1
|
Mayo P, Pascual J, Crisman E, Domínguez C, López MG, León R. Innovative pathological network-based multitarget approaches for Alzheimer's disease treatment. Med Res Rev 2024; 44:2367-2419. [PMID: 38678582 DOI: 10.1002/med.22045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/02/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is a major health threat globally. Its prevalence is forecasted to exponentially increase during the next 30 years due to the global aging population. Currently, approved drugs are merely symptomatic, being ineffective in delaying or blocking the relentless disease advance. Intensive AD research describes this disease as a highly complex multifactorial disease. Disclosure of novel pathological pathways and their interconnections has had a major impact on medicinal chemistry drug development for AD over the last two decades. The complex network of pathological events involved in the onset of the disease has prompted the development of multitarget drugs. These chemical entities combine pharmacological activities toward two or more drug targets of interest. These multitarget-directed ligands are proposed to modify different nodes in the pathological network aiming to delay or even stop disease progression. Here, we review the multitarget drug development strategy for AD during the last decade.
Collapse
Affiliation(s)
- Paloma Mayo
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Jorge Pascual
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Cristina Domínguez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| |
Collapse
|
2
|
Pan J, Yao Q, Wang Y, Chang S, Li C, Wu Y, Shen J, Yang R. The role of PI3K signaling pathway in Alzheimer's disease. Front Aging Neurosci 2024; 16:1459025. [PMID: 39399315 PMCID: PMC11466886 DOI: 10.3389/fnagi.2024.1459025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating progressively neurodegenerative disease. The best-characterized hallmark of AD, which is marked by behavioral alterations and cognitive deficits, is the aggregation of deposition of amyloid-beta (Aβ) and hyper-phosphorylated microtubule-associated protein Tau. Despite decades of experimental progress, the control rate of AD remains poor, and more precise deciphering is needed for potential therapeutic targets and signaling pathways involved. In recent years, phosphoinositide 3-kinase (PI3K) and Akt have been recognized for their role in the neuroprotective effect of various agents, and glycogen synthase kinase 3 (GSK3), a downstream enzyme, is also crucial in the tau phosphorylation and Aβ deposition. An overview of the function of PI3K/Akt pathway in the pathophysiology of AD is provided in this review, along with a discussion of recent developments in the pharmaceuticals and herbal remedies that target the PI3K/Akt signaling pathway. In conclusion, despite the challenges and hurdles, cumulative findings of novel targets and agents in the PI3K/Akt signaling axis are expected to hold promise for advancing AD prevention and treatment.
Collapse
Affiliation(s)
- Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Qi Yao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yankai Wang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Suyan Chang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Chenlong Li
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yongjiang Wu
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Jianhong Shen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Riyun Yang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| |
Collapse
|
3
|
Kim DY, Kim SM, Han IO. Chronic rapid eye movement sleep deprivation aggravates the pathogenesis of Alzheimer's disease by decreasing brain O-GlcNAc cycling in mice. J Neuroinflammation 2024; 21:180. [PMID: 39044290 PMCID: PMC11264383 DOI: 10.1186/s12974-024-03179-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
This study investigated the role of O-GlcNAc cycling in Alzheimer's disease-related changes in brain pathophysiology induced by chronic REM sleep deprivation (CSD) in mice. CSD increased amyloid beta (Aβ) and p-Tau accumulation and impaired learning and memory (L/M) function. CSD decreased dendritic length and spine density. CSD also increased the intensity of postsynaptic density protein-95 (PSD-95) staining. All of these Alzheimer's disease (AD) pathogenic changes were effectively reversed through glucosamine (GlcN) treatment by enhancing O-GlcNAcylation. Interestingly, the lelvel of O-GlcNAcylated-Tau (O-Tau) exhibited an opposite trend compared to p-Tau, as it was elevated by CSD and suppressed by GlcN treatment. CSD increased neuroinflammation, as indicated by elevated levels of glial fibrillary acidic protein and IBA-1-positive glial cells in the brain, which were suppressed by GlcN treatment. CSD promoted the phosphorylation of GSK3β and led to an upregulation in the expression of endoplasmic reticulum (ER) stress regulatory proteins and genes. These alterations were effectively suppressed by GlcN treatment. Minocycline not only suppressed neuroinflammation induced by CSD, but it also rescued the decrease in O-GlcNAc levels caused by CSD. Minocycline also reduced AD neuropathy without affecting CSD-induced ER stress. Notably, overexpressing O-GlcNAc transferase in the dentate gyrus region of the mouse brain rescued CSD-induced cognitive dysfunction, neuropathy, neuroinflammation, and ER stress responses. Collectively, our findings reveal that dysregulation of O-GlcNAc cycling underlies CSD-induced AD pathology and demonstrate that restoration of OGlcNAcylation protects against CSD-induced neurodegeneration.
Collapse
Affiliation(s)
- Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Sang-Min Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea.
| |
Collapse
|
4
|
Richard SA. Elucidating the pivotal molecular mechanisms, therapeutic and neuroprotective effects of lithium in traumatic brain injury. Brain Behav 2024; 14:e3595. [PMID: 38874089 PMCID: PMC11177180 DOI: 10.1002/brb3.3595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Traumatic brain injury (TBI) refers to damage to brain tissue by mechanical or blunt force via trauma. TBI is often associated with impaired cognitive abilities, like difficulties in memory, learning, attention, and other higher brain functions, that typically remain for years after the injury. Lithium is an elementary light metal that is only utilized in salt form due to its high intrinsic reactivity. This current review discusses the molecular mechanisms and therapeutic and neuroprotective effects of lithium in TBI. METHOD The "Boolean logic" was used to search for articles on the subject matter in PubMed and PubMed Central, as well as Google Scholar. RESULTS Lithium's therapeutic action is extremely complex, involving multiple effects on gene secretion, neurotransmitter or receptor-mediated signaling, signal transduction processes, circadian modulation, as well as ion transport. Lithium is able to normalize multiple short- as well as long-term modifications in neuronal circuits that ultimately result in disparity in cortical excitation and inhibition activated by TBI. Also, lithium levels are more distinct in the hippocampus, thalamus, neo-cortex, olfactory bulb, amygdala as well as the gray matter of the cerebellum following treatment of TBI. CONCLUSION Lithium attenuates neuroinflammation and neuronal toxicity as well as protects the brain from edema, hippocampal neurodegeneration, loss of hemispheric tissues, and enhanced memory as well as spatial learning after TBI.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Medicine, Princefield University, Ho, Ghana
- Institute of Neuroscience, Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Khalifa MKA, Abdel-Sattar SA, Amin OM, Kohaf NA, Zaky HS, Abd El-Fattah MA, Mohammed KHA, Badawi NM, Mansoor I, Eassa HA. Effectiveness of epigallocatechin gallate nanoparticles on the in-vivo treatment of Alzheimer's disease in a rat/mouse model: a systematic review. Daru 2024; 32:319-337. [PMID: 38079104 PMCID: PMC11087435 DOI: 10.1007/s40199-023-00494-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/15/2023] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurological disease that causes memory loss over time. Current therapies are limited and frequently inadequate. Epigallocatechin gallate (EGCG), has antioxidant, anti-inflammatory, antifibrosis, anti-remodeling and tissue-protective qualities that may be effective in treatment of different diseases, including AD. Because of nanoparticles' high surface area, they can enhance solubility, stability, pharmacokinetics and biodistribution, and diminish toxicities. Besides, lipid nanoparticles have a high binding affinity that can enhance the rate of drug transport across BBB. So, EGCG nanoparticles represent a promising treatment for AD. OBJECTIVES This systematic review sought to assess the efficacy of EGCG nanoparticles against AD in rat/mouse models. METHODS Study was conducted in accordance with PRISMA guidelines, and the protocol was registered in PROSPERO. Electronic databases were searched to discover relevant studies published up to October 2022. RESULTS Two studies met the inclusion criteria out of 1338 and were included in this systematic review. Collectively, the results indicate that EGCG has a significant potential for reducing AD pathology and improving cognitive deficits in rat/mouse models. The formulated particles were in the nanometer range, as indicated by TEM, with good particle size control and stability. EGCG nanoparticles showed superior pharmacokinetic characteristics and improved blood-brain barrier permeability, and increased brain bioavailability compared to free EGCG. Additionally, nanoEGCG were more effective in modulating oxidative stress than free formulation and decreased AChE in the cortex and hippocampus of AlCl3-treated rats. CONCLUSION This systematic analysis of the two studies included showed that EGCG nanoparticles are efficacious as a potential therapeutic intervention for AD in rat/mouse models. However, limited number of studies found indicates insufficient data in this research point that requires further investigation by experimental studies.
Collapse
Affiliation(s)
- Maha K A Khalifa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11884, Egypt
| | - Somaia A Abdel-Sattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, 11884, Cairo, Egypt
| | - Omnya M Amin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11884, Egypt
| | - Neveen A Kohaf
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo, 11884, Egypt
| | - Heba S Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, 11884, Cairo, Egypt
| | - Marwa A Abd El-Fattah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11884, Egypt
| | - Kamilia H A Mohammed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11884, Egypt
| | - Noha M Badawi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | | | - Heba A Eassa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11884, Egypt.
- Department of Pharmaceutical Sciences, School of Pharmacy & Physician Assistant Studies, University of Saint Joseph, West Hartford, CT, 06117, USA.
| |
Collapse
|
6
|
Hong Y, Sourander C, Hackl B, Patton JS, John J, Paatero I, Coffey E. Jnk1 and downstream signalling hubs regulate anxiety-like behaviours in a zebrafish larvae phenotypic screen. Sci Rep 2024; 14:11174. [PMID: 38750129 PMCID: PMC11096340 DOI: 10.1038/s41598-024-61337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/04/2024] [Indexed: 05/18/2024] Open
Abstract
Current treatments for anxiety and depression show limited efficacy in many patients, indicating the need for further research into the underlying mechanisms. JNK1 has been shown to regulate anxiety- and depressive-like behaviours in mice, however the effectors downstream of JNK1 are not known. Here we compare the phosphoproteomes from wild-type and Jnk1-/- mouse brains and identify JNK1-regulated signalling hubs. We next employ a zebrafish (Danio rerio) larvae behavioural assay to identify an antidepressant- and anxiolytic-like (AA) phenotype based on 2759 measured stereotypic responses to clinically proven antidepressant and anxiolytic (AA) drugs. Employing machine learning, we classify an AA phenotype from extracted features measured during and after a startle battery in fish exposed to AA drugs. Using this classifier, we demonstrate that structurally independent JNK inhibitors replicate the AA phenotype with high accuracy, consistent with findings in mice. Furthermore, pharmacological targeting of JNK1-regulated signalling hubs identifies AKT, GSK-3, 14-3-3 ζ/ε and PKCε as downstream hubs that phenocopy clinically proven AA drugs. This study identifies AKT and related signalling molecules as mediators of JNK1-regulated antidepressant- and anxiolytic-like behaviours. Moreover, the assay shows promise for early phase screening of compounds with anti-stress-axis properties and for mode of action analysis.
Collapse
Affiliation(s)
- Ye Hong
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Christel Sourander
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Benjamin Hackl
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Jedidiah S Patton
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Jismi John
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Eleanor Coffey
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland.
| |
Collapse
|
7
|
Albeely AM, Williams OOF, Blight CR, Thériault RK, Perreault ML. Sex differences in neuronal oscillatory activity and memory in the methylazoxymethanol acetate model of schizophrenia. Schizophr Res 2024; 267:451-461. [PMID: 38643726 DOI: 10.1016/j.schres.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/28/2023] [Accepted: 04/01/2024] [Indexed: 04/23/2024]
Abstract
The methylazoxymethanol acetate (MAM) rodent model is used to study aspects of schizophrenia. However, numerous studies that have employed this model have used only males, resulting in a dearth of knowledge on sex differences in brain function and behaviour. The purpose of this study was to determine whether differences exist between male and female MAM rats in neuronal oscillatory function within and between the prefrontal cortex (PFC), ventral hippocampus (vHIP) and thalamus, behaviour, and in proteins linked to schizophrenia neuropathology. We showed that female MAM animals exhibited region-specific alterations in theta power, elevated low and high gamma power in all regions, and elevated PFC-thalamus high gamma coherence. Male MAM rats had elevated beta and low gamma power in PFC, and elevated vHIP-thalamus coherence. MAM females displayed impaired reversal learning whereas MAM males showed impairments in spatial memory. Glycogen synthase kinase-3 (GSK-3) was altered in the thalamus, with female MAM rats displaying elevated GSK-3α phosphorylation. Male MAM rats showed higher expression and phosphorylation GSK-3α, and higher expression of GSK-β. Sex-specific changes in phosphorylated Tau levels were observed in a region-specific manner. These findings demonstrate there are notable sex differences in behaviour, oscillatory network function, and GSK-3 signaling in MAM rats, thus highlighting the importance of inclusion of both sexes when using this model to study schizophrenia.
Collapse
Affiliation(s)
- Abdalla M Albeely
- Department of Molecular and Cellular Biology, University of Guelph, Ontario, Canada
| | | | - Colin R Blight
- Department of Molecular and Cellular Biology, University of Guelph, Ontario, Canada
| | | | | |
Collapse
|
8
|
Lu YL, Su J, Li JW, Xu WR. A molecular container providing supramolecular protection against acetylcholine hydrolysis. Org Biomol Chem 2024; 22:1634-1638. [PMID: 38323382 DOI: 10.1039/d4ob00024b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline, often attributed to the deficiency of acetylcholine, which can undergo hydrolysis by acetylcholinesterase (AChE) within the biological milieu. Here, we report a supramolecular strategy that takes advantage of confinement effects to inhibit such a hydrolysis process, shedding some light on AD therapy. A water-soluble and bowl-shaped molecule, hexacarboxylated tribenzotriquinacene (TBTQ-C6), was employed to shield acetylcholine (G1) from enzymatic degradation through host-guest binding interactions. Our study revealed highly efficient host-guest interactions with a binding ratio of 1 : 3, resulting in a significant reduction in acetylcholine hydrolysis from 91.1% to 7.4% in the presence of AChE under otherwise identical conditions. Furthermore, TBTQ-C6 showed potential for attenuating the degradation of butyrylcholine (G2) by butyrylcholinesterase (BChE). The broader implications of this study extend to the potential use of molecular containers in various biochemical and pharmacological applications, opening new avenues for research in the field of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Long Lu
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, PR China.
| | - Jing Su
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, PR China.
| | - Jian-Wei Li
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland.
| | - Wen-Rong Xu
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, PR China.
| |
Collapse
|
9
|
Rinauro DJ, Chiti F, Vendruscolo M, Limbocker R. Misfolded protein oligomers: mechanisms of formation, cytotoxic effects, and pharmacological approaches against protein misfolding diseases. Mol Neurodegener 2024; 19:20. [PMID: 38378578 PMCID: PMC10877934 DOI: 10.1186/s13024-023-00651-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/17/2023] [Indexed: 02/22/2024] Open
Abstract
The conversion of native peptides and proteins into amyloid aggregates is a hallmark of over 50 human disorders, including Alzheimer's and Parkinson's diseases. Increasing evidence implicates misfolded protein oligomers produced during the amyloid formation process as the primary cytotoxic agents in many of these devastating conditions. In this review, we analyze the processes by which oligomers are formed, their structures, physicochemical properties, population dynamics, and the mechanisms of their cytotoxicity. We then focus on drug discovery strategies that target the formation of oligomers and their ability to disrupt cell physiology and trigger degenerative processes.
Collapse
Affiliation(s)
- Dillon J Rinauro
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Fabrizio Chiti
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| | - Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY, 10996, USA.
| |
Collapse
|
10
|
Shaik MG, Joshi SV, Akunuri R, Rana P, Rahman Z, Polomoni A, Yaddanapudi VM, Dandekar MP, Srinivas N. Small molecule inhibitors of NLRP3 inflammasome and GSK-3β in the management of traumatic brain injury: A review. Eur J Med Chem 2023; 259:115718. [PMID: 37573828 DOI: 10.1016/j.ejmech.2023.115718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Traumatic brain injury (TBI) is a debilitating mental condition which causes physical disability and morbidity worldwide. TBI may damage the brain by direct injury that subsequently triggers a series of neuroinflammatory events. The activation of NLRP3 inflammasome and dysregulated host immune system has been documented in various neurological disorders such as TBI, ischemic stroke and multiple sclerosis. The activation of NLRP3 post-TBI increases the production of pro-inflammatory cytokines and caspase-1, which are major drivers of neuroinflammation and apoptosis. Similarly, GSK-3β regulates apoptosis through tyrosine kinase and canonical Wnt signalling pathways. Thus, therapeutic targeting of NLRP3 inflammasome and GSK-3β has emerged as promising strategies for regulating the post-TBI neuroinflammation and neurobehavioral disturbances. In this review, we discuss the identification & development of several structurally diverse and pharmacologically interesting small molecule inhibitors for targeting the NLRP3 inflammasome and GSK-3β in the management of TBI.
Collapse
Affiliation(s)
- Mahammad Ghouse Shaik
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Swanand Vinayak Joshi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ravikumar Akunuri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India; Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Preeti Rana
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India
| | - Anusha Polomoni
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India.
| | - Nanduri Srinivas
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India.
| |
Collapse
|
11
|
Pan S, Lan Y, Chen B, Zhou Y, Ying X, Hua Y. Tanshinone IIA changed the amniotic fluid volume and regulated expression of AQP1 and AQP3 in amniotic epithelium cells: a promising drug treating abnormal amniotic fluid volume. Mol Med 2023; 29:83. [PMID: 37386378 DOI: 10.1186/s10020-023-00687-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 06/13/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Many studies have confirmed the association of aquaporins (AQPs) with abnormal amniotic fluid volume (AFV). In our previous experiments, we found that Tanshinone IIA was able to regulate the expression of AQP1 and AQP3. However, the exact mechanism by which Tanshinone IIA regulates AQPs protein expression and its effect on AFV remains unclear. The purpose of this study was to investigate the effects of Tanshinone IIA on AFV and the possible molecular mechanism of regulation of AQP1 and AQP3. METHODS The expression of AQPs protein in the amniotic membranes was compared between pregnant women with normal pregnancy and those with isolated oligohydramnios. The AQP1 knockout (AQP1-KO) mice and wild-type (WT) mice were treated with saline or Tanshinone IIA (10 mg/kg) at 13.5GD and 16.5GD. Human amniotic epithelium cells (hAECs) from pregnant women with normal AFV and isolated oligohydramnios were incubated with 35 μmmol/L Tanshinone IIA or 25 mmol/L LiCl [inhibitor of glycogen synthetic kinase 3β (GSK-3β)]. The protein expressions of AQPs, GSK-3β, phospho-GSK-3β (Ser9) in fetal membranes of mice and human amniotic epithelium cells were detected by western blotting. RESULTS The expression of AQP1 protein in the amniotic membrane of isolated oligohydramnios was increased compared with normal pregnancy. The AFV in AQP1-KO mice is higher than that in WT mice. In wild-type mice, AFV in Tanshinone IIA group was significantly higher than that in control group, and AQP1 protein expression was significantly lower than that in control group, but in AQP1 knockout mice, Tanshinone IIA reduced amniotic fluid volume and AQP3 protein expression at 16.5GD. Tanshinone IIA reduced AQP1, AQP3 and p-GSK-3β (Ser9) protein expression in normal hAECs, and this effect was inhibited by LiCl. In hAECs with oligohydramnios, the down-regulation of AQP1 and up-regulation of AQP3 by Tanshinone IIA was independent of GSK-3β signaling pathway. CONCLUSIONS Tanshinone IIA may increase AFV in normal pregnancy by downregulating AQP1 protein expression in the fetal membranes, which may be associated with p-GSK-3β signaling pathway. But a larger AFV in AQP1-KO mice was significantly attenuated by Tanshinone IIA, which may be related to AQP3. Tanshinone IIA is a promising drug for the treatment of amniotic fluid abnormality.
Collapse
Affiliation(s)
- Shuangjia Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yehui Lan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Baoyi Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yujia Zhou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xinxin Ying
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ying Hua
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
12
|
Yan P, Liu J, Ma H, Feng Y, Cui J, Bai Y, Huang X, Zhu Y, Wei S, Lai J. Effects of glycogen synthase kinase-3β activity inhibition on cognitive, behavioral, and hippocampal ultrastructural deficits in adulthood associated with adolescent methamphetamine exposure. Front Mol Neurosci 2023; 16:1129553. [PMID: 36949769 PMCID: PMC10025487 DOI: 10.3389/fnmol.2023.1129553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Objective Glycogen synthase kinase-3β (GSK3β) has been implicated in the maintenance of synaptic plasticity, memory process, and psychostimulant-induced behavioral effects. Hyperactive GSK3β in the Cornu Ammonis 1 (CA1) subregion of the dorsal hippocampus (DHP) was associated with adolescent methamphetamine (METH) exposure-induced behavioral and cognitive deficits in adulthood. This study aimed to evaluate the possible therapeutic effects of GSK3β inhibition in adulthood on adolescent METH exposure-induced long-term neurobiological deficits. Methods Adolescent male mice were treated with METH from postnatal day (PND) 45-51. In adulthood, three intervention protocols (acute lithium chloride systemic administration, chronic lithium chloride systemic administration, and chronic SB216763 administration within CA1) were used for GSK3β activity inhibition. The effect of GSK3β intervention on cognition, behavior, and GSK3β activity and synaptic ultrastructure in the DHP CA1 subregion were detected in adulthood. Results In adulthood, all three interventions reduced adolescent METH exposure-induced hyperactivity (PND97), while only chronic systemic and chronic within CA1 administration ameliorated the induced impairments in spatial (PND99), social (PND101) and object (PND103) recognition memory. In addition, although three interventions reversed the aberrant GSK3β activity in the DHP CA1 subregion (PND104), only chronic systemic and chronic within CA1 administration rescued adolescent METH exposure-induced synaptic ultrastructure changes in the DHP CA1 subregion (PND104) in adulthood. Conclusion Rescuing synaptic ultrastructural abnormalities in the dHIP CA1 subregion by chronic administration of a GSK3β inhibitor may be a suitable therapeutic strategy for the treatment of behavioral and cognitive deficits in adulthood associated with adolescent METH abuse.
Collapse
Affiliation(s)
- Peng Yan
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Jincen Liu
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Haotian Ma
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Yue Feng
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Jingjing Cui
- Forensic Identification Institute, The Fourth People’s Hospital of Yancheng, Yancheng, China
| | - Yuying Bai
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Xin Huang
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Yongsheng Zhu
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Shuguang Wei
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Shuguang Wei,
| | - Jianghua Lai
- NHC Key Laboratory of Forensic Science, School of Forensic Sciences, Xi’an Jiaotong University, Xi’an, China
- Jianghua Lai,
| |
Collapse
|
13
|
Hottin C, Perron M, Roger JE. GSK3 Is a Central Player in Retinal Degenerative Diseases but a Challenging Therapeutic Target. Cells 2022; 11:cells11182898. [PMID: 36139472 PMCID: PMC9496697 DOI: 10.3390/cells11182898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. Although recent works strongly suggest that inhibiting GSK3 might also be a promising therapeutic approach for retinal degenerative diseases, its full potential is still under-evaluated. In this review, we summarize the literature on the role of GSK3 on the main cellular functions reported as deregulated during retinal degeneration, such as glucose homeostasis which is critical for photoreceptor survival, or oxidative stress, a major component of retinal degeneration. We also discuss the interest in targeting GSK3 for its beneficial effects on inflammation, for reducing neovascularization that occurs in some retinal dystrophies, or for cell-based therapy by enhancing Müller glia cell proliferation in diseased retina. Together, although GSK3 inhibitors hold promise as therapeutic agents, we highlight the complexity of targeting such a multitasked kinase and the need to increase our knowledge of the impact of reducing GSK3 activity on these multiple cellular pathways and biological processes.
Collapse
Affiliation(s)
- Catherine Hottin
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
14
|
Wang N, Liu W, Zhou L, Liu W, Liang X, Liu X, Xu Z, Zhong T, Wu Q, Jiao X, Chen J, Ning X, Jiang X, Zhao Q. Design, Synthesis, and Biological Evaluation of Notopterol Derivatives as Triple Inhibitors of AChE/BACE1/GSK3β for the Treatment of Alzheimer's Disease. ACS OMEGA 2022; 7:32131-32152. [PMID: 36120034 PMCID: PMC9476211 DOI: 10.1021/acsomega.2c03368] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/19/2022] [Indexed: 06/15/2023]
Abstract
The pathogenesis of Alzheimer's disease (AD) is very complex, and there are many hypotheses. Therefore, the development of a multi-target-directed-ligand may be an effective therapeutic strategy. Our previous study showed that notopterol (a natural product from Notopterygium) is a dual BACE1/GSK3β inhibitor. In this study, we designed and synthesized 48 notopterol derivatives with furacoumarin as a scaffold in order to enhance their balanced AChE/BACE1/GSK3β inhibitory activity. Fortunately, 1c showed effective inhibitory activity against AChE (58.7% at 1.0 μM), BACE1 (48.3% at 20 μM), and GSK3β (40.3% at 10 μM). Furthermore, 1c showed good blood-brain barrier penetrability, suitable bioavailability, and oral safety. More importantly, 1c could ameliorate the impaired learning and memory in Aβ-induced AD mice. In conclusion, we reported the triple inhibitor of AChE/BACE1/GSK3β lead compounds based on a furocoumarin scaffold of notopterol for the first time, which provides a potential new strategy for the treatment of AD.
Collapse
Affiliation(s)
- Nan Wang
- Department
of Pharmacy, General Hospital of Northern
Theater Command, Shenyang110840, People’s Republic
of China
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Wenjie Liu
- School
of Traditional Chinese Materia Medica, Shenyang
Pharmaceutical University, Shenyang110016, People’s Republic
of China
| | - Lijun Zhou
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Wenwu Liu
- School
of Traditional Chinese Materia Medica, Shenyang
Pharmaceutical University, Shenyang110016, People’s Republic
of China
| | - Xu Liang
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Xin Liu
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Zihua Xu
- Department
of Pharmacy, General Hospital of Northern
Theater Command, Shenyang110840, People’s Republic
of China
| | - Tianming Zhong
- Department
of Pharmacy, General Hospital of Northern
Theater Command, Shenyang110840, People’s Republic
of China
| | - Qiong Wu
- Department
of Pharmacy, General Hospital of Northern
Theater Command, Shenyang110840, People’s Republic
of China
| | - Xinming Jiao
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Jiangxia Chen
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Xinyue Ning
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Xiaowen Jiang
- Department
of Pharmacy, General Hospital of Northern
Theater Command, Shenyang110840, People’s Republic
of China
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
- Key
Laboratory of Structure-Based Drug Design & Discovery, Ministry
of Education, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Qingchun Zhao
- Department
of Pharmacy, General Hospital of Northern
Theater Command, Shenyang110840, People’s Republic
of China
- School
of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| |
Collapse
|
15
|
Bahlakeh G, Rahbarghazi R, Abedelahi A, Sadigh-Eteghad S, Karimipour M. Neurotrophic factor-secreting cells restored endogenous hippocampal neurogenesis through the Wnt/β-catenin signaling pathway in AD model mice. Stem Cell Res Ther 2022; 13:343. [PMID: 35883119 PMCID: PMC9327342 DOI: 10.1186/s13287-022-03024-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/28/2022] [Indexed: 12/26/2022] Open
Abstract
Background Impairment in neurogenesis correlates with memory and cognitive dysfunction in AD patients. In the recent decade, therapies with stem cell bases are growing and proved to be efficient. This study is a preliminary attempt to explore the impact of NTF-SCs on hippocampal neurogenesis mediated by the Wnt/β-catenin signaling cascade in AD-like mouse brain parenchyma. Methods The BALB/c mice were divided into four groups: Control, AD +Vehicle, AD+ TF-SCs-CM and AD+NTF-SCs (n = 10). For AD induction, 100 µM Aβ1-42 was injected into lateral ventricles. The AD-like model was confirmed via passive avoidance test and Thioflavin-S staining 21 days following Aβ injection. Next, NTF-SCs were differentiated from ADMSCs, and both NTF-SCs and supernatant (NTF-SCs-CM) were injected into the hippocampus after AD confirmation. Endogenous neural stem cells (NSCs) proliferation capacity was assessed after 50 mg/kbW BrdU injection for 4 days using immunofluorescence (IF) staining. The percent of BrdU/Nestin and BrdU/NeuN positive NSCs were calculated. Real-time RT-PCR was used to detect genes related to the Wnt/β-catenin signaling cascade. The spatial learning and memory alternation was evaluated using the Morris water maze (MWM). Results Data showed the reduction in escape latency over 5 days in the AD mice compared to the control group. The administration of NTF-SCs and NTF-SCs-CM increased this value compared to the AD-Vehicle group. Both NTF-SCs and NTF-SCs-CM were the potential to reduce the cumulative distance to the platform in AD mice compared to the AD-Vehicle group. The time spent in target quadrants was ameliorated following NTF-SCs and NTF-SCs-CM transplantation followed by an improved MWM performance. IF imaging revealed the increase in BrdU/Nestin+ and BrdU/NeuN+ in AD mice that received NTF-SCs and NTF-SCs-CM, indicating enhanced neurogenesis. Based on real-time PCR analysis, the expression of PI3K, Akt, MAPK, ERK, Wnt, and β-catenin was upregulated and coincided with the suppression of GSK-3β after injection of NTF-SCs-CM and NTF-SCs. In this study, NTF-SCs had superior effects in AD mice that received NTF-SCs compared to NTF-SCs-CM. Conclusions The activation of Wnt/β-catenin pathway via NTF-SCs can be touted as a possible therapeutic approach to restore neurogenesis in AD mice.
Collapse
Affiliation(s)
- Gozal Bahlakeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Zhou Q, Li S, Li M, Ke D, Wang Q, Yang Y, Liu GP, Wang XC, Liu E, Wang JZ. Human tau accumulation promotes glycogen synthase kinase-3β acetylation and thus upregulates the kinase: A vicious cycle in Alzheimer neurodegeneration. EBioMedicine 2022; 78:103970. [PMID: 35339896 PMCID: PMC8956943 DOI: 10.1016/j.ebiom.2022.103970] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/12/2022] [Accepted: 03/13/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Glycogen synthase kinase-3β (GSK-3β) is one of the most effective kinases in promoting tau hyperphosphorylation and accumulation in Alzheimer's disease (AD). However, it is not clear how GSK-3β activity is regulated during AD progression. METHODS We firstly used mass spectrometry to identify the acetylation site of GSK-3β, and then established the cell and animal models of GSK-3β acetylation. Next, we conducted molecular, cell biological and behavioral tests. Finally, we designed a peptide to test whether blocking tau-mediated GSK-3β acetylation could be beneficial to AD. FINDINGS We found that GSK-3β protein levels increased in the brains of AD patients and the transgenic mice. Overexpressing tau increased GSK-3β protein level with increased acetylation and decreased ubiquitination-related proteolysis. Tau could directly acetylate GSK-3β at K15 both in vitro and in vivo. K15-acetylation inhibited ubiquitination-associated proteolysis of GSK-3β and changed its activity-dependent phosphorylation, leading to over-activation of the kinase. GSK-3β activation by K15-acetylation in turn exacerbated the AD-like pathologies. Importantly, competitively inhibiting GSK-3β K15-acetylation by a novel-designed peptide remarkably improved cognitive impairment and the AD-like pathologies in 3xTg-AD mice. INTERPRETATION Tau can directly acetylate GSK-3β at K15 which reveals a vicious cycle between tau hyperphosphorylation and GSK-3β activation. FUNDING This study was supported in parts by grants from Science and Technology Committee of China (2016YFC1305800), Hubei Province (2018ACA142), Natural Science Foundation of China (91949205, 82001134, 31730035, 81721005), Guangdong Provincial Key S&T Program (018B030336001).
Collapse
Affiliation(s)
- Qiuzhi Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shihong Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengzhu Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Enjie Liu
- Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China.
| |
Collapse
|
17
|
Gomaa AA, Farghaly HS, Ahmed AM, El-Mokhtar MA, Hemida FK. Advancing combination treatment with cilostazol and caffeine for Alzheimer's disease in high fat-high fructose-STZ induced model of amnesia. Eur J Pharmacol 2022; 921:174873. [DOI: 10.1016/j.ejphar.2022.174873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 11/25/2022]
|
18
|
Natural products as novel scaffolds for the design of glycogen synthase kinase 3β inhibitors. Expert Opin Drug Discov 2022; 17:377-396. [PMID: 35262427 DOI: 10.1080/17460441.2022.2043845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The different and relevant roles of GSK-3 are of critical importance since they deal with development, metabolic homeostasis, cell polarity and fate, neuronal growth and differentiation as well as modulation of apoptotic potential. Given their involvement with different diseases, many investigations have been undertaken with the aim of discovering new and promising inhibitors for this target. In this context, atural products represent an invaluable source of active molecules. AREAS COVERED In order to overcome issues such as poor pharmacokinetic properties or efficacy, frequently associated with natural compounds, different GSK-3β inhibitors belonging to alkaloid or flavonoid classes have been subjected to structural modifications in order to obtain more potent and safer compounds. Herein, the authors report the results obtained from studies where natural compounds have been used as hits with the aim of providing new kinase inhibitors endowed with a better inhibitory profile. EXPERT OPINION Structurally modification of natural scaffolds is a proven approach taking advantage of their pharmacological characteristics. Indeed, whatever the strategy adopted is and, despite the limitations associated with the structural complexity of natural products, the authors recommend the use of natural scaffolds as a promising strategy for the discovery of novel and potent GSK-3β inhibitors.
Collapse
|
19
|
Lee Y, Bortolotto ZA, Bradley CA, Sanderson TM, Zhuo M, Kaang BK, Collingridge GL. The GSK-3 Inhibitor CT99021 Enhances the Acquisition of Spatial Learning and the Accuracy of Spatial Memory. Front Mol Neurosci 2022; 14:804130. [PMID: 35153671 PMCID: PMC8829050 DOI: 10.3389/fnmol.2021.804130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK-3) is a Ser/Thr protein kinase that regulates many cellular processes, including synaptic plasticity. Previously, we reported that inhibition of GSK-3 prevents the induction of one of the major forms of synaptic plasticity, N-methyl-D-aspartate receptor (NMDAR)-dependent long-term depression (LTD), in hippocampal slices. In the present study, we have investigated the effects of inhibiting GSK-3 on learning and memory in healthy naïve animals. Systemic administration of a highly selective GSK-3 inhibitor, CT99021, reversibly blocked NMDAR-dependent LTD in the CA1 region of the hippocampus in anesthetized adult mice. In behavioral tasks, CT99021 had no effect on locomotor activity, anxiety, hippocampus-dependent contextual fear memory, and hippocampus-dependent reversal learning. However, CT99021 facilitated the rate of learning in the Morris water maze (MWM) and T-maze and enhanced the accuracy of long-term spatial memory in the MWM. These findings suggest that GSK-3 regulates the accuracy of spatial memory acquisition and recall.
Collapse
Affiliation(s)
- Yeseul Lee
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Zuner A. Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Clarrisa A. Bradley
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Genes and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Thomas M. Sanderson
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Min Zhuo
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Bong-Kiun Kaang
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- *Correspondence: Bong-Kiun Kaang,
| | - Graham L. Collingridge
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
- Graham L. Collingridge,
| |
Collapse
|
20
|
Arciniegas Ruiz SM, Eldar-Finkelman H. Glycogen Synthase Kinase-3 Inhibitors: Preclinical and Clinical Focus on CNS-A Decade Onward. Front Mol Neurosci 2022; 14:792364. [PMID: 35126052 PMCID: PMC8813766 DOI: 10.3389/fnmol.2021.792364] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022] Open
Abstract
The protein kinase, GSK-3, participates in diverse biological processes and is now recognized a promising drug discovery target in treating multiple pathological conditions. Over the last decade, a range of newly developed GSK-3 inhibitors of diverse chemotypes and inhibition modes has been developed. Even more conspicuous is the dramatic increase in the indications that were tested from mood and behavior disorders, autism and cognitive disabilities, to neurodegeneration, brain injury and pain. Indeed, clinical and pre-clinical studies were largely expanded uncovering new mechanisms and novel insights into the contribution of GSK-3 to neurodegeneration and central nerve system (CNS)-related disorders. In this review we summarize new developments in the field and describe the use of GSK-3 inhibitors in the variety of CNS disorders. This remarkable volume of information being generated undoubtedly reflects the great interest, as well as the intense hope, in developing potent and safe GSK-3 inhibitors in clinical practice.
Collapse
|
21
|
Insulin-Induced Recurrent Hypoglycemia Up-Regulates Glucose Metabolism in the Brain Cortex of Chemically Induced Diabetic Rats. Int J Mol Sci 2021; 22:ijms222413470. [PMID: 34948265 PMCID: PMC8708764 DOI: 10.3390/ijms222413470] [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: 10/29/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetes is a chronic metabolic disease that seriously compromises human well-being. Various studies highlight the importance of maintaining a sufficient glucose supply to the brain and subsequently safeguarding cerebral glucose metabolism. The goal of the present work is to clarify and disclose the metabolic alterations induced by recurrent hypoglycemia in the context of long-term hyperglycemia to further comprehend the effects beyond brain harm. To this end, chemically induced diabetic rats underwent a protocol of repeatedly insulin-induced hypoglycemic episodes. The activity of key enzymes of glycolysis, the pentose phosphate pathway and the Krebs cycle was measured by spectrophotometry in extracts or isolated mitochondria from brain cortical tissue. Western blot analysis was used to determine the protein content of glucose and monocarboxylate transporters, players in the insulin signaling pathway and mitochondrial biogenesis and dynamics. We observed that recurrent hypoglycemia up-regulates the activity of mitochondrial hexokinase and Krebs cycle enzymes (namely, pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase and succinate dehydrogenase) and the protein levels of mitochondrial transcription factor A (TFAM). Both insults increased the nuclear factor erythroid 2–related factor 2 (NRF2) protein content and induced divergent effects in mitochondrial dynamics. Insulin-signaling downstream pathways were found to be down-regulated, and glycogen synthase kinase 3 beta (GSK3β) was found to be activated through both decreased phosphorylation at Ser9 and increased phosphorylation at Y216. Interestingly, no changes in the levels of cAMP response element-binding protein (CREB), which plays a key role in neuronal plasticity and memory, were caused by hypoglycemia and/or hyperglycemia. These findings provide experimental evidence that recurrent hypoglycemia, in the context of chronic hyperglycemia, has the capacity to evoke coordinated adaptive responses in the brain cortex that will ultimately contribute to sustaining brain cell health.
Collapse
|
22
|
Zhang H, Han Y, Zhang L, Jia X, Niu Q. The GSK-3β/β-Catenin Signaling-Mediated Brain-Derived Neurotrophic Factor Pathway Is Involved in Aluminum-Induced Impairment of Hippocampal LTP In Vivo. Biol Trace Elem Res 2021; 199:4635-4645. [PMID: 33462795 DOI: 10.1007/s12011-021-02582-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/06/2021] [Indexed: 12/20/2022]
Abstract
The neurotoxic effects of aluminum (Al) are associated with the impairment of synaptic plasticity, the biological basis of learning and memory, the major form of which is long-term potentiation (LTP). The canonical glycogen synthase kinase-3β (GSK-3β)/β-catenin signaling-mediated brain-derived neurotrophic factor (BDNF) pathway has been suggested to play important roles in memory. Thus, Al may affect LTP through this pathway. In this study, a Sprague-Dawley rat model of neurotoxicity was established through intracerebroventricular (i.c.v.) injection of aluminum maltol (Al(mal)3), which was achieved by preimplantation of a cannula into the lateral ventricle. The rats in the control and Al-treated groups received a daily injection of SB216763, an inhibitor of GSK-3β. Electrophysiology and western blot analysis were used to investigate the regulatory effect of the GSK-3β/β-catenin signaling-mediated BDNF pathway on LTP impairment induced by Al(mal)3. The results confirmed that i.c.v. injection of Al(mal)3 significantly suppressed the field excitatory postsynaptic potential (fEPSP) amplitude, as indicated by a decrease in BDNF protein expression, which was accompanied by dose-dependent decreases in β-catenin protein expression and the phosphorylation of GSK-3β at Ser9. Rats that received SB216763, a GSK-3β inhibitor, exhibited higher fEPSP amplitudes than control rats. Furthermore, SB216763 treatment upregulated the hippocampal protein expression of BDNF and β-catenin while increasing the ratio of p-GSK-3β/GSK-3β. From the perspective of the identified β-catenin-BDNF axis, Al impairs hippocampal LTP, possibly through the GSK-3β/β-catenin signaling-mediated BDNF pathway.
Collapse
Affiliation(s)
- Huifang Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China
| | - Yingchao Han
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China
| | - Ling Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China
| | - Xiaofang Jia
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China.
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
23
|
Xu QQ, Shaw PC, Hu Z, Yang W, Ip SP, Xian YF, Lin ZX. Comparison of the chemical constituents and anti-Alzheimer's disease effects of Uncaria rhynchophylla and Uncaria tomentosa. Chin Med 2021; 16:110. [PMID: 34706756 PMCID: PMC8555092 DOI: 10.1186/s13020-021-00514-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/06/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Uncaria tomentosa, which has similar chemical constituents with Uncaria rhynchophylla, has been reported to alleviate cognitive impairments in Alzheimer's disease (AD) animal models. This study aimed to compare the chemical constituents and anti-AD effect of the ethanol extracts of U. tomentosa (UTE) and U. rhynchophylla (URE). METHODS The high-performance liquid chromatography (HPLC) was used to compare the chemical constituents of UTE and URE. Streptozotocin (STZ) was intracerebroventricularly (ICV) injected into adult male Sprague-Dawley (SD) rats to establish AD model. UTE (400 mg/kg) or URE (400 mg/kg) was administrated intragastrically once daily to the rats for 6 consecutive weeks. Morris water maze (MWM) test was conducted to assess the neurological functions in the STZ-induced AD rats. The brain tissues of the rats were harvested for further biochemical assay. RESULTS The MWM test results showed both UTE and URE could significantly improve the learning and memory impairments induced by STZ in rats. Both UTE and URE could significantly inhibit the hyperphosphorylation of tau protein, reduce the elevated levels of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α), enhance activities of antioxidant enzymes (SOD, CAT and GPx) and increase the protein expression of HO-1. In addition, UTE could decrease the malondialdehyde (MDA) level. Furthermore, both UTE and URE significantly enhanced Akt activation, down regulated the activation of glycogen synthase kinase 3β (GSK-3β), and induced the nuclear translocation of Nrf2 in the STZ-induced AD rats. CONCLUSIONS UTE and URE contained similar chemical constituents. We found for the first time that both of them could ameliorate cognitive deficits in the STZ-induced AD rats. The underlying molecular mechanism involve suppression of tau hyperphosphorylation, anti-oxidant and anti-neuroinflammation via modulating Akt (Ser473)/GSK3β (Ser9)-mediated Nrf2 activation. These findings amply implicate that both of UTE and URE are worthy of being developed clinically into pharmaceutical treatment for AD.
Collapse
Affiliation(s)
- Qing-Qing Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Pang Chui Shaw
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zhen Hu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Wen Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Siu-Po Ip
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| |
Collapse
|
24
|
Westmark PR, Garrone B, Ombrato R, Milanese C, Di Giorgio FP, Westmark CJ. Testing Fmr1 KO Phenotypes in Response to GSK3 Inhibitors: SB216763 versus AFC03127. Front Mol Neurosci 2021; 14:751307. [PMID: 34690696 PMCID: PMC8529056 DOI: 10.3389/fnmol.2021.751307] [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: 07/31/2021] [Accepted: 09/15/2021] [Indexed: 11/15/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a proline-directed serine-threonine kinase that is associated with several neurological disorders, including Alzheimer’s disease and fragile X syndrome (FXS). We tested the efficacy of a novel GSK3 inhibitor AFC03127, which was developed by Angelini Pharma, in comparison to the metabotropic glutamate receptor 5 inhibitor 2-Methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP) and the GSK3 inhibitor SB216763 in in vivo and in vitro assays in Fmr1KO mice, a mouse model useful for the study of FXS. The in vivo assay tested susceptibility to audiogenic-induced seizures (AGS) whereas the in vitro assays assessed biomarker expression and dendritic spine length and density in cultured primary neurons as a function of drug dose. MPEP and SB216763 attenuated AGS in Fmr1KO mice, whereas AFC03127 did not. MPEP and AFC03127 significantly reduced dendritic expression of amyloid-beta protein precursor (APP). All drugs rescued spine length and the ratio of mature dendritic spines. Spine density was not statistically different between vehicle and GSK3 inhibitor-treated cells. The drugs were tested over a wide concentration range in the in vitro assays to determine dose responses. A bell-shaped dose response decrease in APP expression was observed in response to AFC03127, which was more effective than SB216763. These findings confirm previous studies demonstrating differential effects of various GSK3 inhibitors on AGS propensity in Fmr1KO mice and confirm APP as a downstream biomarker that is responsive to GSK3 activity.
Collapse
Affiliation(s)
- Pamela R Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
| | | | | | | | | | - Cara J Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States.,Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
25
|
Reilley DJ, Arraf Z, Alexandrova AN. Contrasting Effects of Inhibitors Li + and Be 2+ on Catalytic Cycle of Glycogen Synthase Kinase-3β. J Phys Chem B 2021; 125:9480-9489. [PMID: 34404214 DOI: 10.1021/acs.jpcb.1c05099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ionic lithium shows rare effectiveness for treating bipolar disorder and is a potential drug for neurodegenerative diseases. Unfortunately, lithium suffers from significant drawbacks, mainly a narrow therapeutic window. Among the targets of lithium, glycogen synthase kinase 3β (GSK-3β) may be responsible for its therapeutic effects. The development of alternative, selective inhibitors of this kinase could prevent lithium side effects, but such efforts have met little success so far. An atomistic understanding of Li+ inhibition and the GSK-3β phosphorylation reaction would therefore facilitate the development of new drugs. In this study, we use extensive sampling of catalytic states with our mixed quantum-classical dynamics method QM/DMD and binding affinities from a competitive metal affinity (CMA) approach to expand the atomistic picture of Li+ GSK-3β inhibition. We compare Li+ action with Be2+ and find our results in agreement with in vitro kinetics studies. Ultimately, our simulations show that Li+ inhibition is driven by decreasing the phosphorylation reaction rate, rather than reducing catalytic turnover through tight binding to different GSK-3β states like Be2+ inhibition. The effect of these metals derive from electrostatic differences and especially their smaller atomic radii compared to the native Mg2+ and thus provide insight for the development of GSK-3β inhibitors based on other paradigms.
Collapse
Affiliation(s)
- David J Reilley
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095-1569, United States
| | - Zaher Arraf
- Department of Education in Technology and Science, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Anastassia N Alexandrova
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095-1569, United States.,California NanoSystems Institute, Los Angeles California 90095-1569, United States
| |
Collapse
|
26
|
Telias M, Ben-Yosef D. Pharmacological Manipulation of Wnt/β-Catenin Signaling Pathway in Human Neural Precursor Cells Alters Their Differentiation Potential and Neuronal Yield. Front Mol Neurosci 2021; 14:680018. [PMID: 34421534 PMCID: PMC8371257 DOI: 10.3389/fnmol.2021.680018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/15/2021] [Indexed: 11/13/2022] Open
Abstract
The canonical Wnt/β-catenin pathway is a master-regulator of cell fate during embryonic and adult neurogenesis and is therefore a major pharmacological target in basic and clinical research. Chemical manipulation of Wnt signaling during in vitro neuronal differentiation of stem cells can alter both the quantity and the quality of the derived neurons. Accordingly, the use of Wnt activators and blockers has become an integral part of differentiation protocols applied to stem cells in recent years. Here, we investigated the effects of the glycogen synthase kinase-3β inhibitor CHIR99021, which upregulates β-catenin agonizing Wnt; and the tankyrase-1/2 inhibitor XAV939, which downregulates β-catenin antagonizing Wnt. Both drugs and their potential neurogenic and anti-neurogenic effects were studied using stable lines human neural precursor cells (hNPCs), derived from embryonic stem cells, which can be induced to generate mature neurons by chemically-defined conditions. We found that Wnt-agonism by CHIR99021 promotes induction of neural differentiation, while also reducing cell proliferation and survival. This effect was not synergistic with those of pro-neural growth factors during long-term neuronal differentiation. Conversely, antagonism of Wnt by XAV939 consistently prevented neuronal progression of hNPCs. We show here how these two drugs can be used to manipulate cell fate and how self-renewing hNPCs can be used as reliable human in vitro drug-screening platforms.
Collapse
Affiliation(s)
- Michael Telias
- Wolfe PGD-SC Lab, Racine IVF Unit, Department of Cell and Developmental Biology, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Medical School, Tel-Aviv University, Tel Aviv, Israel
| | - Dalit Ben-Yosef
- Wolfe PGD-SC Lab, Racine IVF Unit, Department of Cell and Developmental Biology, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Medical School, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
27
|
Role of a Heat Shock Transcription Factor and the Major Heat Shock Protein Hsp70 in Memory Formation and Neuroprotection. Cells 2021; 10:cells10071638. [PMID: 34210082 PMCID: PMC8305005 DOI: 10.3390/cells10071638] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
Heat shock proteins (Hsps) represent the most evolutionarily ancient, conserved, and universal system for protecting cells and the whole body from various types of stress. Among Hsps, the group of proteins with a molecular weight of 70 kDa (Hsp70) plays a particularly important role. These proteins are molecular chaperones that restore the native conformation of partially denatured proteins after exposure to proteotoxic forms of stress and are critical for the folding and intracellular trafficking of de novo synthesized proteins under normal conditions. Hsp70s are expressed at high levels in the central nervous system (CNS) of various animals and protect neurons from various types of stress, including heat shock, hypoxia, and toxins. Numerous molecular and behavioral studies have indicated that Hsp70s expressed in the CNS are important for memory formation. These proteins contribute to the folding and transport of synaptic proteins, modulate signaling cascades associated with synaptic activation, and participate in mechanisms of neurotransmitter release. In addition, HSF1, a transcription factor that is activated under stress conditions and mediates Hsps transcription, is also involved in the transcription of genes encoding many synaptic proteins, whose levels are increased in neurons under stress and during memory formation. Thus, stress activates the molecular mechanisms of memory formation, thereby allowing animals to better remember and later avoid potentially dangerous stimuli. Finally, Hsp70 has significant protective potential in neurodegenerative diseases. Increasing the level of endogenous Hsp70 synthesis or injecting exogenous Hsp70 reduces neurodegeneration, stimulates neurogenesis, and restores memory in animal models of ischemia and Alzheimer’s disease. These findings allow us to consider recombinant Hsp70 and/or Hsp70 pharmacological inducers as potential drugs for use in the treatment of ischemic injury and neurodegenerative disorders.
Collapse
|
28
|
Novel treatments for autism spectrum disorder based on genomics and systems biology. Pharmacol Ther 2021; 230:107939. [PMID: 34174273 DOI: 10.1016/j.pharmthera.2021.107939] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder with a complex underlying genetic architecture. There are currently no known pharmacologic treatments for the core ASD symptoms of social deficits and restricted/ repetitive behavior. However, there are dozens of clinical trials currently underway that are testing the impact of novel and existing agents on core and associated symptoms in ASD. METHODS We present a narrative synthesis of the historical and contemporary challenges to drug discovery in ASD. We then provide an overview of novel treatments currently under investigation from a genomics and systems biology perspective. RESULTS Data driven network and cluster analyses suggest alterations in transcriptional regulation, chromatin remodelling, synaptic transmission, neuropeptide signalling, and/or immunological mechanisms may contribute to or underlie the development of ASD. Agents and upcoming trials targeting each of the above listed systems are reviewed. CONCLUSION Identifying effective pharmacologic treatments for the core and associated symptom domains in ASD will require further collaboration and innovation in the areas of outcome measurement, biomarker research, and genomics, as well as systematic efforts to identify and treat subgroups of individuals with ASD who may be differentially responsive to specific treatments.
Collapse
|
29
|
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
|
30
|
Kim JH, Lee J, Choi WH, Park S, Park SH, Lee JH, Lim SM, Mun JY, Cho HS, Han D, Suh YH, Lee MJ. CHIP-mediated hyperubiquitylation of tau promotes its self-assembly into the insoluble tau filaments. Chem Sci 2021; 12:5599-5610. [PMID: 34168795 PMCID: PMC8179656 DOI: 10.1039/d1sc00586c] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/05/2021] [Indexed: 01/08/2023] Open
Abstract
The tau protein is a highly soluble and natively unfolded protein. Under pathological conditions, tau undergoes multiple post-translational modifications (PTMs) and conformational changes to form insoluble filaments, which are the proteinaceous signatures of tauopathies. To dissect the crosstalk among tau PTMs during the aggregation process, we phosphorylated and ubiquitylated recombinant tau in vitro using GSK3β and CHIP, respectively. The resulting phospho-ub-tau contained conventional polyubiquitin chains with lysine 48 linkages, sufficient for proteasomal degradation, whereas unphosphorylated ub-tau species retained only one-three ubiquitin moieties. Mass-spectrometric analysis of in vitro reconstituted phospho-ub-tau revealed seven additional ubiquitylation sites, some of which are known to stabilize tau protofilament stacking in the human brain with tauopathy. When the ubiquitylation reaction was prolonged, phospho-ub-tau transformed into insoluble hyperubiquitylated tau species featuring fibrillar morphology and in vitro seeding activity. We developed a small-molecule inhibitor of CHIP through biophysical screening; this effectively suppressed tau ubiquitylation in vitro and delayed its aggregation in cultured cells including primary cultured neurons. Our biochemical findings point to a "multiple-hit model," where sequential events of tau phosphorylation and hyperubiquitylation function as a key driver of the fibrillization process, thus indicating that targeting tau ubiquitylation may be an effective strategy to alleviate the course of tauopathies.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department of Biomedical Sciences, Seoul National University Graduate School Seoul 03080 Korea +82 2-744-4534 +82 2-740-8254
| | - Jeeyoung Lee
- Department of Biomedical Sciences, Seoul National University Graduate School Seoul 03080 Korea +82 2-744-4534 +82 2-740-8254
| | - Won Hoon Choi
- Department of Biomedical Sciences, Seoul National University Graduate School Seoul 03080 Korea +82 2-744-4534 +82 2-740-8254
| | - Seoyoung Park
- Department of Biochemistry & Molecular Biology, Neuroscience Research Institute, Seoul National University College of Medicine Seoul 03080 Korea
| | - Seo Hyeong Park
- Department of Biomedical Sciences, Seoul National University Graduate School Seoul 03080 Korea +82 2-744-4534 +82 2-740-8254
| | - Jung Hoon Lee
- Department of Biochemistry & Molecular Biology, Neuroscience Research Institute, Seoul National University College of Medicine Seoul 03080 Korea
| | - Sang Min Lim
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology Seoul 02792 Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute Daegu 41062 Korea
| | - Hyun-Soo Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University Seoul 03722 Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital Seoul 03080 Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, Seoul National University Graduate School Seoul 03080 Korea +82 2-744-4534 +82 2-740-8254
- Department of Biochemistry & Molecular Biology, Neuroscience Research Institute, Seoul National University College of Medicine Seoul 03080 Korea
| | - Min Jae Lee
- Department of Biomedical Sciences, Seoul National University Graduate School Seoul 03080 Korea +82 2-744-4534 +82 2-740-8254
- Department of Biochemistry & Molecular Biology, Neuroscience Research Institute, Seoul National University College of Medicine Seoul 03080 Korea
| |
Collapse
|
31
|
Duchon A, Del Mar Muniz Moreno M, Martin Lorenzo S, Silva de Souza MP, Chevalier C, Nalesso V, Meziane H, Loureiro de Sousa P, Noblet V, Armspach JP, Brault V, Herault Y. Multi-influential genetic interactions alter behaviour and cognition through six main biological cascades in Down syndrome mouse models. Hum Mol Genet 2021; 30:771-788. [PMID: 33693642 PMCID: PMC8161522 DOI: 10.1093/hmg/ddab012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is the most common genetic form of intellectual disability caused by the presence of an additional copy of human chromosome 21 (Hsa21). To provide novel insights into genotype–phenotype correlations, we used standardized behavioural tests, magnetic resonance imaging and hippocampal gene expression to screen several DS mouse models for the mouse chromosome 16 region homologous to Hsa21. First, we unravelled several genetic interactions between different regions of chromosome 16 and how they contribute significantly to altering the outcome of the phenotypes in brain cognition, function and structure. Then, in-depth analysis of misregulated expressed genes involved in synaptic dysfunction highlighted six biological cascades centred around DYRK1A, GSK3β, NPY, SNARE, RHOA and NPAS4. Finally, we provide a novel vision of the existing altered gene–gene crosstalk and molecular mechanisms targeting specific hubs in DS models that should become central to better understanding of DS and improving the development of therapies.
Collapse
Affiliation(s)
- Arnaud Duchon
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Maria Del Mar Muniz Moreno
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Sandra Martin Lorenzo
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Marcia Priscilla Silva de Souza
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Claire Chevalier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Valérie Nalesso
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Hamid Meziane
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | | | - Vincent Noblet
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Jean-Paul Armspach
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Veronique Brault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France.,Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| |
Collapse
|
32
|
Mechanisms and Therapeutic Implications of GSK-3 in Treating Neurodegeneration. Cells 2021; 10:cells10020262. [PMID: 33572709 PMCID: PMC7911291 DOI: 10.3390/cells10020262] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders are spreading worldwide and are one of the greatest threats to public health. There is currently no adequate therapy for these disorders, and therefore there is an urgent need to accelerate the discovery and development of effective treatments. Although neurodegenerative disorders are broad ranging and highly complex, they may share overlapping mechanisms, and thus potentially manifest common targets for therapeutic interventions. Glycogen synthase kinase-3 (GSK-3) is now acknowledged to be a central player in regulating mood behavior, cognitive functions, and neuron viability. Indeed, many targets controlled by GSK-3 are critically involved in progressing neuron deterioration and disease pathogenesis. In this review, we focus on three pathways that represent prominent mechanisms linking GSK-3 with neurodegenerative disorders: cytoskeleton organization, the mammalian target of rapamycin (mTOR)/autophagy axis, and mitochondria. We also consider the challenges and opportunities in the development of GSK-3 inhibitors for treating neurodegeneration.
Collapse
|
33
|
Davies MP, Benitez R, Perez C, Jakupovic S, Welsby P, Rzepecka K, Alder J, Davidson C, Martinez A, Hayes JM. Structure-Based Design of Potent Selective Nanomolar Type-II Inhibitors of Glycogen Synthase Kinase-3β. J Med Chem 2021; 64:1497-1509. [PMID: 33499592 DOI: 10.1021/acs.jmedchem.0c01568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
For the first time, the in silico design, screening, and in vitro validation of potent GSK-3β type-II inhibitors are presented. In the absence of crystallographic evidence for a DFG-out GSK-3β activation loop conformation, computational models were designed using an adapted DOLPHIN approach and a method consisting of Prime loop refinement, induced-fit docking, and molecular dynamics. Virtual screening of the Biogenics subset from the ZINC database led to an initial selection of 20 Phase I compounds revealing two low micromolar inhibitors in an isolated enzyme assay. Twenty more analogues (Phase II compounds) related to the hit [pyrimidin-2-yl]amino-furo[3,2-b]furyl-urea scaffold were selected for structure-activity relationship analysis. The Phase II studies led to five highly potent nanomolar inhibitors, with compound 23 (IC50 =0.087 μM) > 100 times more potent than the best Phase I inhibitor, and selectivity for GSK-3β inhibition compared to homologous kinases was observed. Ex vivo experiments (SH-SY5Y cell lines) for tau hyperphosphorylation revealed promising neuroprotective effects at low micromolar concentrations. The type-II inhibitor design has been unraveled as a potential route toward more clinically effective GSK-3β inhibitors.
Collapse
Affiliation(s)
- Matthew P Davies
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Rocio Benitez
- Centro de Investigaciones Biologicas, CSIC, Avenida Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Concepción Perez
- Instituto de Quimica Medica, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Sven Jakupovic
- AnalytiCon Discovery GmbH, Hermannswerder Haus 17, 14473 Potsdam, Germany
| | - Philip Welsby
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Klaudia Rzepecka
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Jane Alder
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Colin Davidson
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Ana Martinez
- Centro de Investigaciones Biologicas, CSIC, Avenida Ramiro de Maeztu 9, 28040 Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | - Joseph M Hayes
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| |
Collapse
|
34
|
Zanni G, Goto S, Fragopoulou AF, Gaudenzi G, Naidoo V, Di Martino E, Levy G, Dominguez CA, Dethlefsen O, Cedazo-Minguez A, Merino-Serrais P, Stamatakis A, Hermanson O, Blomgren K. Lithium treatment reverses irradiation-induced changes in rodent neural progenitors and rescues cognition. Mol Psychiatry 2021; 26:322-340. [PMID: 31723242 PMCID: PMC7815512 DOI: 10.1038/s41380-019-0584-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/13/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022]
Abstract
Cranial radiotherapy in children has detrimental effects on cognition, mood, and social competence in young cancer survivors. Treatments harnessing hippocampal neurogenesis are currently of great relevance in this context. Lithium, a well-known mood stabilizer, has both neuroprotective, pro-neurogenic as well as antitumor effects, and in the current study we introduced lithium treatment 4 weeks after irradiation. Female mice received a single 4 Gy whole-brain radiation dose on postnatal day (PND) 21 and were randomized to 0.24% Li2CO3 chow or normal chow from PND 49 to 77. Hippocampal neurogenesis was assessed on PND 77, 91, and 105. We found that lithium treatment had a pro-proliferative effect on neural progenitors, but neuronal integration occurred only after it was discontinued. Also, the treatment ameliorated deficits in spatial learning and memory retention observed in irradiated mice. Gene expression profiling and DNA methylation analysis identified two novel factors related to the observed effects, Tppp, associated with microtubule stabilization, and GAD2/65, associated with neuronal signaling. Our results show that lithium treatment reverses irradiation-induced loss of hippocampal neurogenesis and cognitive impairment even when introduced long after the injury. We propose that lithium treatment should be intermittent in order to first make neural progenitors proliferate and then, upon discontinuation, allow them to differentiate. Our findings suggest that pharmacological treatment of cognitive so-called late effects in childhood cancer survivors is possible.
Collapse
Affiliation(s)
- Giulia Zanni
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden.
- Department of Developmental Neuroscience, New York State Psychiatric Institute, Columbia University, 1051 Riverside, New York, NY, 10032, USA.
| | - Shinobu Goto
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 467-8601, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Japan
| | - Adamantia F Fragopoulou
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden
| | - Giulia Gaudenzi
- Department of Neuroscience, Karolinska Institutet, Biomedicum, 171 77, Stockholm, Sweden
- Department of Protein Science, Division of Nanobiotechnology, KTH Royal Institute of Technology, Science for Life Laboratory, 171 21, Stockholm, Sweden
| | - Vinogran Naidoo
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden
- Department of Human Biology, Faculty of Health Sciences, Anzio Road Observatory, 7925, University of Cape Town, Cape Town, South Africa
| | - Elena Di Martino
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden
| | - Gabriel Levy
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden
- Ludwig Institute for Cancer Research, Brussels Branch, Avenue Hippocrate 75, 1200, Brussels, Belgium
| | - Cecilia A Dominguez
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden
| | - Olga Dethlefsen
- National Bioinformatics Infrastructure Sweden (NIBIS), Science for Life Laboratory (SciLifeLab), Svante Arrhenius väg 16C, 106 91, Stockholm, Sweden
- Department of Biochemistry and Biophysics (DBB), Stockholm University, Svante Arrhenius väg 16C, 106 91, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, BioClinicum J9:20, 171 64, Stockholm, Sweden
| | - Paula Merino-Serrais
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, BioClinicum J9:20, 171 64, Stockholm, Sweden
| | - Antonios Stamatakis
- Biology-Biochemistry Lab, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Papadiamantopoulou 123, Goudi, 11527, Athens, Greece
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Biomedicum, 171 77, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, 171 64, Stockholm, Sweden.
- Pediatric Oncology, Karolinska University Hospital, Eugeniavägen 23, 171 64, Stockholm, Sweden.
| |
Collapse
|
35
|
Kalli ΕG. The Effect of Nutrients on Alzheimer’s Disease Biomarkers: A Metabolomic Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:301-308. [DOI: 10.1007/978-3-030-78787-5_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
Moidunny S, Benneyworth MA, Titus DJ, Beurel E, Kolli U, Meints J, Jalodia R, Ramakrishnan S, Atkins CM, Roy S. Glycogen synthase kinase-3 inhibition rescues sex-dependent contextual fear memory deficit in human immunodeficiency virus-1 transgenic mice. Br J Pharmacol 2020; 177:5658-5676. [PMID: 33080056 DOI: 10.1111/bph.15288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE A significant number of HIV-1 patients on antiretroviral therapy develop HIV-associated neurocognitive disorders (HAND). Evidence indicate that biological sex may regulate HAND pathogenesis, but the mechanisms remain unknown. We investigated synaptic mechanisms associated with sex differences in HAND, using the HIV-1-transgenic 26 (Tg26) mouse model. EXPERIMENTAL APPROACH Contextual- and cue-dependent memories of male and female Tg26 mice and littermate wild type mice were assessed in a fear conditioning paradigm. Hippocampal electrophysiology, immunohistochemistry, western blot, qRT-PCR and ELISA techniques were used to investigate cellular, synaptic and molecular impairments. KEY RESULTS Cue-dependent memory was unaltered in male and female Tg26 mice, when compared to wild type mice. Male, but not female, Tg26 mice showed deficits in contextual fear memory. Consistently, only male Tg26 mice showed depressed hippocampal basal synaptic transmission and impaired LTP induction in area CA1. These deficits in male Tg26 mice were independent of hippocampal neuronal loss and microglial activation but were associated with increased HIV-1 long terminal repeat mRNA expression, reduced hippocampal synapsin-1 protein, reduced BDNF mRNA and protein, reduced AMPA glutamate receptor (GluA1) phosphorylation levels and increased glycogen synthase kinase 3 (GSK3) activity. Importantly, selective GSK3 inhibition using 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione increased levels of synapsin-1, BDNF and phosphorylated-GluA1 proteins, restored hippocampal basal synaptic transmission and LTP, and improved contextual fear memory in male Tg26 mice. CONCLUSION AND IMPLICATIONS Sex-dependent impairments in contextual fear memory and synaptic plasticity in Tg26 mice are associated with increased GSK3 activity. This implicates GSK3 inhibition as a potential therapeutic strategy to improve cognition in HIV-1 patients.
Collapse
Affiliation(s)
- Shamsudheen Moidunny
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - David J Titus
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.,The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eleonore Beurel
- Department of Psychiatry and Behavioral Sciences, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Udhghatri Kolli
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joyce Meints
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Richa Jalodia
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sundaram Ramakrishnan
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
37
|
Vaidya GN, Rana P, Venkatesh A, Chatterjee DR, Contractor D, Satpute DP, Nagpure M, Jain A, Kumar D. Paradigm shift of "classical" HDAC inhibitors to "hybrid" HDAC inhibitors in therapeutic interventions. Eur J Med Chem 2020; 209:112844. [PMID: 33143937 DOI: 10.1016/j.ejmech.2020.112844] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
'Epigenetic' regulation of genes via post-translational modulation of proteins is the current mainstay approach for the disease therapies, particularly explored in the Histone Deacetylase (HDAC) class of enzymes. Mainly sight saw in cancer chemotherapeutics, HDAC inhibitors have also found a promising role in other diseases (neurodegenerative disorders, cardiovascular diseases, and viral infections) and successfully entered in various combination therapies (pre-clinical/clinical stages). The prevalent flexibility in the structural design of HDAC inhibitors makes them easily tuneable to merge with other pharmacophore modules for generating multi-targeted single hybrids as a novel tactic to overcome drawbacks of polypharmacy. Herein, we reviewed the putative role of prevalent HDAC hybrids inhibitors in the current and prospective stage as a translational approach to overcome the limitations of the existing conventional drug candidates (parent molecule) when used either alone (drug resistance, solubility issues, adverse side effects, selectivity profile) or in combination (pharmacokinetic interactions, patient compliance) for treating various diseases.
Collapse
Affiliation(s)
- Gargi Nikhil Vaidya
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Pooja Rana
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Ashwini Venkatesh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Deep Rohan Chatterjee
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Darshan Contractor
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Dinesh Parshuram Satpute
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Mithilesh Nagpure
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Alok Jain
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India; Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi, India.
| | - Dinesh Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
38
|
Carullo G, Federico S, Relitti N, Gemma S, Butini S, Campiani G. Retinitis Pigmentosa and Retinal Degenerations: Deciphering Pathways and Targets for Drug Discovery and Development. ACS Chem Neurosci 2020; 11:2173-2191. [PMID: 32589402 DOI: 10.1021/acschemneuro.0c00358] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Inherited retinal diseases (IRDs) are a group of retinopathies generally caused by genetic mutations. Retinitis pigmentosa (RP) represents one of the most studied IRDs. RP leads to intense vision loss or blindness resulting from the degeneration of photoreceptor cells. To date, RP is mainly treated with palliative supplementation of vitamin A and retinoids, gene therapies, or surgical interventions. Therefore, a pharmacologically based therapy is an urgent need requiring a medicinal chemistry approach, to validate molecular targets able to deal with retinal degeneration. This Review aims at outlining the recent research efforts in identifying new drug targets for RP, especially focusing on the neuroprotective role of the Wnt/β-catenin/GSK3β pathway and apoptosis modulators (in particular PARP-1) but also on growth factors such as VEGF and BDNF. Furthermore, the role of spatiotemporally expressed G protein-coupled receptors (GPR124) in the retina and the emerging function of histone deacetylase inhibitors in promoting retinal neuroprotection will be discussed.
Collapse
Affiliation(s)
- Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018−2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Stefano Federico
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018−2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018−2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018−2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018−2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018−2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
39
|
Intranasal insulin ameliorates cognitive impairment in a rat model of Parkinson's disease through Akt/GSK3β signaling pathway. Life Sci 2020; 259:118159. [PMID: 32763288 DOI: 10.1016/j.lfs.2020.118159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022]
Abstract
AIMS Parkinson's disease dementia (PDD) is one of the most common non-motor symptoms of advanced Parkinson's disease (PD). This study aimed to determine whether intranasal insulin has protective effects on cognition in the rat PD model induced by 6-hydroxylase dopamine (6-OHDA) through the insulin signaling pathway. MATERIALS AND METHODS The rats were given intranasal insulin administration for six weeks after unilateral medial forebrain bundle (MFB) injection of 6-OHDA. Then a series of cognitive-behavioral tests, immunofluorescence, and immunoblotting was performed on the rats. KEY FINDINGS The results demonstrated that the injection of 6-OHDA in the unilateral MFB damaged working memory and long-term habituation of rats in the T-maze rewarded alternation test and hole-board test. Besides, rats with unilateral 6-OHDA injury performed poorly in terms of escape latency and average speed during the hidden platform training phase rather than in the probe trial of the Morris Water Maze (MWM) test. Immunofluorescence results showed that unilateral 6-OHDA injury in MFB led to the massive death of ipsilateral-substantia nigra (SN) tyrosine hydroxylase (TH)-positive neurons. Western blot results further indicated that 6-OHDA-induced necrosis of ipsilateral-SN dopaminergic neurons reduced the levels of p-Akt (Ser473) and p-GSK3β (Ser9) in the ipsilateral-hippocampus. SIGNIFICANCE These findings provide a solid evidence base for the relationship between PD cognitive impairment and insulin signaling pathways.
Collapse
|
40
|
Dohare P, Kidwai A, Kaur J, Singla P, Krishna S, Klebe D, Zhang X, Hevner R, Ballabh P. GSK3β Inhibition Restores Impaired Neurogenesis in Preterm Neonates With Intraventricular Hemorrhage. Cereb Cortex 2020; 29:3482-3495. [PMID: 30192926 DOI: 10.1093/cercor/bhy217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/30/2018] [Indexed: 01/25/2023] Open
Abstract
Intraventricular hemorrhage (IVH) is a common complication of prematurity in infants born at 23-28 weeks of gestation. Survivors exhibit impaired growth of the cerebral cortex and neurodevelopmental sequeale, but the underlying mechanism(s) are obscure. Previously, we have shown that neocortical neurogenesis continues until at least 28 gestational weeks. This renders the prematurely born infants vulnerable to impaired neurogenesis. Here, we hypothesized that neurogenesis is impaired by IVH, and that signaling through GSK3β, a critical intracellular kinase regulated by Wnt and other pathways, mediates this effect. These hypotheses were tested observationally in autopsy specimens from premature infants, and experimentally in a premature rabbit IVH model. Significantly, in premature infants with IVH, the number of neurogenic cortical progenitor cells was reduced compared with infants without IVH, indicating acutely decreased neurogenesis. This finding was corroborated in the rabbit IVH model, which further demonstrated reduction of upper layer cortical neurons after longer survival. Both the acute reduction of neurogenic progenitors, and the subsequent decrease of upper layer neurons, were rescued by treatment with AR-A014418, a specific inhibitor of GSK3β. Together, these results indicate that IVH impairs late stages of cortical neurogenesis, and suggest that treatment with GSK3β inhibitors may enhance neurodevelopment in premature infants with IVH.
Collapse
Affiliation(s)
- Preeti Dohare
- Department of Pediatrics.,Dominick P. Purpura Department of Neuroscience
| | | | | | | | | | | | | | - Robert Hevner
- Department of Pathology, 9500 Gilman Dr, UCSD, La Jolla, CA, USA
| | - Praveen Ballabh
- Department of Pediatrics.,Dominick P. Purpura Department of Neuroscience.,Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
41
|
Beneficial effects of low-dose lithium on cognitive ability and pathological alteration of Alzheimer's disease transgenic mice model. Neuroreport 2020; 31:943-951. [PMID: 32639272 DOI: 10.1097/wnr.0000000000001499] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lithium has been shown to delay the progression of Alzheimer's disease to reduce the prevalence of dementia. However, its narrow therapeutic index and numerous toxic effects at conventional dosage limited its long-term use to older subjects. Here, we tested the effect of low-dose lithium on cognitive impairment and pathology alterations in a mouse model of Alzheimer's disease, the amyloid precursor protein/presenilin-1 (APP/PS1) transgenic mouse. We found that both chronic and acute administration of lithium dose-dependently increased in blood and brain tissues. Long-term administration of low-dose lithium does not affect the body weight of APP/PS1 mice, but can significantly improve spatial memory of APP/PS1 mice. Pathologically, it also reduced β-amyloid plague and p-tau levels. Therefore, our results show that long-term low-dose lithium can ameliorate cognitive dysfunction and pathological alterations of Alzheimer's disease transgenic mice, and provide a theoretical basis for the further application of low-dose lithium in Alzheimer's disease clinical treatment.
Collapse
|
42
|
GSK-3-TSC axis governs lysosomal acidification through autophagy and endocytic pathways. Cell Signal 2020; 71:109597. [DOI: 10.1016/j.cellsig.2020.109597] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022]
|
43
|
Monaco SA, Matamoros AJ, Gao WJ. Conditional GSK3β deletion in parvalbumin-expressing interneurons potentiates excitatory synaptic function and learning in adult mice. Prog Neuropsychopharmacol Biol Psychiatry 2020; 100:109901. [PMID: 32113851 DOI: 10.1016/j.pnpbp.2020.109901] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 01/03/2023]
Abstract
Glycogen synthase kinase 3β (GSK3β) has gained interest regarding its involvement in psychiatric and neurodegenerative disorders. Recently GSK3 inhibitors were highlighted as promising rescuers of cognitive impairments for a gamut of CNS disorders. Growing evidence supports that fast-spiking parvalbumin (PV) interneurons are critical regulators of cortical computation. Albeit, how excitatory receptors on PV interneurons are regulated and how this affects cognitive function remains unknown. To address these questions, we have generated a novel triple-transgenic conditional mouse with GSK3β genetically deleted from PV interneurons. PV-GSK3β-/- resulted in increased excitability and augmented excitatory synaptic strength in prefrontal PV interneurons. More importantly, these synaptic changes are correlated with accelerated learning with no changes in locomotion and sociability. Our study, for the first time, examined how GSK3β activity affects learning capability via regulation of PV interneurons. This study provides a novel insight into how GSK3β may contribute to disorders afflicted by cognitive deficits.
Collapse
Affiliation(s)
- Sarah A Monaco
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 W School House Ln, Philadelphia, PA 19129, United States of America
| | - Andrew J Matamoros
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, United States of America; Deparment of Pathology and Laboratory Medicine, University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104-4238, United States of America
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 W School House Ln, Philadelphia, PA 19129, United States of America.
| |
Collapse
|
44
|
Fan X, Zhao Z, Wang D, Xiao J. Glycogen synthase kinase-3 as a key regulator of cognitive function. Acta Biochim Biophys Sin (Shanghai) 2020; 52:219-230. [PMID: 32147679 DOI: 10.1093/abbs/gmz156] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/16/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a highly conserved and multifunctional serine/threonine protein kinase widely distributed in eukaryotic cells. GSK-3 is originally thought to be an enzyme that regulates glycogen synthesis. It was subsequently found that GSK-3 influences many critical cellular functions, such as cell structure, neural plasticity, gene expression, and neuronal survival. Recently, GSK-3 has been found to be associated with cognition, and its dysregulation leads to cognitive impairments in many diseases, including Alzheimer's disease, diabetes, depression, Parkinson's disease, and others. In this review, we summarized the current knowledge about the structure of GSK-3, the regulation of GSK-3 activity, and its role in cognitive function and cognitive-related disease.
Collapse
Affiliation(s)
- Xuhong Fan
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Zhenyu Zhao
- Department of Anesthesiology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410000, China
| | - Deming Wang
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Ji Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang 421001, China
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
45
|
Bendová Z, Pačesová D, Novotný J. The day-night differences in ERK1/2, GSK3β activity and c-Fos levels in the brain, and the responsiveness of various brain structures to morphine. J Comp Neurol 2020; 528:2471-2495. [PMID: 32170720 DOI: 10.1002/cne.24906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/17/2020] [Accepted: 03/09/2020] [Indexed: 11/12/2022]
Abstract
As with other drugs or pharmaceuticals, opioids differ in their rewarding or analgesic effects depending on when they are applied. In the previous study, we have demonstrated the day/night difference in the sensitivity of the major circadian clock in the suprachiasmatic nucleus to a low dose of morphine, and showed the bidirectional effect of morphine on pERK1/2 and pGSK3β levels in the suprachiasmatic nucleus depending on the time of administration. The main aim of this study was to identify other brain structures that respond differently to morphine depending on the time of its administration. Using immunohistochemistry, we identified 44 structures that show time-of-day specific changes in c-Fos level and activity of ERK1/2 and GSK3β kinases in response to a single dose of 1 mg/kg morphine. Furthermore, comparison among control groups revealed the differences in the spontaneous levels of all markers with a generally higher level during the night, that is, in the active phase of the day. We thus provide further evidence for diurnal variations in the activity of brain regions outside the suprachiasmatic nucleus indicated by the temporal changes in the molecular substrate. We suggest that these changes are responsible for generating diurnal variation in the reward behavior or analgesic effect of opioid administration.
Collapse
Affiliation(s)
- Zdeňka Bendová
- Faculty of Science, Charles University, Prague, Czech Republic.,Department of Sleep Medicine and Chronobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Dominika Pačesová
- Faculty of Science, Charles University, Prague, Czech Republic.,Department of Sleep Medicine and Chronobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Jiří Novotný
- Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
46
|
Li M, Xia M, Chen W, Wang J, Yin Y, Guo C, Li C, Tang X, Zhao H, Tan Q, Chen Y, Jia Z, Liu X, Feng H. Lithium treatment mitigates white matter injury after intracerebral hemorrhage through brain-derived neurotrophic factor signaling in mice. Transl Res 2020; 217:61-74. [PMID: 31951826 DOI: 10.1016/j.trsl.2019.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 01/04/2023]
Abstract
Intracerebral hemorrhage (ICH), a subtype of stroke with high morbidity and mortality, occurs mainly in the basal ganglia and causes white matter injury (WMI), resulting in severe motor dysfunction and poor prognosis in patients. The preservation of the white matter around the hematoma is crucial for motor function recovery, but there is currently no effective treatment for WMI following ICH. Lithium has been widely used for the treatment of bipolar disorder for decades. Although the protective effects of lithium on neurodegenerative diseases and cerebral trauma have been studied in recent years, whether it can be used to alleviate WMI after ICH remains to be researched. The results of this study revealed that ICH caused significant functional and pathological abnormalities in mice. After LiCl was administered to mice with ICH, behavioural performance and electrophysiological functions were improved and ICH-induced white matter pathological injury, including myelin sheath and axonal degeneration, was ameliorated. Furthermore, LiCl treatment decreased the death of mature oligodendrocytes (OLGs) in ICH mice, which may have been attributed to the enhanced expression of brain-derived neurotrophic factor (BDNF) regulated by the LiCl-induced inhibition of glycogen synthase kinase-3β (GSK-3β). The decreased death of OLGs was closely associated with decreased destruction of the myelin sheath and alleviated degradation of the axons. In summary, this study suggests that the protective effect of lithium on WMI after ICH might be related to an increased level of BDNF and that LiCl treatment may be a potential therapeutic method to palliate WMI after ICH.
Collapse
Affiliation(s)
- Mingxi Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Min Xia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Weixiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Jie Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yi Yin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Chao Guo
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Chengcheng Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xiaoqin Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Hengli Zhao
- Department of Neurology, The Second Medical Central, Chinese PLA (People's Liberation Army) General Hospital, Beijing, PR China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; State Key Laboratory of Trauma, Burn, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Zhengcai Jia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China.
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; State Key Laboratory of Trauma, Burn, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China.
| |
Collapse
|
47
|
Cicvaric A, Sachernegg HM, Stojanovic T, Symmank D, Smani T, Moeslinger T, Uhrin P, Monje FJ. Podoplanin Gene Disruption in Mice Promotes in vivo Neural Progenitor Cells Proliferation, Selectively Impairs Dentate Gyrus Synaptic Depression and Induces Anxiety-Like Behaviors. Front Cell Neurosci 2020; 13:561. [PMID: 32009902 PMCID: PMC6974453 DOI: 10.3389/fncel.2019.00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Podoplanin (Pdpn), a brain-tumor-related glycoprotein identified in humans and animals, is endogenously expressed in several organs critical for life support such as kidney, lung, heart and brain. In the brain, Pdpn has been identified in proliferative nestin-positive adult neural progenitor cells and in neurons of the neurogenic hippocampal dentate gyrus (DG), a structure associated to anxiety, critical for learning and memory functions and severely damaged in people with Alzheimer's Disease (AD). The in vivo role of Pdpn in adult neurogenesis and anxiety-like behavior remained however unexplored. Using mice with disrupted Pdpn gene as a model organism and applying combined behavioral, molecular biological and electrophysiological assays, we here show that the absence of Pdpn selectively impairs long-term synaptic depression in the neurogenic DG without affecting the CA3-Schaffer's collateral-CA1 synapses. Pdpn deletion also enhanced the proliferative capacity of DG neural progenitor cells and diminished survival of differentiated neuronal cells in vitro. In addition, mice with podoplanin gene disruption showed increased anxiety-like behaviors in experimentally validated behavioral tests as compared to wild type littermate controls. Together, these findings broaden our knowledge on the molecular mechanisms influencing hippocampal synaptic plasticity and neurogenesis in vivo and reveal Pdpn as a novel molecular target for future studies addressing general anxiety disorder and synaptic depression-related memory dysfunctions.
Collapse
Affiliation(s)
- Ana Cicvaric
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannah M. Sachernegg
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Dörte Symmank
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville (IBiS)/University of Seville/CIBERCV, Seville, Spain
| | - Thomas Moeslinger
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
48
|
cGMP signaling pathway in hepatic encephalopathy neuroinflammation and cognition. Int Immunopharmacol 2019; 79:106082. [PMID: 31869775 DOI: 10.1016/j.intimp.2019.106082] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/05/2019] [Accepted: 11/24/2019] [Indexed: 12/25/2022]
Abstract
Hepatic encephalopathy (HE) is a complex neuropsychiatric syndrome that results from liver failure and is characterized by a wide range of symptoms such as alteration in the sleep-waking cycle, neuromuscular coordination, mood, and cognition. The deregulation of nitric oxide (NO)/cyclic guanosine monophosphate (cGMP)/protein kinase G (PKG) signaling pathway is thought to play an important role in the etiology and progression of neurodegenerative diseases, and several studies pointed that the cGMP signaling is impaired in patients with HE and experimental models of chronic hyperammonemia. This review aimed to briefly present the current knowledge of the cGMP signaling pathways in neuroinflammation, neurogenesis, and memory in hepatic encephalopathy and its potential therapeutic role.
Collapse
|
49
|
The selective GSK3 inhibitor, SAR502250, displays neuroprotective activity and attenuates behavioral impairments in models of neuropsychiatric symptoms of Alzheimer's disease in rodents. Sci Rep 2019; 9:18045. [PMID: 31792284 PMCID: PMC6888874 DOI: 10.1038/s41598-019-54557-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) has been identified as a promising target for the treatment of Alzheimer’s disease (AD), where abnormal activation of this enzyme has been associated with hyperphosphorylation of tau proteins. This study describes the effects of the selective GSK3 inhibitor, SAR502250, in models of neuroprotection and neuropsychiatric symptoms (NPS) associated with AD. In P301L human tau transgenic mice, SAR502250 attenuated tau hyperphosphorylation in the cortex and spinal cord. SAR502250 prevented the increase in neuronal cell death in rat embryonic hippocampal neurons following application of the neurotoxic peptide, Aβ25–35. In behavioral studies, SAR502250 improved the cognitive deficit in aged transgenic APP(SW)/Tau(VLW) mice or in adult mice after infusion of Aβ25–35. It attenuated aggression in the mouse defense test battery and improved depressive-like state of mice in the chronic mild stress procedure after 4 weeks of treatment. Moreover, SAR502250 decreased hyperactivity produced by psychostimulants. In contrast, the drug failed to modify anxiety-related behaviors or sensorimotor gating deficit. This profile confirms the neuroprotective effects of GSK3 inhibitors and suggests an additional potential in the treatment of some NPS associated with AD.
Collapse
|
50
|
Pardo M, Cheng Y, Sitbon YH, Lowell JA, Grieco SF, Worthen RJ, Desse S, Barreda-Diaz A. Insulin growth factor 2 (IGF2) as an emergent target in psychiatric and neurological disorders. Review. Neurosci Res 2019; 149:1-13. [PMID: 30389571 DOI: 10.1016/j.neures.2018.10.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/05/2018] [Accepted: 10/29/2018] [Indexed: 12/23/2022]
Abstract
Insulin-like growth factor 2 (IGF2) is abundantly expressed in the central nervous system (CNS). Recent evidence highlights the role of IGF2 in the brain, sustained by data showing its alterations as a common feature across a variety of psychiatric and neurological disorders. Previous studies emphasize the potential role of IGF2 in psychiatric and neurological conditions as well as in memory impairments, targeting IGF2 as a pro-cognitive agent. New research on animal models supports that upcoming investigations should explore IGF2's strong promising role as a memory enhancer. The lack of effective treatments for cognitive disturbances as a result of psychiatric diseases lead to further explore IGF2 as a promising target for the development of new pharmacology for the treatment of memory dysfunctions. In this review, we aim at gathering all recent relevant studies and findings on the role of IGF2 in the development of psychiatric diseases that occur with cognitive problems.
Collapse
Affiliation(s)
- M Pardo
- University of Miami Miller School of Medicine, Department of Neurology, Miami, FL, USA.
| | - Y Cheng
- University of California Los Angeles, Neurology Department, Los Angeles, CA, USA.
| | - Y H Sitbon
- University of Miami Miller School of Medicine, Department of Molecular and Cellular Pharmacology, Miami, FL, USA.
| | - J A Lowell
- University of Miami, Department of Psychiatry & Behavioral Sciences, Miami, FL, USA.
| | - S F Grieco
- University of California, Department of Anatomy and Neurobiology, Irvine, CA, USA.
| | - R J Worthen
- University of Miami, Department of Psychiatry & Behavioral Sciences, Miami, FL, USA.
| | - S Desse
- University of Miami, Department of Psychiatry & Behavioral Sciences, Miami, FL, USA.
| | - A Barreda-Diaz
- University of Miami Miller School of Medicine, Department of Neurology, Miami, FL, USA.
| |
Collapse
|