1
|
Gao D, Yin J, Zhang Y, Li C, Zhao L, Wang L, Song J, Zhang H, Niu Q, Lu X. The Molecular Mechanism of miRNA-195-5p Regulating ERK Involvement in Abnormal Phosphorylation of Tau Protein by Aluminum Maltol in PC12 Cells. J Appl Toxicol 2025. [PMID: 40273949 DOI: 10.1002/jat.4795] [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: 09/28/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 04/26/2025]
Abstract
Although aluminum is ubiquitously present on Earth, it is not necessary for life. Aluminum is a metal element that can induce neurotoxicity. The neurotoxicity of aluminum is mainly caused by the aggregation of abnormally phosphorylated tau protein to form neurofibrillary tangles (NFTs). The phosphorylation of tau is regulated by both kinases and phosphatases. ERK is involved in PHF-type tau hyperphosphorylation. Recent studies have revealed that the interaction between microRNAs (miRNAs) and the ERK/MAPK cascade is related to maintaining the normal function of the nervous system. miR-195 is involved in the early development of AD with a potential impact on cognition. Therefore, we speculate that miRNA-195 may regulate ERK activity, thereby causing hyperphosphorylation of tau protein and neurotoxicity. The purpose of this study was to explore the role of miRNA-195-5p in regulating ERK in the process of aluminum maltol-induced tau hyperphosphorylation. The results showed that aluminum exposure decreased the expression level of miRNA-195-5p and increased the expression of P-ERK, abnormal phosphorylated tau. After inhibiting the activity of ERK, the expression of phosphorylation tau protein decreased. There is an interaction effect between inhibiting the activity of ERK and aluminum exposure on the expression of phosphorylated tau proteins. After the overexpression of miRNA-195-5p, the activity of ERK was inhibited. There is an interaction effect between miRNA-195-5p and aluminum exposure on the expression of phosphorylated tau. In conclusion, miRNA-195-5p regulates ERK involvement in the abnormal phosphorylation of tau protein by Al (mal)3in PC12 cells.
Collapse
Affiliation(s)
- Dan Gao
- Xi'an Baoshihua Changqing Hosptial, Xi'an, Shaanxi, China
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Yin
- Sinopharm Tongmei General Hospital, Shanxi Health Commission Key Laboratory of Nervous System Disease Prevention and Treatment, Datong, Shanxi, China
| | - Yunwei Zhang
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chenyang Li
- The Second Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Le Zhao
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Linping Wang
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Song
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huifang Zhang
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qiao Niu
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoting Lu
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Jin D, Zhang M, Shi L, Liu H. Investigating the Impact of IL-6 and CXCL8 on Neurodegeneration and Cognitive Decline in Alzheimer Disease. Int J Neuropsychopharmacol 2024; 28:pyae038. [PMID: 39223908 PMCID: PMC11781222 DOI: 10.1093/ijnp/pyae038] [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: 05/01/2024] [Accepted: 09/02/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Alzheimer disease (AD) is a progressive neurodegenerative disorder primarily affecting the elderly, characterized by severe cognitive impairment and memory loss. Emerging evidence suggests that neuroinflammation plays a significant role in AD pathogenesis, with cytokines like interleukin-6 (IL-6) and C-X-C motif chemokine ligand 8 (CXCL8) contributing to the disease progression. METHODS We utilized Gene Expression Omnibus datasets to identify IL-6 and CXCL8 as pivotal inflammatory markers in AD. In vitro experiments were conducted using SK-N-BE(2)-M17 and THP-1 cell lines treated with IL-6 and CXCL8 to model AD. Additionally, in vivo tests on Amyloid Precursor Protein/Presenilin 1 (APP/PS1) AD mouse models were performed to assess the impact of these cytokines on cognitive functions and brain pathology. RESULTS The results indicated a significant decrease in cell viability, increased apoptosis, and elevated inflammatory factor secretion following IL-6 and CXCL8 treatment in vitro. In vivo, AD mouse models treated with these cytokines exhibited exacerbated emotional distress, decreased social interaction, impaired cognitive functions, and increased amyloid protein deposition in neural tissues. CONCLUSIONS The study highlights the detrimental effects of IL-6 and CXCL8 on neuronal health and cognitive functions in AD. These findings suggest that targeting these cytokines could offer potential therapeutic interventions for improving patient outcomes in Alzheimer disease.
Collapse
Affiliation(s)
- Dongdong Jin
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Shi
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hengfang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Chen H, Yu C, Liu W, Zhu C, Jiang X, Xu C, Liu W, Huang Y, Xu Z, Zhao Q. Discovery of novel α-carboline derivatives as glycogen synthase kinase-3β inhibitors for the treatment of Alzheimer's disease. Arch Pharm (Weinheim) 2022; 355:e2200156. [PMID: 35836098 DOI: 10.1002/ardp.202200156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease (AD) is a chronic and progressive neurodegenerative disease, characterized by irreversible cognitive impairment, memory loss, and behavioral disturbances, ultimately resulting in death. The critical roles of glycogen synthase kinase-3β (GSK-3β) in tau pathology have also received considerable attention. Based on molecular docking studies, a series of novel α-carboline derivatives were designed, synthesized, and evaluated as GSK-3β inhibitors for their various biological activities. Among them, compound ZCH-9 showed the most potent inhibitory activity against GSK-3β, with an IC50 value of 1.71 ± 0.09 µM. The cytotoxicity assay showed that ZCH-9 had low cytotoxicity toward the cell lines SH-SY5Y, HepG2, and HL-7702. Moreover, Western blot analysis indicated that ZCH-9 effectively inhibited hyperphosphorylation of the tau protein in okadaic acid-treated SH-SY5Y cells. The binding mode between ZCH-9 and GSK-3β was analyzed and further clarified throughout the molecular dynamics simulations. In general, these results suggested that the α-carboline-based small-molecule compounds could serve as potential candidates targeting GSK-3β for the treatment of AD.
Collapse
Affiliation(s)
- Huanhua Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Chong Yu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Wenjie Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Chengze Zhu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaowen Jiang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Chang Xu
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Wenwu Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yaoguang Huang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Zihua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Qingchun Zhao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
4
|
Wadhwa P, Jain P, Jadhav HR. Glycogen Synthase Kinase 3 (GSK3): Its Role and Inhibitors. Curr Top Med Chem 2021; 20:1522-1534. [PMID: 32416693 DOI: 10.2174/1568026620666200516153136] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 12/23/2022]
Abstract
Glycogen Synthase Kinase 3 (GSK3) is one of the Serine/Threonine protein kinases, which has gained a lot of attention for its role in a variety of pathways. It has two isoforms, GSK3α and GSK3β. However, GSK3β is highly expressed in different areas of the brain and has been implicated in Alzheimer's disease as it is involved in tau phosphorylation. Due to its high specificity concerning substrate recognition, GSK3 has been considered as an important target. In the last decade, several GSK3 inhibitors have been reported and two molecules are in clinical trials. This review collates the information published in the last decade about the role of GSK3 in Alzheimer's disease and progress in the development of its inhibitors. Using this collated information, medicinal chemists can strategize and design novel GSK3 inhibitors that could be useful in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Pankaj Wadhwa
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani- 333031, Rajasthan, India
| | - Priti Jain
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani- 333031, Rajasthan, India
| | - Hemant R Jadhav
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani- 333031, Rajasthan, India
| |
Collapse
|
5
|
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
|
6
|
Yuan S, Li H, Yang C, Xie W, Wang Y, Zhang J, Cai Z, Mao Z, Xie W, Lü T. DHA attenuates Aβ-induced necroptosis through the RIPK1/RIPK3 signaling pathway in THP-1 monocytes. Biomed Pharmacother 2020; 126:110102. [PMID: 32199223 DOI: 10.1016/j.biopha.2020.110102] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 01/14/2023] Open
Abstract
Monocytes play a crucial role in Alzheimer's disease (AD), and docosahexaenoic acid (DHA) has a neuroprotective effect for many neurodegenerative diseases. However, mechanisms that regulate monocyte and Aβ protein interaction in AD and the effects of DHA on monocytes in the context of AD are not fully understood. The experiments were designed to further explore possible mechanisms of interaction between monocytes and Aβ plaques. Another objective of this study was to investigate a potential mechanism for Aβ-induced necroptosis involving the activation of MAPK and NF-kB signaling pathways in human THP-1 monocytes, as well as how these pathways might be modulated by DHA. Our findings indicate that Aβ25-35 has a "Hormesis" effect on cell viability and necroptosis in THP-1 cells, and Aβ25-35 influences THP-1 cells differentiation as analyzed by flow cytometry. Pretreatment of THP-1 monocytes with DHA effectively inhibited Aβ-induced activation and markedly suppressed protein expression of necroptosis (RIPK1, RIPK3, MLKL) and pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). Moreover, our findings indicate that Aβ25-35 activated the ERK1/2 and p38 signaling pathways, but not NF-κB/p65 signaling, while pre-treatment with DHA followed by Aβ25-35 treatment suppressed only ERK1/2 signaling. Further study revealed that the expression level of RIPK3 is reduced much more during coadministration with DHA and necrostatin-1 (NEC-1) than administration alone with either of them, indicating that DHA may have additional targets. Meanwhile, this finding indicates that DHA can prevent Aβ-induced necroptosis of THP-1 cells via the RIPK1/RIPK3 signaling pathway. Our results also indicate that DHA treatment restored migration of THP-1 monocytes induced by Aβ25-35, and DHA treatment could be a promising new therapy for AD management.
Collapse
Affiliation(s)
- Shiqi Yuan
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Huan Li
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Canhong Yang
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Wenyi Xie
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Yuanyuan Wang
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Jiafa Zhang
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Zibo Cai
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Zhenlin Mao
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Weibing Xie
- Judicial Identification Center of Southern Medical University, No.1023-1063, Shatai Road South, Guangzhou 510515, PR China
| | - Tianming Lü
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China.
| |
Collapse
|
7
|
Wang Y, Zhang Y, Zhang X, Yang T, Liu C, Wang P. Alcohol Dehydrogenase 1B Suppresses β-Amyloid-Induced Neuron Apoptosis. Front Aging Neurosci 2019; 11:135. [PMID: 31231206 PMCID: PMC6560161 DOI: 10.3389/fnagi.2019.00135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/21/2019] [Indexed: 01/01/2023] Open
Abstract
β-amyloid (Aβ) deposition, neurofibrillary tangles induced by phosphorylation of tau protein, and neuronal apoptosis are pathological hallmarks of Alzheimer’s disease (AD). The dementia rate in alcoholic abusers were found to be higher than in control people. The present study explored the potential roles of alcohol dehydrogenase 1B (ADH1B) in AD pathology by determining the ADH1B levels in AD patient sera, in the hippocampus of APP/PS-1 AD model mice, and in an AD model cell line treated with Aβ1-42. The results show that ADH1B levels decreased significantly both in the serum of AD patients and in the hippocampus of APP/PS-1 AD model mice. In addition, the apoptotic rate was reduced and viability was significantly increased in AD model cells transfected with ADH1B overexpression vector. The levels of the p75 neurotrophin receptor (p75NTR), an Aβ1-42 receptor, were down-regulated in the ADH1B overexpressing AD model cell and up-regulated in cells transfected with the shRNA vector of ADH1B. Protein levels of cleaved caspase-3 and Bax decreased significantly, whereas Bcl-2 levels increased in cells overexpressing ADH1B. The opposite trend was observed for cleaved caspase-3, Bax, and Bcl-2 levels in cells transfected with the shRNA vector of ADH1B. The levels of reactive oxygen species (ROS) were found to be reduced in ADH1B overexpressing cells and increased when cells were transfected with the shRNA vector of ADH1B. These results indicate that ADH1B might be important in the prevention of AD, especially for abusers of alcohol, and a potential new target of AD treatment.
Collapse
Affiliation(s)
- Yaqi Wang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Zhang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaomin Zhang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tingting Yang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chengeng Liu
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Peichang Wang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Fu LL, Zhao XY, Ji LD, Xu J. Okadaic acid (OA): Toxicity, detection and detoxification. Toxicon 2019; 160:1-7. [DOI: 10.1016/j.toxicon.2018.12.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/13/2018] [Accepted: 12/21/2018] [Indexed: 10/27/2022]
|
9
|
Jadhav S, Avila J, Schöll M, Kovacs GG, Kövari E, Skrabana R, Evans LD, Kontsekova E, Malawska B, de Silva R, Buee L, Zilka N. A walk through tau therapeutic strategies. Acta Neuropathol Commun 2019; 7:22. [PMID: 30767766 PMCID: PMC6376692 DOI: 10.1186/s40478-019-0664-z] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/21/2019] [Indexed: 12/18/2022] Open
Abstract
Tau neuronal and glial pathologies drive the clinical presentation of Alzheimer's disease and related human tauopathies. There is a growing body of evidence indicating that pathological tau species can travel from cell to cell and spread the pathology through the brain. Throughout the last decade, physiological and pathological tau have become attractive targets for AD therapies. Several therapeutic approaches have been proposed, including the inhibition of protein kinases or protein-3-O-(N-acetyl-beta-D-glucosaminyl)-L-serine/threonine Nacetylglucosaminyl hydrolase, the inhibition of tau aggregation, active and passive immunotherapies, and tau silencing by antisense oligonucleotides. New tau therapeutics, across the board, have demonstrated the ability to prevent or reduce tau lesions and improve either cognitive or motor impairment in a variety of animal models developing neurofibrillary pathology. The most advanced strategy for the treatment of human tauopathies remains immunotherapy, which has already reached the clinical stage of drug development. Tau vaccines or humanised antibodies target a variety of tau species either in the intracellular or extracellular spaces. Some of them recognise the amino-terminus or carboxy-terminus, while others display binding abilities to the proline-rich area or microtubule binding domains. The main therapeutic foci in existing clinical trials are on Alzheimer's disease, progressive supranuclear palsy and non-fluent primary progressive aphasia. Tau therapy offers a new hope for the treatment of many fatal brain disorders. First efficacy data from clinical trials will be available by the end of this decade.
Collapse
Affiliation(s)
- Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska 9, 845 10, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Jesus Avila
- Centro de Biologia Molecular "Severo Ochoa", Consejo Superior de Investigaciones, Cientificas, Universidad Autonoma de Madrid, C/ Nicolas Cabrera, 1. Campus de Cantoblanco, 28049, Madrid, Spain
- Networking Research Center on Neurodegenerative, Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of, Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Dementia Research Centre, University College London, London, UK
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | - Enikö Kövari
- Department of Mental Health and Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Rostislav Skrabana
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Lewis D Evans
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Eva Kontsekova
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Cracow, Poland
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Luc Buee
- Universite of Lille, Inserm, CHU-Lille, UMRS1172, Alzheimer & Tauopathies, Place de Verdun, 59045, Lille cedex, France.
| | - Norbert Zilka
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia.
| |
Collapse
|
10
|
Huang L, Lin M, Zhong X, Yang H, Deng M. Galangin decreases p‑tau, Aβ42 and β‑secretase levels, and suppresses autophagy in okadaic acid‑induced PC12 cells via an Akt/GSK3β/mTOR signaling‑dependent mechanism. Mol Med Rep 2019; 19:1767-1774. [PMID: 30628698 DOI: 10.3892/mmr.2019.9824] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 10/19/2018] [Indexed: 11/05/2022] Open
Abstract
Okadaic acid (OA)‑induced neurotoxicity may be considered a novel tool used to study Alzheimer's disease (AD) pathology, and may be helpful in the development of a novel therapeutic approach. It has been reported that galangin inhibits β‑site amyloid precursor protein‑cleaving enzyme 1 expression, which is a key enzyme for amyloid β (Aβ) generation and is a potential drug candidate for AD therapy. However, further studies are required to confirm its neuroprotective effects in other AD models. The present study aimed to explore the neuroprotective effects of galangin on OA‑induced neurotoxicity in PC12 cells. The cells were divided into the following groups: Control group, model group (175 nM OA for 48 h) and galangin groups (0.25, 0.5 and 1 µg/ml). Beclin‑1, phosphorylated (p)‑protein kinase B (Akt), p‑glycogen synthase kinase (GSK)3β and p‑mechanistic target of rapamycin (mTOR) expression was also measured in the following PC12 cell groups: Control group, model group, 3‑methyladenine group (5 nM), rapamycin group (100 nM) and galangin group (1 µg/ml). The levels of β‑secretase, Aβ42 and p‑tau were detected by ELISA, Beclin‑1 expression was examined by immunohistochemistry and the protein expression levels of p‑Akt, p‑mTOR p‑GSK3β, and Beclin‑1 were detected by western blotting. Galangin treatment enhanced cell viability in cells treated with OA, and decreased β‑secretase, Aβ42 and p‑tau levels. In addition, it suppressed Beclin‑1 and p‑GSK3β expression, but promoted p‑Akt and p‑mTOR expression by regulating the Akt/GSK3β/mTOR pathway. These results indicated that galangin protected PC12 cells from OA‑induced cytotoxicity and inhibited autophagy via the Akt/GSK3β/mTOR pathway, thus suggesting that it may be considered a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Liping Huang
- School of Chemistry and Chemical Engineering, Lingnan Normal University, Zhanjiang, Guangdong 524048, P.R. China
| | - Mingqin Lin
- College of Pharmacy, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Xiaoqin Zhong
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Hongyan Yang
- College of Pharmacy, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Minzhen Deng
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
11
|
Sciú ML, Sebastián-Pérez V, Martinez-Gonzalez L, Benitez R, Perez DI, Pérez C, Campillo NE, Martinez A, Moyano EL. Computer-aided molecular design of pyrazolotriazines targeting glycogen synthase kinase 3. J Enzyme Inhib Med Chem 2018; 34:87-96. [PMID: 30362380 PMCID: PMC6211276 DOI: 10.1080/14756366.2018.1530223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Numerous studies have highlighted the implications of the glycogen synthase kinase 3 (GSK-3) in several processes associated with Alzheimer's disease (AD). Therefore, GSK-3 has become a crucial therapeutic target for the treatment of this neurodegenerative disorder. Hereby, we report the design and multistep synthesis of ethyl 4-oxo-pyrazolo[4,3-d][1-3]triazine-7-carboxylates and their biological evaluation as GSK-3 inhibitors. Molecular modelling studies allow us to develop this new scaffold optimising the chemical structure. Potential binding mode determination in the enzyme and the analysis of the key features in the catalytic site are also described. Furthermore, the ability of pyrazolotriazinones to cross the blood-brain barrier (BBB) was evaluated by passive diffusion and those who showed great GSK-3 inhibition and permeation to the central nervous system (CNS) showed neuroprotective properties against tau hyperphosphorylation in a cell-based model. These new brain permeable pyrazolotriazinones may be used for key in vivo studies and may be considered as new leads for further optimisation for the treatment of AD.
Collapse
Affiliation(s)
- M Lourdes Sciú
- a Department of Chemical and Physical Biology , Centro de Investigaciones Biológicas (CIB, CSIC) Ramiro de Maeztu , Madrid , Spain.,b INFIQC- Department of Organic Chemistry, School of Chemical Sciences , National University of Córdoba , Córdoba , Argentine
| | - Victor Sebastián-Pérez
- a Department of Chemical and Physical Biology , Centro de Investigaciones Biológicas (CIB, CSIC) Ramiro de Maeztu , Madrid , Spain
| | - Loreto Martinez-Gonzalez
- a Department of Chemical and Physical Biology , Centro de Investigaciones Biológicas (CIB, CSIC) Ramiro de Maeztu , Madrid , Spain
| | - Rocio Benitez
- a Department of Chemical and Physical Biology , Centro de Investigaciones Biológicas (CIB, CSIC) Ramiro de Maeztu , Madrid , Spain
| | - Daniel I Perez
- a Department of Chemical and Physical Biology , Centro de Investigaciones Biológicas (CIB, CSIC) Ramiro de Maeztu , Madrid , Spain
| | | | - Nuria E Campillo
- a Department of Chemical and Physical Biology , Centro de Investigaciones Biológicas (CIB, CSIC) Ramiro de Maeztu , Madrid , Spain
| | - Ana Martinez
- a Department of Chemical and Physical Biology , Centro de Investigaciones Biológicas (CIB, CSIC) Ramiro de Maeztu , Madrid , Spain
| | - E Laura Moyano
- b INFIQC- Department of Organic Chemistry, School of Chemical Sciences , National University of Córdoba , Córdoba , Argentine
| |
Collapse
|
12
|
Anti-neuroinflammatory effects of SLOH in Aβ-induced BV-2 microglial cells and 3xTg-AD mice involve the inhibition of GSK-3β. Neurosci Lett 2018; 687:207-215. [PMID: 30278248 DOI: 10.1016/j.neulet.2018.09.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/06/2018] [Accepted: 09/27/2018] [Indexed: 01/14/2023]
Abstract
Neuroinflammation has been observed in post-mortem Alzheimer's disease (AD) brains which could be due to Aβ interacting with microglia and astrocytes. SLOH, a carbazole-based fluorophore, was shown to bind to Aβ peptides. Herein, we investigated the anti-neuroinflammatory effects of SLOH using a BV-2 microglial cell model and a triple transgenic AD (3xTg-AD) mouse model. BV-2 cells were incubated with Aβ in the presence of SLOH for 24 h. The levels of pro-inflammatory and anti-inflammatory cytokines were determined. Moreover, 3xTg-AD mice were administrated with SLOH (2 mg kg-1) for one month. The mice were then sacrificed and the brains were used to assess the levels of pro-inflammatory, anti-inflammatory cytokines and the activation of ionized calcium-binding adapter molecule 1 (Iba1). BV-2 cell studies suggested that SLOH reduced the production and mRNA levels of pro-inflammatory cytokines TNF-α, IL-1β, COX-2, iNOS, and increased IL-10. Animal study confirmed that SLOH reduced the production of pro-inflammatory cytokines and increased the level of anti-inflammatory cytokine. Moreover, SLOH inhibited the activity of GSK-3β. In 3xTg-AD mouse model, SLOH treatment significantly decreased the number of Iba1-positive cells in mouse brains. Our results demonstrated that SLOH significantly attenuated the neuroinflammation through down-regulating the activity of GSK-3β.
Collapse
|
13
|
SLOH, a carbazole-based fluorophore, mitigates neuropathology and behavioral impairment in the triple-transgenic mouse model of Alzheimer's disease. Neuropharmacology 2018; 131:351-363. [PMID: 29309769 DOI: 10.1016/j.neuropharm.2018.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/08/2017] [Accepted: 01/02/2018] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative dysfunction characterized by memory impairment and brings a heavy burden to old people both in developing and developed countries. Amyloid hypothesis reveals that aggregation and deposition of amyloid plaques are the cause of AD neurodegeneration. SLOH, a carbazole-based fluorophore, is reported to inhibit amyloid beta (Aβ) aggregation in vitro. In the current study, we intended to evaluate the protective effect of SLOH in a triple transgenic AD mouse model (3xTg-AD). 3xTg-AD (10-month-old) were treated with SLOH (0.5, 1 and 2 mg kg-1) for one month via intraperitoneal injection. After treatment, cognitive function was assessed by Morris Water Maze (MWM) and Y-maze tasks. In addition, biochemical estimations were used to examine the degree of Aβ deposition, tau hyperphosphorylation and neuroinflammation in the brains of 3xTg-AD mice. An in vitro study was conducted on human neuroblastoma (SH-SY5Y) cells to determine the activity of SLOH on tau and GSK-3β using western blot and immunofluorescence staining. One month treatment with SLOH significantly ameliorated memory impairments in 3xTg-AD mice in MWM and Y-maze tests. Moreover, SLOH treatment mitigated the level of amyloid plaques, tau hyperphosphorylation and neuroinflammation in the mouse brain. SLOH also reduced tau hyperphosphorylation and down-regulated GSK-3β activity in Aβ induced neurotoxic SH-SY5Y cells. The promising results in mitigating amyloid plaques, tau hyperphosphorylation, neuroinflammation and ameliorating cognitive deficits following one-month treatment suggest that SLOH could be a potential multi-target molecule for the AD treatment.
Collapse
|