory of Cell Biology, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - C Brothag
- Kent State University, Kent, OH, USA
| | - B Regadas-Correia
- CNC.IBILI-Institute for Biomedical Imaging and Life Sciences, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CIBIT-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.,Department Quantitative Methods and Information and Management Systems, Coimbra Business School, Coimbra, Portugal
| | - M Fardilha
- Signal Transduction Laboratory, Institute for Research in Biomedicine-iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | | |
Collapse
|
144
|
McCamphill PK, Stoppel LJ, Senter RK, Lewis MC, Heynen AJ, Stoppel DC, Sridhar V, Collins KA, Shi X, Pan JQ, Madison J, Cottrell JR, Huber KM, Scolnick EM, Holson EB, Wagner FF, Bear MF. Selective inhibition of glycogen synthase kinase 3α corrects pathophysiology in a mouse model of fragile X syndrome. Sci Transl Med 2020; 12:eaam8572. [PMID: 32434848 PMCID: PMC8095719 DOI: 10.1126/scitranslmed.aam8572] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/15/2019] [Accepted: 01/11/2020] [Indexed: 01/06/2023]
Abstract
Fragile X syndrome is caused by FMR1 gene silencing and loss of the encoded fragile X mental retardation protein (FMRP), which binds to mRNA and regulates translation. Studies in the Fmr1-/y mouse model of fragile X syndrome indicate that aberrant cerebral protein synthesis downstream of metabotropic glutamate receptor 5 (mGluR5) signaling contributes to disease pathogenesis, but clinical trials using mGluR5 inhibitors were not successful. Animal studies suggested that treatment with lithium might be an alternative approach. Targets of lithium include paralogs of glycogen synthase kinase 3 (GSK3), and nonselective small-molecule inhibitors of these enzymes improved disease phenotypes in a fragile X syndrome mouse model. However, the potential therapeutic use of GSK3 inhibitors has been hampered by toxicity arising from inhibition of both α and β paralogs. Recently, we developed GSK3 inhibitors with sufficient paralog selectivity to avoid a known toxic consequence of dual inhibition, that is, increased β-catenin stabilization. We show here that inhibition of GSK3α, but not GSK3β, corrected aberrant protein synthesis, audiogenic seizures, and sensory cortex hyperexcitability in Fmr1-/y mice. Although inhibiting either paralog prevented induction of NMDA receptor-dependent long-term depression (LTD) in the hippocampus, only inhibition of GSK3α impaired mGluR5-dependent and protein synthesis-dependent LTD. Inhibition of GSK3α additionally corrected deficits in learning and memory in Fmr1-/y mice; unlike mGluR5 inhibitors, there was no evidence of tachyphylaxis or enhanced psychotomimetic-induced hyperlocomotion. GSK3α selective inhibitors may have potential as a therapeutic approach for treating fragile X syndrome.
Collapse
Affiliation(s)
- Patrick K McCamphill
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Laura J Stoppel
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rebecca K Senter
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael C Lewis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Arnold J Heynen
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David C Stoppel
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vinay Sridhar
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX 75390, USA
| | - Katie A Collins
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xi Shi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jon Madison
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffrey R Cottrell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kimberly M Huber
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX 75390, USA
| | - Edward M Scolnick
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Edward B Holson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Florence F Wagner
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Mark F Bear
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
145
|
Bertrand FE. The cross-talk of NOTCH and GSK-3 signaling in colon and other cancers. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118738. [PMID: 32389646 DOI: 10.1016/j.bbamcr.2020.118738] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 12/14/2022]
Abstract
The GSK-3 kinases, GSK-3α and GSK-3β, have a central role in regulating multiple cellular processes such as glycogen synthesis, insulin signaling, cell proliferation and apoptosis. GSK-3β is the most well studied, and was originally described for its role in regulating glycogen synthase. GSK-3β has been studied as a participant in the oncogenic process in a variety of cancers due to its intersection with the PTEN/PI3K/AKT and RAS/RAF/MEK/ERK pathways. Dysregulated signaling through the Notch family of receptors can also promote oncogenesis. Normal Notch receptor signaling regulates cell fate determination in stem cell pools. GSK-3β and Notch share similar targets such β-catenin and the WNT pathway. WNT and β-catenin are involved in several oncogenic processes including those of the colon. In addition, GSK-3β may directly regulate aspects of Notch signaling. This review describes how crosstalk between GSK-3β and Notch can promote oncogenesis, using colon cancer as the primary example.
Collapse
Affiliation(s)
- Fred E Bertrand
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
146
|
Prossomariti A, Piazzi G, Alquati C, Ricciardiello L. Are Wnt/β-Catenin and PI3K/AKT/mTORC1 Distinct Pathways in Colorectal Cancer? Cell Mol Gastroenterol Hepatol 2020; 10:491-506. [PMID: 32334125 PMCID: PMC7369353 DOI: 10.1016/j.jcmgh.2020.04.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023]
Abstract
Wnt/β-catenin and phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin complex 1 (PI3K/AKT/mTORC1) pathways both are critically involved in colorectal cancer (CRC) development, although they are implicated in the modulation of distinct oncogenic mechanisms. In homeostatic and pathologic conditions, these pathways show a fine regulation based mainly on feedback mechanisms, and are connected at multiple levels involving both upstream and downstream common effectors. The ability of the Wnt/β-catenin and PI3K/AKT/mTORC1 pathways to reciprocally control themselves represents one of the main resistance mechanisms to selective inhibitors in CRC, leading to the hypothesis that in specific settings, particularly in cancer driven by genetic alterations in Wnt/β-catenin signaling, the relationship between Wnt/β-catenin and PI3K/AKT/mTORC1 pathways could be so close that they should be considered as a unique therapeutic target. This review provides an update on the Wnt/β-catenin and PI3K/AKT/mTORC1 pathway interconnections in CRC, describing the main molecular players and the potential implications of combined inhibitors as an approach for CRC chemoprevention and treatment.
Collapse
Affiliation(s)
- Anna Prossomariti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy,Center for Applied Biomedical Research, S. Orsola Hospital, University of Bologna, Bologna, Italy,Anna Prossomariti, PhD, Center for Applied Biomedical Research, S. Orsola Hospital, Via Massarenti 9, 40138, Bologna, Italy. fax: (39) 051-2143902.
| | - Giulia Piazzi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy,Center for Applied Biomedical Research, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Chiara Alquati
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy,Center for Applied Biomedical Research, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy,Center for Applied Biomedical Research, S. Orsola Hospital, University of Bologna, Bologna, Italy,Correspondence Address correspondence to: Luigi Ricciardiello, MD, Department of Medical and Surgical Sciences, Via Massarenti 9, 40138, Bologna, Italy. fax: (39) 051-2143381
| |
Collapse
|
147
|
Abstract
The finding of "glycogen synthase kinase-3" (GSK-3) was initially identified as a protein kinase that phosphorylate and inhibited glycogen synthase. However, it was soon discovered that GSK-3 also has significant impact in regulation of truly astonishing number of critical intracellular signaling pathways ranging from regulation of cell growth, neurology, heart failure, diabetes, aging, inflammation, and cancer. Recent studies have validated the feasibility of targeting GSK-3 for its vital therapeutic potential to maintain normal myocardial homeostasis, conversely, its loss is incompatible with life as it can abrupt cell cycle and endorse fatal cardiomyopathy. The current study focuses on its expanding therapeutic action in myocardial tissue, concentrating primarily on its role in diabetes-associated cardiac complication, apoptosis and metabolism, heart failure, cardiac hypertrophy, and myocardial infarction. The current report also includes the finding of our previous investigation that has shown the impact of GSK-3β inhibitor against diabetes-associated myocardial injury and experimentally induced myocardial infarction. We have also discussed some recent identified GSK-3β inhibitors for their cardio-protective potential. The crosstalk of various underlying mechanisms that highlight the significant role of GSK-3β in myocardial pathophysiology have been discussed in the present report. For these literatures, we will rely profoundly on our previous studies and those of others to reconcile some of the deceptive contradictions in the literature.
Collapse
|
148
|
Han Y, Zhu L, Wu W, Zhang H, Hu W, Dai L, Yang Y. Small Molecular Immune Modulators as Anticancer Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:547-618. [PMID: 32185725 DOI: 10.1007/978-981-15-3266-5_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
After decades of intense effort, immune checkpoint inhibitors have been conclusively demonstrated to be effective in cancer treatments and thus are revolutionizing the concepts in the treatment of cancers. Immuno-oncology has arrived and will play a key role in cancer treatment in the foreseeable future. However, efforts to find novel methods to improve the immune response to cancer have not ceased. Small-molecule approaches offer inherent advantages over biologic immunotherapies since they can cross cell membranes, penetrate into tumor tissue and tumor microenvironment more easily, and are amenable to be finely controlled than biological agents, which may help reduce immune-related adverse events seen with biologic therapies and provide more flexibility for the combination use with other therapies and superior clinical benefit. On the one hand, small-molecule therapies can modulate the immune response to cancer by restoring the antitumor immunity, promoting more effective cytotoxic lymphocyte responses, and regulating tumor microenvironment, either directly or epigenetically. On the other hand, the combination of different mechanisms of small molecules with antibodies and other biologics demonstrated admirable synergistic effect in clinical settings for cancer treatment and may expand antibodies' usefulness for broader clinical applications. This chapter provides an overview of small-molecule immunotherapeutic approaches either as monotherapy or in combination for the treatment of cancer.
Collapse
Affiliation(s)
- Yongxin Han
- Lapam Capital LLC., 17C1, Tower 2, Xizhimenwai Street, Xicheng District, Beijing, 100044, China.
| | - Li Zhu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Wei Wu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Hui Zhang
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Wei Hu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Liguang Dai
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Yanqing Yang
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| |
Collapse
|
149
|
Marineau A, Khan KA, Servant MJ. Roles of GSK-3 and β-Catenin in Antiviral Innate Immune Sensing of Nucleic Acids. Cells 2020; 9:cells9040897. [PMID: 32272583 PMCID: PMC7226782 DOI: 10.3390/cells9040897] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 12/23/2022] Open
Abstract
The rapid activation of the type I interferon (IFN) antiviral innate immune response relies on ubiquitously expressed RNA and DNA sensors. Once engaged, these nucleotide-sensing receptors use distinct signaling modules for the rapid and robust activation of mitogen-activated protein kinases (MAPKs), the IκB kinase (IKK) complex, and the IKK-related kinases IKKε and TANK-binding kinase 1 (TBK1), leading to the subsequent activation of the activator protein 1 (AP1), nuclear factor-kappa B (NF-κB), and IFN regulatory factor 3 (IRF3) transcription factors, respectively. They, in turn, induce immunomodulatory genes, allowing for a rapid antiviral cellular response. Unlike the MAPKs, the IKK complex and the IKK-related kinases, ubiquitously expressed glycogen synthase kinase 3 (GSK-3) α and β isoforms are active in unstimulated resting cells and are involved in the constitutive turnover of β-catenin, a transcriptional coactivator involved in cell proliferation, differentiation, and lineage commitment. Interestingly, studies have demonstrated the regulatory roles of both GSK-3 and β-catenin in type I IFN antiviral innate immune response, particularly affecting the activation of IRF3. In this review, we summarize current knowledge on the mechanisms by which GSK-3 and β-catenin control the antiviral innate immune response to RNA and DNA virus infections.
Collapse
Affiliation(s)
- Alexandre Marineau
- Faculty of Pharmacy, Université de Montréal, Montréal, QC H3C3J7, Canada;
| | - Kashif Aziz Khan
- Department of Biology, York University, Toronto, ON M3J1P3, Canada;
| | - Marc J. Servant
- Faculty of Pharmacy, Université de Montréal, Montréal, QC H3C3J7, Canada;
- Réseau Québécois de Recherche sur les Médicaments (RQRM), Montréal, QC H3T1C5, Canada
- Correspondence: ; Tel.: +1-514-343-7966
| |
Collapse
|
150
|
GSK3: A Kinase Balancing Promotion and Resolution of Inflammation. Cells 2020; 9:cells9040820. [PMID: 32231133 PMCID: PMC7226814 DOI: 10.3390/cells9040820] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
GSK3 has been implicated for years in the regulation of inflammation and addressed in a plethora of scientific reports using a variety of experimental (disease) models and approaches. However, the specific role of GSK3 in the inflammatory process is still not fully understood and controversially discussed. Following a detailed overview of structure, function, and various regulatory levels, this review focusses on the immunoregulatory functions of GSK3, including the current knowledge obtained from animal models. Its impact on pro-inflammatory cytokine/chemokine profiles, bacterial/viral infections, and the modulation of associated pro-inflammatory transcriptional and signaling pathways is discussed. Moreover, GSK3 contributes to the resolution of inflammation on multiple levels, e.g., via the regulation of pro-resolving mediators, the clearance of apoptotic immune cells, and tissue repair processes. The influence of GSK3 on the development of different forms of stimulation tolerance is also addressed. Collectively, the role of GSK3 as a kinase balancing the initiation/perpetuation and the amelioration/resolution of inflammation is highlighted.
Collapse
|