1
|
Zueva I, Belyaev G, Petrov K. Disease-modifying effect of donepezil on APP/PS1 mice at different stages of Alzheimer's disease. Mol Cell Biochem 2025:10.1007/s11010-025-05310-2. [PMID: 40399637 DOI: 10.1007/s11010-025-05310-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 05/09/2025] [Indexed: 05/23/2025]
Abstract
Despite Alzheimer's disease (AD) representing a significant global health concern, disease-modifying therapeutic options remain elusive. The use of animal models of the disease to develop drugs intended for the treatment of AD does not always predict their efficacy in clinical trials. Our research demonstrates the benefits of a drug-withdrawal approach to screening AD-modifying compounds, focussing on β-amyloid (Aβ)-related pathological changes in APP/PS1 transgenic mice at different stages of the disease. To assess the efficacy of this approach, we examined the AD-modifying effect of donepezil as a reference drug. A significant cognitive decline exhibited by APP/PS1 transgenic mice from 8.4 months of age was accompanied by progressive accumulation of Aβ plaques, decreased synaptophysin and vesicular acetylcholine transporter immunoexpression. Donepezil had a disease-modifying effect, slowing the deterioration of all the pathological markers studied when treatment was started in a pre-symptomatic stage of AD. However, in the group of mice with advanced stage of AD, such disease-modifying effects were not evident.
Collapse
Affiliation(s)
- Irina Zueva
- Arbuzov Institute of Organic and Physical Chemistry, Federal Research Center "Kazan Scientific Center of the Russian Academy of Sciences", Kazan, Russia.
| | - Grigory Belyaev
- Arbuzov Institute of Organic and Physical Chemistry, Federal Research Center "Kazan Scientific Center of the Russian Academy of Sciences", Kazan, Russia
| | - Konstantin Petrov
- Arbuzov Institute of Organic and Physical Chemistry, Federal Research Center "Kazan Scientific Center of the Russian Academy of Sciences", Kazan, Russia
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
2
|
Iyer K, Tenchov R, Sasso JM, Ralhan K, Jotshi J, Polshakov D, Maind A, Zhou QA. Rare Diseases, Spotlighting Amyotrophic Lateral Sclerosis, Huntington's Disease, and Myasthenia Gravis: Insights from Landscape Analysis of Current Research. Biochemistry 2025; 64:1698-1719. [PMID: 40169538 PMCID: PMC12004453 DOI: 10.1021/acs.biochem.4c00722] [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: 10/27/2024] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Rare diseases are a diverse group of disorders that, despite each individual condition's rarity, collectively affect a significant portion of the global population. Currently approximately 10,000 rare diseases exist globally, with 80% of these diseases being identified as having genetic origins. In this Review, we examine data from the CAS Content Collection to summarize scientific progress in the area of rare diseases. We examine the publication landscape in the area in an effort to provide insights into current advances and developments. We then discuss the evolution of key concepts in the field, genetic associations, as well as the major technologies and development pipelines of rare disease treatments. We focus our attention on three specific rare diseases: (i) amyotrophic lateral sclerosis, a terminal neurodegenerative disease affecting the central nervous system resulting in progressive loss of motor neurons that control voluntary muscles; (ii) Huntington's disease, another terminal neurodegenerative disease that causes progressive degeneration of nerve cells in the brain, with a wide impact on a person's functional abilities; and (iii) myasthenia gravis, a chronic autoimmune synaptopathy leading to skeletal muscle weakness. While the pathogenesis of these rare diseases is being elucidated, there is neither a cure nor preventative treatment available, only symptomatic treatment. The objective of the paper is to provide a broad overview of the evolving landscape of current knowledge on rare diseases and specifically on the biology and genetics of the three spotlighted diseases, to outline challenges and evaluate growth opportunities, an aim to further efforts in solving the remaining challenges.
Collapse
Affiliation(s)
- Kavita
A. Iyer
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Dmitrii Polshakov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Ankush Maind
- ACS
International India Pvt. Ltd., Pune 411044, India
| | | |
Collapse
|
3
|
Wong-Guerra M, Montano-Peguero Y, Hernández-Enseñat D, Ramírez-Sánchez J, Mondelo-Rodríguez A, Padrón-Yaquis AS, García-Alfonso E, Fonseca-Fonseca LA, Nuñez-Figueredo Y. Mitochondrial protective properties exerted by JM-20 in a dementia model induced by intracerebroventricular administration of streptozotocin in mice. Behav Brain Res 2025; 480:115385. [PMID: 39667646 DOI: 10.1016/j.bbr.2024.115385] [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: 08/21/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Mitochondrial dysfunction and brain insulin resistance have been related to Alzheimer's disease (AD) development. Streptozotocin (STZ) is commonly employed to disrupt glucose and insulin metabolism, even causing cognitive impairment in animal models. We aimed at studying the protective effect of JM-20 on STZ-induced memory impairment and brain mitochondrial dysfunction. METHODS Male C57Bl6 mice received 3 mg/kg STZ intracerebroventricularly and JM-20 (0.25 mg/kg or 4 mg/kg) was administered daily by gastric gavage. Episodic memory was evaluated through Y-maze, novel object recognition, and Morris water maze. Endogenous antioxidant systems (catalase and superoxide dismutase activities), total sulfhydryl groups, malondialdehyde levels were also studied and acetylcholinesterase (AChE) activity were assessed in the prefrontal cortex (PC) and hippocampus (HO). RESULTS demonstrated that STZ injection impaired recognition and spatial learning and memory and oxygen flow in all mitochondrial respiration states. Additionally, STZ increased AChE, superoxide dismutase, and catalase activity in the PC but not in HO tissue. A neuroprotective effect of JM-20 on STZ-induced memory decline, and mitochondrial dysfunction was observed, suggesting an important causal interaction. In addition, JM-20 was able to decreased AChE enzyme hyperactivity, rescued endogenous antioxidant systems, and prevented histologically observed neuronal damage CONCLUSION: Our results indicate that JM-20 protects against STZ-induced impairment in brain bioenergetic metabolism and memory, confirming its potential as a candidate for treating neurodegenerative disorders associated with mitochondrial dysfunction like AD.
Collapse
Affiliation(s)
- Maylin Wong-Guerra
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba; Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, USACH, Alameda, Santiago 3363, Chile
| | - Yanay Montano-Peguero
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba; Facultad de Ciencias Químicas y Farmacéuticas, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Casilla 233, Santiago, Chile
| | - Daniela Hernández-Enseñat
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Jeney Ramírez-Sánchez
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Abel Mondelo-Rodríguez
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Alejandro Saúl Padrón-Yaquis
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Enrique García-Alfonso
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba
| | - Luis Arturo Fonseca-Fonseca
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba.
| | - Yanier Nuñez-Figueredo
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos, La Habana 10600, Cuba.
| |
Collapse
|
4
|
Chaturvedi A, Sharma V, Rawal RK, Singh M, Singh V. Sustainable protocol for Cu-catalysed A 3-coupling under solvent-free conditions. Org Biomol Chem 2025; 23:854-863. [PMID: 39625717 DOI: 10.1039/d4ob01728e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
A Cu-catalyzed three-component cascade reaction has been developed, involving ortho-alkynylaryl aldehydes, terminal alkynes and aliphatic/aromatic amines or diamines. This diversity oriented methodology successfully delivered a rich library of 72 molecules in good to excellent yields (yields up to 99%) through the application of an A3-coupling reaction. This method is green, straightforward to execute, requires a short reaction time (2 min-4 h), does not require solvents or harsh or inert conditions, i.e. can be performed in open air, and utilizes only a small amount of a cheap and readily available catalyst (2.5 to 10 mol% CuI). It proficiently produced a variety of biphenylacetylene tethered propargylamines, alkyne tethered dihydroisoquinolines, and N-fused benzimidazoles with excellent regioselectivity.
Collapse
Affiliation(s)
- Anjali Chaturvedi
- Department of Chemistry, Central University of Punjab, Bathinda, 151004, Punjab, India.
- Department of Biological and Chemical Science Baba Farid College, Muktsar Road, Bathinda, Punjab 151001, Punjab, India.
| | - Vishal Sharma
- Department of Chemistry, Central University of Punjab, Bathinda, 151004, Punjab, India.
| | - Ravindra K Rawal
- Chemical Sciences and Technology Division, CSIR-North East Institute of Science and Technology (NEIST), Jorhat, 785006, Assam, India
| | - Manpreet Singh
- Department of Biological and Chemical Science Baba Farid College, Muktsar Road, Bathinda, Punjab 151001, Punjab, India.
| | - Virender Singh
- Department of Chemistry, Central University of Punjab, Bathinda, 151004, Punjab, India.
| |
Collapse
|
5
|
Kashish, Hossain MJ, Khan S. N-Heterocyclic Silylene-Cu(I) Complexes as Efficient Catalysts for Three-Component Coupling Reactions. Chem Asian J 2025; 20:e202401058. [PMID: 39440901 DOI: 10.1002/asia.202401058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/25/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024]
Abstract
We have explored NHSi-supported copper(I) complexes as active and highly efficient catalysts for A3 (aldehyde-alkyne-amine) and KA2 (ketone-alkyne-amine) three-component coupling reactions to synthesize propargyl amines in excellent yields with high TOF under solvent-and additive-free conditions.
Collapse
Affiliation(s)
- Kashish
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Md Jabed Hossain
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Shabana Khan
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| |
Collapse
|
6
|
Vahid ZF, Eskandani M, Dadashi H, Vandghanooni S, Rashidi MR. Recent advances in potential enzymes and their therapeutic inhibitors for the treatment of Alzheimer's disease. Heliyon 2024; 10:e40756. [PMID: 39717593 PMCID: PMC11664286 DOI: 10.1016/j.heliyon.2024.e40756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024] Open
Abstract
Alzheimer's disease (AD), a chronic neurodegenerative disease, is clinically characterized by loss of memory and learning ability among other neurological deficits. Amyloid plaques, hyperphosphorylated tau protein, and neurofibrillary tangles involve in AD etiology. Meanwhile, enzymes and their inhibitors have become the focus of research in AD treatment. In this review, the molecular mechanisms involved in the pathogenesis of AD were overviewed and various enzymes such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), β-secretase, γ-secretase, monoamine oxidase (MAO), and receptor of advanced glycation end products (RAGE) were highlighted as potential targets for AD treatment. Several hybrid molecules with essential substructures derived from various chemotypes have demonstrated desired pharmacological activity. It is envisioned that the development of new drugs that inhibit enzymes involved in AD is a future trend in the management of the disease.
Collapse
Affiliation(s)
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Dadashi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Medicinal Chemistry Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Qiang RR, Xiang Y, Zhang L, Bai XY, Zhang D, Li YJ, Yang YL, Liu XL. Ferroptosis: A new strategy for targeting Alzheimer's disease. Neurochem Int 2024; 178:105773. [PMID: 38789042 DOI: 10.1016/j.neuint.2024.105773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a complex pathogenesis, which involves the formation of amyloid plaques and neurofibrillary tangles. Many recent studies have revealed a close association between ferroptosis and the pathogenesis of AD. Factors such as ferroptosis-associated iron overload, lipid peroxidation, disturbances in redox homeostasis, and accumulation of reactive oxygen species have been found to contribute to the pathological progression of AD. In this review, we explore the mechanisms underlying ferroptosis, describe the link between ferroptosis and AD, and examine the reported efficacy of ferroptosis inhibitors in treating AD. Finally, we discuss the potential challenges to ferroptosis inhibitors use in the clinic, enabling their faster use in clinical treatment.
Collapse
Affiliation(s)
| | - Yang Xiang
- College of Physical Education, Yan'an University, Shaanxi, 716000, China
| | - Lei Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Yang Jing Li
- School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xiao Long Liu
- School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
8
|
Cummings JL, Osse AML, Kinney JW, Cammann D, Chen J. Alzheimer's Disease: Combination Therapies and Clinical Trials for Combination Therapy Development. CNS Drugs 2024; 38:613-624. [PMID: 38937382 PMCID: PMC11258156 DOI: 10.1007/s40263-024-01103-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
Alzheimer's disease (AD) is a complex multifaceted disease. Recently approved anti-amyloid monoclonal antibodies slow disease progression by approximately 30%, and combination therapy appears necessary to prevent the onset of AD or produce greater slowing of cognitive and functional decline. Combination therapies may address core features, non-specific co-pathology commonly occurring in patients with AD (e.g., inflammation), or non-AD pathologies that may co-occur with AD (e.g., α-synuclein). Combination therapies may be advanced through co-development of more than one new molecular entity or through add-on strategies including an approved agent plus a new molecular entity. Addressing add-on combination therapy is currently urgent since patients on anti-amyloid monoclonal antibodies may be included in clinical trials for experimental agents. Phase 1 information must be generated for each agent in combination drug development. Phase 2 and Phase 3 of add-on therapies may contrast the new molecular entity, the approved agent as standard of care, and the combination. More complex development programs including standard or modified combinatorial designs are required for co-development of two or more new molecular entities. Biomarkers are markedly affected by anti-amyloid monoclonal antibodies, and these effects must be anticipated in add-on trials. Examining target engagement biomarkers and comparing the magnitude and sequence of biomarker changes in those receiving more than one therapy, compared with those on monotherapy, may be informative. Using network-based medicine approaches, computational strategies may identify rational combinations using disease and drug effect network mapping.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV, Las Vegas, NV, USA.
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
- , 1380 Opal Valley Street, Henderson, NV, 89052, USA.
| | - Amanda M Leisgang Osse
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV, Las Vegas, NV, USA
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jefferson W Kinney
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV, Las Vegas, NV, USA
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| |
Collapse
|
9
|
He H, Xie J, Huang D, Zhang M, Zhao X, Ying Y, Wang J. DRTerHGAT: A drug repurposing method based on the ternary heterogeneous graph attention network. J Mol Graph Model 2024; 130:108783. [PMID: 38677034 DOI: 10.1016/j.jmgm.2024.108783] [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: 01/09/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Drug repurposing is an effective method to reduce the time and cost of drug development. Computational drug repurposing can quickly screen out the most likely associations from large biological databases to achieve effective drug repurposing. However, building a comprehensive model that integrates drugs, proteins, and diseases for drug repurposing remains challenging. This study proposes a drug repurposing method based on the ternary heterogeneous graph attention network (DRTerHGAT). DRTerHGAT designs a novel protein feature extraction process consisting of a large-scale protein language model and a multi-task autoencoder, so that protein features can be extracted accurately and efficiently from amino acid sequences. The ternary heterogeneous graph of drug-protein-disease comprehensively considering the relationships among the three types of nodes, including three homogeneous and three heterogeneous relationships. Based on the graph and the extracted protein features, the deep features of the drugs and the diseases are extracted by graph convolutional networks (GCN) and heterogeneous graph node attention networks (HGNA). In the experiments, DRTerHGAT is proven superior to existing advanced methods and DRTerHGAT variants. DRTerHGAT's powerful ability for drug repurposing is also demonstrated in Alzheimer's disease.
Collapse
Affiliation(s)
- Hongjian He
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Jiang Xie
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China.
| | - Dingkai Huang
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Mengfei Zhang
- The School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Xuyu Zhao
- School of Life Sciences,Shanghai University, Shanghai, China
| | - Yiwei Ying
- School of Life Sciences,Shanghai University, Shanghai, China
| | - Jiao Wang
- School of Life Sciences,Shanghai University, Shanghai, China.
| |
Collapse
|
10
|
Kumar S, Mahajan A, Ambatwar R, Khatik GL. Recent Advancements in the Treatment of Alzheimer's Disease: A Multitarget-directed Ligand Approach. Curr Med Chem 2024; 31:6032-6062. [PMID: 37861025 DOI: 10.2174/0109298673264076230921065945] [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: 05/25/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and one of the leading causes of progressive dementia, affecting 50 million people worldwide. Many pathogenic processes, including amyloid β aggregation, tau hyperphosphorylation, oxidative stress, neuronal death, and deterioration of the function of cholinergic neurons, are associated with its progression. The one-compound-one-target treatment paradigm was unsuccessful in treating AD due to the multifaceted nature of Alzheimer's disease. The recent development of multitarget-directed ligand research has been explored to target the complementary pathways associated with the disease. We aimed to find the key role and progress of MTDLs in treating AD; thus, we searched for the past ten years of literature on "Pub- Med", "ScienceDirect", "ACS" and "Bentham Science" using the keywords neurodegenerative diseases, Alzheimer's disease, and multitarget-directed ligands. The literature was further filtered based on the quality of work and relevance to AD. Thus, this review highlights the current advancement and advantages of multitarget-directed ligands over traditional single-targeted drugs and recent progress in their development to treat AD.
Collapse
Affiliation(s)
- Sumit Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Amol Mahajan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Ramesh Ambatwar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Gopal L Khatik
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| |
Collapse
|
11
|
Fišar Z. Linking the Amyloid, Tau, and Mitochondrial Hypotheses of Alzheimer's Disease and Identifying Promising Drug Targets. Biomolecules 2022; 12:1676. [PMID: 36421690 PMCID: PMC9687482 DOI: 10.3390/biom12111676] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/23/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023] Open
Abstract
Damage or loss of brain cells and impaired neurochemistry, neurogenesis, and synaptic and nonsynaptic plasticity of the brain lead to dementia in neurodegenerative diseases, such as Alzheimer's disease (AD). Injury to synapses and neurons and accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles are considered the main morphological and neuropathological features of AD. Age, genetic and epigenetic factors, environmental stressors, and lifestyle contribute to the risk of AD onset and progression. These risk factors are associated with structural and functional changes in the brain, leading to cognitive decline. Biomarkers of AD reflect or cause specific changes in brain function, especially changes in pathways associated with neurotransmission, neuroinflammation, bioenergetics, apoptosis, and oxidative and nitrosative stress. Even in the initial stages, AD is associated with Aβ neurotoxicity, mitochondrial dysfunction, and tau neurotoxicity. The integrative amyloid-tau-mitochondrial hypothesis assumes that the primary cause of AD is the neurotoxicity of Aβ oligomers and tau oligomers, mitochondrial dysfunction, and their mutual synergy. For the development of new efficient AD drugs, targeting the elimination of neurotoxicity, mutual potentiation of effects, and unwanted protein interactions of risk factors and biomarkers (mainly Aβ oligomers, tau oligomers, and mitochondrial dysfunction) in the early stage of the disease seems promising.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague, Czech Republic
| |
Collapse
|
12
|
Zheng L, Qin X, Wang J, Zhang M, An Q, Xu J, Qu X, Cao X, Niu B. Discovery of MAO-B Inhibitor with Machine Learning, Topomer CoMFA, Molecular Docking and Multi-Spectroscopy Approaches. Biomolecules 2022; 12:biom12101470. [PMID: 36291679 PMCID: PMC9599443 DOI: 10.3390/biom12101470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 12/05/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia and is a serious disruption to normal life. Monoamine oxidase-B (MAO-B) is an important target for the treatment of AD. In this study, machine learning approaches were applied to investigate the identification model of MAO-B inhibitors. The results showed that the identification model for MAO-B inhibitors with K-nearest neighbor(KNN) algorithm had a prediction accuracy of 94.1% and 88.0% for the 10-fold cross-validation test and the independent test set, respectively. Secondly, a quantitative activity prediction model for MAO-B was investigated with the Topomer CoMFA model. Two separate cutting mode approaches were used to predict the activity of MAO-B inhibitors. The results showed that the cut model with q2 = 0.612 (cross-validated correlation coefficient) and r2 = 0.824 (non-cross-validated correlation coefficient) were determined for the training and test sets, respectively. In addition, molecular docking was employed to analyze the interaction between MAO-B and inhibitors. Finally, based on our proposed prediction model, 1-(4-hydroxyphenyl)-3-(2,4,6-trimethoxyphenyl)propan-1-one (LB) was predicted as a potential MAO-B inhibitor and was validated by a multi-spectroscopic approach including fluorescence spectra and ultraviolet spectrophotometry.
Collapse
Affiliation(s)
- Linfeng Zheng
- School of Life Science, Shanghai University, Shanghai 200444, China
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiangyang Qin
- Department of Chemistry, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Jiao Wang
- School of Life Science, Shanghai University, Shanghai 200444, China
| | - Mengying Zhang
- School of Life Science, Shanghai University, Shanghai 200444, China
| | - Quanlin An
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200444, China
| | - Jinzhi Xu
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200444, China
| | - Xiaosheng Qu
- National Engineering Laboratory of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, China
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200444, China
- Correspondence: (X.C.); (B.N.)
| | - Bing Niu
- School of Life Science, Shanghai University, Shanghai 200444, China
- Correspondence: (X.C.); (B.N.)
| |
Collapse
|
13
|
Zhang H, Wang Y, Wang Y, Li X, Wang S, Wang Z. Recent advance on carbamate-based cholinesterase inhibitors as potential multifunctional agents against Alzheimer's disease. Eur J Med Chem 2022; 240:114606. [PMID: 35858523 DOI: 10.1016/j.ejmech.2022.114606] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD), as the fourth leading cause of death among the elderly worldwide, has brought enormous challenge to the society. Due to its extremely complex pathogeneses, the development of multi-target directed ligands (MTDLs) becomes the major strategy for combating AD. Carbamate moiety, as an essential building block in the development of MTDLs, exhibits structural similarity to neurotransmitter acetylcholine (ACh) and has piqued extensive attention in discovering multifunctional cholinesterase inhibitors. To date, numerous preclinical studies demonstrate that carbamate-based cholinesterase inhibitors can prominently increase the level of ACh and improve cognition impairments and behavioral deficits, providing a privileged strategy for the treatment of AD. Based on the recent research focus on the novel cholinesterase inhibitors with multiple biofunctions, this review aims at summarizing and discussing the most recent studies excavating the potential carbamate-based MTDLs with cholinesterase inhibition efficacy, to accelerate the pace of pleiotropic cholinesterase inhibitors for coping AD.
Collapse
Affiliation(s)
- Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Yuying Wang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Yuqing Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Xuelin Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Shuzhi Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
14
|
Kumar B, Dwivedi AR, Arora T, Raj K, Prashar V, Kumar V, Singh S, Prakash J, Kumar V. Design, Synthesis, and Pharmacological Evaluation of N-Propargylated Diphenylpyrimidines as Multitarget Directed Ligands for the Treatment of Alzheimer's Disease. ACS Chem Neurosci 2022; 13:2122-2139. [PMID: 35797244 DOI: 10.1021/acschemneuro.2c00132] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Alzheimer's disease (AD), a multifactorial complex neural disorder, is categorized with progressive memory loss and cognitive impairment as main clinical features. The multitarget directed ligand (MTDL) strategy is explored for the treatment of multifactorial diseases such as cancer and AD. Herein, we report the synthesis and screening of 24 N-propargyl-substituted diphenylpyrimidine derivatives as MTDLs against acetylcholine/butyrylcholine esterases and monoamine oxidase enzymes. In this series, VP1 showed the most potent MAO-B inhibitory activity with an IC50 value of 0.04 ± 0.002 μM. VP15 with an IC50 value of 0.04 ± 0.003 μM and a selectivity index of 626 (over BuChE) displayed the most potent AChE inhibitory activity in this series. In the reactive oxygen species (ROS) inhibition studies, VP1 reduced intercellular ROS levels in SH-SY5Y cells by 36%. This series of compounds also exhibited potent neuroprotective potential against 6-hydroxydopamine-induced neuronal damage in SH-SY5Y cells with up to 90% recovery. In the in vivo studies in the rats, the hydrochloride salt of VP15 was orally administered and found to cross the blood-brain barrier and reach the target site. VP15·HCl significantly attenuated the spatial memory impairment and improved the cognitive deficits in the mice. This series of compounds were found to be irreversible inhibitors and showed no cytotoxicity against neuronal cells. In in silico studies, the compounds attained thermodynamically stable orientation with complete occupancy at the active site of the receptors. Thus, N-propargyl-substituted diphenylpyrimidines displayed drug-like characteristics and have the potential to be developed as MTDLs for the effective treatment of AD.
Collapse
Affiliation(s)
- Bhupinder Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India.,Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, G.T Road, Moga, Punjab 142001, India
| | - Ashish Ranjan Dwivedi
- Laboratory of Organic and Medicinal Chemistry, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Tania Arora
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Khadga Raj
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, G.T Road, Moga, Punjab 142001, India
| | - Vikash Prashar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Vijay Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, G.T Road, Moga, Punjab 142001, India
| | - Jyoti Prakash
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Vinod Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab, Bathinda, Punjab 151401, India
| |
Collapse
|
15
|
Jana A, Bhattacharjee A, Das SS, Srivastava A, Choudhury A, Bhattacharjee R, De S, Perveen A, Iqbal D, Gupta PK, Jha SK, Ojha S, Singh SK, Ruokolainen J, Jha NK, Kesari KK, Ashraf GM. Molecular Insights into Therapeutic Potentials of Hybrid Compounds Targeting Alzheimer's Disease. Mol Neurobiol 2022; 59:3512-3528. [PMID: 35347587 PMCID: PMC9148293 DOI: 10.1007/s12035-022-02779-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/21/2022] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is one of the most complex progressive neurological disorders involving degeneration of neuronal connections in brain cells leading to cell death. AD is predominantly detected among elder people (> 65 years), mostly diagnosed with the symptoms of memory loss and cognitive dysfunctions. The multifarious pathogenesis of AD comprises the accumulation of pathogenic proteins, decreased neurotransmission, oxidative stress, and neuroinflammation. The conventional therapeutic approaches are limited to symptomatic benefits and are ineffective against disease progression. In recent years, researchers have shown immense interest in the designing and fabrication of various novel therapeutics comprised of naturally isolated hybrid molecules. Hybrid therapeutic compounds are developed from the combination of pharmacophores isolated from bioactive moieties which specifically target and block various AD-associated pathogenic pathways. The method of designing hybrid molecules has numerous advantages over conventional multitarget drug development methods. In comparison to in silico high throughput screening, hybrid molecules generate quicker results and are also less expensive than fragment-based drug development. Designing hybrid-multitargeted therapeutic compounds is thus a prospective approach in developing an effective treatment for AD. Nevertheless, several issues must be addressed, and additional researches should be conducted to develop hybrid therapeutic compounds for clinical usage while keeping other off-target adverse effects in mind. In this review, we have summarized the recent progress on synthesis of hybrid compounds, their molecular mechanism, and therapeutic potential in AD. Using synoptic tables, figures, and schemes, the review presents therapeutic promise and potential for the development of many disease-modifying hybrids into next-generation medicines for AD.
Collapse
Affiliation(s)
- Ankit Jana
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Arkadyuti Bhattacharjee
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Avani Srivastava
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Akshpita Choudhury
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Rahul Bhattacharjee
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed To Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Swagata De
- Department of English, DDE Unit, The University of Burdwan, GolapbagBurdwan, West Bengal, 713104, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, 15551, Al Ain, United Arab Emirates
| | - Sandeep Kumar Singh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, 00076, Espoo, Finland
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, 00076, Espoo, Finland.
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia. .,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
16
|
Prachayasittikul V, Pingaew R, Prachayasittikul S, Prachayasittikul V. 8-Hydroxyquinolines: A Promising Pharmacophore Potentially Developed as Disease-Modifying Agents for Neurodegenerative Diseases: A Review. HETEROCYCLES 2022. [DOI: 10.3987/rev-22-sr(r)6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
17
|
Akıncıoğlu A, Göksu S, Naderi A, Akıncıoğlu H, Kılınç N, Gülçin İ. Cholinesterases, carbonic anhydrase inhibitory properties and in silico studies of novel substituted benzylamines derived from dihydrochalcones. Comput Biol Chem 2021; 94:107565. [PMID: 34474201 DOI: 10.1016/j.compbiolchem.2021.107565] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A series of novel urea, sulfamide and N,N-dipropargyl substituted benzylamines were synthesized from dihydrochalcones. The synthesized compounds were evaluated for their cholinesterases and carbonic anhydrase inhibitory actions. The known dihydrochalcones were converted into four new benzylamines via reductive amination. N,N-Dipropargylamines, ureas and sulfamides were synthesized following the reactions of benzylamines with propargyl bromide, N,N-dimethyl sulfamoyl chloride and N,N-dimethyl carbamoyl chloride. The novel substituted benzylamines derived from dihydrochalcones were evaluated against some enzymes such as human erythrocyte carbonic anhydrase I and II isoenzymes (hCA I and hCA II), acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). The novel substituted benzylamines derived from dihydrochalcones exhibited Ki values in the range of 0.121-1.007 nM on hCA I, and 0.077-0.487 nM on hCA II closely related to several pathological processes. On the other hand, Ki values were found in the range of 0.112-0.558 nM on AChE, 0.061-0.388 nM on BChE. As a result, novel substituted benzylamines derived from dihydrochalcones showed potent inhibitory profiles against indicated metabolic enzymes. In addition, Induced-Fit Docking (IFD) simulations and ADME prediction studies have also been carried out to elucidate the inhibition mechanisms and drug-likeness of the synthesized compounds. Therefore, these results can make significant contributions to the treatment of some global diseases, especially Alzheimer's diseases and glaucoma, and the development of new drugs.
Collapse
Affiliation(s)
- Akın Akıncıoğlu
- Agri Ibrahim Cecen University, Central Researching Laboratory, 04100 Agri, Turkey
| | - Süleyman Göksu
- Atatürk University, Faculty of Science, Department of Chemistry, Erzurum, Turkey.
| | - Ali Naderi
- Atatürk University, Faculty of Science, Department of Chemistry, Erzurum, Turkey
| | - Hülya Akıncıoğlu
- Agri Ibrahim Cecen University, Faculty of Arts and Science, Agri, Turkey
| | - Namık Kılınç
- Igdir University, Vocational School of Health Services, Department of Medical Services and Techniques, Igdir, Turkey
| | - İlhami Gülçin
- Atatürk University, Faculty of Science, Department of Chemistry, Erzurum, Turkey
| |
Collapse
|
18
|
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: 21] [Impact Index Per Article: 5.3] [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
|
19
|
Song IU, Im JJ, Jeong H, Na SH, Chung YA. Possible neuroprotective effects of rasagiline in Alzheimer's disease: a SPECT study. Acta Radiol 2021; 62:784-790. [PMID: 32646230 DOI: 10.1177/0284185120940264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The current lack of effective treatments for Alzheimer's disease (AD) and the rapidly increasing burden of the disease highlight the urgent need to find new treatments. Despite accumulating evidence of the beneficial effects of rasagiline in neurodegenerative diseases such as Parkinson's disease, the effects of rasagiline on the brains of patients with AD have not been elucidated. PURPOSE To examine the effects of rasagiline on regional cerebral flow (rCBF) in patients with AD using single photon emission computed tomography (SPECT). MATERIAL AND METHODS Among 22 patients with AD, 11 patients received adjunctive rasagiline at 1 mg/day in conjunction with acetylcholinesterase inhibitors (AChEI); 11 patients were only treated with AChEI for about 1.6 years. All patients underwent brain technetium-99m hexamethylpropylene amine oxime SPECT scans and clinical assessments at baseline and follow-up visits. Annual percent changes in rCBF were compared between the groups in a voxel-wise manner. RESULTS SPECT analysis revealed that the rasagiline-treated group showed more increased rCBF in the cingulate gyrus, inferior frontal gyrus, putamen, and thalamus compared to the comparison group (P < 0.005). CONCLUSION We demonstrated that adjunctive rasagiline treatment may have beneficial effects on brain perfusion in patients with AD, suggesting potential neuroprotective effects.
Collapse
Affiliation(s)
- In-Uk Song
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jooyeon Jamie Im
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Nuclear Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyeonseok Jeong
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Nuclear Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Hee Na
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong-An Chung
- Department of Nuclear Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
20
|
Manujyothi R, Aneeja T, Anilkumar G. Solvent-free synthesis of propargylamines: an overview. RSC Adv 2021; 11:19433-19449. [PMID: 35479216 PMCID: PMC9033675 DOI: 10.1039/d1ra03324g] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Propargylamines are a class of compounds with many pharmaceutical and biological properties. A green approach to synthesize such compounds is very relevant. This review aims to describe the solvent-free synthetic approaches towards propargylamines via A3 and KA2 coupling reactions covering the literature up to 2021.
Collapse
Affiliation(s)
- Ravi Manujyothi
- Institute for Integrated Programmes and Research in Basic Sciences (IIRBS), Mahatma Gandhi University Priyadarsini Hills P O Kottayam Kerala 686560 India +91-481-2731036
| | - Thaipparambil Aneeja
- School of Chemical Sciences, Mahatma Gandhi University Priyadarsini Hills P O Kottayam Kerala 686560 India
| | - Gopinathan Anilkumar
- Institute for Integrated Programmes and Research in Basic Sciences (IIRBS), Mahatma Gandhi University Priyadarsini Hills P O Kottayam Kerala 686560 India +91-481-2731036
- School of Chemical Sciences, Mahatma Gandhi University Priyadarsini Hills P O Kottayam Kerala 686560 India
- Advanced Molecular Materials Research Centre (AMMRC), Mahatma Gandhi University Priyadarsini Hills P O Kottayam Kerala 686560 India
| |
Collapse
|
21
|
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
|
22
|
Ramos E, Lajarín-Cuesta R, Arribas RL, García-Frutos EM, González-Lafuente L, Egea J, de Los Ríos C, Romero A. In Silico Prediction of the Toxic Potential of Neuroprotective Bifunctional Molecules Based on Chiral N-Propargyl-1,2-amino Alcohol Derivatives. Chem Res Toxicol 2021; 34:1245-1249. [PMID: 33635058 PMCID: PMC8478334 DOI: 10.1021/acs.chemrestox.0c00519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
N-Propargylamines
are useful synthetic scaffolds
for the synthesis of bioactive molecules, and in addition, they possess
important pharmacological activities. We obtained several neuroprotective
molecules, chiral 1,2-amino alcohols and 1,2-diamines, able to reduce
by almost 70% the rotenone and oligomycin A-induced damage in SH-SY5Y
cells. Furthermore, some molecules assessed also counteracted the
toxicity evoked by the Ser/Thr phosphatase inhibitor okadaic acid.
Before extrapolating these data to preclinical studies, we analyze
the molecules through an in silico prediction system
to detect carcinogenicity risk or other toxic effects. In light of
these promising results, these molecules may be considered as a lead
family of neuroprotective and relatively safe compounds.
Collapse
Affiliation(s)
- Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rocío Lajarín-Cuesta
- Health Research Institute, Clinical Pharmacology Service, University Hospital La Princesa, Autonomous University of Madrid, 28006 Madrid, Spain.,Institute Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Raquel L Arribas
- Health Research Institute, Clinical Pharmacology Service, University Hospital La Princesa, Autonomous University of Madrid, 28006 Madrid, Spain.,Institute Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Eva M García-Frutos
- Materials Science Factory,Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Javier Egea
- Health Research Institute, Clinical Pharmacology Service, University Hospital La Princesa, Autonomous University of Madrid, 28006 Madrid, Spain.,Institute Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain.,Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, 28040 Madrid, Spain
| | - Cristóbal de Los Ríos
- Health Research Institute, Clinical Pharmacology Service, University Hospital La Princesa, Autonomous University of Madrid, 28006 Madrid, Spain.,Institute Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
23
|
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
|
24
|
Kumar D, Ambasta RK, Kumar P. Ubiquitin biology in neurodegenerative disorders: From impairment to therapeutic strategies. Ageing Res Rev 2020; 61:101078. [PMID: 32407951 DOI: 10.1016/j.arr.2020.101078] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/24/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022]
Abstract
The abnormal accumulation of neurotoxic proteins is the typical hallmark of various age-related neurodegenerative disorders (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis and Multiple sclerosis. The anomalous proteins, such as Aβ, Tau in Alzheimer's disease and α-synuclein in Parkinson's disease, perturb the neuronal physiology and cellular homeostasis in the brain thereby affecting the millions of human lives across the globe. Here, ubiquitin proteasome system (UPS) plays a decisive role in clearing the toxic metabolites in cells, where any aberrancy is widely reported to exaggerate the neurodegenerative pathologies. In spite of well-advancement in the ubiquitination research, their molecular markers and mechanisms for target-specific protein ubiquitination and clearance remained elusive. Therefore, this review substantiates the role of UPS in the brain signaling and neuronal physiology with their mechanistic role in the NDD's specific pathogenic protein clearance. Moreover, current and future promising therapies are discussed to target UPS-mediated neurodegeneration for better public health.
Collapse
|
25
|
Uddin MS, Kabir MT, Rahman MM, Mathew B, Shah MA, Ashraf GM. TV 3326 for Alzheimer's dementia: a novel multimodal ChE and MAO inhibitors to mitigate Alzheimer's-like neuropathology. ACTA ACUST UNITED AC 2020; 72:1001-1012. [PMID: 32149402 DOI: 10.1111/jphp.13244] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/30/2020] [Accepted: 02/09/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders and a well-recognized cause of dementia with ageing. In this review, we have represented the ChE and MAO inhibitory potential of TV 3326 against AD based on current scientific evidence. KEY FINDINGS The aetiology of AD is quite complex and not completely understood. However, it has been observed that AD involves the deposition of abnormal amyloid beta (Aβ), along with hyperphosphorylation of tau, oxidative stress, low acetylcholine (ACh) level and biometal dyshomeostasis. Due to the complex nature of AD aetiology, active research is required in the areas of development of multitarget drugs with 2 or more complementary biological functions, as they might represent significant progress in the AD treatment. Interestingly, it has been found that TV 3326 (i.e. ladostigil) is regarded as a novel therapeutic agent since it has the potential to cause inhibition of monoamine oxidase (MAO) A and B, and acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) in the brain. Furthermore, it has the capacity to reverse memory impairments, which further suggests the ability of this drug to elevate cholinergic activity in the brain. SUMMARY TV 3326 can avert oxidative-nitrative stress and gliosis. It has also been confirmed that TV 3326 contains neuroprotective and anti-apoptotic properties. Therefore, this distinctive combined inhibition of ChE and MAO along with its neuroprotective property makes TV 3326 a useful drug in the treatment of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Md Motiar Rahman
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
26
|
Kaur P, Kumar B, Gurjar KK, Kumar R, Kumar V, Kumar R. Metal- and Solvent-Free Multicomponent Decarboxylative A 3-Coupling for the Synthesis of Propargylamines: Experimental, Computational, and Biological Investigations. J Org Chem 2020; 85:2231-2241. [PMID: 31877044 DOI: 10.1021/acs.joc.9b02806] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Decarboxylative A3-coupling of ortho-hydroxybenzaldehydes, secondary amines, and alkynoic acids is performed under catalyst and solvent-free conditions. The developed methodology provided a waste-free method for the synthesis of hydroxylated propargylamines which are versatile precursors for various bioactive heterocyclic scaffolds. The experimental and density functional theory studies revealed that the in situ-formed ortho-quinonoid intermediate (formed from ortho-hydroxybenzaldehyde and amine) undergoes a concerted Eschweiler-Clarke type decarboxylation with alkynoic acids. The synthesized compounds were evaluated for MAO-A, MAO-B, and AChE inhibitory activities as potential drug candidates for the treatment of various neurological disorders. Compound 4f was found to be the most potent and selective MAO-B (high selectivity over MAO-A) and AChE inhibitor in the series with IC50 values of 4.27 ± 0.07 and 0.79 ± 0.03 μM, respectively.
Collapse
|
27
|
Kundu S, Kayet A, Baidya R, Satyanarayana L, Maiti DK. Nanofibrils of a Cu II-Thiophenyltriazine-Based Porous Polymer: A Diverse Heterogeneous Nanocatalyst. ACS OMEGA 2020; 5:394-405. [PMID: 31956787 PMCID: PMC6964281 DOI: 10.1021/acsomega.9b02904] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/15/2019] [Indexed: 06/10/2023]
Abstract
Herein, we report knitting of a thiophenyltriazine-based porous organic polymer (TTPOP) with high surface area and high abundance of nitrogen and sulfur sites, synthesized through a simple one-step Friedel-Crafts reaction of 2,4,6-tri(thiophen-2-yl)-1,3,5-triazine and formaldehyde dimethyl acetal in the presence of anhydrous FeCl3, and thereafter grafting of Cu(OAc)2·H2O in the porous polymer framework to achieve the potential catalyst (CuII-TTPOP). TTPOP and CuII-TTPOP were characterized thoroughly utilizing solid-state 13C-CP MAS NMR, Fourier transform infrared, wide-angle powder X-ray diffraction, thermogravimetric analysis, and X-ray photoelectron spectroscopy and surface imaging by transmission electron microscopy and field emission scanning electron microscopy. The porosity of the nanomaterials was observed in the surface imaging and verified through conducting N2 gas adsorption techniques. Keeping in mind the tremendous importance of C-C and C-N coupling and cyclization processes, the newly synthesized CuII-TTPOP was employed successfully for a wide range of organic catalytic transformations under mild conditions to afford directly valuable diindolylmethanes and spiro-analogues, phthalimidines, propargyl amines, and their sugar-based chiral compounds with high yields using readily available substrates. The highly stable new heterogeneous catalyst showed outstanding sustainability, robustness, simple separation, and recyclability.
Collapse
Affiliation(s)
- Sudipta
K. Kundu
- Department
of Chemistry, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Anirban Kayet
- Department
of Chemistry, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Ramlal Baidya
- Department
of Chemistry, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Lanka Satyanarayana
- Analytical
Department, CSIR-Indian Institute of Chemical
Technology, Uppal Road, Hyderabad 500007, India
| | - Dilip K. Maiti
- Department
of Chemistry, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| |
Collapse
|
28
|
Uddin MS, Kabir MT, Rahman MH, Alim MA, Rahman MM, Khatkar A, Al Mamun A, Rauf A, Mathew B, Ashraf GM. Exploring the Multifunctional Neuroprotective Promise of Rasagiline Derivatives for Multi-Dysfunctional Alzheimer's Disease. Curr Pharm Des 2020; 26:4690-4698. [PMID: 32250219 DOI: 10.2174/1381612826666200406075044] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/19/2020] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD) is a chronic, age-related, and irreversible brain disorder that typically develops slowly and gets worse over time. The potent auspicious drug candidate for the treatment of AD is supposed to perform the simultaneous modulation of several targets linked to AD. The new therapeutic approach involves drug candidates that are designed to act on multiple targets and have various pharmacological properties. This trend has triggered the development of various multimodal drugs including TV-3326 (i.e. ladostigil) and M-30 (i.e. a new multitarget iron chelator). TV-3326 combines the neurorestorative/neuroprotective effects of the cholinesterase (ChE) inhibitory activity of rivastigmine with rasagiline (a selective monoamine oxidase-B inhibitor and novel antiparkinsonian agent) in a single molecule. M-30, the second derivative of rasagiline, was developed by combining the propargyl moiety of rasagiline into the skeleton of VK-28 (i.e. a novel brain permeable neuroprotective iron chelator). It has been revealed that both the compounds possess anti-AD effects and therefore, the clinical development is directed to the treatment of this type of neurodegenerative diseases (NDs). In this article, we have reviewed the neuroprotective molecular mechanisms and multimodal effects of TV-3326 and M-30.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Md Habibur Rahman
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Md Abdul Alim
- Department of Chemistry, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Graduate School of Innovative Life Science, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Md Motiar Rahman
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Anurag Khatkar
- Laboratory for Preservation Technology and Enzyme Inhibition Studies, Faculty of Pharmaceutical
Sciences, Maharshi Dayanand University, Rohtak, India
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University,
Jeddah, Saudi Arabia
| |
Collapse
|
29
|
Velazquez R, Meechoovet B, Ow A, Foley C, Shaw A, Smith B, Oddo S, Hulme C, Dunckley T. Chronic Dyrk1 Inhibition Delays the Onset of AD-Like Pathology in 3xTg-AD Mice. Mol Neurobiol 2019; 56:8364-8375. [PMID: 31240602 DOI: 10.1007/s12035-019-01684-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 06/17/2019] [Indexed: 01/14/2023]
Abstract
There is a critical need for new treatment approaches that can slow or prevent the progression of Alzheimer's disease (AD). Targets that act simultaneously on multiple relevant pathways could have significant therapeutic potential. Dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1a) phosphorylates both amyloid precursor protein (APP) and tau. Dyrk1a is upregulated in post-mortem brains of AD patients, and such elevated expression is associated with cognitive deficits. We previously demonstrated that small molecule inhibition of Dyrk1 is well-tolerated and reduces amyloid plaques and pathological forms of tau in 3xTg-AD mice if administered after formation of these pathologies. However, while insoluble forms of hyperphosphorylated tau were reduced by Dyrk1 inhibition, overt neurofibrillary tangle (NFT) pathology remained unchanged. Herein, we specifically test the hypothesis that inhibition of Dyrk1 prior to NFT formation will delay the onset of pathology. 3xTg-AD mice were treated chronically, beginning at 6 months of age, prior to NFT pathology. Mice were dosed daily for either 3 or 6 months and amyloid and tau pathology were assessed. We show that chronic Dyrk1 inhibition reduces insoluble forms of amyloid beta peptides (Aβ) and hyper-phosphorylated tau long-term and that these reductions are associated with dramatic delay in the onset of both amyloid plaques and NFTs. In addition, we show that DYR219, a potent and selective small molecule Dyrk1 inhibitor, induces degradation of Dyrk1a protein, likely contributing to the efficacy of this small molecule approach in vivo. Collectively, these results suggest that therapeutic strategies targeting tau phosphorylation will show the greatest effect if administered very early in the pathogenesis of AD.
Collapse
Affiliation(s)
- R Velazquez
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - B Meechoovet
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - A Ow
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - C Foley
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - A Shaw
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - B Smith
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - S Oddo
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - C Hulme
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Travis Dunckley
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA.
| |
Collapse
|
30
|
Wang N, Qiu P, Cui W, Yan X, Zhang B, He S. Recent Advances in Multi-target Anti-Alzheimer Disease Compounds (2013 Up to the Present). Curr Med Chem 2019; 26:5684-5710. [DOI: 10.2174/0929867326666181203124102] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/03/2018] [Accepted: 11/03/2018] [Indexed: 12/21/2022]
Abstract
:
Since the last century, when scientists proposed the lock-and-key model, the discovery of
drugs has focused on the development of drugs acting on single target. However, single-target drug
therapies are not effective to complex diseases with multi-factorial pathogenesis. Moreover, the
combination of single-target drugs readily causes drug resistance and side effects. In recent years,
multi-target drugs have increasingly been represented among FDA-approved drugs. Alzheimer’s
Disease (AD) is a complex and multi-factorial disease for which the precise molecular mechanisms
are still not fully understood. In recent years, rational multi-target drug design methods, which combine
the pharmacophores of multiple drugs, have been increasingly applied in the development of
anti-AD drugs. In this review, we give a brief description of the pathogenesis of AD and provide
detailed discussions about the recent development of chemical structures of anti-AD agents (2013 up
to present) that have multiple targets, such as amyloid-β peptide, Tau protein, cholinesterases,
monoamine oxidase, β-site amyloid-precursor protein-cleaving enzyme 1, free radicals, metal ions
(Fe2+, Cu2+, Zn2+) and so on. In this paper, we also added some novel targets or possible pathogenesis
which have been reported in recent years for AD therapy. We hope that these findings may provide
new perspectives for the pharmacological treatment of AD.
Collapse
Affiliation(s)
- Ning Wang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Panpan Qiu
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Wei Cui
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Xiaojun Yan
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Bin Zhang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Shan He
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
31
|
González JF, Alcántara AR, Doadrio AL, Sánchez-Montero JM. Developments with multi-target drugs for Alzheimer's disease: an overview of the current discovery approaches. Expert Opin Drug Discov 2019; 14:879-891. [PMID: 31165654 DOI: 10.1080/17460441.2019.1623201] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Alzheimer's disease (AD), the most common type of dementia among older adults, is a chronic neurodegenerative pathology that causes a progressive loss of cognitive functioning with a decline of rational skills. It is well known that AD is multifactorial, so there are many different pharmacological targets that can be pursued. Areas covered: The authors highlight the strategic value of privileged scaffolds in a multi-target lead compound generation against AD, exploring the concept of multi-target design, with a special emphasis on hybrid compounds. Hence, the most promising building blocks for designing and synthesizing hybrid anti-AD drugs are shown, while also presenting the more advanced hybrid compounds. Expert opinion: The available therapeutic arsenal for AD, designed under the traditional paradigm of 'one-drug/one target/one-disease', is based on the inhibition of brain acetylcholinesterase (AChE) to increase acetylcholine (ACh) levels. However, this classical approach has not been sufficiently effective when used to treat any multifactor-depending pathology (cancer, diabetes or AD). The multi-target drug concept has been quickly adopted by medicinal chemists. The basic research developments reported in recent years are a solid foundation that will pave the way for the construction of future AD therapeutics.
Collapse
Affiliation(s)
- Juan F González
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Andrés R Alcántara
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Antonio L Doadrio
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Jose María Sánchez-Montero
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
32
|
|
33
|
Sharma P, Srivastava P, Seth A, Tripathi PN, Banerjee AG, Shrivastava SK. Comprehensive review of mechanisms of pathogenesis involved in Alzheimer's disease and potential therapeutic strategies. Prog Neurobiol 2018; 174:53-89. [PMID: 30599179 DOI: 10.1016/j.pneurobio.2018.12.006] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/04/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022]
Abstract
AD is a progressive neurodegenerative disorder and a leading cause of dementia in an aging population worldwide. The enormous challenge which AD possesses to global healthcare makes it as urgent as ever for the researchers to develop innovative treatment strategies to fight this disease. An in-depth analysis of the extensive available data associated with the AD is needed for a more comprehensive understanding of underlying molecular mechanisms and pathophysiological pathways associated with the onset and progression of the AD. The currently understood pathological and biochemical manifestations include cholinergic, Aβ, tau, excitotoxicity, oxidative stress, ApoE, CREB signaling pathways, insulin resistance, etc. However, these hypotheses have been criticized with several conflicting reports for their involvement in the disease progression. Several issues need to be addressed such as benefits to cost ratio with cholinesterase therapy, the dilemma of AChE selectivity over BChE, BBB permeability of peptidic BACE-1 inhibitors, hurdles related to the implementation of vaccination and immunization therapy, and clinical failure of candidates related to newly available targets. The present review provides an insight to the different molecular mechanisms involved in the development and progression of the AD and potential therapeutic strategies, enlightening perceptions into structural information of conventional and novel targets along with the successful applications of computational approaches for the design of target-specific inhibitors.
Collapse
Affiliation(s)
- Piyoosh Sharma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pavan Srivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Ankit Seth
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Prabhash Nath Tripathi
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Anupam G Banerjee
- 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
|
34
|
Youdim MBH. Monoamine oxidase inhibitors, and iron chelators in depressive illness and neurodegenerative diseases. J Neural Transm (Vienna) 2018; 125:1719-1733. [PMID: 30341696 DOI: 10.1007/s00702-018-1942-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/11/2018] [Indexed: 12/11/2022]
Abstract
In early 1920s, tyramine oxidase was discovered that metabolized tyramine and in 1933 Blaschko demonstrated that this enzyme also metabolized adrenaline, noradrenaline and dopamine. Zeller gave it the name monoamine oxidase (MAO) to distinguish it from the enzyme that oxidatively deaminated diamines. MAO was recognized as an enzyme of crucial interest to pharmacologists because it catalyzed the major inactivation pathway for the catecholamines (and, later, 5-hydroxytryptamine, as well). Within the few decade, the inhibitors of MAO were discovered and introduced for the treatment of depressive illness which was established clinically. However, the first clinical use exposed serious side effects, pharmacological interest in, and investigation of, MAO continued, resulting in the characterization of two forms, distinct forms, MAO-A and -B, and selective inhibitors for them. Selective inhibitors of MAO-B (selegiline, rasagiline and safinamide) have found a therapeutic role in the treatment of Parkinson's disease and reversible inhibitors of MAO-A offered antidepressant activity without the serious side effects of the earlier nonselective MAO inhibitors. Subsequent molecular pharmacological have also generated the concept of neuroprotection, reflecting the possibility of slowing, halting and maybe reversing, neurodegeneration in Parkinson's or Alzheimer's diseases. Increased levels of oxidative stress through the accumulation of iron in the Parkinsonian and Alzheimer brains has been suggested to be critical for the initiation and progress of neurodegeneration. Selective inhibition of brain MAO could contribute importantly to lowering such stress, preventing the formation of hydrogen peroxide. Interaction of Iron with hydrogen peroxide and lead to Fenton reaction and production of the most reactive radical, namely hydroxyl radical. There are complex interactions between free iron levels in brain and MAO, and cascade of neurotoxic events may have practical outcomes for depressive disorders and neurodegenerative diseases. As consequence recent novel therapeutic drugs for neurodegenerative diseases has led to the development of multi target drugs, that possess selective brain MAO A and B inhibitory moiety, iron chelating and antioxidant activities and the ability to increase brain levels of endogenous neurotrophins, such as BDNF, GDNF VEGF and erythropoietin and induce mitochondrial biogenesis.
Collapse
Affiliation(s)
- Moussa B H Youdim
- Technion-Bruce Rappaport Faculty of Medicine, Rappaport Family Research Institute, Haifa, Israel. .,, Yokneam, Israel.
| |
Collapse
|
35
|
Synthesis and Cytoprotective Characterization of 8-Hydroxyquinoline Betti Products. Molecules 2018; 23:molecules23081934. [PMID: 30072653 PMCID: PMC6222637 DOI: 10.3390/molecules23081934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/31/2018] [Accepted: 07/31/2018] [Indexed: 12/20/2022] Open
Abstract
The 8-hydroxyquinoline pharmacophore scaffold has been shown to possess a range of activities as metal chelation, enzyme inhibition, cytotoxicity, and cytoprotection. Based on our previous findings we set out to optimize the scaffold for cytoprotective activity for its potential application in central nervous system related diseases. A 48-membered Betti-library was constructed by the utilization of formic acid mediated industrial-compatible coupling with sets of aromatic primary amines such as anilines, oxazoles, pyridines, and pyrimidines, with (hetero)aromatic aldehydes and 8-hydroxiquinoline derivatives. After column chromatography and re-crystallization, the corresponding analogues were obtained in yields of 13–90%. The synthesized analogs were optimized with the utilization of a cytoprotection assay with chemically induced oxidative stress, and the most active compounds were further tested in orthogonal assays, a real time cell viability method, a fluorescence-activated cell sorting (FACS)-based assay measuring mitochondrial membrane potential changes, and gene expression analysis. The best candidates showed potent, nanomolar activity in all test systems and support the need for future studies in animal models of central nervous system (CNS) disorders.
Collapse
|
36
|
Agatonovic-Kustrin S, Kettle C, Morton DW. A molecular approach in drug development for Alzheimer's disease. Biomed Pharmacother 2018; 106:553-565. [PMID: 29990843 DOI: 10.1016/j.biopha.2018.06.147] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/20/2018] [Accepted: 06/27/2018] [Indexed: 01/08/2023] Open
Abstract
An increase in dementia numbers and global trends in population aging across the world prompts the need for new medications to treat the complex biological dysfunctions, such as neurodegeneration associated with dementia. Alzheimer's disease (AD) is the most common form of dementia. Cholinergic signaling, which is important in cognition, is slowly lost in AD, so the first line therapy is to treat symptoms with acetylcholinesterase inhibitors to increase levels of acetylcholine. Out of five available FDA-approved AD medications, donepezil, galantamine and rivastigmine are cholinesterase inhibitors while memantine, a N-methyl d-aspartate (NMDA) receptor antagonist, blocks the effects of high glutamate levels. The fifth medication consists of a combination of donepezil and memantine. Although these medications can reduce and temporarily slow down the symptoms of AD, they cannot stop the damage to the brain from progressing. For a superior therapeutic effect, multi-target drugs are required. Thus, a Multi-Target-Directed Ligand (MTDL) strategy has received more attention by scientists who are attempting to develop hybrid molecules that simultaneously modulate multiple biological targets. This review highlights recent examples of the MTDL approach and fragment based strategy in the rational design of new potential AD medications.
Collapse
Affiliation(s)
- Snezana Agatonovic-Kustrin
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia; School of Pharmacy and Applied Science, La Trobe Institute for Molecular Sciences, La Trobe University, Edwards Rd., Bendigo, 3550, Australia.
| | - Christine Kettle
- School of Pharmacy and Applied Science, La Trobe Institute for Molecular Sciences, La Trobe University, Edwards Rd., Bendigo, 3550, Australia
| | - David W Morton
- School of Pharmacy and Applied Science, La Trobe Institute for Molecular Sciences, La Trobe University, Edwards Rd., Bendigo, 3550, Australia
| |
Collapse
|
37
|
Novel oxindole derivatives prevent oxidative stress-induced cell death in mouse hippocampal HT22 cells. Neuropharmacology 2018; 135:242-252. [DOI: 10.1016/j.neuropharm.2018.03.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/16/2018] [Accepted: 03/14/2018] [Indexed: 01/18/2023]
|
38
|
Matsuda S, Nakagawa Y, Tsuji A, Kitagishi Y, Nakanishi A, Murai T. Implications of PI3K/AKT/PTEN Signaling on Superoxide Dismutases Expression and in the Pathogenesis of Alzheimer's Disease. Diseases 2018; 6:E28. [PMID: 29677102 PMCID: PMC6023281 DOI: 10.3390/diseases6020028] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease is a neurodegenerative sickness, where the speed of personal disease progression differs prominently due to genetic and environmental factors such as life style. Alzheimer’s disease is described by the construction of neuronal plaques and neurofibrillary tangles composed of phosphorylated tau protein. Mitochondrial dysfunction may be a noticeable feature of Alzheimer’s disease and increased production of reactive oxygen species has long been described. Superoxide dismutases (SODs) protect from excess reactive oxygen species to form less reactive hydrogen peroxide. It is suggested that SODs can play a protective role in neurodegeneration. In addition, PI3K/AKT pathway has been shown to play a critical role on the neuroprotection and inhibiting apoptosis via the enhancing expression of the SODs. This pathway appears to be crucial in Alzheimer’s disease because it is related to the tau protein hyper-phosphorylation. Dietary supplementation of several ordinary compounds may provide a novel therapeutic approach to brain disorders by modulating the function of the PI3K/AKT pathway. Understanding these systems may offer a better efficacy of new therapeutic approaches. In this review, we summarize recent progresses on the involvement of the SODs and PI3K/AKT pathway in neuroprotective signaling against Alzheimer’s disease.
Collapse
Affiliation(s)
- Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Yukie Nakagawa
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Ai Tsuji
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Atsuko Nakanishi
- Department of Food and Nutrition, Faculty of Contemporary Human Life Science, Tezukayama University, Nara 631-8501, Japan.
| | - Toshiyuki Murai
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan.
| |
Collapse
|
39
|
Kumar B, Kumar M, Dwivedi AR, Kumar V. Synthesis, Biological Evaluation and Molecular Modeling Studies of Propargyl-Containing 2,4,6-Trisubstituted Pyrimidine Derivatives as Potential Anti-Parkinson Agents. ChemMedChem 2018. [DOI: 10.1002/cmdc.201700589] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Bhupinder Kumar
- Department of Pharmaceutical Sciences and Natural Products; Central University of Punjab; Mansa Road Bathinda Punjab 151001 India
| | - Mohit Kumar
- Department of Pharmaceutical Sciences and Natural Products; Central University of Punjab; Mansa Road Bathinda Punjab 151001 India
| | - Ashish Ranjan Dwivedi
- Department of Pharmaceutical Sciences and Natural Products; Central University of Punjab; Mansa Road Bathinda Punjab 151001 India
| | - Vinod Kumar
- Department of Pharmaceutical Sciences and Natural Products; Central University of Punjab; Mansa Road Bathinda Punjab 151001 India
| |
Collapse
|
40
|
Stefanachi A, Leonetti F, Pisani L, Catto M, Carotti A. Coumarin: A Natural, Privileged and Versatile Scaffold for Bioactive Compounds. Molecules 2018; 23:E250. [PMID: 29382051 PMCID: PMC6017103 DOI: 10.3390/molecules23020250] [Citation(s) in RCA: 333] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 12/22/2022] Open
Abstract
Many naturally occurring substances, traditionally used in popular medicines around the world, contain the coumarin moiety. Coumarin represents a privileged scaffold for medicinal chemists, because of its peculiar physicochemical features, and the versatile and easy synthetic transformation into a large variety of functionalized coumarins. As a consequence, a huge number of coumarin derivatives have been designed, synthesized, and tested to address many pharmacological targets in a selective way, e.g., selective enzyme inhibitors, and more recently, a number of selected targets (multitarget ligands) involved in multifactorial diseases, such as Alzheimer's and Parkinson's diseases. In this review an overview of the most recent synthetic pathways leading to mono- and polyfunctionalized coumarins will be presented, along with the main biological pathways of their biosynthesis and metabolic transformations. The many existing and recent reviews in the field prompted us to make some drastic selections, and therefore, the review is focused on monoamine oxidase, cholinesterase, and aromatase inhibitors, and on multitarget coumarins acting on selected targets of neurodegenerative diseases.
Collapse
Affiliation(s)
- Angela Stefanachi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Leonardo Pisani
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Marco Catto
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Angelo Carotti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| |
Collapse
|
41
|
Shehzadi SA, Saeed A, Lemière F, Maes BUW, Abbaspour Tehrani K. Zinc(II)-Catalyzed Synthesis of Propargylamines by Coupling Aldimines and Ketimines with Alkynes. European J Org Chem 2018. [DOI: 10.1002/ejoc.201701567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Syeda Aaliya Shehzadi
- Organic Synthesis; Department of Chemistry; University of Antwerp; Groenenborgerlaan 171 2020 Antwerp Belgium
| | - Aamer Saeed
- Department of Chemistry; Quaid-I-Azam University; 45320 Islamabad Pakistan
| | - Filip Lemière
- Biomolecular & Analytical Mass Spectrometry and Center for Proteomics; Department of Chemistry; University of Antwerp; 2020 Antwerp Belgium
| | - Bert U. W. Maes
- Organic Synthesis; Department of Chemistry; University of Antwerp; Groenenborgerlaan 171 2020 Antwerp Belgium
| | - Kourosch Abbaspour Tehrani
- Organic Synthesis; Department of Chemistry; University of Antwerp; Groenenborgerlaan 171 2020 Antwerp Belgium
| |
Collapse
|
42
|
Kumar A, Sharma A. Computational Modeling of Multi-target-Directed Inhibitors Against Alzheimer’s Disease. NEUROMETHODS 2018. [DOI: 10.1007/978-1-4939-7404-7_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
43
|
Marinelli L, Fornasari E, Di Stefano A, Turkez H, Arslan ME, Eusepi P, Ciulla M, Cacciatore I. (R)-α-Lipoyl-Gly-l-Pro-l-Glu dimethyl ester as dual acting agent for the treatment of Alzheimer's disease. Neuropeptides 2017; 66:52-58. [PMID: 28993014 DOI: 10.1016/j.npep.2017.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/05/2017] [Accepted: 09/25/2017] [Indexed: 12/17/2022]
Abstract
In this study, effects of LA-GPE (R-α-Lipoyl-Gly-l-Pro-l-Glu dimethyl ester) and GPE (Gly-L-Pro-L-Glu) on the cytotoxic action of Aβ1-42 were tested with differentiated human neuroblastoma SH-SY5Y cells as cellular Alzheimer model via measurements of mitochondrial viability (MTT assay) and lactate dehydrogenase release (LDH assay). Effects of LA-GPE and GPE on acetylcholinesterase (AChE) activity, total antioxidant capacity (TAC) and total oxidative status (TOS) levels, and neural cell apoptosis and necrosis were also determined. In addition, biological safety of these novel formulations was evaluated in human blood cells using different cytotoxicity and genotoxicity assays. Our results indicated that both compounds could block Aβ1-42 induced cell death. LA-GPE reduced Aβ-induced AChE activity and oxidative stress, suggesting it as a multifunctional compound potentially valuable for the treatment of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Lisa Marinelli
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, Chieti Scalo (CH), Italy
| | - Erika Fornasari
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, Chieti Scalo (CH), Italy
| | - Antonio Di Stefano
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, Chieti Scalo (CH), Italy
| | - Hasan Turkez
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, Chieti Scalo (CH), Italy; Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25240 Erzurum, Turkey
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25240 Erzurum, Turkey
| | - Piera Eusepi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, Chieti Scalo (CH), Italy
| | - Michele Ciulla
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, Chieti Scalo (CH), Italy
| | - Ivana Cacciatore
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, Chieti Scalo (CH), Italy.
| |
Collapse
|
44
|
Shehzadi SA, Saeed A. Cu(I)-catalyzed Green Synthesis of Propargyl Amines Decorated with Carbazole Moiety by A3-Coupling. J CHIN CHEM SOC-TAIP 2017. [DOI: 10.1002/jccs.201700061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
| | - Aamer Saeed
- Department of Chemistry; Quaid-I-Azam University; Islamabad 45320 Pakistan
| |
Collapse
|
45
|
8-Hydroxyquinolines in medicinal chemistry: A structural perspective. Eur J Med Chem 2016; 120:252-74. [DOI: 10.1016/j.ejmech.2016.05.007] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 01/12/2023]
|
46
|
Issa NT, Kruger J, Wathieu H, Raja R, Byers SW, Dakshanamurthy S. DrugGenEx-Net: a novel computational platform for systems pharmacology and gene expression-based drug repurposing. BMC Bioinformatics 2016; 17:202. [PMID: 27151405 PMCID: PMC4857427 DOI: 10.1186/s12859-016-1065-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 04/29/2016] [Indexed: 12/12/2022] Open
Abstract
Background The targeting of disease-related proteins is important for drug discovery, and yet target-based discovery has not been fruitful. Contextualizing overall biological processes is critical to formulating successful drug-disease hypotheses. Network pharmacology helps to overcome target-based bottlenecks through systems biology analytics, such as protein-protein interaction (PPI) networks and pathway regulation. Results We present a systems polypharmacology platform entitled DrugGenEx-Net (DGE-NET). DGE-NET predicts empirical drug-target (DT) interactions, integrates interaction pairs into a multi-tiered network analysis, and ultimately predicts disease-specific drug polypharmacology through systems-based gene expression analysis. Incorporation of established biological network annotations for protein target-disease, −signaling pathway, −molecular function, and protein-protein interactions enhances predicted DT effects on disease pathophysiology. Over 50 drug-disease and 100 drug-pathway predictions are validated. For example, the predicted systems pharmacology of the cholesterol-lowering agent ezetimibe corroborates its potential carcinogenicity. When disease-specific gene expression analysis is integrated, DGE-NET prioritizes known therapeutics/experimental drugs as well as their contra-indications. Proof-of-concept is established for immune-related rheumatoid arthritis and inflammatory bowel disease, as well as neuro-degenerative Alzheimer’s and Parkinson’s diseases. Conclusions DGE-NET is a novel computational method that predicting drug therapeutic and counter-therapeutic indications by uniquely integrating systems pharmacology with gene expression analysis. DGE-NET correctly predicts various drug-disease indications by linking the biological activity of drugs and diseases at multiple tiers of biological action, and is therefore a useful approach to identifying drug candidates for re-purposing. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1065-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naiem T Issa
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA
| | - Jordan Kruger
- Department of Biochemistry & Molecular Biology, Georgetown University, Washington DC, 20057, USA
| | - Henri Wathieu
- Georgetown University Medical Center, Washington DC, 20057, USA
| | - Rajarajan Raja
- George Mason University, 4400 University Dr, Fairfax, VA, 22030, USA
| | - Stephen W Byers
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA.,Department of Biochemistry & Molecular Biology, Georgetown University, Washington DC, 20057, USA
| | - Sivanesan Dakshanamurthy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA. .,Department of Biochemistry & Molecular Biology, Georgetown University, Washington DC, 20057, USA.
| |
Collapse
|
47
|
Propargylamine as functional moiety in the design of multifunctional drugs for neurodegenerative disorders: MAO inhibition and beyond. Future Med Chem 2016; 7:609-29. [PMID: 25921401 DOI: 10.4155/fmc.15.12] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Much progress has been made in designing analogues that can potentially confer neuroprotection against debilitating neurodegenerative disorders, yet the multifactorial pathogenesis of this cluster of diseases remains a stumbling block for the successful design of an 'ultimate' drug. However, with the growing popularity of the "one drug, multiple targets" paradigm, many researchers have successfully synthesized and evaluated drug-like molecules incorporating a propargylamine function that shows potential to serve as multifunctional drugs or multitarget-directed ligands. It is the aim of this review to highlight the reported activities of these propargylamine derivatives and their prospect to serve as drug candidates for the treatment of neurodegenerative disorders.
Collapse
|
48
|
Silva TLD, Rambo RS, Rampon DDS, Radatz CS, Benvenutti EV, Russowsky D, Schneider PH. Covalently immobilized indium(III) composite (In/SiO2) as highly efficient reusable catalyst for A3-coupling of aldehydes, alkynes and amines under solvent-free conditions. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.molcata.2015.01.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
49
|
Fang J, Jiang F, Li J, Zhu Y. Rationale for the use of multifunctional drugs as neuroprotective agents for glaucoma. Neural Regen Res 2015; 7:313-8. [PMID: 25806075 PMCID: PMC4353106 DOI: 10.3969/j.issn.1673-5374.2012.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 11/02/2011] [Indexed: 11/18/2022] Open
Abstract
Glaucoma, the leading cause globally of irreversible blindness, is a neurodegenerative disease characterized by progressive retinal ganglion cell death. To date, no drug has been shown to prevent the retinal ganglion cell loss associated with glaucoma. Multiple mechanisms lead to ganglion cell death in glaucoma, suggesting that a neuroprotectant that has a single mode of action, like memantine, would have a limited positive effect at slowing down ganglion cell death. Conversely, simultaneously targeting several factors may be the best therapeutic approach to improve outcomes. Multifunctional drugs are fast gaining acceptance as a strategy for the treatment of complex disorders of the central nervous system, such as Parkinson's disease, Alzheimer's disease and other progressive neurodegenerative diseases. In this paper, we review the current literature on multifunctional drugs and propose a rationale for the use of multifunctional drugs in glaucomatous optic neuropathy.
Collapse
Affiliation(s)
- Jiahua Fang
- Department of Ophthalmology, Jingzhou First Hospital, Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Fagang Jiang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Jingbo Li
- Department of Ophthalmology, Jingzhou First Hospital, Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Yanhua Zhu
- Department of Ophthalmology, Jingzhou First Hospital, Yangtze University, Jingzhou 434000, Hubei Province, China
| |
Collapse
|
50
|
Song Y, Xu H, Chen W, Zhan P, Liu X. 8-Hydroxyquinoline: a privileged structure with a broad-ranging pharmacological potential. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00284a] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
An overview of the broad-ranging pharmacological applications of 8-HQ derivatives.
Collapse
Affiliation(s)
- Yu'ning Song
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
| | - Hao Xu
- Department of Breast and Thyroid Surgery
- Shandong Provincial Hospital Affiliated to Shandong University
- Jinan
- P. R. China
| | - Wenmin Chen
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
| | - Peng Zhan
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
| | - Xinyong Liu
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
| |
Collapse
|