1
|
Guo J, Zhu Y, Zhi J, Lou Q, Bai R, He Y. Antioxidants in anti-Alzheimer's disease drug discovery. Ageing Res Rev 2025; 107:102707. [PMID: 40021094 DOI: 10.1016/j.arr.2025.102707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Oxidative stress is widely recognized as a key contributor to the pathogenesis of Alzheimer's disease (AD). While not the sole factor, it is closely linked to critical pathological features, such as the formation of senile plaques and neurofibrillary tangles. The development of agents with antioxidant properties has become an area of growing interest in AD research. Between 2015 and 2024, several antioxidant-targeted drugs for AD progressed to clinical trials, with increasing attention to the evaluation of antioxidant properties during their development. Oxidative stress plays a pivotal role in linking various AD hypotheses, underscoring its importance in understanding the disease mechanisms. Despite this, comprehensive reviews addressing advancements in AD drug development from the perspective of antioxidant capacity remain limited, hindering the design of novel compounds. This review aims to explore the mechanistic relationship between oxidative stress and AD, summarize methods for assessing antioxidant capacity, and provide an overview of antioxidant compounds with anti-AD properties reported over the past decade. The goal is to offer strategies for identifying effective antioxidant-based therapies for AD and to deepen our understanding of the role of oxidative stress in AD pathology.
Collapse
Affiliation(s)
- Jianan Guo
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China; Central Laboratory and Precision Medicine Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China; Jinhua Key Laboratory of Cancer Nutrition and Metabolism Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China.
| | - Yalan Zhu
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China
| | - Jia Zhi
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Qiuwen Lou
- Central Laboratory and Precision Medicine Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China; Jinhua Key Laboratory of Cancer Nutrition and Metabolism Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China
| | - Renren Bai
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China.
| | - Yiling He
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province 321000, PR China.
| |
Collapse
|
2
|
Manavi MA, Nourhashemi M, Emami S, Fathian Nasab MH, Dehnavi F, Küçükkılınç TT, Foroumadi A, Sharifzadeh M, Khoobi M. Lipoic acid scaffold applications in the design of multitarget-directed ligands against Alzheimer's disease. Bioorg Chem 2025; 157:108241. [PMID: 39922042 DOI: 10.1016/j.bioorg.2025.108241] [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: 11/03/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Alzheimer's disease (AD) is becoming a fast-growing public health problem which can result in psychological problems as well as loss of speech, language, short-term memory, and motor coordination. Many medications were developed and produced to treat AD, however due to the complexity of the pathology involved in the illness, many of these medications often failed in clinical or preclinical studies. The main issue with the current anti-AD medications is their low efficacy since they use a single target. Multi-target-directed ligands (MTDLs) based on "one molecule; multiple targets" have been introduced to address these two fundamental issues. MTDLs have demonstrated improved efficacy and safety since they regulate many biological targets simultaneously. Alpha-lipoic acid (LA), a natural molecule with distinct properties, is a viable scaffold for developing new MTDLs in treating many neurodegenerative diseases, particularly AD. It is a key mitochondrial enzymes' cofactor and an organic molecule with disulfide functionality. It also has potent antioxidant characteristics that enhance mitochondrial activity. Considering the neuroprotective and anti-inflammatory effects of LA, various hybrids of LA with tacrine, rivastigmine, coumarin and chromone, ibuprofen, melatonin, niacin have been synthesized and biologically evaluated as the MTDLs. In this article, we review the design of LA-based hybrids or conjugates, their biological activities, and structure-activity relationship studies, to develop new MTDLs in the field of AD pharmacotherapy.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Nourhashemi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran; Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Hosein Fathian Nasab
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Dehnavi
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176-14411, Iran
| | - Mehdi Khoobi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Shaaban AE, Ali AR, Ayyad SN, Badria FA. Multi-target directed ligands inspired natural products as an effective approach for the treatment of complex chronic health disorders. Bioorg Chem 2025; 154:108075. [PMID: 39708551 DOI: 10.1016/j.bioorg.2024.108075] [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: 10/01/2024] [Revised: 11/30/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Complex diseases involve multifaceted etiological components, which limit the effectiveness of conventional targeted therapies. Therefore, standard medicinal treatments often face significant challenges and failures when addressing these disease conditions. Furthermore, the growing interest in multidrug resistance (MDR), the occurrence of adverse drug reactions related to use traditional approaches, and the limited clinical efficacy of single-target drug therapy have increased the demand for innovative drug treatments. In this rapidly evolving era, the exploration of multi-target directed ligands (MTDLs) derived from natural products has granted us access to a wide range of compounds with medicinal properties. The allure of these MTDLs lies in their unique ability to minimize side effects from using two medicinal agents, establishing them as the preferred choice for drug developers. MTDLs have been recognized for their extraordinary capacity to collectively hinder multiple pathways implicated in the development of intricate diseases by merging or linking active molecules obtained from these sources. This review delves into promising MTDLs derived from natural products, which modulates diverse biological pathways implicated in complex diseased conditions particularly Alzheimer's disease, diabetes, cardiac disorders and inflammatory conditions.
Collapse
Affiliation(s)
- Amira E Shaaban
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt
| | - Ahmed R Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Seif N Ayyad
- Department of Organic Chemistry, Faculty of Science, Damietta University, New Damietta 34511, Egypt
| | - Farid A Badria
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
4
|
Pravin NJ, Kavalapure RS, Alegaon SG, Gharge S, Ranade SD. Indoles as promising Therapeutics: A review of recent drug discovery efforts. Bioorg Chem 2025; 154:108092. [PMID: 39740309 DOI: 10.1016/j.bioorg.2024.108092] [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: 10/18/2024] [Revised: 12/07/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
Indole, a fundamental heterocyclic core, has emerged as a cornerstone in the medicinal chemistry due to its diverse biological activities and structural versatility. This aromatic compound, present in natural as well as synthetic compounds, offers a versatile platform for the drug discovery. By strategically incorporating functional groups or pharmacophores, researchers can tailor indole-derivatives to target a wide range of diseases. This review delves into the multifaceted applications of indole derivatives, highlighting their potential as therapeutic agents for cancer, diabetes, depression, Alzheimer's diseases, Parkinson's disease, etc. emphasizing how indole derivatives can enhance potency and selectivity. By understanding the structure-activity relationship of indole compounds, scientists can develop innovative drug candidates with improved therapeutic profiles. The review highlights the diverse nature of indole-based derivatives along with the structure-activity relationshipThe current review comprehensively covers the advancements and developments in the field over the past seven years, specifically from 2017 to 2024. This timeframe was selected to provide an up-to-date and thorough analysis of recent progress, capturing significant trends, breakthroughs, and emerging insights within the domain. By focusing on this period, the review ensures relevance and highlights the evolving landscape of research, offering a detailed synthesis of key findings and their implications for future studies.
Collapse
Affiliation(s)
- Naik Jui Pravin
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590 010, Karnataka, India
| | - Rohini S Kavalapure
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590 010, Karnataka, India.
| | - Shankar G Alegaon
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590 010, Karnataka, India
| | - Shankar Gharge
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590 010, Karnataka, India
| | - Shriram D Ranade
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590 010, Karnataka, India
| |
Collapse
|
5
|
Ciaglia T, Miranda MR, Di Micco S, Vietri M, Smaldone G, Musella S, Di Sarno V, Auriemma G, Sardo C, Moltedo O, Pepe G, Bifulco G, Ostacolo C, Campiglia P, Manfra M, Vestuto V, Bertamino A. Neuroprotective Potential of Indole-Based Compounds: A Biochemical Study on Antioxidant Properties and Amyloid Disaggregation in Neuroblastoma Cells. Antioxidants (Basel) 2024; 13:1585. [PMID: 39765912 PMCID: PMC11673510 DOI: 10.3390/antiox13121585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/15/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
Based on the established neuroprotective properties of indole-based compounds and their significant potential as multi-targeted therapeutic agents, a series of synthetic indole-phenolic compounds was evaluated as multifunctional neuroprotectors. Each compound demonstrated metal-chelating properties, particularly in sequestering copper ions, with quantitative analysis revealing approximately 40% chelating activity across all the compounds. In cellular models, these hybrid compounds exhibited strong antioxidant and cytoprotective effects, countering reactive oxygen species (ROS) generated by the Aβ(25-35) peptide and its oxidative byproduct, hydrogen peroxide, as demonstrated by quantitative analysis showing on average a 25% increase in cell viability and a reduction in ROS levels to basal states. Further analysis using thioflavin T fluorescence assays, circular dichroism, and computational studies indicated that the synthesized derivatives effectively promoted the self-disaggregation of the Aβ(25-35) fragment. Taken together, these findings suggest a unique profile of neuroprotective actions for indole-phenolic derivatives, combining chelating, antioxidant, and anti-aggregation properties, which position them as promising compounds for the development of multifunctional agents in Alzheimer's disease therapy. The methods used provide reliable in vitro data, although further in vivo validation and assessment of blood-brain barrier penetration are needed to confirm therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Simone Di Micco
- European Biomedical Research Institute of Salerno (EBRIS), Via Salvatore de Renzi 50, 84125 Salerno, Italy;
| | - Mariapia Vietri
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giulia Auriemma
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carla Sardo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| |
Collapse
|
6
|
Campos-Prieto L, García-Rey A, Sotelo E, Mallo-Abreu A. Multicomponent reactions driving the discovery and optimization of agents targeting central nervous system pathologies. Beilstein J Org Chem 2024; 20:3151-3173. [PMID: 39669443 PMCID: PMC11635293 DOI: 10.3762/bjoc.20.261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024] Open
Abstract
The ongoing quest to discover effective treatments for diseases remains a significant challenge for the scientific community. Multicomponent reactions (MCRs) have emerged as powerful tools in accelerating drug discovery, enabling the rapid generation of chemical libraries with high diversity in a time-efficient and environmentally sustainable manner. In this review, we focus on central nervous system (CNS) disorders, particularly Alzheimer's disease, Parkinson's disease, schizophrenia, depression, and epilepsy, where MCRs have contributed to the development of promising ligands in recent years. Rather than providing an exhaustive overview, this review aims to highlight key studies that address major CNS pathologies, relevant drug targets, and various MCR approaches. We have carefully selected representative articles and apologize to the authors whose important contributions may not be included. By concentrating on these pivotal studies, we strive to offer a clear and concise perspective on current research trends and breakthroughs in this field.
Collapse
Affiliation(s)
- Lucía Campos-Prieto
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Aitor García-Rey
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eddy Sotelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ana Mallo-Abreu
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Av. Diagonal 643, E-08028 Barcelona, Spain
| |
Collapse
|
7
|
Galvani F, Cammarota M, Vacondio F, Rivara S, Boscia F. Protective Activity of Melatonin Combinations and Melatonin-Based Hybrid Molecules in Neurodegenerative Diseases. J Pineal Res 2024; 76:e70008. [PMID: 39582467 PMCID: PMC11586835 DOI: 10.1111/jpi.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
The identification of protective agents for the treatment of neurodegenerative diseases is the mainstay therapeutic goal to modify the disease course and arrest the irreversible disability progression. Pharmacological therapies synergistically targeting multiple pathogenic pathways, including oxidative stress, mitochondrial dysfunction, and inflammation, are prime candidates for neuroprotection. Combination or synergistic therapy with melatonin, whose decline correlates with altered sleep/wake cycle and impaired glymphatic "waste clearance" system in neurodegenerative diseases, has a great therapeutic potential to treat inflammatory neurodegenerative states. Despite the protective outcomes observed in preclinical studies, mild or poor outcomes were observed in clinical settings, suggesting that melatonin combinations promoting synergistic actions at appropriate doses might be more suitable to treat multifactorial neurodegenerative disorders. In this review, we first summarize the key melatonin actions and pathways contributing to cell protection and its therapeutic implication in Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We remark the major controversies in the field, mostly generated by the lack of a common consensus for the optimal dosing, molecular targets, and toxicity. Then, we review the literature investigating the efficacy of melatonin combinations with approved or investigational neuroprotective agents and of melatonin-containing hybrid molecules, both in vitro and in animal models of AD, PD, and MS, as well as the efficacy of add-on melatonin in clinical settings. We highlight the rationale for such melatonin combinations with a focus on the comparison with single-agent treatment and on the assays in which an additive or a synergistic effect has been achieved. We conclude that a better characterization of the mechanisms underlying such melatonin synergistic actions under neuroinflammation at appropriate doses needs to be tackled to advance successful clinical translation of neuroprotective melatonin combination therapies or melatonin-based hybrid molecules.
Collapse
Affiliation(s)
| | - Mariarosaria Cammarota
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of MedicineFederico II University of NaplesNaplesItaly
| | | | - Silvia Rivara
- Department of Food and DrugUniversity of ParmaParmaItaly
| | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of MedicineFederico II University of NaplesNaplesItaly
| |
Collapse
|
8
|
Mayo P, Pascual J, Crisman E, Domínguez C, López MG, León R. Innovative pathological network-based multitarget approaches for Alzheimer's disease treatment. Med Res Rev 2024; 44:2367-2419. [PMID: 38678582 DOI: 10.1002/med.22045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/02/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is a major health threat globally. Its prevalence is forecasted to exponentially increase during the next 30 years due to the global aging population. Currently, approved drugs are merely symptomatic, being ineffective in delaying or blocking the relentless disease advance. Intensive AD research describes this disease as a highly complex multifactorial disease. Disclosure of novel pathological pathways and their interconnections has had a major impact on medicinal chemistry drug development for AD over the last two decades. The complex network of pathological events involved in the onset of the disease has prompted the development of multitarget drugs. These chemical entities combine pharmacological activities toward two or more drug targets of interest. These multitarget-directed ligands are proposed to modify different nodes in the pathological network aiming to delay or even stop disease progression. Here, we review the multitarget drug development strategy for AD during the last decade.
Collapse
Affiliation(s)
- Paloma Mayo
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Jorge Pascual
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Cristina Domínguez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| |
Collapse
|
9
|
Malek R, Sałat K, Totoson P, Karcz T, Refouvelet B, Skrzypczak-Wiercioch A, Maj M, Simakov A, Martin H, Siwek A, Szałaj N, Godyń J, Panek D, Więckowska A, Jozwiak K, Demougeot C, Kieć-Kononowicz K, Chabchoub F, Iriepa I, Marco-Contelles J, Ismaili L. Discovery of New Highly Potent Histamine H 3 Receptor Antagonists, Calcium Channel Blockers, and Acetylcholinesterase Inhibitors. ACS Chem Neurosci 2024; 15:3363-3383. [PMID: 39208251 DOI: 10.1021/acschemneuro.4c00341] [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] [Indexed: 09/04/2024] Open
Abstract
At present, one of the most promising strategies to tackle the complex challenges posed by Alzheimer's disease (AD) involves the development of novel multitarget-directed ligands (MTDLs). To this end, we designed and synthesized nine new MTDLs using a straightforward and cost-efficient one-pot Biginelli three-component reaction. Among these newly developed compounds, one particular small molecule, named 3e has emerged as a promising MTDL. This compound effectively targets critical biological factors associated with AD, including the simultaneous inhibition of cholinesterases (ChEs), selective antagonism of H3 receptors, and blocking voltage-gated calcium channels. Additionally, compound 3e exhibited remarkable neuroprotective activity against H2O2 and Aβ1-40, and effectively restored cognitive function in AD mice treated with scopolamine in the novel object recognition task, confirming that this compound could provide a novel and innovative therapeutic approach for the effective treatment of AD.
Collapse
Affiliation(s)
- Rim Malek
- Université de Franche-Comté, INSERM, UMR 1322 LINC, F-25000 Besançon, France
- Laboratory of Applied Chemistry: Heterocycles, Lipids and Polymers, Faculty of Sciences of Sfax, University of Sfax, B. P 802, Sfax 3000, Tunisia
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków 30-688, Poland
| | - Perle Totoson
- Université de Franche-Comté, EFS, INSERM, UMR 1098 RIGHT, F-25000 Besançon, France
| | - Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Bernard Refouvelet
- Université de Franche-Comté, INSERM, UMR 1322 LINC, F-25000 Besançon, France
| | - Anna Skrzypczak-Wiercioch
- University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, 24/28 Mickiewicz St., Kraków 30-059, Poland
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, Lublin 20-093, Poland
| | - Alexey Simakov
- Université de Franche-Comté, EFS, INSERM, UMR 1098 RIGHT, F-25000 Besançon, France
| | - Helene Martin
- Université de Franche-Comté, EFS, INSERM, UMR 1098 RIGHT, F-25000 Besançon, France
| | - Agata Siwek
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Krzysztof Jozwiak
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, Lublin 20-093, Poland
| | - Celine Demougeot
- Université de Franche-Comté, EFS, INSERM, UMR 1098 RIGHT, F-25000 Besançon, France
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Fakher Chabchoub
- Laboratory of Applied Chemistry: Heterocycles, Lipids and Polymers, Faculty of Sciences of Sfax, University of Sfax, B. P 802, Sfax 3000, Tunisia
| | - Isabel Iriepa
- Universidad de Alcalá. Departamento de Química Orgánica y Química Inorgánica, Alcalá de Henares, Madrid 28805, Spain
- Instituto de Investigación Química Andrés M. del Río (IQAR), Universidad de Alcalá, Alcalá de Henares, Madrid 28805, Spain, Grupo DISCOBAC, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM)
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC), C/ Juan de la Cierva 3, Madrid 28006, Spain
- CIBER, ISCIII, Center for Biomedical Network Research on Rare Diseases (CIBERER), Madrid 28006, Spain
| | - Lhassane Ismaili
- Université de Franche-Comté, INSERM, UMR 1322 LINC, F-25000 Besançon, France
| |
Collapse
|
10
|
Singh G, Kumar S, Panda SR, Kumar P, Rai S, Verma H, Singh YP, Kumar S, Srikrishna S, Naidu VGM, Modi G. Design, Synthesis, and Biological Evaluation of Ferulic Acid-Piperazine Derivatives Targeting Pathological Hallmarks of Alzheimer's Disease. ACS Chem Neurosci 2024; 15:2756-2778. [PMID: 39076038 DOI: 10.1021/acschemneuro.4c00130] [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] [Indexed: 07/31/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia and is characterized by low levels of acetyl and butyrylcholine, increased oxidative stress, inflammation, accumulation of metals, and aggregations of Aβ and tau proteins. Current treatments for AD provide only symptomatic relief without impacting the pathological hallmarks of the disease. In our ongoing efforts to develop naturally inspired novel multitarget molecules for AD, through extensive medicinal chemistry efforts, we have developed 13a, harboring the key functional groups to provide not only symptomatic relief but also targeting oxidative stress, able to chelate iron, inhibiting NLRP3, and Aβ1-42 aggregation in various AD models. 13a exhibited promising anticholinesterase activity against AChE (IC50 = 0.59 ± 0.19 μM) and BChE (IC50 = 5.02 ± 0.14 μM) with excellent antioxidant properties in DPPH assay (IC50 = 5.88 ± 0.21 μM) over ferulic acid (56.49 ± 0.62 μM). The molecular docking and dynamic simulations further corroborated the enzyme inhibition studies and confirmed the stability of these complexes. Importantly, in the PAMPA-BBB assay, 13a turned out to be a promising molecule that can efficiently cross the blood-brain barrier. Notably, 13a also exhibited iron-chelating properties. Furthermore, 13a effectively inhibited self- and metal-induced Aβ1-42 aggregation. It is worth mentioning that 13a demonstrated no symptom of cytotoxicity up to 30 μM concentration in PC-12 cells. Additionally, 13a inhibited the NLRP3 inflammasome and mitigated mitochondrial-induced reactive oxygen species and mitochondrial membrane potential damage triggered by LPS and ATP in HMC-3 cells. 13a could effectively reduce mitochondrial and cellular reactive oxygen species (ROS) in the Drosophila model of AD. Finally, 13a was found to be efficacious in reversing memory impairment in a scopolamine-induced AD mouse model in the in vivo studies. In ex vivo assessments, 13a notably modulates the levels of superoxide, catalase, and malondialdehyde along with AChE and BChE. These findings revealed that 13a holds promise as a potential candidate for further development in AD management.
Collapse
Affiliation(s)
- Gourav Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Rm # 123, Varanasi 221005, India
| | - Sunil Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Rm # 123, Varanasi 221005, India
| | - Samir Ranjan Panda
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781032, India
| | - Prabhat Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sanskriti Rai
- Department of Biochemistry, Institute of Sciences, Banaras Hindu University, Varanasi 201005, India
| | - Himanshu Verma
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Rm # 123, Varanasi 221005, India
| | - Yash Pal Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Rm # 123, Varanasi 221005, India
| | - Saroj Kumar
- Department of Biochemistry, Institute of Sciences, Banaras Hindu University, Varanasi 201005, India
| | - Saripella Srikrishna
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781032, India
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Rm # 123, Varanasi 221005, India
| |
Collapse
|
11
|
Bon L, Banaś A, Dias I, Melo-Marques I, Cardoso SM, Chaves S, Santos MA. New Multitarget Rivastigmine-Indole Hybrids as Potential Drug Candidates for Alzheimer's Disease. Pharmaceutics 2024; 16:281. [PMID: 38399339 PMCID: PMC10892719 DOI: 10.3390/pharmaceutics16020281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia with no cure so far, probably due to the complexity of this multifactorial disease with diverse processes associated with its origin and progress. Several neuropathological hallmarks have been identified that encourage the search for new multitarget drugs. Therefore, following a multitarget approach, nine rivastigmine-indole (RIV-IND) hybrids (5a1-3, 5b1-3, 5c1-3) were designed, synthesized and evaluated for their multiple biological properties and free radical scavenging activity, as potential multitarget anti-AD drugs. The molecular docking studies of these hybrids on the active center of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) suggest their capacity to act as dual enzyme inhibitors with probable greater disease-modifying impact relative to AChE-selective FDA-approved drugs. Compounds 5a3 (IC50 = 10.9 µM) and 5c3 (IC50 = 26.8 µM) revealed higher AChE inhibition than the parent RIV drug. Radical scavenging assays demonstrated that all the hybrids containing a hydroxyl substituent in the IND moiety (5a2-3, 5b2-3, 5c2-3) have good antioxidant activity (EC50 7.8-20.7 µM). The most effective inhibitors of Aβ42 self-aggregation are 5a3, 5b3 and 5c3 (47.8-55.5%), and compounds 5b2 and 5c2 can prevent the toxicity induced by Aβ1-42 to cells. The in silico evaluation of the drug-likeness of the hybrids also showed that all the compounds seem to have potential oral availability. Overall, within this class of RIV-IND hybrids, 5a3 and 5c3 appear as lead compounds for anti-AD drug candidates, deserving further investigation.
Collapse
Affiliation(s)
- Leo Bon
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (L.B.); (A.B.); (I.D.)
| | - Angelika Banaś
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (L.B.); (A.B.); (I.D.)
| | - Inês Dias
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (L.B.); (A.B.); (I.D.)
| | - Inês Melo-Marques
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (I.M.-M.); (S.M.C.)
| | - Sandra M. Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (I.M.-M.); (S.M.C.)
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sílvia Chaves
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (L.B.); (A.B.); (I.D.)
| | - M. Amélia Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal; (L.B.); (A.B.); (I.D.)
| |
Collapse
|
12
|
Egbujor MC. Sulfonamide Derivatives: Recent Compounds with Potent Anti-alzheimer's Disease Activity. Cent Nerv Syst Agents Med Chem 2024; 24:82-104. [PMID: 38275073 DOI: 10.2174/0118715249278489231128042135] [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: 09/14/2023] [Revised: 10/27/2023] [Accepted: 11/06/2023] [Indexed: 01/27/2024]
Abstract
Facile synthetic procedures and broad spectrum of biological activities are special attributes of sulfonamides. Sulfonamide derivatives have demonstrated potential as a class of compounds for the treatment of Alzheimer's disease (AD). Recent sulfonamide derivatives have been reported as prospective anti-AD agents, with a focus on analogues that significantly inhibit the function of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) enzymes and exhibit remarkable antioxidant and anti-inflammatory properties, all of which are critical for the treatment of AD. Sulfonamide- mediated activation of nuclear factor erythroid 2-related factor 2 (NRF2), a key regulator of the endogenous antioxidant response, has also been suggested as a potential therapeutic approach in AD. Additionally, it has been discovered that a number of sulfonamide derivatives show selectivity for the β- and γ-secretase enzymes and a significant reduction of amyloid B (Aβ) aggregation, which have been implicated in AD. The comparative molecular docking of benzenesulfonamide and donepezil, an AD reference drug showed comparable anti-AD activities. These suggest that sulfonamide derivatives may represent a new class of drugs for the treatment of AD. Thus, the current review will focus on recent studies on the chemical synthesis and evaluation of the anti-AD properties, molecular docking, pharmacological profile, and structure-activity relationship (SAR) of sulfonamide derivatives, as well as their potential anti-AD mechanisms of action. This paper offers a thorough assessment of the state of the art in this field of study and emphasizes the potential of sulfonamide derivatives synthesized during the 2012-2023 period as a new class of compounds for the treatment of AD.
Collapse
|
13
|
Wei W, Jing L, Tian Y, Więckowska A, Kang D, Meng B, Panek D, Godyń J, Góral I, Song Y, Liu X, Zhan P. Multifunctional agents against Alzheimer's disease based on oxidative stress: Polysubstituted pyrazine derivatives synthesized by multicomponent reactions. Bioorg Med Chem 2023; 96:117535. [PMID: 37956505 DOI: 10.1016/j.bmc.2023.117535] [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: 09/12/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
As Alzheimer's disease (AD) is a neurodegenerative disease with a complex pathogenesis, the exploration of multi-target drugs may be an effective strategy for AD treatment. Multifunctional small molecular agents can be obtained by connecting two or more active drugs or privileged pharmacophores by multicomponent reactions (MCRs). In this paper, two series of polysubstituted pyrazine derivatives with multifunctional moieties were designed as anti-AD agents and synthesized by Passerini-3CR and Ugi-4CR. Since the oxidative stress plays an important role in the pathological process of AD, the antioxidant activities of the newly synthesized compounds were first evaluated. Subsequently, selected active compounds were further screened in a series of AD-related bioassays, including Aβ1-42 self-aggregation and deaggregation, BACE-1 inhibition, metal chelation, and protection of SH-SY5Y cells from H2O2-induced oxidative damage. Compound A3B3C1 represented the best one with multifunctional potencies. Mechanism study showed that A3B3C1 acted on Nrf2/ARE signaling pathway, thus increasing the expression of related antioxidant proteins NQO1 and HO-1 to normal cell level. Furthermore, A3B3C1 showed good in vitro human plasma and liver microsome stability, indicating a potential for further development as multifunctional anti-AD agent.
Collapse
Affiliation(s)
- Wenxiu Wei
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Lanlan Jing
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Ye Tian
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China; Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| | - Anna Więckowska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Bairu Meng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Dawid Panek
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Justyna Godyń
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Izabella Góral
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Yuning Song
- Department of Clinical Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong 250012, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
14
|
Singh YP, Kumar H. Tryptamine: A privileged scaffold for the management of Alzheimer's disease. Drug Dev Res 2023; 84:1578-1594. [PMID: 37675624 DOI: 10.1002/ddr.22111] [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: 04/10/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Alzheimer's disease (AD) is a chronic and irreversible neurodegenerative disease associated with aging. It is characterized by the progressive loss of memory and other cognitive functions. Although the exact etiology of AD is not well explored, several factors, such as the deposition of amyloid-β (Aβ) plaques, hyperphosphorylation of tau protein, presence of low levels of acetylcholine, and generation of oxidative stress, are key mediators in the progression of AD. Currently, the clinical treatment options for AD are limited and are based on cholinesterase (ChE) inhibitors (e.g., donepezil, rivastigmine, and galantamine), N-methyl- d-aspartic acid receptor antagonists (e.g., memantine), and the recently approved Aβ modulator (e.g., aducanumab). Tryptamine (2-(1H-indol-3-yl)ethan-1-amine) is a small molecule that contains an indole nucleus and an ethylamine side chain. It is also the active metabolite of tryptophan. It possesses a wide range of biological activities related to neurodegenerative disorders, such as ChE inhibition, Aβ aggregation inhibition, antioxidant effects, monoamine-oxidase inhibition, and neuroprotection. Several tryptamine-based hybrid analogs are currently being investigated as multifunctional agents for the development of novel hybrids for AD treatment. Thus, this review article aims to provide in-depth insights into the research progress and strategies for designing multifunctional agents used in Alzheimer's therapy.
Collapse
Affiliation(s)
- Yash P Singh
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Harish Kumar
- Government College of Pharmacy, Shimla, Himachal Pradesh, India
- Department of Technical Education Vocational and Industrial Training, Sunder Nagar, Himachal Pradesh, India
| |
Collapse
|
15
|
Dakhlaoui I, Bernard PJ, Pietrzak D, Simakov A, Maj M, Refouvelet B, Béduneau A, Cornu R, Jozwiak K, Chabchoub F, Iriepa I, Martin H, Marco-Contelles J, Ismaili L. Exploring the Potential of Sulfonamide-Dihydropyridine Hybrids as Multitargeted Ligands for Alzheimer's Disease Treatment. Int J Mol Sci 2023; 24:ijms24119742. [PMID: 37298693 DOI: 10.3390/ijms24119742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease that has a heavy social and economic impact on all societies and for which there is still no cure. Multitarget-directed ligands (MTDLs) seem to be a promising therapeutic strategy for finding an effective treatment for this disease. For this purpose, new MTDLs were designed and synthesized in three steps by simple and cost-efficient procedures targeting calcium channel blockade, cholinesterase inhibition, and antioxidant activity. The biological and physicochemical results collected in this study allowed us the identification two sulfonamide-dihydropyridine hybrids showing simultaneous cholinesterase inhibition, calcium channel blockade, antioxidant capacity and Nrf2-ARE activating effect, that deserve to be further investigated for AD therapy.
Collapse
Affiliation(s)
- Imen Dakhlaoui
- Laboratoire LINC UR 481, Pôle de Chimie Médicinale, Université de Franche-Comté, F-25000 Besançon, France
- Laboratory of Applied Chemistry, Heterocycles, Lipids and Polymers, Faculty of Sciences of Sfax, University of Sfax, B. P 802, Sfax 3000, Tunisia
| | - Paul J Bernard
- Laboratoire LINC UR 481, Pôle de Chimie Médicinale, Université de Franche-Comté, F-25000 Besançon, France
| | - Diana Pietrzak
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland
| | - Alexey Simakov
- PEPITE EA4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland
| | - Bernard Refouvelet
- Laboratoire LINC UR 481, Pôle de Chimie Médicinale, Université de Franche-Comté, F-25000 Besançon, France
| | - Arnaud Béduneau
- PEPITE EA4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Raphaël Cornu
- PEPITE EA4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Krzysztof Jozwiak
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland
| | - Fakher Chabchoub
- Laboratory of Applied Chemistry, Heterocycles, Lipids and Polymers, Faculty of Sciences of Sfax, University of Sfax, B. P 802, Sfax 3000, Tunisia
| | - Isabel Iriepa
- Department of Organic Chemistry and Inorganic Chemistry, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33,6, 28871 Alcalá de Henares, Spain
| | - Helene Martin
- PEPITE EA4267, Université de Franche-Comté, F-25000 Besançon, France
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Lhassane Ismaili
- Laboratoire LINC UR 481, Pôle de Chimie Médicinale, Université de Franche-Comté, F-25000 Besançon, France
| |
Collapse
|
16
|
Milanesi LH, Rossato DR, Rosa JLO, D'avila LF, Metz VG, Rampelotto CR, Pereira VG, Schaffazick SR, de Bona da Silva C, Burger ME. Ferulic acid-loaded nanostructure prevents morphine reinstatement: the involvement of dopamine system, NRF2, and ΔFosB in the striatum brain area of rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023:10.1007/s00210-023-02420-w. [PMID: 36790483 DOI: 10.1007/s00210-023-02420-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
Morphine is among the most powerful analgesics and pain-relieving agents. However, its addictive properties limit their medical use because patients may be susceptible to abuse and reinstatement. Morphine addiction occurs because of dopamine release in the mesolimbic brain area, implying in an increase in oxidative stress. Ferulic acid (FA), a phenolic phytochemical found in a variety of foods, has been reported to exert antioxidant and neuroprotective effects; however, its low bioavailability makes its nano-encapsulated form a promising alternative. This study aimed to evaluate the protective effects of a novel nanosystem with FA on morphine reinstatement and the consequent molecular neuroadaptations and oxidative status in the mesolimbic region. Rats previously exposed to morphine in conditioned place preference (CPP) paradigm were treated with ferulic acid-loaded nanocapsules (FA-Nc) or nonencapsulated FA during morphine-preference extinction. Following the treatments, animals were re-exposed to morphine to induce the reinstatement. While morphine-preference extinction was comparable among all experimental groups, FA-Nc treatment prevented morphine reinstatement. In the dorsal striatum, while morphine exposure increased lipid peroxidation (LP) and reactive species (RS), FA-Nc decreased LP and FA decreased RS levels. Morphine exposure increased the dopaminergic markers (D1R, D3R, DAT) and ΔFosB immunoreactivity in the ventral striatum; however, FA-Nc treatment decreased D1R, D3R, and ΔFosB and increased D2R, DAT, and NRF2. In conclusion, FA-Nc treatment prevented the morphine reinstatement, promoted antioxidant activity, and modified the dopaminergic neurotransmission, NRF2, and ΔFosB, what may indicate a neuroprotective and antioxidant role of this nanoformulation.
Collapse
Affiliation(s)
- Laura Hautrive Milanesi
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | | | - Jéssica Leandra Oliveira Rosa
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | - Lívia Ferraz D'avila
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | - Vinícia Garzella Metz
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | - Camila Reck Rampelotto
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Av, Roraima 1000, Santa Maria, RS, Brazil
| | | | | | - Cristiane de Bona da Silva
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Av, Roraima 1000, Santa Maria, RS, Brazil
| | - Marilise E Burger
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil. .,Departamento de Fisiologia E Farmacologia, UFSM, Santa Maria, RS, 97105-900, Brazil. .,Graduation Program of Pharmacology, Physiology and Pharmacology Department, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
17
|
Chen LL, Fan YG, Zhao LX, Zhang Q, Wang ZY. The metal ion hypothesis of Alzheimer's disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem 2023; 131:106301. [PMID: 36455485 DOI: 10.1016/j.bioorg.2022.106301] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/13/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD), characterized by the β-amyloid protein (Aβ) deposition and tau hyperphosphorylation, is the most common dementia with uncertain etiology. The clinical trials of Aβ monoclonal antibody drugs have almost failed, giving rise to great attention on the other etiologic hypothesis regarding AD such as metal ions dysmetabolism and chronic neuroinflammation. Mounting evidence revealed that the metal ions (iron, copper, and zinc) were dysregulated in the susceptible brain regions of AD patients, which was highly associated with Aβ deposition, tau hyperphosphorylation, neuronal loss, as well as neuroinflammation. Further studies uncovered that iron, copper and zinc could not only enhance the production of Aβ but also directly bind to Aβ and tau to promote their aggregations. In addition, the accumulation of iron and copper could respectively promote ferroptosis and cuproptosis. Therefore, the metal ion chelators were recognized as promising agents for treating AD. This review comprehensively summarized the effects of metal ions on the Aβ dynamics and tau phosphorylation in the progression of AD. Furthermore, taking chronic neuroinflammation contributes to the progression of AD, we also provided a summary of the mechanisms concerning metal ions on neuroinflammation and highlighted the metal ion chelators may be potential agents to alleviate neuroinflammation under the condition of AD. Nevertheless, more investigations regarding metal ions on neuroinflammation should be taken into practice, and the effects of metal ion chelators on neuroinflammation should gain more attention. Running title: Metal chelators against neuroinflammation.
Collapse
Affiliation(s)
- Li-Lin Chen
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Qi Zhang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
18
|
Deng H, Xu Q, Guo HY, Huang X, Chen F, Jin L, Quan ZS, Shen QK. Application of cinnamic acid in the structural modification of natural products: A review. PHYTOCHEMISTRY 2023; 206:113532. [PMID: 36470328 DOI: 10.1016/j.phytochem.2022.113532] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/24/2022] [Accepted: 11/26/2022] [Indexed: 06/17/2023]
Abstract
Natural products can generally exhibit a variety of biological activities, but most show mediocre performance in preliminary activity evaluation. Natural products often require structural modification to obtain promising lead compounds. Cinnamic acid (CA) is readily available and has diverse biological activities and low cytotoxicity. Introducing CA into natural products may improve their performance, enhance biological activity, and reduce toxic side effect. Herein, we aimed to discuss related applications of CA in the structural modification of natural products and provide a theoretical basis for future derivatization and drug development of natural products. Published articles, web databases (PubMed, Science Direct, SCI Finder, and CNKI), and clinical trial websites (https://clinicaltrials.gov/) related to natural products and CA derivatives were included in the discussion. Based on the inclusion criteria, 128 studies were selected and discussed herein. Screening natural products of CA derivatives allowed for classification by their biological activities. The full text is organized according to the biological activities of the derivatives, with the following categories: anti-tumor, neuroprotective, anti-diabetic, anti-microbial, anti-parasitic, anti-oxidative, anti-inflammatory, and other activities. The biological activity of each CA derivative is discussed in detail. Notably, most derivatives exhibited enhanced biological activity and reduced cytotoxicity compared with the lead compound. CA has various advantages and can be widely used in the synthesis of natural product derivatives to enhance the properties of drug candidates or lead compounds.
Collapse
Affiliation(s)
- Hao Deng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Qian Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Xing Huang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Fener Chen
- Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai, 200433, China
| | - Lili Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
19
|
Du Z, Liu C, Liu Z, Song H, Scott P, Du X, Ren J, Qu X. In vivo visualization of enantioselective targeting of amyloid and improvement of cognitive function by clickable chiral metallohelices. Chem Sci 2023; 14:506-513. [PMID: 36741518 PMCID: PMC9847640 DOI: 10.1039/d2sc05897a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/26/2022] [Indexed: 11/30/2022] Open
Abstract
The pathogenesis of Alzheimer's disease (AD) is closely related to several contributing factors, especially amyloid-β (Aβ) aggregation. Bioorthogonal reactions provide a general, facile, and robust route for the localization and derivatization of Aβ-targeted agents. Herein, a pair of chiral alkyne-containing metallohelices (ΛA and ΔA) were demonstrated to enantioselectively target and modulate Aβ aggregation, which has been monitored in triple-transgenic AD model mice and proved to improve cognitive function. Compared with its enantiomer ΔA, ΛA performed better in blocking Aβ fibrillation, relieving Aβ-triggered toxicity, and recovering memory deficits in vivo. Moreover, clickable ΛA could act as a functional module for subsequent visualization and versatile modification of amyloid via bioorthogonal reaction. As a proof-of-concept, thioflavin T, tacrine, and magnetic nanoparticles were conjugated with ΛA to realize Aβ photo-oxygenation, acetylcholinesterase inhibition, and Aβ clearance, respectively. This proof-of-principle work provided new insights into the biolabeling and bioconjugation of multifunctional metallosupramolecules through click reactions for AD therapy.
Collapse
Affiliation(s)
- Zhi Du
- Laboratory of Chemical Biology, State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Chun Liu
- Laboratory of Chemical Biology, State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230029 China
| | - Zhenqi Liu
- Laboratory of Chemical Biology, State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230029 China
| | - Hualong Song
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Peter Scott
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Xiubo Du
- College of Life Sciences and Oceanography, Shenzhen University Shenzhen 518060 P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology, State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230029 China
| | - Xiaogang Qu
- Laboratory of Chemical Biology, State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230029 China
| |
Collapse
|
20
|
Bubley A, Erofeev A, Gorelkin P, Beloglazkina E, Majouga A, Krasnovskaya O. Tacrine-Based Hybrids: Past, Present, and Future. Int J Mol Sci 2023; 24:ijms24021717. [PMID: 36675233 PMCID: PMC9863713 DOI: 10.3390/ijms24021717] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder which is characterized by β-amyloid (Aβ) aggregation, τ-hyperphosphorylation, and loss of cholinergic neurons. The other important hallmarks of AD are oxidative stress, metal dyshomeostasis, inflammation, and cell cycle dysregulation. Multiple therapeutic targets may be proposed for the development of anti-AD drugs, and the "one drug-multiple targets" strategy is of current interest. Tacrine (THA) was the first clinically approved cholinesterase (ChE) inhibitor, which was withdrawn due to high hepatotoxicity. However, its high potency in ChE inhibition, low molecular weight, and simple structure make THA a promising scaffold for developing multi-target agents. In this review, we summarized THA-based hybrids published from 2006 to 2022, thus providing an overview of strategies that have been used in drug design and approaches that have resulted in significant cognitive improvements and reduced hepatotoxicity.
Collapse
Affiliation(s)
- Anna Bubley
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
| | - Alexaner Erofeev
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Peter Gorelkin
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Elena Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
| | - Alexander Majouga
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Olga Krasnovskaya
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
- Correspondence:
| |
Collapse
|
21
|
Current Pharmacotherapy and Multi-Target Approaches for Alzheimer's Disease. Pharmaceuticals (Basel) 2022; 15:ph15121560. [PMID: 36559010 PMCID: PMC9781592 DOI: 10.3390/ph15121560] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by decreased synaptic transmission and cerebral atrophy with appearance of amyloid plaques and neurofibrillary tangles. Cognitive, functional, and behavioral alterations are commonly associated with the disease. Different pathophysiological pathways of AD have been proposed, some of which interact and influence one another. Current treatment for AD mainly involves the use of therapeutic agents to alleviate the symptoms in AD patients. The conventional single-target treatment approaches do not often cause the desired effect in the disease due to its multifactorial origin. Thus, multi-target strategies have since been undertaken, which aim to simultaneously target multiple targets involved in the development of AD. In this review, we provide an overview of the pathogenesis of AD and the current drug therapies for the disease. Additionally, rationales of the multi-target approaches and examples of multi-target drugs with pharmacological actions against AD are also discussed.
Collapse
|
22
|
Ramesh M, Govindaraju T. Multipronged diagnostic and therapeutic strategies for Alzheimer's disease. Chem Sci 2022; 13:13657-13689. [PMID: 36544728 PMCID: PMC9710308 DOI: 10.1039/d2sc03932j] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and a major contributor to dementia cases worldwide. AD is clinically characterized by learning, memory, and cognitive deficits. The accumulation of extracellular amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) of tau are the pathological hallmarks of AD and are explored as targets for clinical diagnosis and therapy. AD pathology is poorly understood and there are no fully approved diagnosis and treatments. Notwithstanding the gap, decades of research in understanding disease mechanisms have revealed the multifactorial nature of AD. As a result, multipronged and holistic approaches are pertinent to targeting multiple biomarkers and targets for developing effective diagnosis and therapeutics. In this perspective, recent developments in Aβ and tau targeted diagnostic and therapeutic tools are discussed. Novel indirect, combination, and circulating biomarkers as potential diagnostic targets are highlighted. We underline the importance of multiplexing and multimodal detection of multiple biomarkers to generate biomarker fingerprints as a reliable diagnostic strategy. The classical therapeutics targeting Aβ and tau aggregation pathways are described with bottlenecks in the strategy. Drug discovery efforts targeting multifaceted toxicity involving protein aggregation, metal toxicity, oxidative stress, mitochondrial damage, and neuroinflammation are highlighted. Recent efforts focused on multipronged strategies to rationally design multifunctional modulators targeting multiple pathological factors are presented as future drug development strategies to discover potential therapeutics for AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| |
Collapse
|
23
|
Pasquereau S, Galais M, Bellefroid M, Pachón Angona I, Morot-Bizot S, Ismaili L, Van Lint C, Herbein G. Ferulic acid derivatives block coronaviruses HCoV-229E and SARS-CoV-2 replication in vitro. Sci Rep 2022; 12:20309. [PMID: 36434137 PMCID: PMC9700709 DOI: 10.1038/s41598-022-24682-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 11/18/2022] [Indexed: 11/27/2022] Open
Abstract
A novel coronavirus, SARS-CoV-2, emerged in China at the end of 2019 causing a large global outbreak. As treatments are of the utmost importance, drugs with broad anti-coronavirus activity embody a rich and rapid drug discovery landscape, where candidate drug compounds could be identified and optimized. To this end, we tested ten small-molecules with chemical structures close to ferulic acid derivatives (FADs) (n = 8), caffeic acid derivatives (CAFDs) (n = 1) and carboxamide derivatives (CAMDs) (n = 1) for their ability to reduce HCoV-229E replication, another member of the coronavirus family. Among these ten drugs tested, five of them namely MBA112, MBA33, MBA27-1, OS4-1 and MBA108-1 were highly cytotoxic and did not warrant further testing. In contrast, we observed a moderate cytotoxicity for two of them, MBA152 and 5c. Three drugs, namely MBA140, LIJ2P40, and MBA28 showed lower cytotoxicity. These candidates were then tested for their antiviral propreties against HCoV-229E and SARS-CoV2 replication. We first observed encouraging results in HCoV-229E. We then measured a reduction of the viral SARS-CoV2 replication by 46% with MBA28 (EC50 > 200 µM), by 58% with MBA140 (EC50 = 176 µM), and by 82% with LIJ2P40 (EC50 = 66.5 µM). Overall, the FAD LIJ2P40 showed a reduction of the viral titer on SARS-CoV-2 up to two logs with moderate cytotoxicity which opens the door to further evaluation to fight Covid-19.
Collapse
Affiliation(s)
- Sébastien Pasquereau
- grid.7459.f0000 0001 2188 3779Pathogens and Inflammation/EPILAB Laboratory, EA 4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), Besançon, France
| | - Mathilde Galais
- grid.4989.c0000 0001 2348 0746Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium
| | - Maxime Bellefroid
- grid.4989.c0000 0001 2348 0746Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium
| | - Irene Pachón Angona
- grid.493090.70000 0004 4910 6615Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, Besançon, France
| | | | - Lhassane Ismaili
- grid.493090.70000 0004 4910 6615Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, Besançon, France
| | - Carine Van Lint
- grid.4989.c0000 0001 2348 0746Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium
| | - Georges Herbein
- grid.7459.f0000 0001 2188 3779Pathogens and Inflammation/EPILAB Laboratory, EA 4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), Besançon, France ,grid.411158.80000 0004 0638 9213Department of Virology, CHU Besançon, Besançon, France
| |
Collapse
|
24
|
Hybridization of Aminoadamantanes with Cinnamic Acid Analogues and Elucidation of Their Antioxidant Profile. J CHEM-NY 2022. [DOI: 10.1155/2022/7582587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A series of seventeen cinnamic acid hybrids (4ai–ci) were obtained through an amidation of aminoadamantanes (amantadine, rimantadine, and memantine) with mixed anhydride generated from different substituted cinnamic acid and ethyl chloroformate. 1H NMR, 13C NMR, IR, and HRMS were used for the confirmation of the structures of the synthesized hybrids. Moreover, the antioxidant profiles of amides were estimated as per five different in vitro methods: 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2,2′-azinobis-3-ethylbenzothiazoline-6-sulfonic acid cation radical (ABTS⁺), ferric reducing antioxidant power (FRAP), cupric reducing antioxidant capacity (CUPRAC) assay, and inhibition of Fe(III)/asc induced lipid peroxidation (LP) in brain homogenate. For comparison, caffeic acid (CaffA), known as a potent naturally occurring antioxidant, was used as a reference compound in our study. The results revealed that the most prominent antioxidant activity was demonstrated by compound 4b2, with excellent CUPRAC, FRAP, scavenging ABTS+˙ potential, and inhibition of Fe/asc–induced LP, followed by 4c6 > 4a6 > CaffA > 4c5 and 4a5 > 4a7. Overall, the results suggest that the hybrids (4b2, 4c6, and 4a6) consisting of a caffeoyl moiety and lipophilic adamantane core endow the molecules with the higher antioxidant activity than their parent compound (caffeic acid), especially against LP. Thus, these promising antioxidants could have beneficial effects in various pathological conditions, where oxidative stress is implicated.
Collapse
|
25
|
Multicomponent reactions as a privileged tool for multitarget-directed ligand strategies in Alzheimer's disease therapy. Future Med Chem 2022; 14:1583-1606. [PMID: 36263996 DOI: 10.4155/fmc-2022-0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Among neurodegenerative pathologies affecting the older population, Alzheimer's disease is the most common type of dementia and leads to neurocognitive and behavioral disorders. It is a complex and progressive age-related multifactorial disease characterized by a series of highly interconnected pathophysiological processes. Within the last decade, the multitarget-directed ligand strategy has emerged as a viable approach to developing complex molecules that exhibit several pharmacophores which can target the different enzymes and receptors involved in the pathogenesis of the disease. Herein, we focus on using multicomponent reactions such as Hantzsch, Biginelli and Ugi to develop these biologically active multitopic ligands.
Collapse
|
26
|
Recent advance on pleiotropic cholinesterase inhibitors bearing amyloid modulation efficacy. Eur J Med Chem 2022; 242:114695. [PMID: 36044812 DOI: 10.1016/j.ejmech.2022.114695] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 12/15/2022]
Abstract
Due to the hugely important roles of neurotransmitter acetylcholine (ACh) and amyloid-β (Aβ) in the pathogenesis of Alzheimer's disease (AD), the development of multi-target directed ligands (MTDLs) focused on cholinesterase (ChE) and Aβ becomes one of the most attractive strategies for combating AD. To date, numerous preclinical studies toward multifunctional conjugates bearing ChE inhibition and anti-Aβ aggregation have been reported. Noteworthily, most of the reported multifunctional cholinesterase inhibitors are carbamate-based compounds due to the initial properties of carbamate moiety. However, because their easy hydrolysis in vivo and the instability of the compound-enzyme conjugate, the mechanism of action of these compounds is rare. Thus, non-carbamate compounds are of great need for developing novel cholinesterase inhibitors. Besides, given that Aβ accumulation begins to occur 10-15 years before AD onset, modulating Aβ is ineffective only in inhibiting its aggregation but not eliminate the already accumulated Aβ if treatment is started when the patient has been diagnosed as AD. Considering the limitation of current Aβ accumulation modulators in ameliorating cognitive deficits and ineffectiveness of ChE inhibitors in blocking disease progression, the development of a practically valuable strategy with multiple pharmaceutical properties including ChE inhibition and Aβ modulation for treating AD is indispensable. In this review, we focus on summarizing the scaffold characteristics of reported non-carbamate cholinesterase inhibitors with Aβ modulation since 2020, and understanding the ingenious multifunctional drug design ideas to accelerate the pace of obtaining more efficient anti-AD drugs in the future.
Collapse
|
27
|
Guo J, Cheng M, Liu P, Cao D, Luo J, Wan Y, Fang Y, Jin Y, Xie SS, Liu J. A multi-target directed ligands strategy for the treatment of Alzheimer's disease: Dimethyl fumarate plus Tranilast modified Dithiocarbate as AChE inhibitor and Nrf2 activator. Eur J Med Chem 2022; 242:114630. [PMID: 35987018 DOI: 10.1016/j.ejmech.2022.114630] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) possessed intricate pathogenesis. Currently, multi-targeted drugs were considered to have the potential to against AD by simultaneously triggering molecules in functionally complementary pathways. Hence, a series of molecules based on the pharmacophoric features of Dimethyl fumarate, Tranilast, and Dithiocarbate were designed and synthesized. These compounds showed significant AChE inhibitory activity in vitro. Among them, compound 4c2 displayed the mighty inhibitory activity to hAChE (IC50 = 0.053 μM) and held the ability to cross the BBB. Kinetic study and molecular docking pointed out that 4c2 bound well into the active sites of hAChE, forming steady and sturdy interactions with key residues in hAChE. Additionally, 4c2 as an Nrf2 activator could promote the nuclear translocation of Nrf2 protein and induce the expressions of Nrf2-dependent enzymes HO-1, NQO1, and GPX4. Moreover, 4c2 rescued BV-2 cells from H2O2-induced injury and inhibited ROS accumulation. For the anti-neuroinflammatory potential of 4c2, we observed that 4c2 could lower the levels of pro-inflammatory cytokines (NO, IL-6 and TNF-α) and suppressed the expressions of iNOS and COX-2. In particular, 4c2 was well tolerated in mice (2500 mg/kg, p.o.) and efficaciously recovered the memory impairment in a Scopolamine-induced mouse model. Overall, these results highlighted that 4c2 was a promising multi-targeted agent for treating AD.
Collapse
Affiliation(s)
- Jie Guo
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Maojun Cheng
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Peng Liu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Duanyuan Cao
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Jinchong Luo
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Yang Wan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Yuanying Fang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Sai-Sai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China.
| | - Jing Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China.
| |
Collapse
|
28
|
Nguyen TH, Tran PT, Pham NQA, Hoang VH, Hiep DM, Ngo ST. Identifying Possible AChE Inhibitors from Drug-like Molecules via Machine Learning and Experimental Studies. ACS OMEGA 2022; 7:20673-20682. [PMID: 35755364 PMCID: PMC9219098 DOI: 10.1021/acsomega.2c00908] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/27/2022] [Indexed: 05/30/2023]
Abstract
Acetylcholinesterase (AChE) is one of the most important drug targets for Alzheimer's disease (AD) treatment. In this work, a machine learning model was trained to rapidly and accurately screen large chemical databases for the potential inhibitors of AChE. The obtained results were then validated via in vitro enzyme assay. Moreover, atomistic simulations including molecular docking and molecular dynamics simulations were then used to understand molecular insights into the binding process of ligands to AChE. In particular, two compounds including benzyl trifluoromethyl ketone and trifluoromethylstyryl ketone were indicated as highly potent inhibitors of AChE because they established IC50 values of 0.51 and 0.33 μM, respectively. The obtained IC50 of two compounds is significantly lower than that of galantamine (2.10 μM). The predicted log(BB) suggests that the compounds may be able to traverse the blood-brain barrier. A good agreement between computational and experimental studies was observed, indicating that the hybrid approach can enhance AD therapy.
Collapse
Affiliation(s)
- Trung Hai Nguyen
- Laboratory
of Theoretical and Computational Biophysics, Advanced Institute of
Materials Science, Ton Duc Thang
University, Ho Chi Minh City, Vietnam
- Faculty
of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Phuong-Thao Tran
- Hanoi
University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 008404, Vietnam
| | - Ngoc Quynh Anh Pham
- Faculty
of Chemical Engineering, Ho Chi Minh City
University of Technology (HCMUT), Ho Chi Minh City 700000, Vietnam
| | - Van-Hai Hoang
- Faculty
of Pharmacy, Phenikka University, Hanoi 008404, Vietnam
- Phenikka
Institute for Advanced Study, Phenikka University, Hanoi 008404, Vietnam
| | - Dinh Minh Hiep
- Department
of Agriculture and Rural Development, Ho Chi Minh City 700000, Vietnam
| | - Son Tung Ngo
- Laboratory
of Theoretical and Computational Biophysics, Advanced Institute of
Materials Science, Ton Duc Thang
University, Ho Chi Minh City, Vietnam
- Faculty
of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
29
|
George N, Jawaid Akhtar M, Al Balushi KA, Alam Khan S. Rational drug design strategies for the development of promising multi-target directed indole hybrids as Anti-Alzheimer agents. Bioorg Chem 2022; 127:105941. [PMID: 35714473 DOI: 10.1016/j.bioorg.2022.105941] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is a neurological disorder that leads to dementia i.e., progressive memory loss accompanied with worsening of thinking ability of an individual. The cause of AD is not fully understood but it progresses with age where brain cells gradually die over time. According to the World Health Organization (WHO), currently 50 million people worldwide are affected by dementia and 60-70% of the cases belong to AD. Cumulative research over the past few decades have shown that molecules that act at a single target possess limited efficacy since these investigational drugs are not able to act against complex pathologies and thus do not provide permanent cure. Designing of multi-target directed ligands (MTDLs) appears to be more beneficial and a rational approach to treat chronic complex diseases including neurodegenerative diseases. Recently, MTDLs are being extensively researched by the medicinal chemists for the development of drugs for the treatment of various multifactorial diseases. Indole is one of the privileged scaffolds which is considered as an essential mediator between the gut-brain axis because of its neuroprotective, anti-inflammatory, β-amyloid anti-aggregation and antioxidant activities. Herein, we have reviewed the potential of some indole-hybrids acting at multiple targets in the pathogenesis of AD. We have reviewed research articles from the year 2014-2021 from various scientific databases and highlighted the synthetic strategies, mechanisms of neuroprotection, toxicity, structure activity relationships and molecular docking studies of various indole-hybrid derivatives. This literature review of published data on indole derivatives indicated that developing indole hybrids have improved the pharmacokinetic profile with lower toxicity, provided synergistic effect, helped to develop more potent compounds and prevented drug-drug interactions. It is evident that this class of compounds have potential to inhibit multiple enzymes targets involved in the pathogenesis of AD and therefore indole hybrids as MTDLs may play an important role in the development of anti-AD molecules.
Collapse
Affiliation(s)
- Namy George
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman
| | - Md Jawaid Akhtar
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman
| | - Khalid A Al Balushi
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman
| | - Shah Alam Khan
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman.
| |
Collapse
|
30
|
Thai QM, Pham TNH, Hiep DM, Pham MQ, Tran PT, Nguyen TH, Ngo ST. Searching for AChE inhibitors from natural compounds by using machine learning and atomistic simulations. J Mol Graph Model 2022; 115:108230. [DOI: 10.1016/j.jmgm.2022.108230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/14/2022]
|
31
|
Lachowicz JI, Lecca LI, Meloni F, Campagna M. Metals and Metal-Nanoparticles in Human Pathologies: From Exposure to Therapy. Molecules 2021; 26:6639. [PMID: 34771058 PMCID: PMC8587420 DOI: 10.3390/molecules26216639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/13/2023] Open
Abstract
An increasing number of pathologies correlates with both toxic and essential metal ions dyshomeostasis. Next to known genetic disorders (e.g., Wilson's Disease and β-Thalassemia) other pathological states such as neurodegeneration and diabetes are characterized by an imbalance of essential metal ions. Metal ions can enter the human body from the surrounding environment in the form of free metal ions or metal-nanoparticles, and successively translocate to different tissues, where they are accumulated and develop distinct pathologies. There are no characteristic symptoms of metal intoxication, and the exact diagnosis is still difficult. In this review, we present metal-related pathologies with the most common onsets, biomarkers of metal intoxication, and proper techniques of metal qualitative and quantitative analysis. We discuss the possible role of drugs with metal-chelating ability in metal dyshomeostasis, and present recent advances in therapies of metal-related diseases.
Collapse
Affiliation(s)
| | | | | | - Marcello Campagna
- Division of Occupational Medicine, Department of Medical Sciences and Public Health, University of Cagliari, 09048 Monserrato, CA, Italy; (J.I.L.); (L.I.L.); (F.M.)
| |
Collapse
|
32
|
Synthesis of new Hantzsch adducts showing Ca 2+ channel blockade capacity, cholinesterase inhibition and antioxidant power. Future Med Chem 2021; 13:1717-1729. [PMID: 34402661 DOI: 10.4155/fmc-2021-0176] [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: 01/08/2023] Open
Abstract
Background: Alzheimer's disease is a chronic neurodegenerative chronic disease with a heavy social and economic impact in our developed societies, which still lacks an efficient therapy. Method: This paper describes the Hantzsch multicomponent synthesis of twelve alkyl hexahydro-quinoline-3-carboxylates, 4a-l, along with the evaluation of their Ca2+ channel blockade capacity, cholinesterase inhibition and antioxidant power. Results: Compound 4l showed submicromolar inhibition of butyrylcholinesterase, Ca2+ channel antagonism and an antioxidant effect. Conclusion: Compound 4l is an interesting compound that deserves further investigation for Alzheimer's disease therapy.
Collapse
|
33
|
Kador PF, Salvi R. Multifunctional Redox Modulators Protect Auditory, Visual, and Cognitive Function. Antioxid Redox Signal 2021; 36:1136-1157. [PMID: 34162214 PMCID: PMC9221172 DOI: 10.1089/ars.2021.0129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/26/2022]
Abstract
Significance: Oxidative stress contributes to vision, hearing and neurodegenerative disorders. Currently, no treatments prevent these disorders; therefore, there is an urgent need for redox modulators that can prevent these disorders. Recent Advances: Oxidative stress is associated with the generation of reactive oxygen species (ROS) and reactive nitrogen species, metal dyshomeostasis, and mitochondrial dysfunction. Here, we discuss the role that oxidative stress and metal dyshomeostasis play in hearing loss, visual impairments, and neurodegeneration and discuss the benefits of a new class of multifunctional redox modulators (MFRMs) that suppress sensory and neural degeneration. MFRMs not only reduce free radicals but also independently bind transition metals associated with the generation of hydroxyl radicals. The MFRMs redistribute zinc from neurotoxic amyloid beta zinc (Aβ:Zn) complexes to the cytoplasm, facilitating the degradation of Aβ plaques by matrix metalloprotease-2 (MMP-2). Although MFRMs bind copper (Cu1+, Cu2+), iron (Fe2+, Fe3+), zinc (Zn2+), and manganese (Mn2+), they do not deplete free cytoplasmic Zn+2 and they protect mitochondria from Mn+2-induced dysfunction. Oral administration of MFRMs reduce ROS-induced cataracts, protect the retina from light-induced degeneration, reduce neurotoxic Aβ:Zn plaque formation, and protect auditory hair cells from noise-induced hearing loss. Critical Issues: Regulation of redox balance is essential for clinical efficacy in maintaining sensory functions. Future Directions: Future use of these MFRMs requires additional pharmacokinetic, pharmacodynamics, and toxicological data to bring them into widespread clinical use. Additional animal studies are also needed to determine whether MFRMs can prevent neurodegeneration, dementia, and other forms of vision and hearing loss.
Collapse
Affiliation(s)
- Peter F. Kador
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
34
|
Scheiner M, Hoffmann M, He F, Poeta E, Chatonnet A, Monti B, Maurice T, Decker M. Selective Pseudo-irreversible Butyrylcholinesterase Inhibitors Transferring Antioxidant Moieties to the Enzyme Show Pronounced Neuroprotective Efficacy In Vitro and In Vivo in an Alzheimer's Disease Mouse Model. J Med Chem 2021; 64:9302-9320. [PMID: 34152756 DOI: 10.1021/acs.jmedchem.1c00534] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A series of multitarget-directed ligands (MTDLs) was designed by functionalizing a pseudo-irreversible butyrylcholinesterase (BChE) inhibitor. The obtained hybrids were investigated in vitro regarding their hBChE and hAChE inhibition, their enzyme kinetics, and their antioxidant physicochemical properties (DPPH, ORAC, metal chelating). In addition, in vitro assays were applied to investigate antioxidant effects using murine hippocampal HT22 cells and immunomodulatory effects on the murine microglial N9 cell line. The MTDLs retained their antioxidative properties compared to the parent antioxidant-moieties in vitro and the inhibition of hBChE was maintained in the submicromolar range. Representative compounds were tested in a pharmacological Alzheimer's disease (AD) mouse model and demonstrated very high efficacy at doses as low as 0.1 mg/kg. The most promising compound was also tested in BChE-/- mice and showed reduced efficacy. In vivo neuroprotection by BChE inhibition can be effectively enhanced by incorporation of structurally diverse antioxidant moieties.
Collapse
Affiliation(s)
- Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Matthias Hoffmann
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Feng He
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Eleonora Poeta
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Arnaud Chatonnet
- DMEM, University of Montpellier, INRAE, 34060 Montpellier, France
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, 34095 Montpellier, France
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
35
|
Yang GX, Sun JM, Zheng LL, Zhang L, Li J, Gan HX, Huang Y, Huang J, Diao XX, Tang Y, Wang R, Ma L. Twin drug design, synthesis and evaluation of diosgenin derivatives as multitargeted agents for the treatment of vascular dementia. Bioorg Med Chem 2021; 37:116109. [PMID: 33780813 DOI: 10.1016/j.bmc.2021.116109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/06/2021] [Indexed: 12/21/2022]
Abstract
A novel series of multitargeted molecules were designed and synthesized by combining the pharmacological role of cholinesterase inhibitor and antioxidant of steroid as potential ligands for the treatment of Vascular Dementia (VD). The oxygen-glucose deprivation (OGD) model was used to evaluate these molecules, among which the most potent compound ML5 showed the highest activity. Firstly, ML5 showed appropriate inhibition of cholinesterases (ChEs) at orally 15 mg/kg in vivo. The further test revealed that ML5 promoted the nuclear translocation of Nrf2. Furthermore, ML5 has significant neuroprotective effect in vivo model of bilateral common carotid artery occlusion (BCCAO), significantly increasing the expression of Nrf2 protein in the cerebral cortex. In the molecular docking research, we predicted the ML5 combined with hAChE and Keap1. Finally, compound ML5 displayed normal oral absorption and it was nontoxic at 500 mg/kg, po, dose. We can draw the conclusion that ML5 could be considered as a new potential compound for VD treatment.
Collapse
Affiliation(s)
- Gui-Xiang Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jia-Min Sun
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Lu-Lu Zheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Li Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jie Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hai-Xian Gan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yan Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xing-Xing Diao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
36
|
Ismaili L, Monnin J, Etievant A, Arribas RL, Viejo L, Refouvelet B, Soukup O, Janockova J, Hepnarova V, Korabecny J, Kucera T, Jun D, Andrys R, Musilek K, Baguet A, García-Frutos EM, De Simone A, Andrisano V, Bartolini M, de los Ríos C, Marco-Contelles J, Haffen E. (±)- BIGI-3h: Pentatarget-Directed Ligand combining Cholinesterase, Monoamine Oxidase, and Glycogen Synthase Kinase 3β Inhibition with Calcium Channel Antagonism and Antiaggregating Properties for Alzheimer's Disease. ACS Chem Neurosci 2021; 12:1328-1342. [PMID: 33797877 DOI: 10.1021/acschemneuro.0c00803] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multitarget-directed ligands (MTDLs) are considered a promising therapeutic strategy to address the multifactorial nature of Alzheimer's disease (AD). Novel MTDLs have been designed as inhibitors of human acetylcholinesterases/butyrylcholinesterases, monoamine oxidase A/B, and glycogen synthase kinase 3β and as calcium channel antagonists via the Biginelli multicomponent reaction. Among these MTDLs, (±)-BIGI-3h was identified as a promising new hit compound showing in vitro balanced activities toward the aforementioned recognized AD targets. Additional in vitro studies demonstrated antioxidant effects and brain penetration, along with the ability to inhibit the aggregation of both τ protein and β-amyloid peptide. The in vivo studies have shown that (±)-BIGI-3h (10 mg/kg intraperitoneally) significantly reduces scopolamine-induced cognitive deficits.
Collapse
Affiliation(s)
- Lhassane Ismaili
- Neurosciences intégratives et cliniques EA 481, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Julie Monnin
- Neurosciences intégratives et cliniques EA 481, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Adeline Etievant
- Neurosciences intégratives et cliniques EA 481, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Raquel L. Arribas
- Servicio de Farmacología Clínica, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/Diego de León, 62, 28006 Madrid, Spain
- Instituto Teofilo Hernando, Universidad Autónoma de Madrid, C/Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Lucía Viejo
- Servicio de Farmacología Clínica, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/Diego de León, 62, 28006 Madrid, Spain
- Instituto Teofilo Hernando, Universidad Autónoma de Madrid, C/Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Bernard Refouvelet
- Neurosciences intégratives et cliniques EA 481, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Ondrej Soukup
- Biomedical Research Center, University Hospital Hradec Kralove, Czech Republic, University of Defence, 50003 Hradec Kralove, Czech Republic
| | - Jana Janockova
- Biomedical Research Center, University Hospital Hradec Kralove, Czech Republic, University of Defence, 50003 Hradec Kralove, Czech Republic
| | - Vendula Hepnarova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, 66210 Brno, Czech Republic
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove, Czech Republic, University of Defence, 50003 Hradec Kralove, Czech Republic
| | - Tomas Kucera
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, 66210 Brno, Czech Republic
| | - Daniel Jun
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, 66210 Brno, Czech Republic
| | - Rudolf Andrys
- Faculty of Science, Department of Chemistry, University Hradec Kralove, Rokitanskeho 62, 50003 Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Faculty of Science, Department of Chemistry, University Hradec Kralove, Rokitanskeho 62, 50003 Hradec Kralove, Czech Republic
| | - Aurelie Baguet
- Université Bourgogne Franche Comté, INSERM, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - Eva M. García-Frutos
- Instituto de Ciencia de Materiales de Madrid, CSIC, Cantoblanco, 28049 Madrid, Spain
| | - Angela De Simone
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Corso di Augusto, 237, 47921 Rimini, Italy
| | - Vincenza Andrisano
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Corso di Augusto, 237, 47921 Rimini, Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Cristóbal de los Ríos
- Servicio de Farmacología Clínica, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/Diego de León, 62, 28006 Madrid, Spain
- Instituto Teofilo Hernando, Universidad Autónoma de Madrid, C/Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry, Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Emmanuel Haffen
- Neurosciences intégratives et cliniques EA 481, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| |
Collapse
|
37
|
Zhou LC, Liang YF, Huang Y, Yang GX, Zheng LL, Sun JM, Li Y, Zhu FL, Qian HW, Wang R, Ma L. Design, synthesis, and biological evaluation of diosgenin-indole derivatives as dual-functional agents for the treatment of Alzheimer's disease. Eur J Med Chem 2021; 219:113426. [PMID: 33848787 DOI: 10.1016/j.ejmech.2021.113426] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/17/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023]
Abstract
The complex pathogenesis of Alzheimer's disease (AD) has become a major obstacle in its treatment. An effective approach is to develop multifunctional agents that simultaneously target multiple pathological processes. Here, a series of diosgenin-indole compounds were designed, synthesized and evaluated for their neuroprotective effects against H2O2 (hydrogen peroxide), 6-OHDA (6-hydroxydopamine) and Aβ (beta amyloid) damages. Preliminary structure-activities relationship revealed that the introduction of indole fragment and electron-donating group at C-5 on ring indole could be beneficial for neuroprotective activities. Results indicated that compound 5b was the most promising candidate against cellular damage induced by H2O2 (52.9 ± 1.9%), 6-OHDA (38.4 ± 2.4%) and Aβ1-42 (54.4 ± 2.7%). Molecular docking study suggested the affinity for 5b bound to Aβ1-42 was -40.59 kcal/mol, which revealed the strong binding affinity of 5b to Aβ1-42. The predicted values of brain/blood partition coefficient (-0.733) and polar surface area (85.118 Å2) indicated the favorable abilities of BBB permeation and absorption of 5b. In addition, 5b significantly decreased ROS (reactive oxygen species) production induced by H2O2. In the following in vivo experiment, 5b obviously attenuated memory and learning impairments of Aβ-injected mice. In summary, compound 5b could be considered as a promising dual-functional neuroprotective agent against AD.
Collapse
Affiliation(s)
- Li-Cheng Zhou
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Ying-Fan Liang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yi Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Gui-Xiang Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Lu-Lu Zheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Jia-Min Sun
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Fu-Li Zhu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - He-Wen Qian
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China.
| |
Collapse
|
38
|
Moni L, Banfi L, Basso A, Mori A, Risso F, Riva R, Lambruschini C. A Thorough Study on the Photoisomerization of Ferulic Acid Derivatives. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Lisa Moni
- Department of Chemistry and Industrial Chemistry University of Genova Via Dodecaneso, 31 16146 Genova Italy
| | - Luca Banfi
- Department of Chemistry and Industrial Chemistry University of Genova Via Dodecaneso, 31 16146 Genova Italy
| | - Andrea Basso
- Department of Chemistry and Industrial Chemistry University of Genova Via Dodecaneso, 31 16146 Genova Italy
| | - Alessia Mori
- Department of Chemistry and Industrial Chemistry University of Genova Via Dodecaneso, 31 16146 Genova Italy
| | - Federica Risso
- Department of Chemistry and Industrial Chemistry University of Genova Via Dodecaneso, 31 16146 Genova Italy
| | - Renata Riva
- Department of Chemistry and Industrial Chemistry University of Genova Via Dodecaneso, 31 16146 Genova Italy
| | - Chiara Lambruschini
- Department of Chemistry and Industrial Chemistry University of Genova Via Dodecaneso, 31 16146 Genova Italy
| |
Collapse
|
39
|
Singh YP, Rai H, Singh G, Singh GK, Mishra S, Kumar S, Srikrishna S, Modi G. A review on ferulic acid and analogs based scaffolds for the management of Alzheimer's disease. Eur J Med Chem 2021; 215:113278. [PMID: 33662757 DOI: 10.1016/j.ejmech.2021.113278] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/30/2021] [Accepted: 01/30/2021] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is an age-related multifactorial neurodegenerative disorder characterized by severe central cholinergic neuronal loss, gradually contributing to cognitive dysfunction and impaired motor activity, resulting in the brain's cell death at the later stages of AD. Although the etiology of AD is not well understood, however, several factors such as oxidative stress, deposition of amyloid-β (Aβ) peptides to form Aβ plaques, intraneuronal accumulation of hyperphosphorylated tau protein, and low level of acetylcholine are thought to play a major role in the pathogenesis of AD. There is practically no drug for AD treatment that can address the basic factors responsible for the neurodegeneration and slow down the disease progression. The currently available therapies for AD in the market focus on providing only symptomatic relief without addressing the aforesaid basic factors responsible for the neurodegeneration. Ferulic acid (FA) is a phenol derivative from natural sources and serves as a potential pharmacophore that exerts multiple pharmacological properties such as antioxidant, neuroprotection, Aβ aggregation modulation, and anti-inflammatory. Several FA based hybrid analogs are under investigation as a multi-target directed ligand (MTDLs) to develop novel hybrid compounds for the treatment of AD. In the present review article, we are focused on the critical pathogenic factors responsible for the onset of AD followed by the developments of FA pharmacophore-based hybrids compounds as a novel multifunctional therapeutic agent to address the limitations associated with available treatment for AD. The rationale behind the development of these compounds and their pharmacological activities in particular to their ChE inhibition (ChEI), neuroprotection, antioxidant property, Aβ aggregation modulation, and metal chelation ability, are discussed in detail. We have also discussed the discovery of caffeic and cinnamic acids based MTDLs for AD. This review paper provides an in-depth insight into the research progress and current status of these novel therapeutics in AD and prospects for developing a druggable molecule with desired pharmacological affinity and reduced toxicity for the management of AD.
Collapse
Affiliation(s)
- Yash Pal Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Himanshu Rai
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Gourav Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Gireesh Kumar Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar Gaya, 824236, India
| | - Sunil Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - S Srikrishna
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
40
|
Zhu H, Dronamraju V, Xie W, More SS. Sulfur-containing therapeutics in the treatment of Alzheimer's disease. Med Chem Res 2021; 30:305-352. [PMID: 33613018 PMCID: PMC7889054 DOI: 10.1007/s00044-020-02687-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022]
Abstract
Sulfur is widely existent in natural products and synthetic organic compounds as organosulfur, which are often associated with a multitude of biological activities. OBenzothiazole, in which benzene ring is fused to the 4,5-positions of the thiazolerganosulfur compounds continue to garner increasing amounts of attention in the field of medicinal chemistry, especially in the development of therapeutic agents for Alzheimer's disease (AD). AD is a fatal neurodegenerative disease and the primary cause of age-related dementia posing severe societal and economic burdens. Unfortunately, there is no cure for AD. A lot of research has been conducted on sulfur-containing compounds in the context of AD due to their innate antioxidant potential and some are currently being evaluated in clinical trials. In this review, we have described emerging trends in the field, particularly the concept of multi-targeting and formulation of disease-modifying strategies. SAR, pharmacological targets, in vitro/vivo ADMET, efficacy in AD animal models, and applications in clinical trials of such sulfur compounds have also been discussed. This article provides a comprehensive review of organosulfur-based AD therapeutic agents and provides insights into their future development.
Collapse
Affiliation(s)
- Haizhou Zhu
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Venkateshwara Dronamraju
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
41
|
Zhou L, Shi X, Yin H, Huang Y, Wang R, Ma L. Design, Synthesis and Biological Evaluation of Nobiletin Derivatives as Multifunctional Agents for the Treatment of Alzheimer's Disease. ChemistrySelect 2021. [DOI: 10.1002/slct.202004239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Licheng Zhou
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Ximeng Shi
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Huanhuan Yin
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Yi Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| |
Collapse
|
42
|
Gonzalez A. Antioxidants and Neuron-Astrocyte Interplay in Brain Physiology: Melatonin, a Neighbor to Rely on. Neurochem Res 2021; 46:34-50. [PMID: 31989469 DOI: 10.1007/s11064-020-02972-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
This manuscript is a review focused onto the role of astrocytes in the protection of neurons against oxidative stress and how melatonin can contribute to the maintenance of brain homeostasis. The first part of the review is dedicated to the dependence of neurons on astrocytes by terms of survival under oxidative stress conditions. Additionally, the effects of melatonin against oxidative stress in the brain and its putative role in the protection against diseases affecting the brain are highlighted. The effects of melatonin on the physiology of neurons and astrocytes also are reviewed.
Collapse
Affiliation(s)
- Antonio Gonzalez
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, 10003, Cáceres, Spain.
| |
Collapse
|
43
|
Bautista‐Aguilera ÓM, Ismaili L, Iriepa I, Diez‐Iriepa D, Chabchoub F, Marco‐Contelles J, Pérez M. Tacrines as Therapeutic Agents for Alzheimer's Disease. V. Recent Developments. CHEM REC 2020; 21:162-174. [DOI: 10.1002/tcr.202000107] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Óscar M. Bautista‐Aguilera
- Departamento de Química Orgánica and Química Inorgánica. Ctra. Madrid-Barcelona Universidad de Alcalá Km. 33, 6 28871 Madrid Spain
| | - Lhassane Ismaili
- Laboratoire de Chimie Organique et Thérapeutique Neurosciences intégratives et cliniques EA 481 Univ. Bourgogne Franche-Comté, UFR Santé 19, rue Ambroise Paré F-25000 Besançon France
| | - Isabel Iriepa
- Departamento de Química Orgánica and Química Inorgánica. Ctra. Madrid-Barcelona Universidad de Alcalá Km. 33, 6 28871 Madrid Spain
- Institute of Chemical Research Andrés M. del Río Alcalá University, 28805-Alcalá de Henares Madrid Spain
| | - Daniel Diez‐Iriepa
- Departamento de Química Orgánica and Química Inorgánica. Ctra. Madrid-Barcelona Universidad de Alcalá Km. 33, 6 28871 Madrid Spain
| | - Fakher Chabchoub
- Laboratoire de Chimie Appliquée: Hétérocycles Corps Gras et Polymères Faculté des Sciences de Sfax Université de Sfax. B. P 802. 3000 Sfax Tunisie
| | - José Marco‐Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC) Juan de la Cierva 3 28006- Madrid Spain
| | - Marta Pérez
- Public Health Department Faculty of Medicine and Nursing University of the Basque Country. Leioa Spain
| |
Collapse
|
44
|
Ortiz CJC, Damasio CM, Pruccoli L, Nadur NF, de Azevedo LL, Guedes IA, Dardenne LE, Kümmerle AE, Tarozzi A, Viegas C. Cinnamoyl-N-Acylhydrazone-Donepezil Hybrids: Synthesis and Evaluation of Novel Multifunctional Ligands Against Neurodegenerative Diseases. Neurochem Res 2020; 45:3003-3020. [PMID: 33079324 DOI: 10.1007/s11064-020-03148-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/25/2020] [Accepted: 10/07/2020] [Indexed: 11/30/2022]
Abstract
A new series of ten multifunctional Cinnamoyl-N-acylhydrazone-donepezil hybrids was synthesized and evaluated as multifunctional ligands against neurodegenerative diseases. The molecular hybridization approach was based on the combination of 1-benzyl-4-piperidine fragment from the anti-Alzheimer AChE inhibitor donepezil (1) and the cinnamoyl subunit from curcumin (2), a natural product with remarkable antioxidant, neuroprotective and anti-inflammatory properties, using a N-acylhydrazone fragment as a spacer subunit. Compounds 4a and 4d showed moderate inhibitory activity towards AChE with IC50 values of 13.04 and 9.1 µM, respectively. In addition, compound 4a and 4d showed a similar predicted binding mode to that observed for donepezil in the molecular docking studies. On the other hand, compounds 4a and 4c exhibited significant radical scavenging activity, showing the best effects on the DPPH test and also exhibited a significant protective neuronal cell viability exposed to t-BuOOH and against 6-OHDA insult to prevent the oxidative stress in Parkinson's disease. Similarly, compound 4c was capable to prevent the ROS formation, with indirect antioxidant activity increasing intracellular GSH levels and the ability to counteract the neurotoxicity induced by both OAβ1-42 and 3-NP. In addition, ADMET in silico prediction indicated that both compounds 4a and 4c did not show relevant toxic effects. Due to their above-mentioned biological properties, compounds 4a and 4c could be explored as lead compounds in search of more effective and low toxic small molecules with multiple neuroprotective effects for neurodegenerative diseases.
Collapse
Affiliation(s)
- Cindy Juliet Cristancho Ortiz
- PeQuiM-Laboratory of Research in Medicinal Chemistry, Federal University of Alfenas, Jovino Fernandes Sales Avenue 2600, Alfenas, MG, 37130-840, Brazil
| | - Caio Miranda Damasio
- PeQuiM-Laboratory of Research in Medicinal Chemistry, Federal University of Alfenas, Jovino Fernandes Sales Avenue 2600, Alfenas, MG, 37130-840, Brazil
| | - Letizia Pruccoli
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto 237, 47921, Rimini, Italy
| | - Nathália Fonseca Nadur
- Laboratory of Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, 21941-902, Brazil
| | - Luciana Luiza de Azevedo
- Laboratory of Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, 21941-902, Brazil
| | | | | | - Arthur Eugen Kümmerle
- Laboratory of Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, 21941-902, Brazil
| | - Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto 237, 47921, Rimini, Italy.
| | - Claudio Viegas
- PeQuiM-Laboratory of Research in Medicinal Chemistry, Federal University of Alfenas, Jovino Fernandes Sales Avenue 2600, Alfenas, MG, 37130-840, Brazil.
| |
Collapse
|
45
|
Pachón Angona I, Martin H, Daniel S, Moraleda I, Bonet A, Wnorowski A, Maj M, Jozwiak K, Iriepa I, Refouvelet B, Marco-Contelles J, Ismaili L. Synthesis of Hantzsch Adducts as Cholinesterases and Calcium Flux inhibitors, Antioxidants and Neuroprotectives. Int J Mol Sci 2020; 21:ijms21207652. [PMID: 33081112 PMCID: PMC7589057 DOI: 10.3390/ijms21207652] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 01/22/2023] Open
Abstract
We report herein the design, synthesis, biological evaluation, and molecular modelling of new inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), able to block Ca+2 channels also showing antioxidant and neuroprotective activities. The new MTDL, dialkyl 2,6-dimethyl-4-(4-((5-aminoalkyl)oxy)phenyl)-1,4-dihydropyridine-3,5-dicarboxylate 3a-p, have been obtained via Hantzsch reaction from appropriate and commercially available precursors. Pertinent biological analysis has prompted us to identify MTDL 3h [dimethyl-4-(4-((5-(4-benzylpiperidin-1-yl)pentyl)oxy)phenyl)-2,6-dimethyl-1,4-dihydropyridine-3,5-dicarboxylate] as an attractive inhibitor of AChE (1.8 μM) and BuChE (2 μM), Ca+2 channel antagonist (47.72% at 10 μM), and antioxidant (2.54 TE) agent, showing significant neuroprotection 28.68% and 38.29% against H2O2, and O/R, respectively, at 0.3 μM, thus being considered a hit-compound for further investigation in our search for anti-Alzheimer's disease agents.
Collapse
Affiliation(s)
- Irene Pachón Angona
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
| | - Helene Martin
- PEPITE EA4267, Laboratoire de Toxicologie Cellulaire, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France; (H.M.); (A.B.)
| | - Solene Daniel
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
| | - Ignacio Moraleda
- Department of Organic Chemistry and Inorganic Chemistry, School Sciences, University of Alcalá, Ctra. Barcelona, Km. 33.6, 28871 Alcalá de Henares, Spain;
| | - Alexandre Bonet
- PEPITE EA4267, Laboratoire de Toxicologie Cellulaire, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France; (H.M.); (A.B.)
| | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Krzysztof Jozwiak
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Isabel Iriepa
- Department of Organic Chemistry and Inorganic Chemistry, School Sciences, University of Alcalá, Ctra. Barcelona, Km. 33.6, 28871 Alcalá de Henares, Spain;
- Correspondence: (I.I.); (B.R.); (L.I.)
| | - Bernard Refouvelet
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
- Correspondence: (I.I.); (B.R.); (L.I.)
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC), Juan de la Cierva, 3, 28006-Madrid, Spain;
| | - Lhassane Ismaili
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
- Correspondence: (I.I.); (B.R.); (L.I.)
| |
Collapse
|
46
|
Wang N, Zhou Y, Zhao L, Wang C, Ma W, Ge G, Wang Y, Ullah I, Muhammad F, Alwayli D, Zhi D, Li H. Ferulic acid delayed amyloid β-induced pathological symptoms by autophagy pathway via a fasting-like effect in Caenorhabditis elegans. Food Chem Toxicol 2020; 146:111808. [PMID: 33045309 DOI: 10.1016/j.fct.2020.111808] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/27/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
The amyloid β (Aβ) generation or aggregation plays a crucial role in Alzheimer's disease (AD). Autophagy agonists, which function as the clearance of Aβ, could be the potential drug candidates against AD. In staple food crops, ferulic acid (FA) is an enormously copious and almost ubiquitous phenolic antioxidant. In the present study, FA significantly inhibited Aβ-induced pathological symptoms of paralysis and hypersensitivity to exogenous serotonin, meanwhile restrained Aβ monomers, oligomers, and deposits in AD C. elegans. FA increased the expression of autophagy reporter LGG-1 and enhanced autophagy flux. However, the autophagy inhibitors abolished the restrictive action of FA on the worm paralysis phenotype. According to these results, FA triggered autophagy and ameliorated Aβ-induced pathological symptoms by the autophagy pathway. Moreover, FA activated the HLH-30 transcription factor to nuclear localization, which acts upstream of autophagy in fasted animals, reduced the level of lipids, but affected nor the growth of E. coli OP50, neither animal food intake behavior. These suggest that FA induced a fasting-like effect to activate the autophagy pathway. Additionally, FA ameliorated poly Q aggregations in Huntington's disease worm. Thus, FA could not only affect AD, broadly but also neurodegenerative diseases characterized by misfolded or aggregated proteins.
Collapse
Affiliation(s)
- Ningbo Wang
- School of Life Sciences, Lanzhou University, China.
| | - Yongtao Zhou
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Clinical Center for Parkinson's Disease, Capital Medical University, Beijing, China.
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, China.
| | - Caiding Wang
- School of Life Sciences, Lanzhou University, China.
| | - Wuli Ma
- School of Life Sciences, Lanzhou University, China.
| | - Guangfei Ge
- School of Life Sciences, Lanzhou University, China.
| | - Yu Wang
- School of Pharmacy, Lanzhou University, China.
| | - Inam Ullah
- School of Life Sciences, Lanzhou University, China.
| | | | | | - Dejuan Zhi
- School of Pharmacy, Lanzhou University, China.
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, China; School of Pharmacy, Lanzhou University, China.
| |
Collapse
|
47
|
Marotta G, Basagni F, Rosini M, Minarini A. Memantine Derivatives as Multitarget Agents in Alzheimer's Disease. Molecules 2020; 25:molecules25174005. [PMID: 32887400 PMCID: PMC7504780 DOI: 10.3390/molecules25174005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/24/2022] Open
Abstract
Memantine (3,5-dimethyladamantan-1-amine) is an orally active, noncompetitive N-methyl-D-aspartate receptor (NMDAR) antagonist approved for treatment of moderate-to-severe Alzheimer’s disease (AD), a neurodegenerative condition characterized by a progressive cognitive decline. Unfortunately, memantine as well as the other class of drugs licensed for AD treatment acting as acetylcholinesterase inhibitors (AChEIs), provide only symptomatic relief. Thus, the urgent need in AD drug development is for disease-modifying therapies that may require approaching targets from more than one path at once or multiple targets simultaneously. Indeed, increasing evidence suggests that the modulation of a single neurotransmitter system represents a reductive approach to face the complexity of AD. Memantine is viewed as a privileged NMDAR-directed structure, and therefore, represents the driving motif in the design of a variety of multi-target directed ligands (MTDLs). In this review, we present selected examples of small molecules recently designed as MTDLs to contrast AD, by combining in a single entity the amantadine core of memantine with the pharmacophoric features of known neuroprotectants, such as antioxidant agents, AChEIs and Aβ-aggregation inhibitors.
Collapse
|
48
|
Triazolopyridopyrimidine: A New Scaffold for Dual-Target Small Molecules for Alzheimer's Disease Therapy. Molecules 2020; 25:molecules25143190. [PMID: 32668671 PMCID: PMC7397274 DOI: 10.3390/molecules25143190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) is multifactorial disease characterized by the accumulation of abnormal extracellular deposits of amyloid-beta (Aβ) peptide, and intracellular neurofibrillary tangles (NFTs), along with dramatic neuronal death and decreased levels of choline acetyltransferase. Given the limited therapeutic success of available drugs, it is urgent to explore all the opportunities available to combat this illness. Among them, the discovery of new heterocyclic scaffolds binding different receptors involved in AD should offer structural diversity and new therapeutic solutions. In this context, this work describes new triazolopyridopyrimidine easily prepared in good yields showing anticholinesterase inhibition and strong antioxidant power, particularly the most balanced: 6-amino-5-(4-methoxyphenyl)-2-phenyl-[1,2,4]triazolo[1′,5′:1,6] pyrido[2,3-d]pyrimidine-4-carbonitrile(3c) with IC50 equal to 1.32 μM against AChE and oxygen radical absorbance capacity (ORAC) value equal to 4.01 Trolox equivalents (TE); thus representing a new and very promising hit-triazolopyridopyrimidine for AD therapy.
Collapse
|
49
|
Synthesis of new ferulic/lipoic/comenic acid-melatonin hybrids as antioxidants and Nrf2 activators via Ugi reaction. Future Med Chem 2020; 11:3097-3108. [PMID: 31838896 DOI: 10.4155/fmc-2019-0191] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: Oxidative stress has been implicated in the pathogenesis of many neurodegenerative diseases, and particularly in Alzheimer's disease. Results: This work describes the Ugi multicomponent synthesis, antioxidant power and Nrf2 pathway induction in antioxidant response element cells of (E)-N-(2-((2-(1H-indol-3-yl)ethyl)amino)-2-oxoethyl)-N-(2-(5-(benzyloxy)-1H-indol-3-yl)ethyl)-3-(4-hydroxy-3-methoxyphenyl)acryl amides 8a-d, N-(2-((2-(1H-indol-3-yl)ethyl)amino)-2-oxoethyl)-N-(2-(5-(benzyloxy)-1H-indol-3-yl)ethyl)-5-(1,2-dithiolan-3-yl)pentanamides 8e-h and N-(2-((2-(1H-indol-3-yl)ethyl)amino)-2-oxoethyl)-N-(2-(5-(benzyloxy)-1H-indol-3-yl)ethyl)-5-hydroxy-4-oxo-4H-pyran-2-carboxamides 8i,j. Conclusion: We have identified compounds 8e and 8g, showing a potent antioxidant capacity, a remarkable neuroprotective effect against the cell death induced by H2O2 in SH-SY5Y cells, and a performing activation of the Nrf2 signaling pathway, as very interesting new antioxidant agents for pathologies that curse with oxidative stress.
Collapse
|
50
|
Huang-Pu-Tong-Qiao Formula Ameliorates Tau Phosphorylation by Inhibiting the CaM-CaMKIV Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8956071. [PMID: 34046075 PMCID: PMC8128057 DOI: 10.1155/2020/8956071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease. It is a chronic, lethal disease in which brain function is severely impaired and neuronal damage is irreversible. Huang-Pu-Tong-Qiao (HPTQ), a formula from traditional Chinese medicine, has been used in the clinical treatment of AD for many years, with remarkable effects. However, the neuroprotective mechanisms of HPTQ in AD have not yet been investigated. In the present study, we used AD models in vivo and in vitro, to investigate both the neuroprotective effect of HPTQ water extracts (HPTQ-W) and the potential mechanisms of this action. For the in vivo study, after HPTQ intervention, the Morris water maze test was used to examine learning and memory in rats. Transmission electron microscopy and immunofluorescence methods were then used to investigate neuronal damage. For the in vitro experiments, rat primary hippocampal neurons were cultured and cell viability was examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Additionally, mRNA levels of CaM, CaMKK, CaMKIV, and tau were examined using qRT-PCR, and protein expression of CaM, CaMKK, p-CaMKIV, and p-tau were examined using western blot. In vivo, we revealed that HPTQ significantly improved learning and memory deficits and attenuated neuronal damage in the AD rat model. Furthermore, in vitro results showed that HPTQ significantly increased cell viability in the AD cell model. We also demonstrated that HPTQ significantly decreased the mRNA levels of CaM, CaMKK, CaMKIV, and tau and significantly decreased the protein expressions of CaM, CaMKK, p-CaMKIV, and p-tau. In conclusion, our results indicated that HPTQ improved cognition and ameliorated neuronal damage in AD models and implicated a reduction in tau phosphorylation caused by inhibition of the CaM-CaMKIV pathway as a possible mechanism.
Collapse
|