1
|
Guo J, Zhu Y, Zhi J, Lou Q, Bai R, He Y. Antioxidants in anti-Alzheimer's disease drug discovery. Ageing Res Rev 2025; 107:102707. [PMID: 40021094 DOI: 10.1016/j.arr.2025.102707] [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: 01/15/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Oxidative stress is widely recognized as a key contributor to the pathogenesis of Alzheimer's disease (AD). While not the sole factor, it is closely linked to critical pathological features, such as the formation of senile plaques and neurofibrillary tangles. The development of agents with antioxidant properties has become an area of growing interest in AD research. Between 2015 and 2024, several antioxidant-targeted drugs for AD progressed to clinical trials, with increasing attention to the evaluation of antioxidant properties during their development. Oxidative stress plays a pivotal role in linking various AD hypotheses, underscoring its importance in understanding the disease mechanisms. Despite this, comprehensive reviews addressing advancements in AD drug development from the perspective of antioxidant capacity remain limited, hindering the design of novel compounds. This review aims to explore the mechanistic relationship between oxidative stress and AD, summarize methods for assessing antioxidant capacity, and provide an overview of antioxidant compounds with anti-AD properties reported over the past decade. The goal is to offer strategies for identifying effective antioxidant-based therapies for AD and to deepen our understanding of the role of oxidative stress in AD pathology.
Collapse
Affiliation(s)
- Jianan Guo
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China; Central Laboratory and Precision Medicine Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China; Jinhua Key Laboratory of Cancer Nutrition and Metabolism Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China.
| | - Yalan Zhu
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China
| | - Jia Zhi
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Qiuwen Lou
- Central Laboratory and Precision Medicine Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China; Jinhua Key Laboratory of Cancer Nutrition and Metabolism Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China
| | - Renren Bai
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China.
| | - Yiling He
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China.
| |
Collapse
|
2
|
Li Y, Zhang Q, Wang X, Liu Z, Chen H, Su Z, Xu Y, Zhang W, Du Y, Tan Z, Huang S, Liu W, Sang Z. Development of novel rivastigmine derivatives as selective BuChE inhibitors for the treatment of AD. Bioorg Chem 2025; 157:108245. [PMID: 39954353 DOI: 10.1016/j.bioorg.2025.108245] [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: 12/13/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder among the elderly, and there are currently no effective treatment options available. Selective inhibition of butyrylcholinesterase (BuChE) has emerged as a promising strategy for AD therapeutics. In this study, we identified compound 6a as a lead candidate derived from the structural modification of rivastigmine. Our findings indicated that 6a acts as a potent selective BuChE inhibitor, demonstrating an IC50 value of 0.33 μM (SI > 151.5). Furthermore, compound 6a displayed favorable neuroprotective properties, along with excellent blood-brain barrier (BBB) permeability and drug-like characteristics. In vivo investigations utilizing an AlCl3-induced zebrafish model of AD revealed that compound 6a significantly improved cognitive function, which was further supported by its ability to mitigate memory impairment induced by scopolamine. Overall, these results highlight compound 6a as a promising selective BuChE inhibitor with potential therapeutic implications for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yuting Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China; School of Life and Health Sciences, Hainan University, Haikou 570228 China
| | - Qiyao Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China; School of Food Science and Engineering, Hainan University, Haikou 570228 China
| | - Xinxin Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China
| | - Zhengwei Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China; College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061 China
| | - Hongsong Chen
- College of Animal Science and Technology. Inner Mongolia Minzu University, Tongliao, Inner Mongolia 028000, China
| | - Zhenhui Su
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China
| | - Yidan Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China
| | - Weichang Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China
| | - Yulu Du
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China
| | - Zhenghuai Tan
- Institute of Traditional Chinese Medicine Pharmacology and Toxicology, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Shuheng Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China.
| | - Wenmin Liu
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061 China.
| | - Zhipei Sang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228 China.
| |
Collapse
|
3
|
Zhao X, Hu Q, Wang X, Li C, Chen X, Zhao D, Qiu Y, Xu H, Wang J, Ren L, Zhang N, Li S, Gong P, Hou Y. Dual-target inhibitors based on acetylcholinesterase: Novel agents for Alzheimer's disease. Eur J Med Chem 2024; 279:116810. [PMID: 39243456 DOI: 10.1016/j.ejmech.2024.116810] [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: 07/15/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly, accounting for 60 %-70 % of cases. At present, the pathogenesis of this condition remains unclear, but the hydrolysis of acetylcholine (ACh) is thought to play a role. Acetylcholinesterase (AChE) can break down ACh transmission from the presynaptic membrane and stop neurotransmitters' excitatory effect on the postsynaptic membrane, which plays a key role in nerve conduction. Acetylcholinesterase inhibitors (AChEIs) can delay the hydrolysis of acetylcholine (ACh), which represents a key strategy for treating AD. Due to its complex etiology, AD has proven challenging to treat. Various inhibitors and antagonists targeting key enzymes and proteins implicated in the disease's pathogenesis have been explored as potential therapeutic agents. These include Glycogen Synthase Kinase 3β (GSK-3β) inhibitors, β-site APP Cleaving Enzyme (BACE-1) inhibitors, Monoamine Oxidase (MAO) inhibitors, Phosphodiesterase inhibitors (PDEs), N-methyl--aspartic Acid (NMDA) antagonists, Histamine 3 receptor antagonists (H3R), Serotonin receptor subtype 4 (5-HT4R) antagonists, Sigma1 receptor antagonists (S1R) and soluble Epoxide Hydrolase (sEH) inhibitors. The drug development strategy of multi-target-directed ligands (MTDLs) offers unique advantages in the treatment of complex diseases. On the one hand, it can synergistically enhance the therapeutic efficacy of single-target drugs. On the other hand, it can also reduce the side effects. In this review, we discuss the design strategy of dual inhibitors based on acetylcholinesterase and the structure-activity relationship of these drugs.
Collapse
Affiliation(s)
- Xingyi Zhao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qiaoguan Hu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiaoqian Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Chunting Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiao Chen
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Dong Zhao
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Yue Qiu
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Haoyu Xu
- Yangtze River Pharmaceutical (Group) CO., Ltd. NO.1 South Yangtze River Road, Taizhou City, Jiangsu Province, 225321, China
| | - Jiaqi Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Le Ren
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Na Zhang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Shuang Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Ping Gong
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Yunlei Hou
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
4
|
Zhang J, Jiang P, Wang S, Li M, Hao Z, Guan W, Pan J, Wu J, Zhang Y, Li H, Chen L, Yang B, Liu Y. Recent advances in the natural product analogues for the treatment of neurodegenerative diseases. Bioorg Chem 2024; 153:107819. [PMID: 39276492 DOI: 10.1016/j.bioorg.2024.107819] [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: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
Neurodegenerative diseases (NDs) represent a hallmark of numerous incapacitating and untreatable conditions, the incidence of which is escalating swiftly, exemplified by Alzheimer's disease and Parkinson's disease. There is an urgent necessity to create pharmaceuticals that exhibit high efficacy and minimal toxicity in order to address these debilitating diseases. The structural complexity and diversity of natural products confer upon them a broad spectrum of biological activities, thereby significantly contributing to the history of drug discovery. Nevertheless, natural products present challenges in drug discovery, including time-consuming separation processes, low content, low bioavailability, and other related issues. To address these challenges, numerous analogs of natural products have been synthesized. This methodology enables the rapid synthesis of analogs of natural products with the potential to serve as lead compounds for drug development, thereby paving the way for the discovery of novel pharmaceuticals. This paper provides a summary of 127 synthetic analogues featuring various natural product structures, including flavonoids, alkaloids, coumarins, phenylpropanoids, terpenoids, polyphenols, and amides. The compounds are categorized based on their efficacy in treating various diseases. Furthermore, this article delves into the structure-activity relationship (SAR) of certain analogues, offering a thorough point of reference for the systematic development of pharmaceuticals aimed at addressing neurodegenerative conditions.
Collapse
Affiliation(s)
- Jinling Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; Research Institute of Medicine & Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Peng Jiang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Shuping Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Mengmeng Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Zhichao Hao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Wei Guan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Juan Pan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Jiatong Wu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Yiqiang Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Hua Li
- Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Yan Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| |
Collapse
|
5
|
Jafni S, Sathya S, Arunkumar M, Kiruthiga C, Jeyakumar M, Murugesh E, Devi KP. Hesperidin Methyl Chalcone reduces extracellular Aβ (25-35) peptide aggregation and fibrillation and also protects Neuro 2a cells from Aβ (25-35) induced neuronal dysfunction. Bioorg Med Chem 2023; 96:117536. [PMID: 38016411 DOI: 10.1016/j.bmc.2023.117536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023]
Abstract
In the present study, we evaluated the neuroprotective potential of Hesperidin Methyl Chalcone (HMC) against the neurotoxicity induced by Aβ(25-35) peptide. HMC demonstrated higher free-radical scavenging activity than Hesperidin in initial cell-free studies. Investigations using the fluorescent dye thioflavin T with Aβ(25-35) peptide showed that HMC has the ability to combat extracellular amyloid aggregation by possessing anti-aggregation property against oligomers and by disaggregating mature fibrils. Also, the results of the molecular simulation studies show that HMC ameliorated oligomer formation. Further, the anti-Alzheimer's property of HMC was investigated in in vitro cell conditions by pre-treating the neuro 2a (N2a) cells with HMC before inducing Aβ(25-35) toxicity. The findings demonstrate that HMC increased cell viability, reduced oxidative stress, prevented macromolecular damage, allayed mitochondrial dysfunction, and exhibited anticholinesterase activity. HMC also reduced Aβ induced neuronal cell death by modulating caspase-3 activity, Bax expression and Bcl2 overexpression, demonstrating that HMC pre-treatment reduced mitochondrial damage and intrinsic apoptosis induced by Aβ(25-35).In silico evaluation against potential AD targets reveal that HMC could be a potent inhibitor of BACE-1, inhibiting the formation of toxic Aβ peptides. Overall, the findings imply that the neuroprotective efficacy of HMC has high prospects for addressing a variety of pathogenic consequences caused by amyloid beta in AD situations and alleviating cognitive impairments.
Collapse
Affiliation(s)
- Sakthivel Jafni
- Department of Biotechnology, Alagappa University, Karaikudi 630003, Tamil Nadu, India
| | - Sethuraman Sathya
- Department of Biotechnology, Alagappa University, Karaikudi 630003, Tamil Nadu, India
| | - Malaisamy Arunkumar
- Transcription Regulation Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | | | - Mahalingam Jeyakumar
- Department of Biotechnology, Alagappa University, Karaikudi 630003, Tamil Nadu, India
| | - Easwaran Murugesh
- Research Scientist, Bioinformatics Centre, GRC - Ganga Hospital, Coimbatore, India
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi 630003, Tamil Nadu, India.
| |
Collapse
|
6
|
Sharma P, Singh M, Singh V, Singh TG, Singh T, Ahmad SF. Recent Development of Novel Aminoethyl-Substituted Chalcones as Potential Drug Candidates for the Treatment of Alzheimer's Disease. Molecules 2023; 28:6579. [PMID: 37764355 PMCID: PMC10534526 DOI: 10.3390/molecules28186579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
No drug on the market, as a single entity, participates in different pathways involved in the pathology of Alzheimer's disease. The current study is aimed at the exploration of multifunctional chalcone derivatives which can act on multiple targets involved in Alzheimer's disease. A series of novel aminoethyl-substituted chalcones have been developed using in silico approaches (scaffold morphing, molecular docking, and ADME) and reported synthetic methods. The synthesized analogs were characterized and evaluated biologically using different in vitro assays against AChE, AGEs, and radical formation. Among all compounds, compound PS-10 was found to have potent AChE inhibitory activity (IC50 = 15.3 nM), even more than the standard drug (IC50 = 15.68 nM). Further, the in vivo evaluation of PS-10 against STZ-induced dementia in rats showed memory improvement (Morris Water Maze test) in rats. Also, PS-10 inhibited STZ-induced brain AChE activity and oxidative stress, further strengthening the observed in vitro effects. Further, the molecular dynamic simulation studies displayed the stability of the PS-10 and AChE complex. The novel aminoethyl-substituted chalcones might be considered potential multifunctional anti-Alzheimer's molecules.
Collapse
Affiliation(s)
- Pratibha Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India (T.G.S.)
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India (T.G.S.)
| | - Varinder Singh
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India (T.G.S.)
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A & M Health Science Center, College Station, TX 77807, USA;
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| |
Collapse
|
7
|
Chen J, Zheng Z, Li M, Cao C, Zhou X, Wang B, Gan X, Huang Z, Liu Y, Huang W, Liang F, Chen K, Zhao Y, Wang X, Wu J, Lin L. Design, synthesis and evaluation of monoketene compounds as novel potential Parkinson's disease agents by suppressing ER stress via AKT. Bioorg Chem 2023; 136:106543. [PMID: 37119784 DOI: 10.1016/j.bioorg.2023.106543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/01/2023]
Abstract
Curcumin is identified that it has the potential to treat Parkinson's disease (PD), but its instability limits its further application in clinic. The mono-carbonyl analogs of curcumin (MACs) with diketene structure can effectively improve its stability, but it is highly toxic. In the present study, a less cytotoxic and more stable monoketene MACs skeleton S2 was obtained, and a series of monoketene MACs were synthesized by combining 4-hydroxy-3‑methoxy groups of curcumin. In the 6-OHDA-induced PD's model in-vitro, some compounds exhibited significant neurotherapeutic effect. The quantitative structure-activity relationship (QSAR) model established by the random forest algorithm (RF) for the cell viability rate of above compounds showed that the statistical results are good (R2 = 0.883507), with strong reliability. Among all compounds, the most active compound A4 played an important role in neuroprotection in the PD models both in vitro and in vivo by activating AKT pathway, and then inhibiting the apoptosis of cells caused by endoplasmic reticulum (ER) stress. In the PD model in-vivo, compound A4 significantly improved survival of dopaminergic neurons and the contents of neurotransmitters. It also enhanced the retention of nigrostriatal function which was better than the effect in the mice treated by Madopar, a classical clinical drug for PD. In summary, we screened out the compound A4 with high stability, less cytotoxic monoketene compounds. And these founding provide evidence that the compound A4 can protect dopaminergic neurons via activating AKT and subsequently suppressing ER stress in PD.
Collapse
Affiliation(s)
- Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhiwei Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mingqi Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chengkun Cao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xuli Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Bozhen Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xin Gan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhicheng Huang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yugang Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 315020, China
| | - Wenting Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 315020, China
| | - Fei Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Keyang Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yeli Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianzhang Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China; Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China.
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
8
|
Sharma P, Singh M. An ongoing journey of chalcone analogues as single and multi-target ligands in the field of Alzheimer's disease: A review with structural aspects. Life Sci 2023; 320:121568. [PMID: 36925061 DOI: 10.1016/j.lfs.2023.121568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Alzheimer's disease (AD) is a chronic and irreversible neurodegenerative disorder with progressive dementia and cognitive impairment. AD poses severe health challenge in elderly people and become one of the leading causes of death worldwide. It possesses complex pathophysiology with several hypotheses (cholinergic hypothesis, amyloid hypothesis, tau hypothesis, oxidative stress, mitochondrial dysfunction etc.). Several attempts have been made for the management of multifactorial AD. Acetylcholinesterase is the only target has been widely explored in the management of AD to the date. The current review set forth the chalcone based natural, semi-synthetic and synthetic compounds in the search of potential anti-Alzheimer's agents. The main highlights of current review emphasizes on chalcone target different enzymes and pathways like Acetylcholinesterase, β-secretase (BACE1), tau proteins, MAO, free radicals, Advanced glycation end Products (AGEs) etc. and their structure activity relationships contributing in the inhibition of above mentioned various targets of AD.
Collapse
Affiliation(s)
- Pratibha Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
9
|
Development of the "hidden" multi-target-directed ligands by AChE/BuChE for the treatment of Alzheimer's disease. Eur J Med Chem 2023; 251:115253. [PMID: 36921526 DOI: 10.1016/j.ejmech.2023.115253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/09/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023]
Abstract
Accumulation of evidences suggested that excessive amounts of AChE and BuChE in the brain of AD patients at the different stage of AD, which could hydrolyze ACh and accelerated Aβ aggregation. To develop new "hidden" multifunctional agents through AChE/BuChE would be a promising strategy to treat AD. To this end, firstly, a series of chalcone derivatives with chelating property was designed and synthesized. The in vitro results showed that compound 3f indicated significant selective MAO-B inhibitory activity (IC50 = 0.67 μM) and remarkable anti-inflammatory property. It also significantly inhibited self-induced Aβ1-42 aggregation and showed remarkable neuroprotective effects on Aβ25-35-induced PC12 cell injury. Furthermore, compound 3f was a selective metal chelator and could inhibit Cu2+-induced Aβ1-42 aggregation. Based on this, the carbamate fragment was introduced to compound 3f to obtain carbamate derivatives. The biological activity results exhibited that compound 4b showed good BBB permeability, good AChE inhibitory potency (IC50 = 5.3 μM), moderate BuChE inhibitory potency (IC50 = 12.4 μM), significant MAO-B inhibitory potency, anti-inflammation potency on LPS-induced BV-2 cells and neuroprotective effects on Aβ25-35-induced PC12 cell injury. Compared with 3f, compound 4b did not show obvious chelation property. Significantly, compound 4b could be activated by AChE/BuChE following inhibition of AChE/BuChE to liberate an active multifunctional chelator 3f, which was consistent with our original intention. More importantly, compounds 3f and 4b presented favorable ADME properties and good stability in artificial gastrointestinal fluid, blood plasma and rat liver microsomes. The in vivo results suggested that compound 4b (0.0195 μg/mL) could significantly improve dyskinesia and reaction capacity of the AlCl3-induced zebrafish AD model by increasing the level of ACh. Together our data suggest that compound 4b was a promising "hidden" multifunctional agent by AChE/BuChE, and this strategy deserved further development for the treatment of AD.
Collapse
|
10
|
Zhong G, Guo J, Pang C, Su D, Tang C, Jing L, Zhang F, He P, Yan Y, Chen Z, Liu J, Jiang N. Novel AP2238-clorgiline hybrids as multi-target agents for the treatment of Alzheimer's disease: Design, synthesis, and biological evaluation. Bioorg Chem 2023; 130:106224. [DOI: 10.1016/j.bioorg.2022.106224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/02/2022]
|
11
|
Design, Synthesis, anticancer evaluation and in silico studies of 2,4,6-trimethoxychalcone derivatives. Saudi Pharm J 2023; 31:65-84. [PMID: 36685294 PMCID: PMC9845116 DOI: 10.1016/j.jsps.2022.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
Chalcone, a common chemical scaffold of many naturally occurring compounds, has been widely used as an effective template for drug discovery due to its broad biological activities. In this study, a series of chalcone derivatives were designed and synthesized based on the hybridization of 1-(2,4,6-trimethoxyphenyl)butan-1-one with chalcone. Interestingly, most of the target compounds exhibited inhibitory effect of tumor cells in vitro. Especially, (E)-3-(5-bromopyridin-2-yl)-1-(2,4,6-trimethoxyphenyl)prop-2-en-1-one (B3) revealed over 10-fold potency than 5-fluorocrail against the Hela and MCF-7 cells with IC50 values of 3.204 and 3.849 μM respectively. Moreover, B3 displayed low toxicity on normal cells. Further experiments indicated that B3 effectively inhibited the proliferation and migration of tumor cells, and promoted their apoptosis. The calculation and prediction of ADME showed that the target compounds may have good pharmacokinetic properties and oral bioavailability. Reverse molecular docking suggested that the possible target of B3 is CDK1. Taken together, these results suggested that B3 appears to be a promising candidate that merits further attention in the development of anticancer drugs.
Collapse
|
12
|
Current Pharmacotherapy and Multi-Target Approaches for Alzheimer's Disease. Pharmaceuticals (Basel) 2022; 15:ph15121560. [PMID: 36559010 PMCID: PMC9781592 DOI: 10.3390/ph15121560] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by decreased synaptic transmission and cerebral atrophy with appearance of amyloid plaques and neurofibrillary tangles. Cognitive, functional, and behavioral alterations are commonly associated with the disease. Different pathophysiological pathways of AD have been proposed, some of which interact and influence one another. Current treatment for AD mainly involves the use of therapeutic agents to alleviate the symptoms in AD patients. The conventional single-target treatment approaches do not often cause the desired effect in the disease due to its multifactorial origin. Thus, multi-target strategies have since been undertaken, which aim to simultaneously target multiple targets involved in the development of AD. In this review, we provide an overview of the pathogenesis of AD and the current drug therapies for the disease. Additionally, rationales of the multi-target approaches and examples of multi-target drugs with pharmacological actions against AD are also discussed.
Collapse
|
13
|
Zhu G, Bai P, Wang K, Mi J, Yang J, Hu J, Ban Y, Xu R, Chen R, Wang C, Tang L, Sang Z. Design, synthesis, and evaluation of novel O-alkyl ferulamide derivatives as multifunctional ligands for treating Alzheimer's disease. J Enzyme Inhib Med Chem 2022; 37:1375-1388. [PMID: 35549612 PMCID: PMC9116242 DOI: 10.1080/14756366.2022.2073442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/05/2022] [Accepted: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
Herein, a series of novel O-alkyl ferulamide derivatives were designed and synthesised through the multi-target-directed ligands (MTDLs) strategy. The biological activities in vitro showed that compounds 5a, 5d, 5e, 5f, and 5h indicated significantly selective MAO-B inhibitory potency (IC50 = 0.32, 0.56, 0.54, 0.73, and 0.86 μM, respectively) and moderate antioxidant activity. Moreover, compounds 5a, 5d, 5e, 5f, and 5h showed potent anti-inflammatory properties, remarkable effects on self-induced Aβ1-42 aggregation, and potent neuroprotective effect on Aβ1-42-induced PC12 cell injury. Furthermore, compounds 5a, 5d, 5e, 5f, and 5h presented good blood-brain barrier permeation in vitro and drug-like properties. More interesting, the PET/CT images with [11C]5f demonstrated that [11C]5f could penetrate the BBB with a high brain uptake and exhibited good brain clearance kinetic property. Therefore, compound 5f would be a promising multi-functional agent for the treatment of AD.
Collapse
Affiliation(s)
- Gaofeng Zhu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Basic Medical Sciences, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Ping Bai
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Keren Wang
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, China
| | - Jing Mi
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, China
| | - Jing Yang
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, China
| | - Jiaqi Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Basic Medical Sciences, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Yujuan Ban
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Basic Medical Sciences, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Ran Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Basic Medical Sciences, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Rui Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Basic Medical Sciences, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Changning Wang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Lei Tang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Basic Medical Sciences, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Zhipei Sang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Basic Medical Sciences, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, China
| |
Collapse
|
14
|
Zhao Z, Yu X, Zhu L, Tan S, Fu W, Wang L, An Y. Synthesis of
α
,
β
‐Unsaturated Ketones with Secondary Alcohols and Aldehydes Catalyzed by Fe(acac)
3. ChemistrySelect 2022. [DOI: 10.1002/slct.202202093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Zhengjia Zhao
- School of Chemistry and Chemical Engineering Liaoning Normal University Dalian 116029 China E-mail: anyue
| | - Xiangzhu Yu
- School of Chemistry and Chemical Engineering Liaoning Normal University Dalian 116029 China E-mail: anyue
| | - Lina Zhu
- School of Chemistry and Chemical Engineering Liaoning Normal University Dalian 116029 China E-mail: anyue
| | - Shangzhi Tan
- School of Chemistry and Chemical Engineering Liaoning Normal University Dalian 116029 China E-mail: anyue
| | - Weiru Fu
- School of Chemistry and Chemical Engineering Liaoning Normal University Dalian 116029 China E-mail: anyue
| | - Lianyue Wang
- School of Chemistry and Chemical Engineering Liaoning Normal University Dalian 116029 China E-mail: anyue
| | - Yue An
- School of Chemistry and Chemical Engineering Liaoning Normal University Dalian 116029 China E-mail: anyue
| |
Collapse
|
15
|
Umar T, Meena RP, Mustehasan, Kumar P, Khan AA. Recent Updates in Development of Small Molecules as Potential Clinical Candidates for Alzheimer's Disease: A Review. Chem Biol Drug Des 2022; 100:674-681. [PMID: 35996229 DOI: 10.1111/cbdd.14133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 12/08/2022]
Abstract
Alzheimer's disease (AD) is one of the prominent causes for disability and lowered quality of life worldwide in elderly population. It has fostered immense burden to AD patients, families and society. Burgeoning progress in the field of pathogenesis over last two decades has persuaded the investigation of novel pharmacological therapeutics that focuses towards the pathophysiological events of AD. Miscellaneous clinical trials, development and testing of interventions aimed at various targets, such as anti-tau and anti-amyloid interventions, neurotransmitter modification, neuroprotection and anti-neuroinflammation interventions, cognitive enhancement, and interventions to palliate behavioral symptoms have been carried out. Despite massive efforts to find disease modifying therapies there lingers a vital need for continuing the advancement in progress of the AD research. This review features the new developments of small molecule compounds that will be beneficial in evolution of new AD therapies. In particular, this review briefly describes summary of mechanistic causes chiefly associated with AD and focuses on medicinal approach via small molecule inhibitors that can manage cognitive impairment and dysfunction and may combat Alzheimer's development.
Collapse
Affiliation(s)
- Tarana Umar
- Central Council for Research in Unani Medicine, 61-65, Institutional Area. Opp. D Block. Janakpuri, New Delhi, India
| | - R P Meena
- Central Council for Research in Unani Medicine, 61-65, Institutional Area. Opp. D Block. Janakpuri, New Delhi, India
| | - Mustehasan
- Central Council for Research in Unani Medicine, 61-65, Institutional Area. Opp. D Block. Janakpuri, New Delhi, India
| | - Pawan Kumar
- Central Council for Research in Unani Medicine, 61-65, Institutional Area. Opp. D Block. Janakpuri, New Delhi, India
| | - Asim Ali Khan
- Central Council for Research in Unani Medicine, 61-65, Institutional Area. Opp. D Block. Janakpuri, New Delhi, India
| |
Collapse
|
16
|
Alzheimer's disease: Updated multi-targets therapeutics are in clinical and in progress. Eur J Med Chem 2022; 238:114464. [DOI: 10.1016/j.ejmech.2022.114464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
|
17
|
Malik YA, Awad TA, Abdalla M, Yagi S, Alhazmi HA, Ahsan W, Albratty M, Najmi A, Muhammad S, Khalid A. Chalcone Scaffolds Exhibiting Acetylcholinesterase Enzyme Inhibition: Mechanistic and Computational Investigations. Molecules 2022; 27:3181. [PMID: 35630658 PMCID: PMC9145706 DOI: 10.3390/molecules27103181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/23/2022] Open
Abstract
This study was aimed to perform the mechanistic investigations of chalcone scaffold as inhibitors of acetylcholinesterase (AChE) enzyme using molecular docking and molecular dynamics simulation tools. Basic chalcones (C1-C5) were synthesized and their in vitro AChE inhibition was tested. Binding interactions were studied using AutoDock and Surflex-Dock programs, whereas the molecular dynamics simulation studies were performed to check the stability of the ligand-protein complex. Good AChE inhibition (IC50 = 22 ± 2.8 to 37.6 ± 0.75 μM) in correlation with the in silico results (binding energies = -8.55 to -8.14 Kcal/mol) were obtained. The mechanistic studies showed that all of the functionalities present in the chalcone scaffold were involved in binding with the amino acid residues at the binding site through hydrogen bonding, π-π, π-cation, π-sigma, and hydrophobic interactions. Molecular dynamics simulation studies showed the formation of stable complex between the AChE enzyme and C4 ligand.
Collapse
Affiliation(s)
- Yossra A. Malik
- Department of Biochemistry, Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Center for Research, Khartoum 11111, Sudan; (Y.A.M.); (T.A.A.)
- Unit of Molecular Genetics, Central Laboratory, Khartoum 11111, Sudan
| | - Talal Ahmed Awad
- Department of Biochemistry, Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Center for Research, Khartoum 11111, Sudan; (Y.A.M.); (T.A.A.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ibn Sina University, Khartoum 10995, Sudan
| | - Mohnad Abdalla
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan 250012, China;
| | - Sakina Yagi
- Department of Botany, Faculty of Science, University of Khartoum, Khartoum 11115, Sudan;
| | - Hassan A. Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia;
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (W.A.); (M.A.); (A.N.)
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (W.A.); (M.A.); (A.N.)
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (W.A.); (M.A.); (A.N.)
| | - Asim Najmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (W.A.); (M.A.); (A.N.)
| | - Shabbir Muhammad
- Department of Chemistry, College of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Asaad Khalid
- Department of Biochemistry, Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Center for Research, Khartoum 11111, Sudan; (Y.A.M.); (T.A.A.)
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia;
| |
Collapse
|
18
|
Deng C, Mi J, Zhou Y, Li X, Liu Z, Sang Z, Li H. Design, synthesis, and biological evaluation of novel 2-acetylphenol-rivastigmine hybrids as potential multifunctional agents for the treatment of Alzheimer’s disease. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Pradhan LK, Sahoo PK, Chauhan S, Das SK. Recent Advances Towards Diagnosis and Therapeutic Fingerprinting for Alzheimer's Disease. J Mol Neurosci 2022; 72:1143-1165. [PMID: 35553375 DOI: 10.1007/s12031-022-02009-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/02/2022] [Indexed: 12/12/2022]
Abstract
Since the report of "a peculiar severe disease process of the cerebral cortex" by Alois Alzheimer in 1906, it was considered to be a rare condition characterized by loss of cognition, memory impairment, and pathological markers such as senile plaques or neurofibrillary tangles (NFTs). Later on, the report was published in the textbook "Psychiatrie" and the disease was named as Alzheimer's disease (AD) and was known to be the consequences of aging; however, owing to its complex etiology, there is no cure for the progressive neurodegenerative disorder. Our current understanding of the mechanisms involved in the pathogenesis of AD is still at the mechanistic level. The treatment strategies applied currently only alleviate the symptoms and co-morbidities. For instance, the available treatments such as the usage of acetylcholinesterase inhibitors and N-methyl D-aspartate antagonists have minimal impact on the disease progression and target the later aspects of the disease. The recent advancements in the last two decades have made us more clearly understand the pathophysiology of the disease which has led to the development of novel therapeutic strategies. This review gives a brief idea about the various facets of AD pathophysiology and its management through modern investigational therapies to give a new direction for development of targeted therapeutic measures.
Collapse
Affiliation(s)
- Lilesh Kumar Pradhan
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India
| | - Pradyumna Kumar Sahoo
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India
| | - Santosh Chauhan
- Autophagy Laboratory, Infectious Disease Biology Division, Institute of Life Sciences, Bhubaneswar-751023, India.
| | - Saroj Kumar Das
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India.
| |
Collapse
|
20
|
Frank A, Hamidi N, Xue F. Regioselective alkylation of 2,4-dihydroxybenzyaldehydes and 2,4-dihydroxyacetophenones. Tetrahedron Lett 2022; 95:153755. [PMID: 35495552 PMCID: PMC9053733 DOI: 10.1016/j.tetlet.2022.153755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We report a cesium bicarbonate-mediated alkylation of 2,4-dihydroxybenzyaldehyde and 2,4-dihydroxyacetophenone to generate 4-alkylated products in acetonitrile at 80 °C with excellent regioselectivity, up to 95% isolated yields, and broad substrate scope.
Collapse
Affiliation(s)
- Aziza Frank
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Negar Hamidi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| |
Collapse
|
21
|
George G, Koyiparambath VP, Sukumaran S, Nair AS, Pappachan LK, Al-Sehemi AG, Kim H, Mathew B. Structural Modifications on Chalcone Framework for Developing New Class of Cholinesterase Inhibitors. Int J Mol Sci 2022; 23:ijms23063121. [PMID: 35328542 PMCID: PMC8953944 DOI: 10.3390/ijms23063121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/18/2022] Open
Abstract
Due to the multifaceted pharmacological activities of chalcones, these scaffolds have been considered one of the most privileged frameworks in the drug discovery process. Structurally, chalcones are α, β-unsaturated carbonyl functionalities with two aryl or heteroaryl units. Amongst the numerous pharmacological activities explored for chalcone derivatives, the development of novel chalcone analogs for the treatment of Alzheimer's disease (AD) is among the research topics of most interest. Chalcones possess numerous advantages, such as smaller molecular size, opportunities for further structural modification thereby altering the physicochemical properties, cost-effectiveness, and convenient synthetic methodology. The present review highlights the recent evidence of chalcones as a privileged structure in AD drug development processes. Different classes of chalcone-derived analogs are summarized for the easy understanding of the previously reported analogs as well as the importance of certain functionalities in exhibiting cholinesterase inhibition. In this way, this review will shed light on the medicinal chemistry fraternity for the design and development of novel promising chalcone candidates for the treatment of AD.
Collapse
Affiliation(s)
- Ginson George
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Vishal Payyalot Koyiparambath
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Sunitha Sukumaran
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Aathira Sujathan Nair
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Leena K. Pappachan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Abdullah G. Al-Sehemi
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Hoon Kim
- Department of Pharmacy, Sunchon National University, Suncheon 57922, Korea
- Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Korea
- Correspondence: (H.K.); (B.M.)
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
- Correspondence: (H.K.); (B.M.)
| |
Collapse
|
22
|
Verma A, Kumar Waiker D, Bhardwaj B, Saraf P, Shrivastava SK. The molecular mechanism, targets, and novel molecules in the treatment of Alzheimer's disease. Bioorg Chem 2021; 119:105562. [PMID: 34952243 DOI: 10.1016/j.bioorg.2021.105562] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/27/2021] [Accepted: 12/12/2021] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurological illness that causes dementia mainly in the elderly. The challenging obstacles related to AD has freaked global healthcare system to encourage scientists in developing novel therapeutic startegies to overcome with the fatal disease. The current treatment therapy of AD provides only symptomatic relief and to some extent disease-modifying effects. The current approach for AD treatment involves designing of cholinergic inhibitors, Aβ disaggregation inducing agents, tau inhibitors and several antioxidants. Hence, extensive research on AD therapy urgently requires a deep understanding of its pathophysiology and exploration of various chemical scaffolds to design and develop a potential drug candidate for the treatment. Various issues linked between disease and therapy need to be considered such as BBB penetration capability, clinical failure and multifaceted pathophisiology requires a proper attention to develop a lead candidate. This review article covers all probable mechanisms including one of the recent areas for investigation i.e., lipid dyshomeostasis, pathogenic involvement of P. gingivalis and neurovascular dysfunction, recently reported molecules and drugs under clinical investigations and approved by FDA for AD treatment. Our summarized information on AD will attract the researchers to understand and explore current status and structural modifications of the recently reported heterocyclic derivatives in drug development for AD therapy.
Collapse
Affiliation(s)
- Akash Verma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Digambar Kumar Waiker
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Bhagwati Bhardwaj
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Poorvi Saraf
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushant K Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India.
| |
Collapse
|
23
|
Sang Z, Song Q, Cao Z, Deng Y, Zhang L. Design, synthesis, and evaluation of chalcone-Vitamin E-donepezil hybrids as multi-target-directed ligands for the treatment of Alzheimer's disease. J Enzyme Inhib Med Chem 2021; 37:69-85. [PMID: 34894968 PMCID: PMC8667902 DOI: 10.1080/14756366.2021.1993845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A novel series of chalcone-Vitamin E-donepezil hybrids was designed and developed based on multitarget-directed ligands (MTDLs) strategy for treating Alzheimer’s disease (AD). The biological results revealed that compound 17f showed good AChE inhibitory potency (ratAChE IC50 = 0.41 µM; eeAChE IC50 = 1.88 µM). Both the kinetic analysis and docking study revealed that 17f was a mixed type AChE inhibitor. 17f was also a good antioxidant (ORAC = 3.3 eq), selective metal chelator and huMAO-B inhibitor (IC50 = 8.8 µM). Moreover, it showed remarkable inhibition of self- and Cu2+-induced Aβ1–42 aggregation with a 78.0 and 93.5% percentage rate at 25 µM, respectively, and disassembled self-induced and Cu2+-induced aggregation of the accumulated Aβ1–42 fibrils with 72.3 and 84.5% disaggregation rate, respectively. More importantly, 17f exhibited a good neuroprotective effect on H2O2-induced PC12 cell injury and presented good blood-brain barrier permeability in vitro. Thus, 17f was a promising multi-target-directed ligand for treating AD.
Collapse
Affiliation(s)
- Zhipei Sang
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, China.,Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qing Song
- Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhongcheng Cao
- Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yong Deng
- Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
24
|
Ceyhun İ, Karaca Ş, Osmaniye D, Sağlık BN, Levent S, Özkay Y, Kaplancıklı ZA. Design and synthesis of novel chalcone derivatives and evaluation of their inhibitory activities against acetylcholinesterase. Arch Pharm (Weinheim) 2021; 355:e2100372. [PMID: 34893996 DOI: 10.1002/ardp.202100372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022]
Abstract
According to the cholinergic hypothesis, an increase in the acetylcholine level in Alzheimer's disease patients relatively slows down the symptoms of the disease. The most commonly used drug, donepezil, is a cholinesterase inhibitor. In this study, 12 new chalcones (2a-l) were designed and synthesized. In biological activity studies, the acetylcholinesterase (AChE) and butyrylcholinesterase inhibitory potentials of all compounds were evaluated using the in vitro Ellman method. The biological evaluation showed that compounds 2d, 2f, 2j, and 2l displayed significant activity against AChE. The compounds 2d, 2f, 2j, and 2l displayed IC50 values of 0.042, 0.024, 0.053, and 0.033 µM against AChE, respectively. The reference drug donepezil (IC50 = 0.021 µM) also displayed significant inhibition of AChE. The inhibitory activities of these compounds for β-amyloid plaque aggregation were investigated. The enzyme kinetic study was performed to observe the effect of the most active compound 2f on the substrate-enzyme relationship, and a mixed-type inhibition of AchE was determined. Further, docking simulation also revealed that these compounds (2d, 2f, 2j, and 2l) interacted with the enzyme active site in a similar manner to donepezil. The most active derivative, compound 2f, interacted with the amino acids Trp286, Phe295, Tyr341, Trp86, and Glu202.
Collapse
Affiliation(s)
- İlçim Ceyhun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Şevval Karaca
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Derya Osmaniye
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Begüm N Sağlık
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Serkan Levent
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Yusuf Özkay
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Zafer A Kaplancıklı
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
25
|
Rehuman N, Oh JM, Nath LR, Khames A, Abdelgawad MA, Gambacorta N, Nicolotti O, Jat R, Kim H, Mathew B. Halogenated Coumarin-Chalcones as Multifunctional Monoamine Oxidase-B and Butyrylcholinesterase Inhibitors. ACS OMEGA 2021; 6:28182-28193. [PMID: 34723016 PMCID: PMC8552465 DOI: 10.1021/acsomega.1c04252] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/24/2021] [Indexed: 02/08/2023]
Abstract
A series of halogenated coumarin-chalcones were synthesized, characterized, and their inhibitory activities against monoamine oxidases (MAOs), acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and β-site amyloid precursor protein cleaving enzyme 1 (BACE-1) were evaluated. Compound CC2 most potently inhibited MAO-B with an IC50 value of 0.51 μM, followed by CC1 (IC50 = 0.69 μM), with a selectivity index (SI) of >78.4 and >58.0, respectively, over MAO-A. However, none of the compounds effectively inhibited MAO-A, AChE, and BChE, except for CC2 and CC3 inhibiting BChE with IC50 values of 7.00 (SI > 5.73 over AChE) and 11.8 μM, respectively. CC1 and CC2 were found to be reversible and competitive inhibitors of MAO-B, with K i values of 0.50 ± 0.06 and 0.53 ± 0.04 μM, respectively, and CC2 was also a reversible and competitive inhibitor of BChE, with a K i value of 2.84 ± 0.09 μM. The parallel artificial membrane permeability assay (PAMPA) method showed that lead candidates can cross the blood-brain barrier (BBB). The in vitro toxicity analysis on the Vero cell line (Normal African green monkey kidney epithelial cells) by MTT confirmed that both CC1 and CC2 were nontoxic up to 100 μg/mL, which is almost equivalent to 100 times of their effective concentration used in biological studies. In addition, CC1 and CC2 attenuated H2O2-induced cellular damage via their reactive oxygen species (ROS) scavenging effect. These results suggest that CC1 and CC2 are selective and competitive inhibitors of MAO-B, and that CC2 is a selective and competitive inhibitor of BChE. Molecular docking studies of lead compounds provided the possible type of interactions in the targeted enzymes. Based on the findings, both compounds, CC1 and CC2, can be considered plausible drug candidates against neurodegenerative disorders.
Collapse
Affiliation(s)
- Nisha
Abdul Rehuman
- Department
of Pharmaceutical Chemistry, Dr. Joseph
Mar Thoma Institute of Pharmaceutical Sciences & Research, Alappuzha, Kerala 690503, India
| | - Jong Min Oh
- Department
of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Lekshmi R. Nath
- Department
of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682, India
| | - Ahmed Khames
- Department
of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Mohamed A. Abdelgawad
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf 72341, Saudi Arabia
| | - Nicola Gambacorta
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Via E. Orabona, 4, I-70125 Bari, Italy
| | - Orazio Nicolotti
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Via E. Orabona, 4, I-70125 Bari, Italy
| | - Rakesh
Kumar Jat
- Department
of Pharmaceutical Chemistry, JJTU University, Jhunjhunu 333001, India
| | - Hoon Kim
- Department
of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Bijo Mathew
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| |
Collapse
|
26
|
Constantinescu T, Lungu CN. Anticancer Activity of Natural and Synthetic Chalcones. Int J Mol Sci 2021; 22:11306. [PMID: 34768736 PMCID: PMC8582663 DOI: 10.3390/ijms222111306] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 11/16/2022] Open
Abstract
Cancer is a condition caused by many mechanisms (genetic, immune, oxidation, and inflammatory). Anticancer therapy aims to destroy or stop the growth of cancer cells. Resistance to treatment is theleading cause of the inefficiency of current standard therapies. Targeted therapies are the most effective due to the low number of side effects and low resistance. Among the small molecule natural compounds, flavonoids are of particular interest for theidentification of new anticancer agents. Chalcones are precursors to all flavonoids and have many biological activities. The anticancer activity of chalcones is due to the ability of these compounds to act on many targets. Natural chalcones, such as licochalcones, xanthohumol (XN), panduretin (PA), and loncocarpine, have been extensively studied and modulated. Modification of the basic structure of chalcones in order to obtain compounds with superior cytotoxic properties has been performed by modulating the aromatic residues, replacing aromatic residues with heterocycles, and obtaining hybrid molecules. A huge number of chalcone derivatives with residues such as diaryl ether, sulfonamide, and amine have been obtained, their presence being favorable for anticancer activity. Modification of the amino group in the structure of aminochalconesis always favorable for antitumor activity. This is why hybrid molecules of chalcones with different nitrogen hetercycles in the molecule have been obtained. From these, azoles (imidazole, oxazoles, tetrazoles, thiazoles, 1,2,3-triazoles, and 1,2,4-triazoles) are of particular importance for the identification of new anticancer agents.
Collapse
Affiliation(s)
- Teodora Constantinescu
- Department of Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University, 400012 Cluj-Napoca, Romania
| | - Claudiu N. Lungu
- Department of Surgery, Country Emergency Hospital Braila, 810249 Braila, Romania
| |
Collapse
|
27
|
Development of novel 2-acetylphenol-O-alkylhydroxyethylamine derivatives as multifunctional agents for Alzheimer’s disease treatment. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02786-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
28
|
Koyiparambath VP, Oh JM, Khames A, Abdelgawad MA, Nair AS, Nath LR, Gambacorta N, Ciriaco F, Nicolotti O, Kim H, Mathew B. Trimethoxylated Halogenated Chalcones as Dual Inhibitors of MAO-B and BACE-1 for the Treatment of Neurodegenerative Disorders. Pharmaceutics 2021; 13:850. [PMID: 34201128 PMCID: PMC8226672 DOI: 10.3390/pharmaceutics13060850] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 02/05/2023] Open
Abstract
Six halogenated trimethoxy chalcone derivatives (CH1-CH6) were synthesized and spectrally characterized. The compounds were further evaluated for their inhibitory potential against monoamine oxidases (MAOs) and β-secretase (BACE-1). Six compounds inhibited MAO-B more effectively than MAO-A, and the 2',3',4'-methoxy moiety in CH4-CH6 was more effective for MAO-B inhibition than the 2',4',6'-methoxy moiety in CH1-CH3. Compound CH5 most potently inhibited MAO-B, with an IC50 value of 0.46 µM, followed by CH4 (IC50 = 0.84 µM). In 2',3',4'-methoxy derivatives (CH4-CH6), the order of inhibition was -Br in CH5 > -Cl in CH4 > -F in CH6 at the para-position in ring B of chalcone. CH4 and CH5 were selective for MAO-B, with selectivity index (SI) values of 15.1 and 31.3, respectively, over MAO-A. CH4 and CH5 moderately inhibited BACE-1 with IC50 values of 13.6 and 19.8 µM, respectively. When CH4 and CH5 were assessed for their cell viability studies on the normal African Green Monkey kidney cell line (VERO) using MTT assays, it was noted that both compounds were found to be safe, and only a slightly toxic effect was observed in concentrations above 200 µg/mL. CH4 and CH5 decreased reactive oxygen species (ROS) levels of VERO cells treated with H2O2, indicating both compounds retained protective effects on the cells by antioxidant activities. All compounds showed high blood brain barrier permeabilities analyzed by a parallel artificial membrane permeability assay (PAMPA). Molecular docking and ADME prediction of the lead compounds provided more insights into the rationale behind the binding and the CNS drug likeness. From non-test mutagenicity and cardiotoxicity studies, CH4 and CH5 were non-mutagenic and non-/weak-cardiotoxic. These results suggest that CH4 and CH5 could be considered candidates for the cure of neurological dysfunctions.
Collapse
Affiliation(s)
- Vishal Payyalot Koyiparambath
- Department of Pharmaceutical Chemistry, AIMS Health Sciences Campus, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi 682041, India; (V.P.K.); (A.S.N.)
| | - Jong Min Oh
- Department of Pharmacy, Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Korea;
| | - Ahmed Khames
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box-11099, Taif 21944, Saudi Arabia;
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62514, Egypt
| | - Aathira Sujathan Nair
- Department of Pharmaceutical Chemistry, AIMS Health Sciences Campus, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi 682041, India; (V.P.K.); (A.S.N.)
| | - Lekshmi R. Nath
- Department of Pharmacogonosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India;
| | - Nicola Gambacorta
- Dipartimento di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Via E. Orabona, 4, I-70125 Bari, Italy; (N.G.); (O.N.)
| | - Fulvio Ciriaco
- Dipartimento di Chimica, Università degli Studi di Bari “Aldo Moro”, Via E. Orabona, 4, I-70125 Bari, Italy;
| | - Orazio Nicolotti
- Dipartimento di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Via E. Orabona, 4, I-70125 Bari, Italy; (N.G.); (O.N.)
| | - Hoon Kim
- Department of Pharmacy, Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Korea;
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, AIMS Health Sciences Campus, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi 682041, India; (V.P.K.); (A.S.N.)
| |
Collapse
|
29
|
Zhang C, Lv Y, Bai R, Xie Y. Structural exploration of multifunctional monoamine oxidase B inhibitors as potential drug candidates against Alzheimer's disease. Bioorg Chem 2021; 114:105070. [PMID: 34126574 DOI: 10.1016/j.bioorg.2021.105070] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
AD is one of the most typical neurodegenerative disorders that suffer many seniors worldwide. Recently, MAO inhibitors have received increasing attention not only for their roles involved in monoamine neurotransmitters metabolism and oxidative stress but also for their additional neuroprotective and neurorescue effects against AD. The curiosity in MAO inhibitors is reviving, and novel MAO-B inhibitors recently developed with ancillary activities (e.g., Aβ aggregation and AChE inhibition, anti-ROS and chelating activities) have been proposed as multitarget drugs foreshadowing a positive outlook for the treatment of AD. The current review describes the recent development of the design, synthesis, and screening of multifunctional ligands based on MAO-B inhibition for AD therapy. Structure-activity relationships and rational design strategies of the synthetic or natural product derivatives (chalcones, coumarins, chromones, and homoisoflavonoids) are discussed.
Collapse
Affiliation(s)
- Changjun Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou, PR China
| | - Yangjing Lv
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, PR China
| | - Renren Bai
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, PR China.
| | - Yuanyuan Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou, PR China; College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, PR China.
| |
Collapse
|
30
|
Kumar B, Thakur A, Dwivedi AR, Kumar R, Kumar V. Multi-Target-Directed Ligands as an Effective Strategy for the Treatment of Alzheimer's Disease. Curr Med Chem 2021; 29:1757-1803. [PMID: 33982650 DOI: 10.2174/0929867328666210512005508] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder, and multiple pathological factors are believed to be involved in the genesis and progression of the disease. A number of hypotheses, including Acetylcholinesterase, Monoamine oxidase, β-Amyloid, Tau protein, etc., have been proposed for the initiation and progression of the disease. At present, acetylcholine esterase inhibitors and memantine (NMDAR antagonist) are the only approved therapies for the symptomatic management of AD. Most of these single-target drugs have miserably failed in the treatment or halting the progression of the disease. Multi-factorial diseases like AD require complex treatment strategies that involve simultaneous modulation of a network of interacting targets. Since the last few years, Multi-Target-Directed Ligands (MTDLs) strategy, drugs that can simultaneously hit multiple targets, is being explored as an effective therapeutic approach for the treatment of AD. In the current review article, the authors have briefly described various pathogenic pathways associated with AD. The importance of Multi-Target-Directed Ligands and their design strategies in recently reported articles have been discussed in detail. Potent leads are identified through various structure-activity relationship studies, and their drug-like characteristics are described. Recently developed promising compounds have been summarized in the article. Some of these MTDLs with balanced activity profiles against different targets have the potential to be developed as drug candidates for the treatment of AD.
Collapse
Affiliation(s)
- Bhupinder Kumar
- Central University of Punjab Department of Pharmaceutical Sciences and Natural Products, India
| | - Amandeep Thakur
- Central University of Punjab Department of Pharmaceutical Sciences and Natural Products, India
| | | | - Rakesh Kumar
- Central University of Punjab, Bathinda, Punjab-151001, India
| | - Vinod Kumar
- Department of Chemistry, Central University of Punjab, Bathinda, Punjab-151001, India
| |
Collapse
|
31
|
Wang K, Shi J, Zhou Y, He Y, Mi J, Yang J, Liu S, Tang X, Liu W, Tan Z, Sang Z. Design, synthesis and evaluation of cinnamic acid hybrids as multi-target-directed agents for the treatment of Alzheimer's disease. Bioorg Chem 2021; 112:104879. [PMID: 33915461 DOI: 10.1016/j.bioorg.2021.104879] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/01/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022]
Abstract
Herein, combining 1,2,3,4-tetrahydroisoquinoline and benzylpiperidine groups into cinnamic acid derivatives, a series of novel cinnamic acid hybrids was rationally designed, synthesized and evaluated by the multi-target-directed ligands (MTDLs) strategy. Hybrid 4e was the most promising one among these hybrids with a reversible huBuChE inhibitor (IC50 = 2.5 μM) and good MAO-B inhibition activity (IC50 = 1.3 μM) and antioxidant potency (ORAC = 0.4 eq). Moreover, compound 4e significantly inhibited self-mediated Aβ1-42 aggregation (65.2% inhibition rate). Compound 4e exhibited remarkable anti-inflammatory propery and neuroprotective effect. Furthermore, compound 4e displayed favourable blood-brain barrier penetration via parallel artificial membrane permeation assay (PAMPA). The obtained results also revealed that compound 4e significantly improved dyskinesia recovery rate and response efficiency on AD model zebrafish. Further, 4e did not show obvious acute toxicity at dose up to 1500 mg/kg in vivo and improved scopolamine-induced memory impairment. Importantly, compound 4e showed good stability in both artificial gastric fluid and artificial intestinal fluid. Therefore, compound 4e presented a promising multi-targeted active molecule for treating AD.
Collapse
Affiliation(s)
- Keren Wang
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Jian Shi
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Yi Zhou
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Ying He
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Jing Mi
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Jing Yang
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Shuang Liu
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Xiangcheng Tang
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Wenmin Liu
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China
| | - Zhenghuai Tan
- Institute of Traditional Chinese Medicine Pharmacology and Toxicology, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China.
| | - Zhipei Sang
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, China.
| |
Collapse
|
32
|
Design, synthesis and evaluation of novel dimethylamino chalcone-O-alkylamines derivatives as potential multifunctional agents against Alzheimer's disease. Eur J Med Chem 2021; 216:113310. [PMID: 33667847 DOI: 10.1016/j.ejmech.2021.113310] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/05/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
A novel series of dimethylamino chalcone-O-alkylamines derivatives was designed and synthesized as multifunctional agents for the treatment of AD. All the target compounds exhibited significant abilities to inhibit and disaggregate Aβ aggregation, and acted as potential selective AChE inhibitors, biometal chelators and selective MAO-B inhibitors. Among these compounds, compound TM-6 showed the greatest inhibitory activity against self-induced Aβ aggregation (IC50 = 0.88 μM) and well disaggregation ability toward self-induced Aβ aggregation (95.1%, 25 μM), the TEM images, molecular docking study and molecular dynamics simulations provided reasonable explanation for its high efficiency, and it was also found to be a remarkable antioxidant (ORAC-FL values of 2.1eq.), the best AChE inhibitor (IC50 = 0.13 μM) and MAO-B inhibitor (IC50 = 1.0 μM), as well as a good neuroprotectant. UV-visual spectrometry and ThT fluorescence assay revealed that compound TM-6 was not only a good biometal chelator by inhibiting Cu2+-induced Aβ aggregation (95.3%, 25 μM) but also could disassemble the well-structured Aβ fibrils (88.1%, 25 μM). Further, TM-6 could cross the blood-brain barrier (BBB) in vitro. More importantly, compound TM-6 did not show any acute toxicity in mice at doses of up to 1000 mg/kg and improved scopolamine-induced memory impairment. Taken together, these data indicated that TM-6, an excellent balanced multifunctional inhibitor, was a potential lead compound for the treatment of AD.
Collapse
|
33
|
Sharma P, Singh M, Mathew B. An Update of Synthetic Approaches and Structure‐Activity Relationships of Various Classes of Human MAO‐B Inhibitors. ChemistrySelect 2021. [DOI: 10.1002/slct.202004188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Pratibha Sharma
- Chitkara College of Pharmacy Chitkara University Punjab India
| | - Manjinder Singh
- Chitkara College of Pharmacy Chitkara University Punjab India
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus Kochi 682 041 India
| |
Collapse
|
34
|
Synthesis and neuroprotective effects of novel chalcone-triazole hybrids. Bioorg Chem 2020; 105:104384. [DOI: 10.1016/j.bioorg.2020.104384] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/16/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023]
|
35
|
Shi J, Zhou Y, Wang K, Ma Q, Wei R, Li Q, Zhao Y, Qiao Z, Liu S, Leng Y, Liu W, Sang Z. Design, synthesis and biological evaluation of Schiff’s base derivatives as multifunctional agents for the treatment of Alzheimer’s disease. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02666-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
36
|
Xue K, Sun G, Zhang Y, Chen X, Zhou Y, Hou J, Long H, Zhang Z, Lei M, Wu W. A new method for the synthesis of chalcone derivatives promoted by PPh3/I2under non-alkaline conditions. SYNTHETIC COMMUN 2020. [DOI: 10.1080/00397911.2020.1847295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Kangsheng Xue
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Guoxiang Sun
- School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, Jiangsu, P. R. China
| | - Yanzhi Zhang
- College of Pharmacy, Dali University, Dali, P. R. China
| | - Xubing Chen
- College of Pharmacy, Dali University, Dali, P. R. China
| | - Yang Zhou
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Jinjun Hou
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Huali Long
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Zijia Zhang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Min Lei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Wanying Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
37
|
Chithiraikumar C, Ponmuthu KV, Harikrishnan M, Malini N, Sepperumal M, Siva A. Efficient base-free asymmetric one-pot synthesis of spiro[indoline-3,3′-pyrrolizin]-2-one derivatives catalyzed by chiral organocatalyst. RESEARCH ON CHEMICAL INTERMEDIATES 2020. [DOI: 10.1007/s11164-020-04303-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
38
|
Phenothiazine‐based chalcones as potential dual‐target inhibitors toward cholinesterases (AChE, BuChE) and monoamine oxidases (MAO‐A, MAO‐B). J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4156] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Sakata RP, Antoniolli G, Lancellotti M, Kawano DF, Guimarães Barbosa E, Almeida WP. Synthesis and biological evaluation of 2'-Aminochalcone: A multi-target approach to find drug candidates to treat Alzheimer's disease. Bioorg Chem 2020; 103:104201. [PMID: 32890999 DOI: 10.1016/j.bioorg.2020.104201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/17/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative process that compromises cognitive functions. The physiopathology of AD is multifactorial and is mainly supported by the cholinergic and amyloid hypotheses, which allows the identification the fundamental role of some markers, such as the enzymes acetylcholinesterase (AChE) and β-secretase (BACE-1), and the β-amyloid peptide (Aβ). In this work, we prepared a series of chalcones and 2'-aminochalcones, which were tested against AChE and BACE-1 enzymes and on the aggregation of Aβ. All compounds inhibited AChE activity with different potencies. We have found that the majority of chalcones having the amino group are able to inhibit BACE-1, which was not observed for chalcones without this group. The most active compound is the one derived from 2,3-dichlorobenzaldeyde, having an IC50 value of 2.71 μM. A molecular docking study supported this result, showing a good interaction of the amino group with aspartic acid residues of the catalytic diade of BACE-1. Thioflavin-T fluorescence emission is reduced in 30 - 40%, when Aβ42 is incubated in the presence of some chalcones under aggregation conditions. In vitro cytotoxicity and in silico prediction of pharmacokinetic properties were also conducted in this study.
Collapse
Affiliation(s)
- Renata P Sakata
- Institute of Chemistry, University of Campinas, Brazil; Porphirio da Paz High School, Campinas, SP, Brazil
| | | | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences, University of Campinas, 200, Candido Portinari, Campinas, SP ZC 13083-871, Brazil
| | - Daniel Fabio Kawano
- Faculty of Pharmaceutical Sciences, University of Campinas, 200, Candido Portinari, Campinas, SP ZC 13083-871, Brazil
| | | | - Wanda P Almeida
- Institute of Chemistry, University of Campinas, Brazil; Faculty of Pharmaceutical Sciences, University of Campinas, 200, Candido Portinari, Campinas, SP ZC 13083-871, Brazil.
| |
Collapse
|
40
|
Manzoor S, Hoda N. A comprehensive review of monoamine oxidase inhibitors as Anti-Alzheimer's disease agents: A review. Eur J Med Chem 2020; 206:112787. [PMID: 32942081 DOI: 10.1016/j.ejmech.2020.112787] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
Monoamine oxidases (MAO-A and MAO-B) are mammalian flavoenzyme, which catalyze the oxidative deamination of several neurotransmitters like norepinephrine, dopamine, tyramine, serotonin, and some other amines. The oxidative deamination produces several harmful side products like ammonia, peroxides, and aldehydes during the biochemical reaction. The concentration of biochemical neurotransmitter alteration in the brain by MAO is directly related with several neurological disorders like Alzheimer's disease and Parkinson's disease (PD). Activated MAO also contributes to the amyloid beta (Aβ) aggregation by two successive cleft β-secretase and γ-secretase of amyloid precursor protein (APP). Additionally, activated MAO is also involved in aggregation of neurofibrillary tangles and cognitive destruction through the cholinergic neuronal damage and disorder of the cholinergic system. MAO inhibition has general anti-Alzheimer's disease effect as a consequence of oxidative stress reduction prompted by MAO enzymes. In this review, we outlined and addressed recent understanding on MAO enzymes such as their structure, physiological function, catalytic mechanism, and possible therapeutic goals in AD. In addition, it also highlights the current development and discovery of potential MAO inhibitors (MAOIs) from various chemical scaffolds.
Collapse
Affiliation(s)
- Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
41
|
Tang YL, Zheng X, Qi Y, Pu XJ, Liu B, Zhang X, Li XS, Xiao WL, Wan CP, Mao ZW. Synthesis and anti-inflammatory evaluation of new chalcone derivatives bearing bispiperazine linker as IL-1β inhibitors. Bioorg Chem 2020; 98:103748. [DOI: 10.1016/j.bioorg.2020.103748] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/20/2022]
|
42
|
Calycosin and 8-O-methylretusin isolated from Maackia amurensis as potent and selective reversible inhibitors of human monoamine oxidase-B. Int J Biol Macromol 2020; 151:441-448. [PMID: 32087226 DOI: 10.1016/j.ijbiomac.2020.02.144] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/09/2020] [Accepted: 02/14/2020] [Indexed: 01/27/2023]
Abstract
Nineteen compounds were isolated from the stems of Maackia amurensis by activity-guided screening for new human monoamine oxidase-B (hMAO-B) inhibitors. Among the compounds isolated, flavonoids calycosin (5) and 8-O-methylretusin (6) were found to potently and selectively inhibit hMAO-B (IC50 = 0.24 and 0.23 μM, respectively) but not hMAO-A with high selectivity index (SI) values (SI = 293.8 and 81.3, respectively). In addition, 5 and 6 reversibly and competitively inhibited hMAO-B with Ki values of 0.057 and 0.054 μM, respectively. A pterocarpan (-)-medicarpin (18) was also observed to strongly inhibit hMAO-B (IC50 = 0.30 μM). Most of the compounds weakly inhibited AChE, except isolupalbigenin (13) (IC50 = 20.6 μM), which suggested 13 be considered a potential dual function inhibitor of MAO-B and AChE. Molecular docking simulation revealed that the binding affinities of 5 and 6 for hMAO-B (both -9.3 kcal/mol) were higher than those for hMAO-A (-7.4 and -7.2 kcal/mol, respectively). Compound 5 was found to interact by hydrogen bonding with hMAO-B at Cys172 residue (distance: 3.250 Å); no hydrogen bonding was predicted between 5 and hMAO-A. These findings suggest that compounds 5 and 6 be considered novel potent, selective, and reversible hMAO-B inhibitors and candidates for the treatment of neurological disorders.
Collapse
|
43
|
Wu Z, Palanimuthu D, Braidy N, Salikin NH, Egan S, Huang MLH, Richardson DR. Novel multifunctional iron chelators of the aroyl nicotinoyl hydrazone class that markedly enhance cellular NAD + /NADH ratios. Br J Pharmacol 2020; 177:1967-1987. [PMID: 31895471 DOI: 10.1111/bph.14963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/17/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is a multifactorial condition leading to cognitive decline and represents a major global health challenge in ageing populations. The lack of effective AD therapeutics led us to develop multifunctional nicotinoyl hydrazones to target several pathological characteristics of AD. EXPERIMENTAL APPROACH We synthesised 20 novel multifunctional agents based on the nicotinoyl hydrazone scaffold, which acts as a metal chelator and a lipophilic delivery vehicle, donating a NAD+ precursor to cells, to target metal dyshomeostasis, oxidative stress, β-amyloid (Aβ) aggregation, and a decrease in the NAD+ /NADH ratio. KEY RESULTS The most promising compound, 6-methoxysalicylaldehyde nicotinoyl hydrazone (SNH6), demonstrated low cytotoxicity, potent iron (Fe)-chelation efficacy, significant inhibition of copper-mediated Aβ aggregation, oxidative stress alleviation, effective donation of NAD+ to NAD-dependent metabolic processes (PARP and sirtuin activity) and enhanced cellular NAD+ /NADH ratios, as well as significantly increased median Caenorhabditis elegans lifespan (to 1.46-fold of the control); partly decreased BACE1 expression, resulting in significantly lower soluble amyloid precursor protein-β (sAPPβ) and Aβ1-40 levels; and favourable blood-brain barrier-permeation properties. Structure-activity relationships demonstrated that the ability of these nicotinoyl hydrazones to increase NAD+ was dependent on the electron-withdrawing or electron-donating substituents on the aldehyde- or ketone-derived moiety. Aldehyde-derived hydrazones containing the ONO donor set and electron-donating groups were required for NAD+ donation and low cytotoxicity. CONCLUSIONS AND IMPLICATIONS The nicotinoyl hydrazones, particularly SNH6, have the potential to act as multifunctional therapeutic agents and delivery vehicles for NAD+ precursors for AD treatment.
Collapse
Affiliation(s)
- Zhixuan Wu
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Duraippandi Palanimuthu
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia.,Schools of Medicine, Huzhou University, Huzhou Central Hospital, Huzhou, China
| | - Nor Hawani Salikin
- School of Biological, Earth and Environmental Sciences, Centre for Marine Science and Innovation, University of New South Wales, Sydney, Australia
| | - Suhelen Egan
- School of Biological, Earth and Environmental Sciences, Centre for Marine Science and Innovation, University of New South Wales, Sydney, Australia
| | - Michael L H Huang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia.,Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
44
|
Salem MA, Ragab A, Askar AA, El-Khalafawy A, Makhlouf AH. One-pot synthesis and molecular docking of some new spiropyranindol-2-one derivatives as immunomodulatory agents and in vitro antimicrobial potential with DNA gyrase inhibitor. Eur J Med Chem 2020; 188:111977. [DOI: 10.1016/j.ejmech.2019.111977] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/07/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
|
45
|
Development of novel carbazole derivatives with effective multifunctional action against Alzheimer’s diseases: Design, synthesis, in silico, in vitro and in vivo investigation. Bioorg Chem 2020; 95:103524. [DOI: 10.1016/j.bioorg.2019.103524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/15/2019] [Accepted: 12/18/2019] [Indexed: 01/04/2023]
|
46
|
Shi CC, Chen TR, Zhang QH, Wei LH, Huang C, Zhu YD, Liu HB, Bai YK, Wang FJ, Guo WZ, Zhang LR, Ge GB. Inhibition of human thrombin by the constituents of licorice: inhibition kinetics and mechanistic insights through in vitro and in silico studies. RSC Adv 2020; 10:3626-3635. [PMID: 35492646 PMCID: PMC9048847 DOI: 10.1039/c9ra09203j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/13/2020] [Indexed: 12/30/2022] Open
Abstract
Thrombin inhibition therapy is a practical strategy to reduce thrombotic and cardiovascular risks via blocking the formation of blood clots. This study aimed to identify naturally occurring thrombin inhibitors from licorice (one of the most popular edible herbs), as well as to investigate their inhibitory mechanisms. Among all tested licorice constituents, licochalcone A was found as the most efficacious agent against human thrombin (IC50 = 7.96 μM). Inhibition kinetic analyses demonstrated that licochalcone A was a mixed inhibitor against thrombin-mediated Z-Gly-Gly-Arg-AMC acetate hydrolysis, with a Ki value of 12.23 μM. Furthermore, mass spectrometry-based chemoproteomic assays and molecular docking simulations revealed that licochalcone A could bind to human thrombin at both exosite I and the catalytic site. In summary, our findings demonstrated that the chalcones isolated from licorice were a new class of direct thrombin inhibitors, also suggesting that licochalcone A was a promising lead compound for developing novel anti-thrombotic agents. Licochalcone A, a bioactive compound from licorice, displayed strong inhibition of thrombin.![]()
Collapse
|
47
|
Zhou Y, Sun W, Peng J, Yan H, Zhang L, Liu X, Zuo Z. Design, synthesis and biological evaluation of novel copper-chelating acetylcholinesterase inhibitors with pyridine and N-benzylpiperidine fragments. Bioorg Chem 2019; 93:103322. [DOI: 10.1016/j.bioorg.2019.103322] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
|