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
|
Azam U, Naseer MM, Rochais C. Analysis of skeletal diversity of multi-target directed ligands (MTDLs) targeting Alzheimer's disease. Eur J Med Chem 2025; 286:117277. [PMID: 39848035 DOI: 10.1016/j.ejmech.2025.117277] [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: 10/11/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/25/2025]
Abstract
Alzheimer's disease (AD) remains a significant healthcare challenge, necessitating innovative therapeutic approaches to address its complex and multifactorial nature. Traditional drug discovery strategies targeting single molecular targets are not sufficient for the effective treatment of AD. In recent years, MTDLs have emerged as promising candidates for AD therapy, aiming to simultaneously modulate multiple pathological targets. Among the various strategies employed in MTDL design, pharmacophore hybridization offers a versatile approach to integrate diverse pharmacophoric features within a single molecular scaffold. This strategy provides access to a wide array of chemical space for the design and development of novel therapeutic agents. This review, therefore, provides a comprehensive overview of skeletal diversity exhibited by MTDLs designed recently for AD therapy based on pharmacophore hybridization approach. A diverse range of pharmacophoric elements and core scaffolds hybridized to construct MTDLs that has the potential to target multiple pathological features of AD including amyloid-beta aggregation, tau protein hyperphosphorylation, cholinergic dysfunction, oxidative stress, and neuroinflammation are discussed. Through the comprehensive analysis and integration of structural insights of key biomolecular targets, this review aims to enhance optimization efforts in MTDL design, ultimately striving towards a comprehensive cure for the multifaceted pathophysiology of the disease.
Collapse
Affiliation(s)
- Uzma Azam
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Moazzam Naseer
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan; Université de Caen Normandie, Normandie Univ., CERMN, 14000, Caen, France.
| | - Christophe Rochais
- Université de Caen Normandie, Normandie Univ., CERMN, 14000, Caen, France.
| |
Collapse
|
3
|
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
|
4
|
Bayraktar G, Bartolini M, Bolognesi ML, Erdoğan MA, Armağan G, Bayır E, Şendemir A, Bagetta D, Alcaro S, Alptüzün V. Novel multifunctional tacrine-donepezil hybrids against Alzheimer's disease: Design synthesis and bioactivity studies. Arch Pharm (Weinheim) 2024; 357:e2300575. [PMID: 38593283 DOI: 10.1002/ardp.202300575] [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: 10/06/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
A series of tacrine-donepezil hybrids were synthesized as potential multifunctional anti-Alzheimer's disease (AD) compounds. For this purpose, tacrine and the benzylpiperidine moiety of donepezil were fused with a hydrazone group to achieve a small library of tacrine-donepezil hybrids. In agreement with the design, all compounds showed inhibitory activity toward both acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) with IC50 values in the low micromolar range. Kinetic studies on the most potent cholinesterase (ChE) inhibitors within the series showed a mixed-type inhibition mechanism on both enzymes. Also, the docking studies indicated that the compounds inhibit ChEs by dual binding site (DBS) interactions. Notably, tacrine-donepezil hybrids also exhibited significant neuroprotection against H2O2-induced cell death in a differentiated human neuroblastoma (SH-SY5Y) cell line at concentrations close to their IC50 values on ChEs and showed high to medium blood-brain barrier (BBB) permeability on human cerebral microvascular endothelial cells (HBEC-5i). Besides, the compounds do not cause remarkable toxicity in a human hepatocellular carcinoma cell line (HepG2) and SH-SY5Y cells. Additionally, the compounds were predicted to also have good bioavailability. Among the tested compounds, H4, H16, H17, and H24 stand out with their biological profile. Taken together, the proposed novel tacrine-donepezil scaffold represents a promising starting point for the development of novel anti-ChE multifunctional agents against AD.
Collapse
Affiliation(s)
- Gülşah Bayraktar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Mumin Alper Erdoğan
- Department of Physiology, Katip Celebi University School of Medicine, Izmir, Turkey
| | - Güliz Armağan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Ece Bayır
- Ege University Central Research Test and Analysis Laboratory Application and Research Center (EGE-MATAL), Ege University, Izmir, Turkey
| | - Aylin Şendemir
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey
| | - Donatella Bagetta
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Catanzaro, Italy
| | - Vildan Alptüzün
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
5
|
Mohamadpour B, Mirazi N, Komaki A, Basir HS, Hosseini A. Protective effects of selegiline against amyloid beta-induced anxiety-like behavior and memory impairment. Brain Behav 2024; 14:e3599. [PMID: 38873869 PMCID: PMC11176901 DOI: 10.1002/brb3.3599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex and common neurodegenerative disorder. The present study aimed to investigate the potential effects of selegiline (SEL) on various aspects of memory performance, anxiety, and oxidative stress in an AD rat model induced by intracerebroventricular injection of amyloid beta1-42 (Aβ1-42). METHODS Oral administration of SEL at a dose of 0.5 mg/kg/day was performed for 30 consecutive days. Following the 30 days, several tests, including the open-field, elevated plus-maze, novel object recognition, Morris water maze, and passive avoidance learning were conducted to assess locomotor activity, anxiety-like behavior, recognition memory, spatial memory, and passive avoidance memory, respectively. RESULTS The results indicate that the induction of AD in rats led to recognition memory, spatial memory, and passive avoidance memory impairments, as well as increased anxiety. Additionally, the AD rats exhibited a decrease in total antioxidant capacity and an increase in total oxidant status levels, suggesting an imbalance in oxidative-antioxidant status. However, the administration of SEL improved memory performance, reduced anxiety, and modulated oxidative-antioxidant status in AD rats. CONCLUSIONS These findings provide evidence that SEL may alleviate anxiety-like behavior and cognitive deficits induced by Aβ through modulation of oxidative-antioxidant status.
Collapse
Affiliation(s)
- Behnam Mohamadpour
- Department of Biology, Faculty of Basic ScienceBu‐Ali Sina UniversityHamedanIran
| | - Naser Mirazi
- Department of Biology, Faculty of Basic ScienceBu‐Ali Sina UniversityHamedanIran
| | - Alireza Komaki
- Neurophysiology Research CenterHamadan University of Medical SciencesHamadanIran
| | - Hamid Shokati Basir
- Department of Biology, Faculty of Basic ScienceBu‐Ali Sina UniversityHamedanIran
| | - Abdolkarim Hosseini
- Faculty of Life Sciences and BiotechnologyShahid Beheshti UniversityTehranIran
| |
Collapse
|
6
|
Suckling CJ. The allure of targets for novel drugs. RSC Med Chem 2024; 15:472-484. [PMID: 38389887 PMCID: PMC10880906 DOI: 10.1039/d3md00621b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/12/2023] [Indexed: 02/24/2024] Open
Abstract
The challenges of bringing new medicines to patients have been extensively discussed and debated, including consideration of the contribution that academic laboratories can make. At the University of Strathclyde, drug discovery has been a continuing focal activity since the 1960s, and in the past 30 years, the author has led or contributed to many projects of different character and for diverse diseases. A feature common to these projects is the extension of concepts of molecular and biological targets in drug discovery research. In mechanistic terms, these have included compounds that are activators and not inhibitors, and in particular multitargeted compounds. With respect to relevance to disease, schizophrenia, pulmonary disfunction, autoimmune, and infectious disease are most relevant. These projects are discussed in the context of classical medicinal chemistry and more recent concepts in and approaches to drug discovery.
Collapse
Affiliation(s)
- Colin J Suckling
- Department of Pure & Applied Chemistry, University of Strathclyde 295 Cathedral Street Glasgow G1 1Xl Scotland UK
| |
Collapse
|
7
|
Luque FJ, Muñoz-Torrero D. Acetylcholinesterase: A Versatile Template to Coin Potent Modulators of Multiple Therapeutic Targets. Acc Chem Res 2024. [PMID: 38333993 PMCID: PMC10882973 DOI: 10.1021/acs.accounts.3c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
ConspectusThe enzyme acetylcholinesterase (AChE) hydrolyzes the neurotransmitter acetylcholine (ACh) at cholinergic synapses of the peripheral and central nervous system. Thus, it is a prime therapeutic target for diseases that occur with a cholinergic deficit, prominently Alzheimer's disease (AD). Working at a rate near the diffusion limit, it is considered one of nature's most efficient enzymes. This is particularly meritorious considering that its catalytic site is buried at the bottom of a 20-Å-deep cavity, which is preceded by a bottleneck with a diameter shorter than that of the trimethylammonium group of ACh, which has to transit through it. Not only the particular architecture and amino acid composition of its active site gorge enable AChE to largely overcome this potential drawback, but it also offers plenty of possibilities for the design of novel inhibitor drug candidates.In this Account, we summarize our different approaches to colonize the vast territory of the AChE gorge in the pursuit of increased occupancy and hence of inhibitors with increased affinity. We pioneered the use of molecular hybridization to design inhibitors with extended binding at the CAS, reaching affinities among the highest reported so far. Further application of molecular hybridization to grow CAS extended binders by attaching a PAS-binding moiety through suitable linkers led to multisite inhibitors that span the whole length of the gorge, reaching the PAS and even interacting with midgorge residues. We show that multisite AChE inhibitors can also be successfully designed the other way around, by starting with an optimized PAS binder and then colonizing the gorge and CAS. Molecular hybridization from a multicomponent reaction-derived PAS binder afforded a single-digit picomolar multisite AChE inhibitor with more than 1.5 million-fold increased potency relative to the initial hit. This illustrates the powerful alliance between molecular hybridization and gorge occupancy for designing potent AChE inhibitors.Beyond AChE, we show that the stereoelectronic requirements imposed by the AChE gorge for multisite binding have a templating effect that leads to compounds that are active in other key biological targets in AD and other neurological and non-neurological diseases, such as BACE-1 and the aggregation of amyloidogenic proteins (β-amyloid, tau, α-synuclein, prion protein, transthyretin, and human islet amyloid polypeptide). The use of known pharmacophores for other targets as the PAS-binding motif enables the rational design of multitarget agents with multisite binding within AChE and activity against a variety of targets or pathological events, such as oxidative stress and the neuroinflammation-modulating enzyme soluble epoxide hydrolase, among others.We hope that our results can contribute to the development of drug candidates that can modify the course of neurodegeneration and may inspire future works that exploit the power of molecular hybridization in other proteins featuring large cavities.
Collapse
Affiliation(s)
- F Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, E-08921 Santa Coloma de Gramenet, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, E-08028 Barcelona, Spain
- Institute of Theoretical and Computational Chemistry (IQTC), University of Barcelona, E-08028 Barcelona, Spain
| | - Diego Muñoz-Torrero
- Institute of Biomedicine (IBUB), University of Barcelona, E-08028 Barcelona, Spain
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
8
|
Kumar S, Mahajan A, Ambatwar R, Khatik GL. Recent Advancements in the Treatment of Alzheimer's Disease: A Multitarget-directed Ligand Approach. Curr Med Chem 2024; 31:6032-6062. [PMID: 37861025 DOI: 10.2174/0109298673264076230921065945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and one of the leading causes of progressive dementia, affecting 50 million people worldwide. Many pathogenic processes, including amyloid β aggregation, tau hyperphosphorylation, oxidative stress, neuronal death, and deterioration of the function of cholinergic neurons, are associated with its progression. The one-compound-one-target treatment paradigm was unsuccessful in treating AD due to the multifaceted nature of Alzheimer's disease. The recent development of multitarget-directed ligand research has been explored to target the complementary pathways associated with the disease. We aimed to find the key role and progress of MTDLs in treating AD; thus, we searched for the past ten years of literature on "Pub- Med", "ScienceDirect", "ACS" and "Bentham Science" using the keywords neurodegenerative diseases, Alzheimer's disease, and multitarget-directed ligands. The literature was further filtered based on the quality of work and relevance to AD. Thus, this review highlights the current advancement and advantages of multitarget-directed ligands over traditional single-targeted drugs and recent progress in their development to treat AD.
Collapse
Affiliation(s)
- Sumit Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Amol Mahajan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Ramesh Ambatwar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Gopal L Khatik
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| |
Collapse
|
9
|
Zaręba P, Łątka K, Mazur G, Gryzło B, Pasieka A, Godyń J, Panek D, Skrzypczak-Wiercioch A, Höfner GC, Latacz G, Maj M, Espargaró A, Sabaté R, Jóźwiak K, Wanner KT, Sałat K, Malawska B, Kulig K, Bajda M. Discovery of novel multifunctional ligands targeting GABA transporters, butyrylcholinesterase, β-secretase, and amyloid β aggregation as potential treatment of Alzheimer's disease. Eur J Med Chem 2023; 261:115832. [PMID: 37837674 DOI: 10.1016/j.ejmech.2023.115832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/12/2023] [Accepted: 09/23/2023] [Indexed: 10/16/2023]
Abstract
Alzheimer's disease (AD) is a global health problem in the medical sector that will increase over time. The limited treatment of AD leads to the search for a new clinical candidate. Considering the multifactorial nature of AD, a strategy targeting number of regulatory proteins involved in the development of the disease is an effective approach. Here, we present a discovery of new multi-target-directed ligands (MTDLs), purposely designed as GABA transporter (GAT) inhibitors, that successfully provide the inhibitory activity against butyrylcholinesterase (BuChE), β-secretase (BACE1), amyloid β aggregation and calcium channel blockade activity. The selected GAT inhibitors, 19c and 22a - N-benzylamide derivatives of 4-aminobutyric acid, displayed the most prominent multifunctional profile. Compound 19c (mGAT1 IC50 = 10 μM, mGAT4 IC50 = 12 μM and BuChE IC50 = 559 nM) possessed the highest hBACE1 and Aβ40 aggregation inhibitory activity (IC50 = 1.57 μM and 99 % at 10 μM, respectively). Additionally, it showed a decrease in both the elongation and nucleation constants of the amyloid aggregation process. In contrast compound 22a represented the highest activity and a mixed-type of eqBuChE inhibition (IC50 = 173 nM) with hBACE1 (IC50 = 9.42 μM), Aβ aggregation (79 % at 10 μM) and mGATs (mGAT1 IC50 = 30 μM, mGAT4 IC50 = 25 μM) inhibitory activity. Performed molecular docking studies described the mode of interactions with GATs and enzymatic targets. In ADMET in vitro studies both compounds showed acceptable metabolic stability and low neurotoxicity. Successfully, compounds 19c and 22a at the dose of 30 mg/kg possessed statistically significant antiamnesic properties in a mouse model of amnesia caused by scopolamine and assessed in the novel object recognition (NOR) task or the passive avoidance (PA) task.
Collapse
Affiliation(s)
- Paula Zaręba
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Kamil Łątka
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Gabriela Mazur
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Beata Gryzło
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Anna Pasieka
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Justyna Godyń
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Dawid Panek
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Anna Skrzypczak-Wiercioch
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Kraków, Mickiewicz 24/28 St., 30-059, Kraków, Poland
| | - Georg C Höfner
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München Butenandtstr., 5-13, 81377, Munich, Germany
| | - Gniewomir Latacz
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, W. Chodzki 4a St., 20-093, Lublin, Poland
| | - Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028, Barcelona, Spain
| | - Raimon Sabaté
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028, Barcelona, Spain
| | - Krzysztof Jóźwiak
- Department of Biopharmacy, Medical University of Lublin, W. Chodzki 4a St., 20-093, Lublin, Poland
| | - Klaus T Wanner
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München Butenandtstr., 5-13, 81377, Munich, Germany
| | - Kinga Sałat
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Barbara Malawska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Katarzyna Kulig
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Marek Bajda
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland.
| |
Collapse
|
10
|
Fronza MG, Alves D, Praticò D, Savegnago L. The neurobiology and therapeutic potential of multi-targeting β-secretase, glycogen synthase kinase 3β and acetylcholinesterase in Alzheimer's disease. Ageing Res Rev 2023; 90:102033. [PMID: 37595640 DOI: 10.1016/j.arr.2023.102033] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Alzheimer's Disease (AD) is the most common form of dementia, affecting almost 50 million of people around the world, characterized by a complex and age-related progressive pathology with projections to duplicate its incidence by the end of 2050. AD pathology has two major hallmarks, the amyloid beta (Aβ) peptides accumulation and tau hyperphosphorylation, alongside with several sub pathologies including neuroinflammation, oxidative stress, loss of neurogenesis and synaptic dysfunction. In recent years, extensive research pointed out several therapeutic targets which have shown promising effects on modifying the course of the disease in preclinical models of AD but with substantial failure when transposed to clinic trials, suggesting that modulating just an isolated feature of the pathology might not be sufficient to improve brain function and enhance cognition. In line with this, there is a growing consensus that an ideal disease modifying drug should address more than one feature of the pathology. Considering these evidence, β-secretase (BACE1), Glycogen synthase kinase 3β (GSK-3β) and acetylcholinesterase (AChE) has emerged as interesting therapeutic targets. BACE1 is the rate-limiting step in the Aβ production, GSK-3β is considered the main kinase responsible for Tau hyperphosphorylation, and AChE play an important role in modulating memory formation and learning. However, the effects underlying the modulation of these enzymes are not limited by its primarily functions, showing interesting effects in a wide range of impaired events secondary to AD pathology. In this sense, this review will summarize the involvement of BACE1, GSK-3β and AChE on synaptic function, neuroplasticity, neuroinflammation and oxidative stress. Additionally, we will present and discuss new perspectives on the modulation of these pathways on AD pathology and future directions on the development of drugs that concomitantly target these enzymes.
Collapse
Affiliation(s)
- Mariana G Fronza
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis (LASOL), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), UFPel, RS, Brazil
| | - Domenico Praticò
- Alzheimer's Center at Temple - ACT, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Lucielli Savegnago
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil.
| |
Collapse
|
11
|
Liu X, Yu C, Su B, Zha D. Synthesis and properties of the kojic acid dimer and its potential for the treatment of Alzheimer's disease. RSC Med Chem 2023; 14:268-276. [PMID: 36846369 PMCID: PMC9945874 DOI: 10.1039/d2md00383j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The kojic acid dimer (KAD) is a metabolite derived from developing cottonseed when contaminated with aflatoxin. The KAD has been shown to exhibit bright greenish-yellow fluorescence, but little else is known about its biological activity. In this study, using kojic acid as a raw material, we developed a four-step synthetic route that achieved the gram-scale preparation of the KAD in approximately 25% total yield. The structure of the KAD was verified by single-crystal X-ray diffraction. The KAD showed good safety in a variety of cells and had a good protective effect in SH-SY5Y cells. At concentrations lower than 50 μM, the KAD was superior to vitamin C in ABTS+ free radical scavenging assay; the KAD resisted the production of reactive oxygen species induced by H2O2 as confirmed by fluorescence microscopy observation and flow cytometry analysis. Notably, the KAD could enhance the superoxide dismutase activity, which might be the mechanism of its antioxidant activity. The KAD also moderately inhibited the deposition of amyloid-β (Aβ) and selectively chelated Cu2+, Zn2+, Fe2+, Fe3+, and Al3+, which are related to the progress of Alzheimer's disease. Based on its good effects in terms of oxidative stress, neuroprotection, inhibition of Aβ deposition, and metal accumulation, the KAD shows potential for the multi-target treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Xueyan Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University Fuzhou 350004 Fujian Province China .,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University China
| | - Chuanyu Yu
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University Fuzhou 350004 Fujian Province China
| | - Biling Su
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University Fuzhou 350004 Fujian Province China
| | - Daijun Zha
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University Fuzhou 350004 Fujian Province China .,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University China
| |
Collapse
|
12
|
Nicolescu RCB, Maylin ZR, Pérez‐Areales FJ, Iegre J, Pandha HS, Asim M, Spring DR. Hybrid Androgen Receptor Inhibitors Outperform Enzalutamide and EPI-001 in in vitro Models of Prostate Cancer Drug Resistance. ChemMedChem 2023; 18:e202200548. [PMID: 36300876 PMCID: PMC10098645 DOI: 10.1002/cmdc.202200548] [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: 10/10/2022] [Revised: 10/26/2022] [Indexed: 01/20/2023]
Abstract
Androgen receptor targeted therapies for prostate cancer have serious limitations in advanced stages of the disease. While resistance to the FDA-approved enzalutamide is extensively documented, novel therapies based on epichlorohydrin scaffolds (EPI) are currently in clinical trials, but display suboptimal pharmacokinetics. Herein, we report the synthesis and biological characterisation of a novel class of compounds designed through covalently linking enzalutamide and EPI-001 through various triazole based linkers. The compounds display an 18 to 53 fold improvement in the cell killing potency towards C4-2b prostate cancer (PCa) cells compared to the gold standards of therapy, enzalutamide and EPI-001. The most promising compounds were proven to exhibit their toxicity exclusively through androgen receptor (AR) mediated pathways. This work sets the basis for the first class of hybrid AR inhibitors which successfully combine two drug moieties - EPI-001 and enzalutamide - into the same molecule.
Collapse
Affiliation(s)
| | - Zoe R. Maylin
- Department of Clinical and Experimental MedicineUniversity of SurreyGuildfordGU2 TXHUK
| | | | - Jessica Iegre
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Hardev S. Pandha
- Department of Clinical and Experimental MedicineUniversity of SurreyGuildfordGU2 TXHUK
| | - Mohammad Asim
- Department of Clinical and Experimental MedicineUniversity of SurreyGuildfordGU2 TXHUK
| | - David R. Spring
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| |
Collapse
|
13
|
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
|
14
|
In vitro and in vivo biological evaluation of newly synthesized multi-target 20(R)-panaxadiol derivatives for treating Alzheimer's disease. Eur J Med Chem 2022; 244:114825. [DOI: 10.1016/j.ejmech.2022.114825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/18/2022]
|
15
|
Chávez-Gutiérrez E, Martínez-Arellanes M, Murillo-López M, Medina-Guzmán MF, Mobarak-Richaud L, Pelcastre-Guzmán K, Quintana-Romero OJ, Ariza-Castolo A, Ayala-Moreno MDR, Salazar JR, Guerra-Araiza C, Rodríguez-Páez L, Pinto-Almazán R, Loza-Mejía MA. In Combo Studies for the Optimization of 5-Aminoanthranilic Acid Derivatives as Potential Multitarget Drugs for the Management of Metabolic Syndrome. Pharmaceuticals (Basel) 2022; 15:1461. [PMID: 36558912 PMCID: PMC9784827 DOI: 10.3390/ph15121461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Metabolic syndrome is a set of risk factors that consist of abdominal obesity, arterial hypertension, alterations in the lipid profile, and hyperglycemia. The current therapeutic strategy includes polypharmacy, using three or more drugs to control each syndrome component. However, this approach has drawbacks that could lead to therapeutic failure. Multitarget drugs are molecules with the ability to act on different targets simultaneously and are an attractive alternative for treating complex diseases such as metabolic syndrome. Previously, we identified a triamide derivative of 5-aminoanthranilic acid that exhibited hypoglycemic, hypolipemic, and antihypertensive activities simultaneously. In the present study, we report the synthesis and in combo evaluation of new derivatives of anthranilic acid, intending to identify the primary structural factors that improve the activity over metabolic syndrome-related parameters. We found that substitution on position 5, incorporation of 3,4-dimethoxyphenyl substituents, and having a free carboxylic acid group lead to the in vitro inhibition of HMG-CoA reductase, and simultaneously the diminution of the serum levels of glucose, triglycerides, and cholesterol in a diet-induced in vivo model.
Collapse
Affiliation(s)
- Edwin Chávez-Gutiérrez
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
- Doctorado en Ciencias en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación Manuel Carpio y Plan de Ayala s/n, Mexico City 11340, Mexico
| | - Matilda Martínez-Arellanes
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
| | - Montserrat Murillo-López
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
| | - María Fernanda Medina-Guzmán
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
| | - Laila Mobarak-Richaud
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
| | - Karen Pelcastre-Guzmán
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
| | - Osvaldo Javier Quintana-Romero
- Department of Chemistry, Center for Research and Advanced Studies, The National Polytechnic Institute (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | - Armando Ariza-Castolo
- Department of Chemistry, Center for Research and Advanced Studies, The National Polytechnic Institute (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico
| | | | - Juan Rodrigo Salazar
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
| | - Christian Guerra-Araiza
- Medical Research Unit in Pharmacology, Specialities Hospital Bernardo Sepúlveda, National Medical Center XXI Century, Social Security Mexican Institute (IMSS), Av. Cuauhtémoc 330, Mexico City 06720, Mexico
| | - Lorena Rodríguez-Páez
- Biochemistry Department, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación Manuel Carpio y Plan de Ayala s/n, Mexico City 11340, Mexico
| | - Rodolfo Pinto-Almazán
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Mexico City 11340, Mexico
| | - Marco A. Loza-Mejía
- Design, Isolation and Synthesis of Bioactive Molecules Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico
| |
Collapse
|
16
|
Álvarez-Berbel I, Espargaró A, Viayna A, Caballero AB, Busquets MA, Gámez P, Luque FJ, Sabaté R. Three to Tango: Inhibitory Effect of Quercetin and Apigenin on Acetylcholinesterase, Amyloid-β Aggregation and Acetylcholinesterase-Amyloid Interaction. Pharmaceutics 2022; 14:2342. [PMID: 36365159 PMCID: PMC9699245 DOI: 10.3390/pharmaceutics14112342] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 10/13/2023] Open
Abstract
One of the pathological hallmarks of Alzheimer's disease (AD) is the formation of amyloid-β plaques. Since acetylcholinesterase (AChE) promotes the formation of such plaques, the inhibition of this enzyme could slow down the progression of amyloid-β aggregation, hence being complementary to the palliative treatment of cholinergic decline. Antiaggregation assays performed for apigenin and quercetin, which are polyphenolic compounds that exhibit inhibitory properties against the formation of amyloid plaques, reveal distinct inhibitory effects of these compounds on Aβ40 aggregation in the presence and absence of AChE. Furthermore, the analysis of the amyloid fibers formed in the presence of these flavonoids suggests that the Aβ40 aggregates present different quaternary structures, viz., smaller molecular assemblies are generated. In agreement with a noncompetitive inhibition of AChE, molecular modeling studies indicate that these effects may be due to the binding of apigenin and quercetin at the peripheral binding site of AChE. Since apigenin and quercetin can also reduce the generation of reactive oxygen species, the data achieved suggest that multitarget catechol-type compounds may be used for the simultaneous treatment of various biological hallmarks of AD.
Collapse
Affiliation(s)
- Irene Álvarez-Berbel
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Antonio Viayna
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy, Institute of Theoretical and Computational Chemistry (IQTCUB) and Institute of Biomedicine (IBUB), Campus Torribera, University of Barcelona, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain
| | - Ana Belén Caballero
- Department of Inorganic and Organic Chemistry, Faculty of Chemistry, Institute of Nanoscience and Nanotechnology (IN2UB) and NanoBIC, University of Barcelona, 08028 Barcelona, Spain
| | - Maria Antònia Busquets
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Patrick Gámez
- Department of Inorganic and Organic Chemistry, Faculty of Chemistry, Institute of Nanoscience and Nanotechnology (IN2UB) and NanoBIC, University of Barcelona, 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies, Passeig Lluís Companys 23, 08010 Barcelona, Spain
| | - Francisco Javier Luque
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy, Institute of Theoretical and Computational Chemistry (IQTCUB) and Institute of Biomedicine (IBUB), Campus Torribera, University of Barcelona, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain
| | - Raimon Sabaté
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
17
|
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: 7] [Impact Index Per Article: 2.3] [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
|
18
|
Zhang Z, Cheng M, Guo J, Wan Y, Wang R, Fang Y, Jin Y, Xie SS, Liu J. Design, synthesis and biological evaluation of novel pyrazolone derivatives as selective butyrylcholinesterase inhibitors with antioxidant activity against Alzheimer's disease. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Codony S, Pont C, Griñán-Ferré C, Di Pede-Mattatelli A, Calvó-Tusell C, Feixas F, Osuna S, Jarné-Ferrer J, Naldi M, Bartolini M, Loza MI, Brea J, Pérez B, Bartra C, Sanfeliu C, Juárez-Jiménez J, Morisseau C, Hammock BD, Pallàs M, Vázquez S, Muñoz-Torrero D. Discovery and In Vivo Proof of Concept of a Highly Potent Dual Inhibitor of Soluble Epoxide Hydrolase and Acetylcholinesterase for the Treatment of Alzheimer's Disease. J Med Chem 2022; 65:4909-4925. [PMID: 35271276 PMCID: PMC8958510 DOI: 10.1021/acs.jmedchem.1c02150] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
With innumerable clinical failures of target-specific drug candidates for multifactorial diseases, such as Alzheimer's disease (AD), which remains inefficiently treated, the advent of multitarget drug discovery has brought a new breath of hope. Here, we disclose a class of 6-chlorotacrine (huprine)-TPPU hybrids as dual inhibitors of the enzymes soluble epoxide hydrolase (sEH) and acetylcholinesterase (AChE), a multitarget profile to provide cumulative effects against neuroinflammation and memory impairment. Computational studies confirmed the gorge-wide occupancy of both enzymes, from the main site to a secondary site, including a so far non-described AChE cryptic pocket. The lead compound displayed in vitro dual nanomolar potencies, adequate brain permeability, aqueous solubility, human microsomal stability, lack of neurotoxicity, and it rescued memory, synaptic plasticity, and neuroinflammation in an AD mouse model, after low dose chronic oral administration.
Collapse
Affiliation(s)
- Sandra Codony
- Laboratory
of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy
and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Caterina Pont
- Laboratory
of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy
and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology
Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry,
Faculty of Pharmacy and Food Sciences, and Institute of Neurosciences, University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Ania Di Pede-Mattatelli
- Department
of Pharmacy and Pharmaceutical Technology and Physical Chemistry,
Faculty of Pharmacy and Food Sciences, and Institute of Theoretical
and Computational Chemistry (IQTCUB), University
of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Carla Calvó-Tusell
- CompBioLab
Group, Departament de Química and Institut de Química
Computacional i Catàlisi (IQCC), Universitat de Girona, C/ Maria Aurèlia Capmany 69, E-17003 Girona, Spain
| | - Ferran Feixas
- CompBioLab
Group, Departament de Química and Institut de Química
Computacional i Catàlisi (IQCC), Universitat de Girona, C/ Maria Aurèlia Capmany 69, E-17003 Girona, Spain
| | - Sílvia Osuna
- CompBioLab
Group, Departament de Química and Institut de Química
Computacional i Catàlisi (IQCC), Universitat de Girona, C/ Maria Aurèlia Capmany 69, E-17003 Girona, Spain,Institució
Catalana de Recerca i Estudis Avançats (ICREA), E-08010 Barcelona, Spain
| | - Júlia Jarné-Ferrer
- Pharmacology
Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry,
Faculty of Pharmacy and Food Sciences, and Institute of Neurosciences, University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Marina Naldi
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Belmeloro, 6, I-40126 Bologna, Italy
| | - Manuela Bartolini
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Belmeloro, 6, I-40126 Bologna, Italy
| | - María Isabel Loza
- BioFarma
Research Group, Centro Singular de Investigación en Medicina
Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Av. de Barcelona s/n, E-15782 Santiago de Compostela, Spain
| | - José Brea
- BioFarma
Research Group, Centro Singular de Investigación en Medicina
Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Av. de Barcelona s/n, E-15782 Santiago de Compostela, Spain
| | - Belén Pérez
- Department
of Pharmacology, Therapeutics and Toxicology, Autonomous University of Barcelona, E-08193 Bellaterra, Spain
| | - Clara Bartra
- Institute
of Biomedical Research of Barcelona, CSIC and Institut d’Investigacions
Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló, 149, E-08036 Barcelona, Spain
| | - Coral Sanfeliu
- Institute
of Biomedical Research of Barcelona, CSIC and Institut d’Investigacions
Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló, 149, E-08036 Barcelona, Spain
| | - Jordi Juárez-Jiménez
- Department
of Pharmacy and Pharmaceutical Technology and Physical Chemistry,
Faculty of Pharmacy and Food Sciences, and Institute of Theoretical
and Computational Chemistry (IQTCUB), University
of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Christophe Morisseau
- Department
of Entomology and Nematology and Comprehensive Cancer Center, University of California, One Shields Avenue, Davis, California 95616, United States
| | - Bruce D. Hammock
- Department
of Entomology and Nematology and Comprehensive Cancer Center, University of California, One Shields Avenue, Davis, California 95616, United States
| | - Mercè Pallàs
- Pharmacology
Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry,
Faculty of Pharmacy and Food Sciences, and Institute of Neurosciences, University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Santiago Vázquez
- Laboratory
of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy
and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain,. Phone: (+34) 934024533
| | - Diego Muñoz-Torrero
- Laboratory
of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy
and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028 Barcelona, Spain,. Phone: (+34) 934024533
| |
Collapse
|
20
|
Singh YP, Kumar N, Priya K, Chauhan BS, Shankar G, Kumar S, Singh GK, Srikrishna S, Garg P, Singh G, Rai G, Modi G. Exploration of Neuroprotective Properties of a Naturally Inspired Multifunctional Molecule (F24) against Oxidative Stress and Amyloid β Induced Neurotoxicity in Alzheimer's Disease Models. ACS Chem Neurosci 2022; 13:27-42. [PMID: 34931800 DOI: 10.1021/acschemneuro.1c00443] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pathological hallmarks of Alzheimer's disease (AD) are manifested as an increase in the level of oxidative stress and aggregation of the amyloid-β protein. In vitro, in vivo, and in silico experiments were designed and carried out with multifunctional cholinergic inhibitor, F24 (EJMC-7a) to explore its neuroprotective effects in AD models. The neuroprotection ability of F24 was tested in SH-SY5Y cells, a widely used neuronal cell line. The pretreatment and subsequent co-treatment of SH-SY5Y cells with different doses of F24 was effective in rescuing the cells from H2O2 induced neurotoxicity. F24 treated cells were found to be effective in the reduction of cellular reactive oxygen species, DNA damage, and Aβ1-42 induced neurotoxicity, which validated its neuroprotective effectiveness. F24 exhibited efficacy in an in vivo Drosophila model by rescuing eye phenotypes from degeneration caused by Aβ toxicity. Further, computational studies were carried out to monitor the interaction between F24 and Aβ1-42 aggregates. The computational studies corroborated our in vitro and in vivo studies suggesting Aβ1-42 aggregation modulation ability of F24. The brain entry ability of F24 was studied in the parallel artificial membrane permeability assay. Finally, F24 was tested at doses of 1 and 2.5 mg/kg in the Morris water maze AD model. The neuroprotective properties shown by F24 strongly suggest that multifunctional features of this molecule provide symptomatic relief and act as a disease-modifying agent in the treatment of AD. The results from our experiments strongly indicated that natural template-based F24 could serve as a lead molecule for further investigation to explore multifunctional therapeutic agents for AD management.
Collapse
Affiliation(s)
- Yash Pal Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Navneet Kumar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160062 Punjab, India
| | - Khushbu Priya
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Brijesh Singh Chauhan
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Gauri Shankar
- 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
| | - Gireesh Kumar Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya, 824236 Bihar, India
| | - Saripella Srikrishna
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160062 Punjab, India
| | - Gourav Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Geeta Rai
- Department of Molecular and Human Genetics, 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
|
21
|
From virtual screening hits targeting a cryptic pocket in BACE-1 to a nontoxic brain permeable multitarget anti-Alzheimer lead with disease-modifying and cognition-enhancing effects. Eur J Med Chem 2021; 225:113779. [PMID: 34418785 DOI: 10.1016/j.ejmech.2021.113779] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/22/2022]
Abstract
Starting from six potential hits identified in a virtual screening campaign directed to a cryptic pocket of BACE-1, at the edge of the catalytic cleft, we have synthesized and evaluated six hybrid compounds, designed to simultaneously reach BACE-1 secondary and catalytic sites and to exert additional activities of interest for Alzheimer's disease (AD). We have identified a lead compound with potent in vitro activity towards human BACE-1 and cholinesterases, moderate Aβ42 and tau antiaggregating activity, and brain permeability, which is nontoxic in neuronal cells and zebrafish embryos at concentrations above those required for the in vitro activities. This compound completely restored short- and long-term memory in a mouse model of AD (SAMP8) relative to healthy control strain SAMR1, shifted APP processing towards the non-amyloidogenic pathway, reduced tau phosphorylation, and increased the levels of synaptic proteins PSD95 and synaptophysin, thereby emerging as a promising disease-modifying, cognition-enhancing anti-AD lead.
Collapse
|
22
|
de Andrade Ramos G, Souza de Oliveira A, Bartolini M, Naldi M, Liparulo I, Bergamini C, Uliassi E, Wu L, Fraser PE, Abreu M, Kiametis AS, Gargano R, Silveira ER, Brand GD, Prchal L, Soukup O, Korábečný J, Bolognesi ML, Soares Romeiro LA. Discovery of sustainable drugs for Alzheimer's disease: cardanol-derived cholinesterase inhibitors with antioxidant and anti-amyloid properties. RSC Med Chem 2021; 12:1154-1163. [PMID: 34355181 PMCID: PMC8293282 DOI: 10.1039/d1md00046b] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/29/2021] [Indexed: 01/21/2023] Open
Abstract
As part of our efforts to develop sustainable drugs for Alzheimer's disease (AD), we have been focusing on the inexpensive and largely available cashew nut shell liquid (CNSL) as a starting material for the identification of new acetylcholinesterase (AChE) inhibitors. Herein, we decided to investigate whether cardanol, a phenolic CNSL component, could serve as a scaffold for improved compounds with concomitant anti-amyloid and antioxidant activities. Ten new derivatives, carrying the intact phenolic function and an aminomethyl functionality, were synthesized and first tested for their inhibitory potencies towards AChE and butyrylcholinesterase (BChE). 5 and 11 were found to inhibit human BChE at a single-digit micromolar concentration. Transmission electron microscopy revealed the potential of five derivatives to modulate Aβ aggregation, including 5 and 11. In HORAC assays, 5 and 11 performed similarly to standard antioxidant ferulic acid as hydroxyl scavenging agents. Furthermore, in in vitro studies in neuronal cell cultures, 5 and 11 were found to effectively inhibit reactive oxygen species production at a 10 μM concentration. They also showed a favorable initial ADME/Tox profile. Overall, these results suggest that CNSL is a promising raw material for the development of potential disease-modifying treatments for AD.
Collapse
Affiliation(s)
- Giselle de Andrade Ramos
- Department of Pharmacy, Health Sciences Faculty, University of Brasília, Campus Universitário Darcy Ribeiro 70910-900 Brasília DF Brazil
| | - Andressa Souza de Oliveira
- Department of Pharmacy, Health Sciences Faculty, University of Brasília, Campus Universitário Darcy Ribeiro 70910-900 Brasília DF Brazil
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, University of Bologna Via Belmeloro 6 40126 Bologna Italy
| | - Marina Naldi
- Department of Pharmacy and Biotechnology, University of Bologna Via Belmeloro 6 40126 Bologna Italy
| | - Irene Liparulo
- Department of Pharmacy and Biotechnology, University of Bologna Via Belmeloro 6 40126 Bologna Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, University of Bologna Via Belmeloro 6 40126 Bologna Italy
| | - Elisa Uliassi
- Department of Pharmacy and Biotechnology, University of Bologna Via Belmeloro 6 40126 Bologna Italy
| | - Ling Wu
- Tanz Centre for Research in Neurodegenerative Diseases and Dept. of Medical Biophysics, University of Toronto Krembil Discovery Tower, 60 Leonard Avenue, 6KD-402 M5T 2S8 Toronto ON Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Dept. of Medical Biophysics, University of Toronto Krembil Discovery Tower, 60 Leonard Avenue, 6KD-402 M5T 2S8 Toronto ON Canada
| | - Monica Abreu
- Physics Institute, University of Brasília, Campus Universitário Darcy Ribeiro 70910-900 Brasília DF Brazil
| | - Alessandra Sofia Kiametis
- Physics Institute, University of Brasília, Campus Universitário Darcy Ribeiro 70910-900 Brasília DF Brazil
| | - Ricardo Gargano
- Physics Institute, University of Brasília, Campus Universitário Darcy Ribeiro 70910-900 Brasília DF Brazil
| | - Edilberto Rocha Silveira
- CENAUREMN, Department of Organic and Inorganic Chemistry, Federal University of Ceará 60021-970 Fortaleza CE Brazil
| | - Guilherme D Brand
- Chemistry Institute, University of Brasília, Campus Universitário Darcy Ribeiro 70910-900 Brasília DF Brazil
| | - Lukas Prchal
- Biomedical Research Centre, University Hospital Hradec Kralove Sokolska 581, 500 05 Hradec Kralove Czech Republic
| | - Ondřej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove Sokolska 581, 500 05 Hradec Kralove Czech Republic
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence Trebesska 1575, 500 01 Hradec Kralove Czech Republic
| | - Jan Korábečný
- Biomedical Research Centre, University Hospital Hradec Kralove Sokolska 581, 500 05 Hradec Kralove Czech Republic
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence Trebesska 1575, 500 01 Hradec Kralove Czech Republic
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, University of Bologna Via Belmeloro 6 40126 Bologna Italy
| | - Luiz Antonio Soares Romeiro
- Department of Pharmacy, Health Sciences Faculty, University of Brasília, Campus Universitário Darcy Ribeiro 70910-900 Brasília DF Brazil
| |
Collapse
|
23
|
Zhang H, Song Q, Yu G, Cao Z, Qiang X, Liu X, Deng Y. Phthalimide-(N-alkylbenzylamine) cysteamide hybrids as multifunctional agents against Alzheimer's disease: Design, synthesis, and biological evaluation. Chem Biol Drug Des 2021; 98:493-500. [PMID: 34143938 DOI: 10.1111/cbdd.13905] [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: 12/21/2020] [Revised: 04/13/2021] [Accepted: 06/06/2021] [Indexed: 11/30/2022]
Abstract
The complex pathogenesis of Alzheimer's disease (AD) calls for multi-target approach for disease treatment. Herein, based on the MTDLs strategy, a series of phthalimide-(N-alkylbenzylamine) cysteamide hybrids were designed, synthesized, and investigated in vitro for the purpose. Most of the target compounds were found to be potential multi-target agents. In vitro results showed that compound 9e was the representative compound in this series, endowed with high EeAChE and HuAChE inhibitory potency (IC50 = 1.55 µm and 2.23 µm, respectively), good inhibitory activity against self-induced Aβ1-42 aggregation (36.08% at 25 µm), and moderate antioxidant capacity (ORAC-FL value was 0.68 Trolox equivalents). Molecular docking studies rationalized the binding mode of 9e in both PAS and CAS of AChE. Moreover, 9e displayed excellent ability to against H2 O2 -induced PC12 cell injury and penetrate BBB. Overall, these results highlighted that compound 9e was an effective and promising multi-target agent for further anti-AD drug development.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qing Song
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Guangjun Yu
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhongcheng Cao
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xiaoming Qiang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xiuxiu Liu
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yong Deng
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Pasieka A, Panek D, Szałaj N, Espargaró A, Więckowska A, Malawska B, Sabaté R, Bajda M. Dual Inhibitors of Amyloid-β and Tau Aggregation with Amyloid-β Disaggregating Properties: Extended In Cellulo, In Silico, and Kinetic Studies of Multifunctional Anti-Alzheimer's Agents. ACS Chem Neurosci 2021; 12:2057-2068. [PMID: 34019757 PMCID: PMC8291496 DOI: 10.1021/acschemneuro.1c00235] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
![]()
In Alzheimer’s
disease, neurons slowly degenerate due to
the accumulation of misfolded amyloid β and tau proteins. In
our research, we performed extended studies directed at amyloid β
and tau aggregation inhibition using in cellulo (Escherichia coli model of protein aggregation), in silico, and in vitro kinetic studies.
We tested our library of 1-benzylamino-2-hydroxyalkyl multifunctional
anti-Alzheimer’s agents and identified very potent dual aggregation
inhibitors. Among the tested derivatives, we selected compound 18, which exhibited a unique profile of biological activity.
This compound was the most potent and balanced dual aggregation inhibitor
(Aβ42 inhibition (inh.) 80.0%, tau inh. 68.3% in
10 μM), with previously reported in vitro inhibitory
activity against hBuChE, hBACE1,
and Aβ (hBuChE IC50 = 5.74 μM; hBACE1 IC50 = 41.6 μM; Aβ aggregation
(aggr.) inh. IC50 = 3.09 μM). In docking studies
for both proteins, we tried to explain the different structural requirements
for the inhibition of Aβ vs tau. Moreover, docking and kinetic
studies showed that compound 18 could inhibit the amyloid
aggregation process at several steps and also displayed disaggregating
properties. These results may help to design the next generations
of dual or selective aggregation inhibitors.
Collapse
Affiliation(s)
- Anna Pasieka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028 Barcelona, Spain
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Raimon Sabaté
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028 Barcelona, Spain
| | - Marek Bajda
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| |
Collapse
|
25
|
Yao H, Uras G, Zhang P, Xu S, Yin Y, Liu J, Qin S, Li X, Allen S, Bai R, Gong Q, Zhang H, Zhu Z, Xu J. Discovery of Novel Tacrine-Pyrimidone Hybrids as Potent Dual AChE/GSK-3 Inhibitors for the Treatment of Alzheimer's Disease. J Med Chem 2021; 64:7483-7506. [PMID: 34024109 DOI: 10.1021/acs.jmedchem.1c00160] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Based on a multitarget strategy, a series of novel tacrine-pyrimidone hybrids were identified for the potential treatment of Alzheimer's disease (AD). Biological evaluation results demonstrated that these hybrids exhibited significant inhibitory activities toward acetylcholinesterase (AChE) and glycogen synthase kinase 3 (GSK-3). The optimal compound 27g possessed excellent dual AChE/GSK-3 inhibition both in terms of potency and equilibrium (AChE: IC50 = 51.1 nM; GSK-3β: IC50 = 89.3 nM) and displayed significant amelioration on cognitive deficits in scopolamine-induced amnesia mice and efficient reduction against phosphorylation of tau protein on Ser-199 and Ser-396 sites in glyceraldehyde (GA)-stimulated differentiated SH-SY5Y cells. Furthermore, compound 27g exhibited eligible pharmacokinetic properties, good kinase selectivity, and moderate neuroprotection against GA-induced reduction in cell viability and neurite damage in SH-SY5Y-derived neurons. The multifunctional profiles of compound 27g suggest that it deserves further investigation as a promising lead for the prospective treatment of AD.
Collapse
Affiliation(s)
- Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Giuseppe Uras
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Pengfei Zhang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Ying Yin
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.,CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Jie Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Shuai Qin
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Xinuo Li
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Stephanie Allen
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Renren Bai
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China
| | - Qi Gong
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Haiyan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| |
Collapse
|
26
|
Pasieka A, Panek D, Jończyk J, Godyń J, Szałaj N, Latacz G, Tabor J, Mezeiova E, Chantegreil F, Dias J, Knez D, Lu J, Pi R, Korabecny J, Brazzolotto X, Gobec S, Höfner G, Wanner K, Więckowska A, Malawska B. Discovery of multifunctional anti-Alzheimer's agents with a unique mechanism of action including inhibition of the enzyme butyrylcholinesterase and γ-aminobutyric acid transporters. Eur J Med Chem 2021; 218:113397. [PMID: 33838585 DOI: 10.1016/j.ejmech.2021.113397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/31/2022]
Abstract
Looking for an effective anti-Alzheimer's agent is very challenging; however, a multifunctional ligand strategy may be a promising solution for the treatment of this complex disease. We herein present the design, synthesis and biological evaluation of novel hydroxyethylamine derivatives displaying unique, multiple properties that have not been previously reported. The original mechanism of action combines inhibitory activity against disease-modifying targets: β-secretase enzyme (BACE1) and amyloid β (Aβ) aggregation, along with an effect on targets associated with symptom relief - inhibition of butyrylcholinesterase (BuChE) and γ-aminobutyric acid transporters (GATs). Among the obtained molecules, compound 36 exhibited the most balanced and broad activity profile (eeAChE IC50 = 2.86 μM; eqBuChE IC50 = 60 nM; hBuChE IC50 = 20 nM; hBACE1 IC50 = 5.9 μM; inhibition of Aβ aggregation = 57.9% at 10 μM; mGAT1 IC50 = 10.96 μM; and mGAT2 IC50 = 19.05 μM). Moreover, we also identified 31 as the most potent mGAT4 and hGAT3 inhibitor (IC50 = 5.01 μM and IC50 = 2.95 μM, respectively), with high selectivity over other subtypes. Compounds 36 and 31 represent new anti-Alzheimer agents that can ameliorate cognitive decline and modify the progress of disease.
Collapse
Affiliation(s)
- Anna Pasieka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Jakub Jończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Julia Tabor
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Eva Mezeiova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Fabien Chantegreil
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - José Dias
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Damijan Knez
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Junfeng Lu
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Rongbiao Pi
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Georg Höfner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Klaus Wanner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
27
|
Shukur KT, Ercetin T, Luise C, Sippl W, Sirkecioglu O, Ulgen M, Coskun GP, Yarim M, Gazi M, Gulcan HO. Design, synthesis, and biological evaluation of new urolithin amides as multitarget agents against Alzheimer's disease. Arch Pharm (Weinheim) 2021; 354:e2000467. [PMID: 33511649 DOI: 10.1002/ardp.202000467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 12/31/2022]
Abstract
A series of urolithin amide (i.e., URO-4-URO-10 and THU-4-THU-10) derivatives was designed and synthesized, and their chemical structures were confirmed with spectroscopic techniques and elemental analysis. The title compounds and synthesis intermediates (THU-1-THU-10 and URO-1-URO-10) were evaluated for their potential to inhibit acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), and monoamine oxidase B (MAO-B). Compounds THU-4 and THU-8 were found to be the most potent inhibitors for the cholinesterases and MAO-B, respectively. The docking studies were also employed to evaluate the binding modes of the most active compounds with AChE, BuChE, and MAO-B. Furthermore, the moderate-to-strong activities of the compounds were also displayed in amyloid-beta inhibition and antioxidant assay systems. The results pointed out that the urolithin scaffold can be employed in drug design studies for the development of multitarget ligands acting on various cascades shown to be important within the pathophysiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Karar T Shukur
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, TR North Cyprus, Turkey
| | - Tugba Ercetin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, TR North Cyprus, Turkey
| | - Chiara Luise
- Institute of Pharmacy, Martin Luther University of Halle-Wittenberg, Halle (Saale), Germany
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin Luther University of Halle-Wittenberg, Halle (Saale), Germany
| | - Okan Sirkecioglu
- Department of Chemistry, Istanbul Technical University, Istanbul, Turkey
| | - Mert Ulgen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Acibadem University, Istanbul, Turkey
| | - Goknil P Coskun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Acibadem University, Istanbul, Turkey
| | - Mine Yarim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Mustafa Gazi
- Faculty of Arts and Science, Eastern Mediterranean University, Famagusta, TR North Cyprus, Turkey
| | - Hayrettin O Gulcan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, TR North Cyprus, Turkey
| |
Collapse
|
28
|
Malafaia D, Albuquerque HMT, Silva AMS. Amyloid-β and tau aggregation dual-inhibitors: A synthetic and structure-activity relationship focused review. Eur J Med Chem 2021; 214:113209. [PMID: 33548635 DOI: 10.1016/j.ejmech.2021.113209] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/16/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is one of the most common types of dementia, especially in elderly, with an increasing number of people suffering from this disease worldwide. There are no available disease-modifying therapies and only four drugs are approved for the relief of symptoms. Currently, the therapeutic approach used for AD treatment is based on single target drugs, which are not capable to stop its progression. To address this issue, multi-target compounds, combining two or more pharmacophores in a single molecular entity, have gained increasing interest to deal with the multiple factors related to AD. The exact cause of AD is not yet completely disclosed, and several hallmarks have been associated to this neurodegenerative disease. Even though, the accumulation of both amyloid-β plaques (Aβ) and neurofibrillary tangles (NFTs) are fully accepted as the main AD hallmarks, being object of lots of research for early-stage diagnosis and pharmacological therapy. In this context, this review summarizes the state-of-the-art in the field of dual-target inhibitors of both Aβ and tau aggregation simultaneously, including the design and synthetic strategy of the dual-target compounds, as well as a brief structure-activity relationships (SAR) analysis.
Collapse
Affiliation(s)
- Daniela Malafaia
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| | - Hélio M T Albuquerque
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal.
| | - Artur M S Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
29
|
Viayna E, Coquelle N, Cieslikiewicz-Bouet M, Cisternas P, Oliva CA, Sánchez-López E, Ettcheto M, Bartolini M, De Simone A, Ricchini M, Rendina M, Pons M, Firuzi O, Pérez B, Saso L, Andrisano V, Nachon F, Brazzolotto X, García ML, Camins A, Silman I, Jean L, Inestrosa NC, Colletier JP, Renard PY, Muñoz-Torrero D. Discovery of a Potent Dual Inhibitor of Acetylcholinesterase and Butyrylcholinesterase with Antioxidant Activity that Alleviates Alzheimer-like Pathology in Old APP/PS1 Mice. J Med Chem 2020; 64:812-839. [PMID: 33356266 DOI: 10.1021/acs.jmedchem.0c01775] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The combination of the scaffolds of the cholinesterase inhibitor huprine Y and the antioxidant capsaicin results in compounds with nanomolar potencies toward human acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) that retain or improve the antioxidant properties of capsaicin. Crystal structures of their complexes with AChE and BChE revealed the molecular basis for their high potency. Brain penetration was confirmed by biodistribution studies in C57BL6 mice, with one compound (5i) displaying better brain/plasma ratio than donepezil. Chronic treatment of 10 month-old APP/PS1 mice with 5i (2 mg/kg, i.p., 3 times per week, 4 weeks) rescued learning and memory impairments, as measured by three different behavioral tests, delayed the Alzheimer-like pathology progression, as suggested by a significantly reduced Aβ42/Aβ40 ratio in the hippocampus, improved basal synaptic efficacy, and significantly reduced hippocampal oxidative stress and neuroinflammation. Compound 5i emerges as an interesting anti-Alzheimer lead with beneficial effects on cognitive symptoms and on some underlying disease mechanisms.
Collapse
Affiliation(s)
- Elisabet Viayna
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Nicolas Coquelle
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS UMR 5075, F-38054 Grenoble, France.,Large Scale Structures Group, Institut Laue-Langevin, F-38042 Grenoble Cedex 9, France
| | | | - Pedro Cisternas
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, P.O. Box 114, 8331150 Santiago, Chile
| | - Carolina A Oliva
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, P.O. Box 114, 8331150 Santiago, Chile
| | - Elena Sánchez-López
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, E-28031 Madrid, Spain
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, E-28031 Madrid, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neuroscience, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University Rovira i Virgili, E-43201 Reus, Spain
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Angela De Simone
- Department of Drug Science and Technology, University of Turin, I-10125 Torino, Italy
| | - Mattia Ricchini
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Marisa Rendina
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Mégane Pons
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA (UMR 6014), 76000 Rouen, France
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, PO Box 3288, 71345 Shiraz, Iran
| | - Belén Pérez
- Department of Pharmacology, Therapeutics and Toxicology, Autonomous University of Barcelona, E-08193 Bellaterra, Barcelona, Spain
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Vincenza Andrisano
- Department for Life Quality Studies, University of Bologna, Corso d'Augusto 237, I-47921 Rimini, Italy
| | - Florian Nachon
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées BP73, 91993 Brétigny sur Orge, France
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées BP73, 91993 Brétigny sur Orge, France
| | - Maria Luisa García
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, E-28031 Madrid, Spain
| | - Antoni Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, E-28031 Madrid, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neuroscience, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Israel Silman
- Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Ludovic Jean
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA (UMR 6014), 76000 Rouen, France
| | - Nibaldo C Inestrosa
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, P.O. Box 114, 8331150 Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, 6200000 Punta Arenas, Chile
| | - Jacques-Philippe Colletier
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS UMR 5075, F-38054 Grenoble, France
| | - Pierre-Yves Renard
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA (UMR 6014), 76000 Rouen, France
| | - Diego Muñoz-Torrero
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| |
Collapse
|