1
|
Ansari MM, Sahu SK, Singh TG, Singh SRJ, Kaur P. Evolving significance of kinase inhibitors in the management of Alzheimer's disease. Eur J Pharmacol 2024; 979:176816. [PMID: 39038637 DOI: 10.1016/j.ejphar.2024.176816] [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: 03/23/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease is a neurodegenerative problem with progressive loss of memory and other cognitive function disorders resulting in the imbalance of neurotransmitter activity and signaling progression, which poses the need of the potential therapeutic target to improve the intracellular signaling cascade brought by kinases. Protein kinase plays a significant and multifaceted role in the treatment of Alzheimer's disease, by targeting pathological mechanisms like tau hyperphosphorylation, neuroinflammation, amyloid-beta production and synaptic dysfunction. In this review, we thoroughly explore the essential protein kinases involved in Alzheimer's disease, detailing their physiological roles, regulatory impacts, and the newest inhibitors and compounds that are progressing into clinical trials. All the findings of studies exhibited the promising role of kinase inhibitors in the management of Alzheimer's disease. However, it still poses the need of addressing current challenges and opportunities involved with this disorder for the future perspective of kinase inhibitors in the management of Alzheimer's disease. Further study includes the development of biomarkers, combination therapy, and next-generation kinase inhibitors with increased potency and selectivity for its future prospects.
Collapse
Affiliation(s)
- Md Mustafiz Ansari
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | | | - Sovia R J Singh
- University Language Centre- Chitkara Business School, Chitkara University, Punjab, India
| | - Paranjeet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
2
|
Hosseininia M, Rostami F, Delphi L, Ghasemzadeh Z, Kouhkan F, Rezayof A. Memory impairment was ameliorated by corticolimbic microinjections of arachidonylcyclopropylamide (ACPA) and miRNA-regulated lentiviral particles in a streptozotocin-induced Alzheimer's rat model. Exp Neurol 2023; 370:114560. [PMID: 37783412 DOI: 10.1016/j.expneurol.2023.114560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/16/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
The present study aimed to investigate the effect of corticolimbic cannabinoid CB1 receptors activity on memory impairment in the intracerebroventricular (ICV)-streptozotocin (STZ) animal model of Alzheimer's like-disease. This study also assessed whether the corticolimbic overexpression of miRNA-137 or -let-7a could increase the endocannabinoids by inhibiting the monoglyceride lipase (MAGL) to ameliorate STZ response. The results showed that ICV microinjection of STZ (3 mg/kg/10 μl) impaired passive avoidance memory retrieval. The chronic microinjection of arachidonylcyclopropylamide (ACPA; 10 ng/0.5 μl), a selective cannabinoid CB1 receptor agonist, into the hippocampal CA1 region, the central amygdala (CeA) or the medial prefrontal cortex (mPFC) ameliorated the amnesic effect of ICV-STZ. Intra-CA1 or -CeA microinjection of ACPA alone did not affect memory retrieval, while its microinjection into the mPFC impaired memory formation. Based on bioinformatics analysis and verification of the MAGL gene, miRNA-137 and -let-7a were chosen to target the expression levels of MAGL in the corticolimbic regions. The chronic corticolimbic microinjection of lentiviral particles containing miRNA-137 or -let-7a ameliorated ICV-STZ-induced memory impairment. The high transfection efficiency was determined for each virus using comparing fluorescent and conventional vision. Corticolimbic overexpression of miRNA-137 or -let-7a decreased the MAGL gene expression that encodes the MAGL enzyme to increase the endocannabinoids. Thus, among the molecular mechanisms and signaling pathways involved in the pathophysiology of Alzheimer's disease (AD), it is worth mentioning the role of endocannabinoids in the corticolimbic regions. CB1 receptor agonists, miRNA-137 or -let-7a, may be potential therapeutic targets against cognitive decline in AD.
Collapse
Affiliation(s)
- Mohammad Hosseininia
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Rostami
- Stem Cell Technology Research Center, P.O. Box: 15856-36473, 15856-36473 Tehran, Iran
| | - Ladan Delphi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Kouhkan
- Stem Cell Technology Research Center, P.O. Box: 15856-36473, 15856-36473 Tehran, Iran.
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
3
|
Lin CH, Hsieh YS, Sun YC, Huang WH, Chen SL, Weng ZK, Lin TH, Wu YR, Chang KH, Huang HJ, Lee GC, Hsieh-Li HM, Lee-Chen GJ. Virtual Screening and Testing of GSK-3 Inhibitors Using Human SH-SY5Y Cells Expressing Tau Folding Reporter and Mouse Hippocampal Primary Culture under Tau Cytotoxicity. Biomol Ther (Seoul) 2023; 31:127-138. [PMID: 35790892 PMCID: PMC9810448 DOI: 10.4062/biomolther.2022.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/05/2022] [Accepted: 05/24/2022] [Indexed: 01/13/2023] Open
Abstract
Glycogen synthase kinase-3β (GSK-3β) is an important serine/threonine kinase that implicates in multiple cellular processes and links with the neurodegenerative diseases including Alzheimer's disease (AD). In this study, structure-based virtual screening was performed to search database for compounds targeting GSK-3β from Enamine's screening collection. Of the top-ranked compounds, 7 primary hits underwent a luminescent kinase assay and a cell assay using human neuroblastoma SH-SY5Y cells expressing Tau repeat domain (TauRD) with pro-aggregant mutation ΔK280. In the kinase assay for these 7 compounds, residual GSK-3β activities ranged from 36.1% to 90.0% were detected at the IC50 of SB-216763. In the cell assay, only compounds VB-030 and VB-037 reduced Tau aggregation in SH-SY5Y cells expressing ΔK280 TauRD-DsRed folding reporter. In SH-SY5Y cells expressing ΔK280 TauRD, neither VB-030 nor VB-037 increased expression of GSK-3α Ser21 or GSK-3β Ser9. Among extracellular signal-regulated kinase (ERK), AKT serine/threonine kinase 1 (AKT), mitogen-activated protein kinase 14 (P38) and mitogen-activated protein kinase 8 (JNK) which modulate Tau phosphorylation, VB-037 attenuated active phosphorylation of P38 Thr180/Tyr182, whereas VB-030 had no effect on the phosphorylation status of ERK, AKT, P38 or JNK. However, both VB-030 and VB-037 reduced endogenous Tau phosphorylation at Ser202, Thr231, Ser396 and Ser404 in neuronally differentiated SH-SY5Y expressing ΔK280 TauRD. In addition, VB-030 and VB-037 further improved neuronal survival and/or neurite length and branch in mouse hippocampal primary culture under Tau cytotoxicity. Overall, through inhibiting GSK-3β kinase activity and/or p-P38 (Thr180/Tyr182), both compounds may serve as promising candidates to reduce Tau aggregation/cytotoxicity for AD treatment.
Collapse
Affiliation(s)
- Chih-Hsin Lin
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Yu-Shao Hsieh
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Ying-Chieh Sun
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Wun-Han Huang
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Shu-Ling Chen
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Zheng-Kui Weng
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Te-Hsien Lin
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan 33378, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan 33378, Taiwan
| | - Hei-Jen Huang
- Department of Nursing, Mackay Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
| | - Guan-Chiun Lee
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan,Corresponding Authors E-mail: (Lee GC), (Hsieh-Li HM), (Lee-Chen GJ), Tel:+886-2-7749-6351 (Lee GC), +886-2-7749-6354 (Hsieh-Li HM), +886-2-7749-6359 (Lee-Chen GJ), Fax:+886-2-2931-2904 (Lee GC), +886-2-2931-2904 (Hsieh-Li HM), +886-2-2931-2904 (Lee-Chen GJ)
| | - Hsiu Mei Hsieh-Li
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan,Corresponding Authors E-mail: (Lee GC), (Hsieh-Li HM), (Lee-Chen GJ), Tel:+886-2-7749-6351 (Lee GC), +886-2-7749-6354 (Hsieh-Li HM), +886-2-7749-6359 (Lee-Chen GJ), Fax:+886-2-2931-2904 (Lee GC), +886-2-2931-2904 (Hsieh-Li HM), +886-2-2931-2904 (Lee-Chen GJ)
| | - Guey-Jen Lee-Chen
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan,Corresponding Authors E-mail: (Lee GC), (Hsieh-Li HM), (Lee-Chen GJ), Tel:+886-2-7749-6351 (Lee GC), +886-2-7749-6354 (Hsieh-Li HM), +886-2-7749-6359 (Lee-Chen GJ), Fax:+886-2-2931-2904 (Lee GC), +886-2-2931-2904 (Hsieh-Li HM), +886-2-2931-2904 (Lee-Chen GJ)
| |
Collapse
|
4
|
Chiu YJ, Lin TH, Chang KH, Lin W, Hsieh-Li HM, Su MT, Chen CM, Sun YC, Lee-Chen GJ. Novel TRKB agonists activate TRKB and downstream ERK and AKT signaling to protect Aβ-GFP SH-SY5Y cells against Aβ toxicity. Aging (Albany NY) 2022; 14:7568-7586. [PMID: 36170028 PMCID: PMC9550238 DOI: 10.18632/aging.204306] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 09/17/2022] [Indexed: 11/28/2022]
Abstract
Decreased BDNF and impaired TRKB signaling contribute to neurodegeneration in Alzheimer’s disease (AD). We have shown previously that coumarin derivative LM-031 enhanced CREB/BDNF/BCL2 pathway. In this study we explored if LM-031 analogs LMDS-1 to -4 may act as TRKB agonists to protect SH-SY5Y cells against Aβ toxicity. By docking computation for binding with TRKB using 7,8-DHF as a control, all four LMDS compounds displayed potential of binding to domain d5 of TRKB. In addition, all four LMDS compounds exhibited anti-aggregation and neuroprotective efficacy on SH-SY5Y cells with induced Aβ-GFP expression. Knock-down of TRKB significantly attenuated TRKB downstream signaling and the neurite outgrowth-promoting effects of these LMDS compounds. Among them, LMDS-1 and -2 were further examined for TRKB signaling. Treatment of ERK inhibitor U0126 or PI3K inhibitor wortmannin decreased p-CREB, BDNF and BCL2 in Aβ-GFP cells, implicating the neuroprotective effects are via activating TRKB downstream ERK, PI3K-AKT and CREB signaling. LMDS-1 and -2 are blood–brain barrier permeable as shown by parallel artificial membrane permeability assay. Our results demonstrate how LMDS-1 and -2 are likely to work as TRKB agonists to exert neuroprotection in Aβ cells, which may shed light on the potential application in therapeutics of AD.
Collapse
Affiliation(s)
- Ya-Jen Chiu
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Te-Hsien Lin
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Ming-Tsan Su
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Ying-Chieh Sun
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| |
Collapse
|
5
|
Hassan NA, Alshamari AK, Hassan AA, Elharrif MG, Alhajri AM, Sattam M, Khattab RR. Advances on Therapeutic Strategies for Alzheimer's Disease: From Medicinal Plant to Nanotechnology. Molecules 2022; 27:4839. [PMID: 35956796 PMCID: PMC9369981 DOI: 10.3390/molecules27154839] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic dysfunction of neurons in the brain leading to dementia. It is characterized by gradual mental failure, abnormal cognitive functioning, personality changes, diminished verbal fluency, and speech impairment. It is caused by neuronal injury in the cerebral cortex and hippocampal area of the brain. The number of individuals with AD is growing at a quick rate. The pathology behind AD is the progress of intraneuronal fibrillary tangles, accumulation of amyloid plaque, loss of cholinergic neurons, and decrease in choline acetyltransferase. Unfortunately, AD cannot be cured, but its progression can be delayed. Various FDA-approved inhibitors of cholinesterase enzyme such as rivastigmine, galantamine, donepezil, and NDMA receptor inhibitors (memantine), are available to manage the symptoms of AD. An exhaustive literature survey was carried out using SciFinder's reports from Alzheimer's Association, PubMed, and Clinical Trials.org. The literature was explored thoroughly to obtain information on the various available strategies to prevent AD. In the context of the present scenario, several strategies are being tried including the clinical trials for the treatment of AD. We have discussed pathophysiology, various targets, FDA-approved drugs, and various drugs in clinical trials against AD. The goal of this study is to shed light on current developments and treatment options, utilizing phytopharmaceuticals, nanomedicines, nutraceuticals, and gene therapy.
Collapse
Affiliation(s)
- Nasser A. Hassan
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; (A.M.A.); (M.S.)
- Synthetic Unit, Department of Photochemistry, Chemical Industries Research Institute, National Research Centre, Cairo 12622, Egypt;
| | - Asma K. Alshamari
- Department of Chemistry, College of Science, Ha’il University, Ha’il 81451, Saudi Arabia;
| | - Allam A. Hassan
- Department of Chemistry, Faculty of Science, Suez University, Suez 43221, Egypt;
- Department of Chemistry, College of Science, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Mohamed G. Elharrif
- Department of Basic Medical Sciences, College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia;
| | - Abdullah M. Alhajri
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; (A.M.A.); (M.S.)
| | - Mohammed Sattam
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; (A.M.A.); (M.S.)
| | - Reham R. Khattab
- Synthetic Unit, Department of Photochemistry, Chemical Industries Research Institute, National Research Centre, Cairo 12622, Egypt;
| |
Collapse
|
6
|
Novel Aurora A Kinase Inhibitor Fangchinoline Enhances Cisplatin-DNA Adducts and Cisplatin Therapeutic Efficacy in OVCAR-3 Ovarian Cancer Cells-Derived Xenograft Model. Int J Mol Sci 2022; 23:ijms23031868. [PMID: 35163790 PMCID: PMC8836832 DOI: 10.3390/ijms23031868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/30/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Aurora A kinase (Aurora A) is a serine/threonine kinase regulating control of multiple events during cell-cycle progression. Playing roles in promoting proliferation and inhibiting cell death in cancer cells leads Aurora A to become a target for cancer therapy. It is overexpressed and associated with a poor prognosis in ovarian cancer. Improving cisplatin therapy outcomes remains an important issue for advanced-stage ovarian cancer treatment, and Aurora A inhibitors may improve it. In the present study, we identified natural compounds with higher docking scores than the known Aurora A ligand through structure-based virtual screening, including the natural compound fangchinoline, which has been associated with anticancer activities but not yet investigated in ovarian cancer. The binding and inhibition of Aurora A by fangchinoline were verified using cellular thermal shift and enzyme activity assays. Fangchinoline reduced viability and proliferation in ovarian cancer cell lines. Combination fangchinoline and cisplatin treatment enhanced cisplatin-DNA adduct levels, and the combination index revealed synergistic effects on cell viability. An in vivo study showed that fangchinoline significantly enhanced cisplatin therapeutic effects in OVCAR-3 ovarian cancer-bearing mice. Fangchinoline may inhibit tumor growth and enhance cisplatin therapy in ovarian cancer. This study reveals a novel Aurora A inhibitor, fangchinoline, as a potentially viable adjuvant for ovarian cancer therapy.
Collapse
|
7
|
Man VH, Lin D, He X, Gao J, Wang J. Joint Computational/Cell-Based Approach for Screening Inhibitors of Tau Oligomerization: A Proof-of-Concept Study. J Alzheimers Dis 2022; 89:107-119. [PMID: 35848028 DOI: 10.3233/jad-220450] [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] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tau assembly produces soluble oligomers and insoluble neurofibrillary tangles, which are neurotoxic to the brain and associated with Alzheimer's and Parkinson's diseases. Therefore, preventing tau aggregation is a promising therapy for those neurodegenerative disorders. OBJECTIVE The aim of this study was to develop a joint computational/cell-based oligomerization protocol for screening inhibitors of tau assembly. METHODS Virtual oligomerization inhibition (VOI) experiment using molecular dynamics simulation was performed to screen potential oligomerization inhibitors of PHF6 hexapeptide. Tau seeding assay, which is directly related to the outcome of therapeutic intervention, was carried out to confirm a ligand's ability in inhibiting tau assembly formation. RESULTS Our protocol was tested on two known compounds, EGCG and Blarcamesine. EGCG inhibited both the aggregation of PHF6 peptide in VOI and tau assembly in tau seeding assay, while Blarcamesine was not a good inhibitor at the two tasks. We also pointed out that good binding affinity to tau aggregates is needed, but not sufficient for a ligand to become a good inhibitor of tau oligomerization. CONCLUSION VOI goes beyond traditional computational inhibitor screening of amyloid aggregation by directly examining the inhibitory ability of a ligand to tau oligomerization. Comparing with the traditional biochemical assays, tau seeding activities in cells is a better indicator for the outcome of a therapeutic intervention. Our hybrid protocol has been successfully validated. It can effectively and efficiently identify the inhibitors of amyloid oligomerization/aggregation processes, thus, facilitate to the drug development of tau-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Viet Hoang Man
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Da Lin
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xibing He
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Gao
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Yang L, Yang G, Chen X, Yang Q, Yao X, Bing Z, Niu Y, Huang L, Yang L. Deep Scoring Neural Network Replacing the Scoring Function Components to Improve the Performance of Structure-Based Molecular Docking. ACS Chem Neurosci 2021; 12:2133-2142. [PMID: 34081851 DOI: 10.1021/acschemneuro.1c00110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Accurate prediction of protein-ligand interactions can greatly promote drug development. Recently, a number of deep-learning-based methods have been proposed to predict protein-ligand binding affinities. However, these methods independently extract the feature representations of proteins and ligands but ignore the relative spatial positions and interaction pairs between them. Here, we propose a virtual screening method based on deep learning, called Deep Scoring, which directly extracts the relative position information and atomic attribute information on proteins and ligands from the docking poses. Furthermore, we use two Resnets to extract the features of ligand atoms and protein residues, respectively, and generate an atom-residue interaction matrix to learn the underlying principles of the interactions between proteins and ligands. This is then followed by a dual attention network (DAN) to generate the attention for two related entities (i.e., proteins and ligands) and to weigh the contributions of each atom and residue to binding affinity prediction. As a result, Deep Scoring outperforms other structure-based deep learning methods in terms of screening performance (area under the receiver operating characteristic curve (AUC) of 0.901 for an unbiased DUD-E version), pose prediction (AUC of 0.935 for PDBbind test set), and generalization ability (AUC of 0.803 for the CHEMBL data set). Finally, Deep Scoring was used to select novel ERK2 inhibitor, and two compounds (D264-0698 and D483-1785) were obtained with potential inhibitory activity on ERK2 through the biological experiments.
Collapse
Affiliation(s)
- Lijuan Yang
- Institute of Modern Physics, Chinese Academy of Science, Lanzhou 730000, China
- School of Physics and Technology, Lanzhou University, Lanzhou 730000, China
- School of Physics, University of Chinese Academy of Science, Beijing 100049, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| | - Guanghui Yang
- Institute of Modern Physics, Chinese Academy of Science, Lanzhou 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| | - Xiaolong Chen
- Institute of Modern Physics, Chinese Academy of Science, Lanzhou 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| | - Qiong Yang
- Institute of Modern Physics, Chinese Academy of Science, Lanzhou 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| | - Xiaojun Yao
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Zhitong Bing
- Institute of Modern Physics, Chinese Academy of Science, Lanzhou 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| | - Yuzhen Niu
- Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo 255049, China
| | - Liang Huang
- School of Physics and Technology, Lanzhou University, Lanzhou 730000, China
| | - Lei Yang
- Institute of Modern Physics, Chinese Academy of Science, Lanzhou 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| |
Collapse
|
9
|
Rai SN, Mishra D, Singh P, Vamanu E, Singh MP. Therapeutic applications of mushrooms and their biomolecules along with a glimpse of in silico approach in neurodegenerative diseases. Biomed Pharmacother 2021; 137:111377. [PMID: 33601145 DOI: 10.1016/j.biopha.2021.111377] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Neurodegenerative diseases (NDs) represent a common neurological pathology that determines a progressive deterioration of the brain or the nervous system. For treating NDs, comprehensive and alternative medicines have attracted scientific researchers' attention recently. Edible mushrooms are essential for preventing several age-based neuronal dysfunctions such as Parkinson's and Alzheimer's diseases. Mushroom such as Grifola frondosa, Lignosus rhinocerotis, Hericium erinaceus, may improve cognitive functions. It has also been reported that edible mushrooms (basidiocarps/mycelia extracts or isolated bioactive compounds) may reduce beta-amyloid-induced neurotoxicity. Medicinal mushrooms are being used for novel and natural compounds that help modulate immune responses and possess anti-cancer, anti-microbial, and anti-oxidant properties. Compounds such as polyphenols, terpenoids, alkaloids, sesquiterpenes, polysaccharides, and metal chelating agents are validated in different ND treatments. This review aims to assess mushrooms' role and their biomolecules utilization for treating different kinds of NDs. The action mechanisms, presented here, including reducing oxidative stress, neuroinflammation, and modulation of acetylcholinesterase activity, protecting neurons or stimulation, and regulating neurotrophins synthesis. We also provide background about neurodegenerative diseases and in-silico techniques of the drug research. High costs associated with experiments and current ethical law imply efficient alternatives with limited cost value. In silico approaches provide an alternative method with low cost that has been successfully implemented to cure ND disorders in recent days. We also describe the applications of computational procedures such as molecular docking, virtual high-throughput screening, molecular dynamic (MD) simulation, quantum-mechanical methods for drug design. They were reported against various targets in NDs.
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Centre of Biotechnology, University of Allahabad, Prayagraj 211002, India.
| | - Divya Mishra
- Centre of Bioinformatics, University of Allahabad, Prayagraj 211002, India.
| | - Payal Singh
- Department of Zoology, MMV, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agronomic Science and Veterinary Medicine, 59 Marasti blvd, 1 district, 011464 Bucharest, Romania.
| | - M P Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj 211002, India.
| |
Collapse
|
10
|
Chiu YJ, Lin SA, Chen WL, Lin TH, Lin CH, Yao CF, Lin W, Wu YR, Chang KH, Lee-Chen GJ, Chen CM. Pathomechanism characterization and potential therapeutics identification for SCA3 targeting neuroinflammation. Aging (Albany NY) 2020; 12:23619-23646. [PMID: 33196459 PMCID: PMC7762503 DOI: 10.18632/aging.103700] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 06/29/2020] [Indexed: 12/12/2022]
Abstract
Polyglutamine (polyQ)-mediated spinocerebellar ataxias (SCA) are caused by mutant genes with expanded CAG repeats encoding polyQ tracts. The misfolding and aggregation of polyQ proteins result in increased reactive oxygen species (ROS) and cellular toxicity. Inflammation is a common manifestation of oxidative stress and inflammatory process further reduces cellular antioxidant capacity. Increase of activated microglia in the pons of SCA type 3 (SCA3) patients suggests the involvement of neuroinflammation in the disease pathogenesis. In this study, we evaluated the anti-inflammatory potentials of indole compound NC009-1, 4-aminophenol-arachidonic acid derivative AM404, quinoline compound VB-037 and chalcone-coumarin derivative LM-031 using human HMC3 microglia and SCA3 ATXN3/Q75-GFP SH-SY5Y cells. The four tested compounds displayed anti-inflammatory activity by suppressing NO, IL-1β, TNF-α and IL-6 production and CD68 expression of IFN-γ-activated HMC3 microglia. In retinoic acid-differentiated ATXN3/Q75-GFP SH-SY5Y cells inflamed with IFN-γ-primed HMC3 conditioned medium, treatment with the tested compounds mitigated the increased caspase 1 activity and lactate dehydrogenase release, reduced polyQ aggregation and ROS and/or promoted neurite outgrowth. Examination of IL-1β- and TNF-α-mediated signaling pathways revealed that the tested compounds decreased IκBα/P65, JNK/JUN and/or P38/STAT1 signaling. The study results suggest the potential of NC009-1, AM404, VB-037 and LM-031 in treating SCA3 and probable other polyQ diseases.
Collapse
Affiliation(s)
- Ya-Jen Chiu
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Shu-An Lin
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Wan-Ling Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Te-Hsien Lin
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chih-Hsin Lin
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| |
Collapse
|
11
|
Wang L, Bharti, Kumar R, Pavlov PF, Winblad B. Small molecule therapeutics for tauopathy in Alzheimer's disease: Walking on the path of most resistance. Eur J Med Chem 2020; 209:112915. [PMID: 33139110 DOI: 10.1016/j.ejmech.2020.112915] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia characterized by presence of extracellular amyloid plaques and intracellular neurofibrillary tangles composed of tau protein. Currently there are close to 50 million people living with dementia and this figure is expected to increase to 75 million by 2030 putting a huge burden on the economy due to the health care cost. Considering the effects on quality of life of patients and the increasing burden on the economy, there is an enormous need of new disease modifying therapies to tackle this disease. The current therapies are dominated by only symptomatic treatments including cholinesterase inhibitors and N-methyl-D-aspartate receptor blockers but no disease modifying treatments exist so far. After several failed attempts to develop drugs against amyloidopathy, tau targeting approaches have been in the main focus of drug development against AD. After an overview of the tauopathy in AD, this review summarizes recent findings on the development of small molecules as therapeutics targeting tau modification, aggregation, and degradation, and tau-oriented multi-target directed ligands. Overall, this work aims to provide a comprehensive and critical overview of small molecules which are being explored as a lead candidate for discovering drugs against tauopathy in AD.
Collapse
Affiliation(s)
- Lisha Wang
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
| | - Bharti
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Rajnish Kumar
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Pavel F Pavlov
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Memory Clinic, Theme Aging, Karolinska University Hospital, 14186, Huddinge, Sweden
| | - Bengt Winblad
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Memory Clinic, Theme Aging, Karolinska University Hospital, 14186, Huddinge, Sweden.
| |
Collapse
|
12
|
Qiu ZC, Zhang Y, Xiao HH, Chui-Wa Poon C, Li XL, Cui JF, Wong MK, Yao XS, Wong MS. 8-prenylgenistein exerts osteogenic effects via ER α and Wnt-dependent signaling pathway. Exp Cell Res 2020; 395:112186. [DOI: 10.1016/j.yexcr.2020.112186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 12/20/2022]
|
13
|
Loeffler DA. AMBAR, an Encouraging Alzheimer's Trial That Raises Questions. Front Neurol 2020; 11:459. [PMID: 32547478 PMCID: PMC7272580 DOI: 10.3389/fneur.2020.00459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
Grifols' recent Alzheimer Management by Albumin Replacement (“AMBAR”) study investigated the effects of plasmapheresis with albumin replacement, plus intravenous immunoglobulin (IVIG) in some subjects, in patients with mild-to-moderate Alzheimer's disease (AD). AMBAR was a phase IIb trial in the United States and a phase III trial in Europe. There were three treatment groups (plasmapheresis with albumin replacement; plasmapheresis with low dose albumin and IVIG; plasmapheresis with high dose albumin and IVIG) and sham-treated controls. Disease progression in pooled treated patients was 66% less than control subjects based on ADAS-Cog scores (p = 0.06) and 52% less based on ADCS-ADL scores (p = 0.03). Moderate AD patients had 61% less progression, based on both ADAS-Cog and ADCS-ADL scores, than their sham-treated counterparts (p-values 0.05 and 0.002), and their CDR-Sb scores declined 53% less than their sham-treated counterparts. However, ADAS-Cog and ADCS-ADL scores were not significantly different between actively-treated and sham-treated mild AD patients, although CDR-Sb scores improved vs. baseline for treated mild AD patients. Patients administered both IVIG and albumin had less reduction in brain glucose metabolism than sham-treated patients. Questions raised by these findings include: what mechanism(s) contributed to slowing of disease progression? Is this approach as effective in mild AD as in moderate AD? Must IVIG be included in the protocol? Does age, sex, or ApoE genotype influence treatment response? Does the protocol increase the risk for amyloid-related imaging abnormalities? How long does disease progression remain slowed post-treatment? A further study should allow this approach to be optimized.
Collapse
Affiliation(s)
- David A Loeffler
- Beaumont Research Institute, Department of Neurology, Beaumont Health, Royal Oak, MI, United States
| |
Collapse
|
14
|
Iraji A, Khoshneviszadeh M, Firuzi O, Khoshneviszadeh M, Edraki N. Novel small molecule therapeutic agents for Alzheimer disease: Focusing on BACE1 and multi-target directed ligands. Bioorg Chem 2020; 97:103649. [PMID: 32101780 DOI: 10.1016/j.bioorg.2020.103649] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/05/2020] [Accepted: 02/03/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder that effects 50 million people worldwide. In this review, AD pathology and the development of novel therapeutic agents targeting AD were fully discussed. In particular, common approaches to prevent Aβ production and/or accumulation in the brain including α-secretase activators, specific γ-secretase modulators and small molecules BACE1 inhibitors were reviewed. Additionally, natural-origin bioactive compounds that provide AD therapeutic advances have been introduced. Considering AD is a multifactorial disease, the therapeutic potential of diverse multi target-directed ligands (MTDLs) that combine the efficacy of cholinesterase (ChE) inhibitors, MAO (monoamine oxidase) inhibitors, BACE1 inhibitors, phosphodiesterase 4D (PDE4D) inhibitors, for the treatment of AD are also reviewed. This article also highlights descriptions on the regulator of serotonin receptor (5-HT), metal chelators, anti-aggregants, antioxidants and neuroprotective agents targeting AD. Finally, current computational methods for evaluating the structure-activity relationships (SAR) and virtual screening (VS) of AD drugs are discussed and evaluated.
Collapse
Affiliation(s)
- Aida Iraji
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsima Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
Wei S, Peng W, Mai Y, Li K, Wei W, Hu L, Zhu S, Zhou H, Jie W, Wei Z, Kang C, Li R, Liu Z, Zhao B, Cai Z. Outer membrane vesicles enhance tau phosphorylation and contribute to cognitive impairment. J Cell Physiol 2019; 235:4843-4855. [PMID: 31663118 DOI: 10.1002/jcp.29362] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/07/2019] [Indexed: 01/18/2023]
Affiliation(s)
- Shouchao Wei
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Wanjuan Peng
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Yingren Mai
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Kanglan Li
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Wei Wei
- Health Department, Gaomi People's Hospital Weifang Medical University Gaomi China
| | - Li Hu
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Shaoping Zhu
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Haihong Zhou
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Wanxin Jie
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Zhuangsheng Wei
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Chenyao Kang
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Ruikai Li
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Zhou Liu
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Bin Zhao
- Department of Neurology, Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral Diseases, Institute of Neurology Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Zhiyou Cai
- Department of Neurology, Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing General Hospital University of Chinese Academy of Sciences Chongqing China
| |
Collapse
|
16
|
Gorantla NV, Das R, Mulani FA, Thulasiram HV, Chinnathambi S. Neem Derivatives Inhibits Tau Aggregation. J Alzheimers Dis Rep 2019; 3:169-178. [PMID: 31259310 PMCID: PMC6597962 DOI: 10.3233/adr-190118] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tau is a phosphoprotein with natively unfolded conformation that functions to stabilize microtubules in axons. Alzheimer’s disease pathology triggers several modifications in tau, which causes it to lose its affinity towards microtubule, thus, leading to microtubule disassembly and loss of axonal integrity. This elicit accumulation of tau as paired helical filaments is followed by stable neurofibrillary tangles formation. A large number of small molecules have been isolated from Azadirachta indica with varied medicinal applications. The intermediate and final limonoids, nimbin and salannin respectively, isolated from Azadirachta indica, were screened against tau aggregation. ThS and ANS fluorescence assay showed the role of intermediate and final limonoids in preventing heparin induced cross-β sheet formation and also decreased hydrophobicity, which are characteristic nature of tau aggregation. Transmission electron microscopy studies revealed that limonoids restricted the aggregation of tau to fibrils; in turn, limonoids led to the formation of short and fragile aggregates. Both the limonoids were non-toxic to HEK293T cells thus, substantiating limonoids as a potential lead in overcoming Alzheimer’s disease.
Collapse
Affiliation(s)
- Nalini V Gorantla
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Rashmi Das
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Fayaj A Mulani
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Hirekodathakallu V Thulasiram
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| |
Collapse
|
17
|
Kaufman MJ, Kanayama G, Hudson JI, Pope HG. Supraphysiologic-dose anabolic-androgenic steroid use: A risk factor for dementia? Neurosci Biobehav Rev 2019; 100:180-207. [PMID: 30817935 PMCID: PMC6451684 DOI: 10.1016/j.neubiorev.2019.02.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 02/06/2023]
Abstract
Supraphysiologic-dose anabolic-androgenic steroid (AAS) use is associated with physiologic, cognitive, and brain abnormalities similar to those found in people at risk for developing Alzheimer's Disease and its related dementias (AD/ADRD), which are associated with high brain β-amyloid (Aβ) and hyperphosphorylated tau (tau-P) protein levels. Supraphysiologic-dose AAS induces androgen abnormalities and excess oxidative stress, which have been linked to increased and decreased expression or activity of proteins that synthesize and eliminate, respectively, Aβ and tau-P. Aβ and tau-P accumulation may begin soon after initiating supraphysiologic-dose AAS use, which typically occurs in the early 20s, and their accumulation may be accelerated by other psychoactive substance use, which is common among non-medical AAS users. Accordingly, the widespread use of supraphysiologic-dose AAS may increase the numbers of people who develop dementia. Early diagnosis and correction of sex-steroid level abnormalities and excess oxidative stress could attenuate risk for developing AD/ADRD in supraphysiologic-dose AAS users, in people with other substance use disorders, and in people with low sex-steroid levels or excess oxidative stress associated with aging.
Collapse
Affiliation(s)
- Marc J Kaufman
- McLean Imaging Center, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA.
| | - Gen Kanayama
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - James I Hudson
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Harrison G Pope
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Jain AK, Karthikeyan C, McIntosh KD, Tiwari AK, Trivedi P, DuttKonar A. Unravelling the potency of 4,5-diamino-4H-1,2,4 triazole-3-thiol derivatives for kinase inhibition using a rational approach. NEW J CHEM 2019. [DOI: 10.1039/c8nj04205e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
This report describes the design of potent kinase inhibitors by simply fine tuning the surroundings of triazole core with diversified derivatization.
Collapse
Affiliation(s)
- Arvind Kumar Jain
- School of Pharmaceutical Sciences
- Rajiv Gandhi Technological University
- Bhopal
- Gandhinagar
- India
| | - C. Karthikeyan
- School of Pharmaceutical Sciences
- Rajiv Gandhi Technological University
- Bhopal
- Gandhinagar
- India
| | - Kyle Douglas McIntosh
- Department of Pharmacology and Experimental Therapeutics
- College of Pharmacy and Pharmaceutical Sciences
- University of Toledo
- USA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics
- College of Pharmacy and Pharmaceutical Sciences
- University of Toledo
- USA
| | - Piyush Trivedi
- School of Pharmaceutical Sciences
- Rajiv Gandhi Technological University
- Bhopal
- Gandhinagar
- India
| | - Anita DuttKonar
- School of Pharmaceutical Sciences
- Rajiv Gandhi Technological University
- Bhopal
- Gandhinagar
- India
| |
Collapse
|
19
|
Huang HJ, Chen SL, Huang HY, Sun YC, Lee GC, Lee-Chen GJ, Hsieh-Li HM, Su MT. Chronic low dose of AM404 ameliorates the cognitive impairment and pathological features in hyperglycemic 3xTg-AD mice. Psychopharmacology (Berl) 2019; 236:763-773. [PMID: 30426182 PMCID: PMC6469654 DOI: 10.1007/s00213-018-5108-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022]
Abstract
RATIONALE Hyperglycemia accelerates the progression of Alzheimer's disease (AD), and GSK3β plays a potential link between AD and hyperglycemia. Therefore, a direct or indirect GSK3β inhibition may have potential to delay the progression of AD. Our previous biochemical assay identified AM404 as a GSK3β inhibitor at high dose (IC50 = 5.353 μM); however, other study suggests that AM404 impaired synaptic plasticity of hippocampus at high dose (10 mg/kg; i.p.). Therefore, the dose and duration of treatment are crucial for the effects of AM404. OBJECTIVE The effects and molecular mechanisms of AM404 at low dose were evaluated from in vitro to in vivo models. METHODS AM404 (0.1-0.5 μM) was tested on tau hyperphosphorylated mouse hippocampal primary cultures treated with Wortmannin (WT) and GF109203X (GFX). Hyperglycemic triple transgenic AD (3×Tg-AD) mice at 6 months old were intraperitoneally injected with AM404 (0.25 mg/kg) for 4 weeks. The spatial learning and memory of mice were measured using the Morris water maze. Mouse brain and serum samples were collected for pathological analyses. RESULTS AM404 (0.5 μM) exhibited significantly augmented neuroprotection toward tau hyperphosphorylation in primary cultures. The chronic systemic administration of AM404 (0.25 mg/kg) attenuated cognitive deficits in hyperglycemic 3×Tg-AD mice. Moreover, chronic low dose of AM404 significantly attenuated Aβ production, tau protein phosphorylation, and inflammation associated with an increase of pS473Akt and pS9-GSK3β. Therefore, AM404 at low dose, not only increased neuroprotection, but also ameliorated cognitive deficit, could be partly by regulating the Akt/GSK3β signaling, which may contribute to downregulation of Aβ, tau hyperphosphorylation, and inflammation in hyperglycemic 3×Tg-AD mice. CONCLUSIONS These results highlight that chronic administration of AM404 at low dose may be through the Akt/GSK3β pathway to ameliorate the impairment in hyperglycemic 3×Tg-AD mice.
Collapse
Affiliation(s)
- Hei-Jen Huang
- Department of Nursing, Mackay Junior College of Medicine, Nursing and Management, Taipei, 11260 Taiwan
| | - Shu-Ling Chen
- 0000 0001 2158 7670grid.412090.eDepartment of Life Science, National Taiwan Normal University, Taipei, 11677 Taiwan
| | - Hsin-Yu Huang
- 0000 0001 2158 7670grid.412090.eDepartment of Life Science, National Taiwan Normal University, Taipei, 11677 Taiwan
| | - Ying-Chieh Sun
- 0000 0001 2158 7670grid.412090.eDepartment of Chemistry, National Taiwan Normal University, Taipei, 11677 Taiwan
| | - Guan-Chiun Lee
- 0000 0001 2158 7670grid.412090.eDepartment of Life Science, National Taiwan Normal University, Taipei, 11677 Taiwan
| | - Guey-Jen Lee-Chen
- 0000 0001 2158 7670grid.412090.eDepartment of Life Science, National Taiwan Normal University, Taipei, 11677 Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan.
| | - Ming-Tsan Su
- Department of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan.
| |
Collapse
|
20
|
Makhouri FR, Ghasemi JB. In Silico Studies in Drug Research Against Neurodegenerative Diseases. Curr Neuropharmacol 2018; 16:664-725. [PMID: 28831921 PMCID: PMC6080098 DOI: 10.2174/1570159x15666170823095628] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 07/24/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Background Neurodegenerative diseases such as Alzheimer's disease (AD), amyotrophic lateral sclerosis, Parkinson's disease (PD), spinal cerebellar ataxias, and spinal and bulbar muscular atrophy are described by slow and selective degeneration of neurons and axons in the central nervous system (CNS) and constitute one of the major challenges of modern medicine. Computer-aided or in silico drug design methods have matured into powerful tools for reducing the number of ligands that should be screened in experimental assays. Methods In the present review, the authors provide a basic background about neurodegenerative diseases and in silico techniques in the drug research. Furthermore, they review the various in silico studies reported against various targets in neurodegenerative diseases, including homology modeling, molecular docking, virtual high-throughput screening, quantitative structure activity relationship (QSAR), hologram quantitative structure activity relationship (HQSAR), 3D pharmacophore mapping, proteochemometrics modeling (PCM), fingerprints, fragment-based drug discovery, Monte Carlo simulation, molecular dynamic (MD) simulation, quantum-mechanical methods for drug design, support vector machines, and machine learning approaches. Results Detailed analysis of the recently reported case studies revealed that the majority of them use a sequential combination of ligand and structure-based virtual screening techniques, with particular focus on pharmacophore models and the docking approach. Conclusion Neurodegenerative diseases have a multifactorial pathoetiological origin, so scientists have become persuaded that a multi-target therapeutic strategy aimed at the simultaneous targeting of multiple proteins (and therefore etiologies) involved in the development of a disease is recommended in future.
Collapse
Affiliation(s)
| | - Jahan B Ghasemi
- Chemistry Department, Faculty of Sciences, University of Tehran, Tehran, Iran
| |
Collapse
|
21
|
Moreira-Silva D, Carrettiero DC, Oliveira ASA, Rodrigues S, Dos Santos-Lopes J, Canas PM, Cunha RA, Almeida MC, Ferreira TL. Anandamide Effects in a Streptozotocin-Induced Alzheimer's Disease-Like Sporadic Dementia in Rats. Front Neurosci 2018; 12:653. [PMID: 30333717 PMCID: PMC6176656 DOI: 10.3389/fnins.2018.00653] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by multiple cognitive deficits including memory and sensorimotor gating impairments as a result of neuronal and synaptic loss. The endocannabinoid system plays an important role in these deficits but little is known about its influence on the molecular mechanism regarding phosphorylated tau (p-tau) protein accumulation - one of the hallmarks of AD -, and on the density of synaptic proteins. Thus, the aim of this study was to investigate the preventive effects of anandamide (N-arachidonoylethanolamine, AEA) on multiple cognitive deficits and on the levels of synaptic proteins (syntaxin 1, synaptophysin and synaptosomal-associated protein, SNAP-25), cannabinoid receptor type 1 (CB1) and molecules related to p-tau degradation machinery (heat shock protein 70, HSP70), and Bcl2-associated athanogene (BAG2) in an AD-like sporadic dementia model in rats using intracerebroventricular (icv) injection of streptozotocin (STZ). Our hypothesis is that AEA could interact with HSP70, modulating the level of p-tau and synaptic proteins, preventing STZ-induced cognitive impairments. Thirty days after receiving bilateral icv injections of AEA or STZ or both, the cognitive performance of adult male Wistar rats was evaluated in the object recognition test, by the escape latency in the elevated plus maze (EPM), by the tone and context fear conditioning as well as in prepulse inhibition tests. Subsequently, the animals were euthanized and their brains were removed for histological analysis or for protein quantification by Western Blotting. The behavioral results showed that STZ impaired recognition, plus maze and tone fear memories but did not affect contextual fear memory and prepulse inhibition. Moreover, AEA prevented recognition and non-associative emotional memory impairments induced by STZ, but did not influence tone fear conditioning. STZ increased the brain ventricular area and this enlargement was prevented by AEA. Additionally, STZ reduced the levels of p-tau (Ser199/202) and increased p-tau (Ser396), although AEA did not affect these alterations. HSP70 was found diminished only by STZ, while BAG2 levels were decreased by STZ and AEA. Synaptophysin, syntaxin and CB1 receptor levels were reduced by STZ, but only syntaxin was recovered by AEA. Altogether, albeit AEA failed to modify some AD-like neurochemical alterations, it partially prevented STZ-induced cognitive impairments, changes in synaptic markers and ventricle enlargement. This study showed, for the first time, that the administration of an endocannabinoid can prevent AD-like effects induced by STZ, boosting further investigations about the modulation of endocannabinoid levels as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Daniel Moreira-Silva
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Daniel C Carrettiero
- Center for Natural and Human Sciences, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Adriele S A Oliveira
- Center for Natural and Human Sciences, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Samanta Rodrigues
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Joyce Dos Santos-Lopes
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Paula M Canas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Rodrigo A Cunha
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Maria C Almeida
- Center for Natural and Human Sciences, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Tatiana L Ferreira
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| |
Collapse
|
22
|
Kim H, Han H. Computer-Aided Multi-Target Management of Emergent Alzheimer's Disease. Bioinformation 2018; 14:167-180. [PMID: 29983487 PMCID: PMC6016757 DOI: 10.6026/97320630014167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) represents an enormous global health burden in terms of human suffering and economic cost. AD management requires a shift from the prevailing paradigm targeting pathogenesis to design and develop effective drugs with adequate success in clinical trials. Therefore, it is of interest to report a review on amyloid beta (Aβ) effects and other multi-targets including cholinesterase, NFTs, tau protein and TNF associated with brain cell death to be neuro-protective from AD. It should be noted that these molecules have been generated either by target-based or phenotypic methods. Hence, the use of recent advancements in nanomedicine and other natural compounds screening tools as a feasible alternative for circumventing specific liabilities is realized. We review recent developments in the design and identification of neuro-degenerative compounds against AD generated using current advancements in computational multi-target modeling algorithms reflected by theragnosis (combination of diagnostic tests and therapy) concern.
Collapse
Affiliation(s)
- Hyunjo Kim
- Department of Medical Informatics, Ajou Medical University Hospital, Suwon, Kyeounggido province, South Korea
| | - Hyunwook Han
- Department of Informatics, School of Medicine, CHA University, Seongnam, South Korea
- Institute of Basic Medical Sciences, School of Medicine, CHA University, Seongnam, South Korea
| |
Collapse
|
23
|
Kumar A, Tiwari A, Sharma A. Changing Paradigm from one Target one Ligand Towards Multi-target Directed Ligand Design for Key Drug Targets of Alzheimer Disease: An Important Role of In Silico Methods in Multi-target Directed Ligands Design. Curr Neuropharmacol 2018; 16:726-739. [PMID: 29542413 PMCID: PMC6080096 DOI: 10.2174/1570159x16666180315141643] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/01/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
Alzheimer disease (AD) is now considered as a multifactorial neurodegenerative disorder and rapidly increasing to an alarming situation and causing higher death rate. One target one ligand hypothesis does not provide complete solution of AD due to multifactorial nature of the disease and one target one drug fails to provide better treatment against AD. Moreover, currently available treatments are limited and most of the upcoming treatments under clinical trials are based on modulating single target. So, the current AD drug discovery research is shifting towards a new approach for a better solution that simultaneously modulates more than one targets in the neurodegenerative cascade. This can be achieved by network pharmacology, multi-modal therapies, multifaceted, and/or the more recently proposed term "multi-targeted designed drugs". Drug discovery project is a tedious, costly and long-term project. Moreover, multi-target AD drug discovery added extra challenges such as the good binding affinity of ligands for multiple targets, optimal ADME/T properties, no/less off-target side effect and crossing of the blood-brain barrier. These hurdles may be addressed by insilico methods for an efficient solution in less time and cost as computational methods successfully applied to single target drug discovery project. Here, we are summarizing some of the most prominent and computationally explored single targets against AD and further, we discussed a successful example of dual or multiple inhibitors for same targets. Moreover, we focused on ligand and structure-based computational approach to design MTDL against AD. However, it is not an easy task to balance dual activity in a single molecule but computational approach such as virtual screening docking, QSAR, simulation and free energy is useful in future MTDLs drug discovery alone or in combination with a fragment-based method. However, rational and logical implementations of computational drug designing methods are capable of assisting AD drug discovery and play an important role in optimizing multi-target drug discovery.
Collapse
Affiliation(s)
- Akhil Kumar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow-226015, (U.P.), India
| | - Ashish Tiwari
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow-226015, (U.P.), India
| | - Ashok Sharma
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow-226015, (U.P.), India
| |
Collapse
|
24
|
Hsu CJ, Hsu WC, Lee DJ, Liu AL, Chang CM, Shih HJ, Huang WH, Lee-Chen GJ, Hsieh-Li HM, Lee GC, Sun YC. Investigation of the bindings of a class of inhibitors with GSK3β kinase using thermodynamic integration MD simulation and kinase assay. Chem Biol Drug Des 2017; 90:272-281. [PMID: 28127860 DOI: 10.1111/cbdd.12946] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 12/09/2016] [Accepted: 01/05/2017] [Indexed: 01/08/2023]
Abstract
GSK3β kinase is a noteworthy target for discovery of the drugs that will be used to treat several diseases. In the effort to identify a new inhibitor lead compound, we utilized thermodynamic integration (TI)-molecular dynamics (MD) simulation and kinase assay to investigate the bindings between GSK3β kinase and five compounds that were analogous to a known inhibitor with an available crystal structure. TI-MD simulations of the first two compounds (analogs 1 and 2) were used for calibration. The computed binding affinities of analogs 1 and 2 agreed well with the experimental results. The rest three compounds (analogs 3-5) were newly obtained from a database search, and their affinity data were newly measured in our labs. TI-MD simulations predicted the binding modes and the computed ΔΔG values have a reasonably good correlation with the experimental affinity data. These newly identified inhibitors appear to be new leads according to our survey of GSK3β inhibitors listed in recent review articles. The predicted binding modes of these compounds should aid in designing new derivatives of these compounds in the future.
Collapse
Affiliation(s)
- Chia-Jen Hsu
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Wen-Chi Hsu
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Der-Jay Lee
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - An-Lun Liu
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Chia-Ming Chang
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Huei-Jhen Shih
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Wun-Han Huang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Guan-Chiun Lee
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ying-Chieh Sun
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
25
|
St-Cyr Giguère F, Attiori Essis S, Chagniel L, Germain M, Cyr M, Massicotte G. The sphingosine-1-phosphate receptor 1 agonist SEW2871 reduces Tau-Ser262 phosphorylation in rat hippocampal slices. Brain Res 2017; 1658:51-59. [DOI: 10.1016/j.brainres.2017.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/13/2022]
|
26
|
Lawson M, Rodrigo J, Baratte B, Robert T, Delehouzé C, Lozach O, Ruchaud S, Bach S, Brion JD, Alami M, Hamze A. Synthesis, biological evaluation and molecular modeling studies of imidazo[1,2-a]pyridines derivatives as protein kinase inhibitors. Eur J Med Chem 2016; 123:105-114. [PMID: 27474927 DOI: 10.1016/j.ejmech.2016.07.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/23/2016] [Accepted: 07/19/2016] [Indexed: 01/04/2023]
Abstract
We report here the synthesis, the biological evaluation and the molecular modeling studies of new imidazo[1,2-a]pyridines derivatives designed as potent kinase inhibitors. This collection was obtained from 2-aminopyridines and 2-bromoacetophenone which afforded final compound in only one step. The bioactivity of this family of new compounds was tested using protein kinase and ATP competition assays. The structure-activity relationship (SAR) revealed that six compounds inhibit DYRK1A and CLK1 at a micromolar range. Docking studies provided possible explanations that correlate with the SAR data. The most active compound 4c inhibits CLK1 (IC50 of 0.7 μM) and DYRK1A (IC50 of 2.6 μM).
Collapse
Affiliation(s)
- Marie Lawson
- BioCIS, Univ. Paris-Sud, CNRS, équipe labellisée Ligue Contre le Cancer, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Jordi Rodrigo
- BioCIS, Univ. Paris-Sud, CNRS, équipe labellisée Ligue Contre le Cancer, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Blandine Baratte
- Sorbonne Universités, UPMC Univ Paris 06, CNRS USR3151, "Protein Phosphorylation and Human Disease" Unit, Plateforme de criblage KISSf, Station Biologique de Roscoff, Place Georges Teissier, 29688, Roscoff, France
| | - Thomas Robert
- Sorbonne Universités, UPMC Univ Paris 06, CNRS USR3151, "Protein Phosphorylation and Human Disease" Unit, Plateforme de criblage KISSf, Station Biologique de Roscoff, Place Georges Teissier, 29688, Roscoff, France
| | - Claire Delehouzé
- Sorbonne Universités, UPMC Univ Paris 06, CNRS USR3151, "Protein Phosphorylation and Human Disease" Unit, Plateforme de criblage KISSf, Station Biologique de Roscoff, Place Georges Teissier, 29688, Roscoff, France
| | - Olivier Lozach
- Sorbonne Universités, UPMC Univ Paris 06, CNRS USR3151, "Protein Phosphorylation and Human Disease" Unit, Plateforme de criblage KISSf, Station Biologique de Roscoff, Place Georges Teissier, 29688, Roscoff, France
| | - Sandrine Ruchaud
- Sorbonne Universités, UPMC Univ Paris 06, CNRS USR3151, "Protein Phosphorylation and Human Disease" Unit, Plateforme de criblage KISSf, Station Biologique de Roscoff, Place Georges Teissier, 29688, Roscoff, France
| | - Stéphane Bach
- Sorbonne Universités, UPMC Univ Paris 06, CNRS USR3151, "Protein Phosphorylation and Human Disease" Unit, Plateforme de criblage KISSf, Station Biologique de Roscoff, Place Georges Teissier, 29688, Roscoff, France
| | - Jean-Daniel Brion
- BioCIS, Univ. Paris-Sud, CNRS, équipe labellisée Ligue Contre le Cancer, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Mouad Alami
- BioCIS, Univ. Paris-Sud, CNRS, équipe labellisée Ligue Contre le Cancer, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Abdallah Hamze
- BioCIS, Univ. Paris-Sud, CNRS, équipe labellisée Ligue Contre le Cancer, Université Paris-Saclay, 92290, Châtenay-Malabry, France.
| |
Collapse
|