1
|
Herrera-Arozamena C, Estrada-Valencia M, García-Díez G, Pérez C, León R, Infantes L, Morales-García JA, Pérez-Castillo A, Del Sastre E, López MG, Rodríguez-Franco MI. Discovery of a potent melatonin-based inhibitor of quinone reductase-2 with neuroprotective and neurogenic properties. Eur J Med Chem 2024; 277:116763. [PMID: 39146834 DOI: 10.1016/j.ejmech.2024.116763] [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: 06/12/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
5-Methoxy-3-(5-methoxyindolin-2-yl)-1H-indole (3), whose structure was unambiguously elucidated by X-ray analysis, was identified as a multi-target compound with potential application in neurodegenerative diseases. It is a low nanomolar inhibitor of QR2 (IC50 = 7.7 nM), with greater potency than melatonin and comparable efficacy to the most potent QR2 inhibitors described to date. Molecular docking studies revealed the potential binding mode of 3 to QR2, which explains its superior potency compared to melatonin. Furthermore, compound 3 inhibits hMAO-A, hMAO-B and hLOX-5 in the low micromolar range and is an excellent ROS scavenger. In phenotypic assays, compound 3 showed neuroprotective activity in a cellular model of oxidative stress damage, it was non-toxic, and was able to activate neurogenesis from neural stem-cell niches of adult mice. These excellent biological properties, together with its both good in silico and in vitro drug-like profile, highlight compound 3 as a promising drug candidate for neurodegenerative diseases.
Collapse
Affiliation(s)
- Clara Herrera-Arozamena
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Martín Estrada-Valencia
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Guillermo García-Díez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Concepción Pérez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Lourdes Infantes
- Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas (IQF-CSIC), C/ Serrano 119, E-28006, Madrid, Spain
| | - José A Morales-García
- Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Avda. Complutense s/n, E-28040, Madrid, Spain
| | - Ana Pérez-Castillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), C/ Arturo Duperier 4, E-28029, Madrid, Spain
| | - Eric Del Sastre
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), C/ Arzobispo Morcillo 4, E-28029, Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), C/ Arzobispo Morcillo 4, E-28029, Madrid, Spain
| | - María Isabel Rodríguez-Franco
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain.
| |
Collapse
|
2
|
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
|
3
|
Angelova VT, Georgiev B, Pencheva T, Pajeva I, Rangelov M, Todorova N, Zheleva-Dimitrova D, Kalcheva-Yovkova E, Valkova IV, Vassilev N, Mihaylova R, Stefanova D, Petrov B, Voynikov Y, Tzankova V. Design, Synthesis, In Silico Studies and In Vitro Evaluation of New Indole- and/or Donepezil-like Hybrids as Multitarget-Directed Agents for Alzheimer's Disease. Pharmaceuticals (Basel) 2023; 16:1194. [PMID: 37765003 PMCID: PMC10534827 DOI: 10.3390/ph16091194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease (AD) is considered a complex neurodegenerative condition which warrants the development of multitargeted drugs to tackle the key pathogenetic mechanisms of the disease. In this study, two novel series of melatonin- and donepezil-based hybrid molecules with hydrazone (3a-r) or sulfonyl hydrazone (5a-l) fragments were designed, synthesized, and evaluated as multifunctional ligands against AD-related neurodegenerative mechanisms. Two lead compounds (3c and 3d) exhibited a well-balanced multifunctional profile, demonstrating intriguing acetylcholinesterase (AChE) inhibition, promising antioxidant activity assessed by DPPH, ABTS, and FRAP methods, as well as the inhibition of lipid peroxidation in the linoleic acid system. Compound 3n, possessing two indole scaffolds, showed the highest activity against butyrylcholinesterase (BChE) and a high selectivity index (SI = 47.34), as well as a pronounced protective effect in H2O2-induced oxidative stress in SH-SY5Y cells. Moreover, compounds 3c, 3d, and 3n showed low neurotoxicity against malignant neuroblastoma cell lines of human (SH-SY5Y) and murine (Neuro-2a) origin, as well as normal murine fibroblast cells (CCL-1) that indicate the in vitro biocompatibility of the experimental compounds. Furthermore, compounds 3c, 3d, and 3n were capable of penetrating the blood-brain barrier (BBB) in the experimental PAMPA-BBB study. The molecular docking showed that compound 3c could act as a ligand to both MT1 and MT2 receptors, as well as to AchE and BchE enzymes. Taken together, those results outline compounds 3c, 3d, and 3n as promising prototypes in the search of innovative compounds for the treatment of AD-associated neurodegeneration with oxidative stress. This study demonstrates that hydrazone derivatives with melatonin and donepezil are appropriate for further development of new AChE/BChE inhibitory agents.
Collapse
Affiliation(s)
- Violina T. Angelova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (I.V.V.); (Y.V.)
| | - Borislav Georgiev
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (B.G.); (N.T.)
| | - Tania Pencheva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (T.P.); (I.P.)
| | - Ilza Pajeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (T.P.); (I.P.)
| | - Miroslav Rangelov
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (M.R.); (N.V.)
| | - Nadezhda Todorova
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (B.G.); (N.T.)
| | | | - Elena Kalcheva-Yovkova
- Faculty of Computer Systems and Techologies, Technical University–Sofia, 1000 Sofia, Bulgaria;
| | - Iva V. Valkova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (I.V.V.); (Y.V.)
| | - Nikolay Vassilev
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (M.R.); (N.V.)
| | - Rositsa Mihaylova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (R.M.); (D.S.); (B.P.); (V.T.)
| | - Denitsa Stefanova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (R.M.); (D.S.); (B.P.); (V.T.)
| | - Boris Petrov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (R.M.); (D.S.); (B.P.); (V.T.)
| | - Yulian Voynikov
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (I.V.V.); (Y.V.)
| | - Virginia Tzankova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (R.M.); (D.S.); (B.P.); (V.T.)
| |
Collapse
|
4
|
Amoroso R, Maccallini C, Bellezza I. Activators of Nrf2 to Counteract Neurodegenerative Diseases. Antioxidants (Basel) 2023; 12:antiox12030778. [PMID: 36979026 PMCID: PMC10045503 DOI: 10.3390/antiox12030778] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/15/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Neurodegenerative diseases are incurable and debilitating conditions that result in progressive degeneration and loss of nerve cells. Oxidative stress has been proposed as one factor that plays a potential role in the pathogenesis of neurodegenerative disorders since neuron cells are particularly vulnerable to oxidative damage. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is strictly related to anti-inflammatory and antioxidative cell response; therefore, its activation and the consequent enhancement of the related cellular pathways have been proposed as a potential therapeutic approach. Several Nrf2 activators with different mechanisms and diverse structures have been reported, but those applied for neurodisorders are still limited. However, in the very last few years, interesting progress has been made, particularly in enhancing the blood-brain barrier penetration, to make Nrf2 activators effective drugs, and in designing Nrf2-based multitarget-directed ligands to affect multiple pathways involved in the pathology of neurodegenerative diseases. The present review gives an overview of the most representative findings in this research area.
Collapse
Affiliation(s)
- Rosa Amoroso
- Department of Pharmacy, University "G.d'Annunzio" of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
| | - Cristina Maccallini
- Department of Pharmacy, University "G.d'Annunzio" of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
| | - Ilaria Bellezza
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant'Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| |
Collapse
|
5
|
8-Amide and 8-carbamate substitution patterns as modulators of 7-hydroxy-4-methylcoumarin's antidepressant profile: Synthesis, biological evaluation and docking studies. Eur J Med Chem 2023; 248:115091. [PMID: 36638711 DOI: 10.1016/j.ejmech.2023.115091] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Psychiatric and neurological disorders affect millions of people worldwide. Currently available treatments may help to improve symptoms, but they cannot cure the diseases. Therefore, there is an urgent need for potent and safe therapeutic solutions. 8-Amide and 8-carbamatecoumarins were synthetized and evaluated as human monoamine oxidase A and B (hMAO-A and hMAO-B) inhibitors. Comparison between both scaffolds has been established, and we hypothesized that the introduction of different substituents can modulate hMAO activity and selectivity. N-(7-Hydroxy-4-methylcoumarin-8-yl)-4-methylbenzamide (9) and ethyl N-(7-hydroxy-4-methylcoumarin-8-yl)carbamate (20) proved to be the most active and selective hMAO-A inhibitors (IC50 = 15.0 nM and IC50 = 22.0 nM, respectively), being compound 9 an irreversible hMAO-A inhibitor twenty-four times more active in vitro than moclobemide, a drug used in the treatment of depression and anxiety. Based on PAMPA assay results, both compounds proved to be good candidates to cross the blood-brain barrier. In addition, these compounds showed non-significant cytotoxicity on neuronal viability assays. Also, the best compound proved to have a t1/2 of 6.84 min, an intrinsic clearance of 195.63 μL min-1 mg-1 protein, and to be chemically stable at pH 3.0, 7.4 and 10.0. Docking studies were performed to better understand the binding affinities and selectivity profiles for both hMAO isoforms. Finally, theoretical drug-like properties calculations corroborate the potential of both scaffolds on the search for new therapeutic solutions for psychiatric disorders as depression.
Collapse
|
6
|
Hamid K, Tran VH, Duke RK, Duke CC. Three Australian Lepidosperma Labill. Species as sources of prenylated and oxyprenylated derivatives of piceatannol, resveratrol and pinosylvin: Melatoninergic binding and inhibition of quinone reductase 2. PHYTOCHEMISTRY 2022; 203:113396. [PMID: 35998831 DOI: 10.1016/j.phytochem.2022.113396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Prenylated and hydroxyprenylated piceatannol, resveratrol and pinosylvin derivatives were isolated from resin produced by three Australian Lepidosperma Labill. Species (Cyperaceae). From L. congestum R.Br. one known compound, 3',5'-bis-prenyl-E-resveratrol, and five undescribed compounds were isolated, 3'-O-prenyl-5'-prenyl-E-piceatannol, 5',6'-bis-prenyl-E-piceatannol, 5'-prenyl-E-piceatannol, 3',5'-bis(3-hydroxy-3-methylbutyl)-E-resveratrol and 3',5'-bis-E-hydroxyprenyl-E-resveratrol. From L. gunnii Boeckeler one undescribed compound was isolated, 3'-E-hydroxyprenyl-5'-Z-hydroxyprenyl-E-resveratrol. From L. laterale R.Br. six undescribed compounds were isolated, 3-O-prenyl-E-pinosylvin, 3-O-Z-hydroxyprenyl-E-pinosylvin, 3'-Z-hydroxyprenyl-E-resveratrol, 3-O-Z-hydroxyprenyl-E-resveratrol, 3-O-Z-hydroxyprenyl-4'-O-methyl-E-resveratrol, and 3-O-prenyl-3'-δ,δ'-dihydroxyprenyl-E-resveratrol. Compounds, including a reference compound 3-O-prenyl-3'-O-methyl-E-piceatannol, were screened in an assay for melatoninergic binding to MT1 and MT2 receptors and binding to QR2/MT3 enzyme, and for inhibition of QR2/MT3 in a functional assay. Strong binding was observed for 3-O-Z-hydroxyprenyl-E-resveratrol with a Ki of 0.022 nM and the strongest inhibition of QR2/MT3 observed was for the reference compound, 3-O-prenyl-3'-O-methyl-E-piceatannol, with an inhibition of 61% at 1 μM and 95% at 10 μM. The three most active binders and inhibitors of QR2/MT3 were found to have a common substructure corresponding to 3-O-prenylresveratrol.
Collapse
Affiliation(s)
- Kaiser Hamid
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Van H Tran
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Rujee K Duke
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Colin C Duke
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.
| |
Collapse
|
7
|
Akhzari M, Barazesh M, Jalili S. Melatonin as an antioxidant agent in disease prevention: A biochemical focus. LETT ORG CHEM 2022. [DOI: 10.2174/1570178619666220325124451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Abstract:
In the recent years, free radicals and oxidative stress have been found to be associated with aging, cancer, atherosclerosis, neurodegenerative disorders, diabetes, and inflammatory diseases. Confirming the role of oxidants in numerous pathological situations including cancer, developing antioxidants as therapeutic platforms is needed. It has been well established that melatonin and its derived metabolites function as endogenous free-radical scavengers and broad spectrum antioxidants. To achieve this function, melatonin can directly detoxify reactive oxygen and reactive nitrogen species and indirectly overexpress antioxidant enzymes while suppressing the activity of pro-oxidant enzymes. Many investigations have also confirmed the role of melatonin and its derivatives in different physiological processes and therapeutic functions such as controlling the circadian rhythm and immune functions. This review aimed to focus on melatonin as a beneficial agent for the stimulation of antioxidant enzymes and inhibition of lipid peroxidation and to evaluate its contribution to protection against oxidative damages. In addition, the clinical application of melatonin in several diseases is discussed. Finally, the safety and efficacy of melatonin in clinical backgrounds is also reviewed.
Collapse
Affiliation(s)
- Morteza Akhzari
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Sajad Jalili
- Department of Orthopedics, Faculty of Medicine, Ahvaz, Jundishapour University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Herrera-Arozamena C, Estrada-Valencia M, López-Caballero P, Pérez C, Morales-García JA, Pérez-Castillo A, Sastre ED, Fernández-Mendívil C, Duarte P, Michalska P, Lombardía J, Senar S, León R, López MG, Rodríguez-Franco MI. Resveratrol-Based MTDLs to Stimulate Defensive and Regenerative Pathways and Block Early Events in Neurodegenerative Cascades. J Med Chem 2022; 65:4727-4751. [PMID: 35245051 PMCID: PMC8958504 DOI: 10.1021/acs.jmedchem.1c01883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
By replacing a phenolic
ring of (E)-resveratrol
with an 1,3,4-oxadiazol-2(3H)-one heterocycle, new
resveratrol-based multitarget-directed ligands (MTDLs) were obtained.
They were evaluated in several assays related to oxidative stress
and inflammation (monoamine oxidases, nuclear erythroid 2-related
factor, quinone reductase-2, and oxygen radical trapping) and then
in experiments of increasing complexity (neurogenic properties and
neuroprotection vs okadaic acid). 5-[(E)-2-(4-Methoxyphenyl)ethenyl]-3-(prop-2-yn-1-yl)-1,3,4-oxadiazol-2(3H)-one (4e) showed a well-balanced MTDL profile:
cellular activation of the NRF2-ARE pathway (CD = 9.83 μM),
selective inhibition of both hMAO-B and QR2 (IC50s = 8.05
and 0.57 μM), and the best ability to promote hippocampal neurogenesis.
It showed a good drug-like profile (positive in vitro central nervous
system permeability, good physiological solubility, no glutathione
conjugation, and lack of PAINS or Lipinski alerts) and exerted neuroprotective
and antioxidant actions in both acute and chronic Alzheimer models
using hippocampal tissues. Thus, 4e is an interesting
MTDL that could stimulate defensive and regenerative pathways and
block early events in neurodegenerative cascades.
Collapse
Affiliation(s)
- Clara Herrera-Arozamena
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006 Madrid, Spain.,Programa de Doctorado en Química Orgánica, Facultad de Química, Universidad Complutense de Madrid, Avda. Complutense s/n, E-28040 Madrid, Spain
| | - Martín Estrada-Valencia
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Patricia López-Caballero
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Concepción Pérez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - José A Morales-García
- Instituto de Investigaciones Biomédicas (CSIC-UAM), C/Arturo Duperier, 4, E-28029 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), C/Valderrebollo 5, E-28031 Madrid, Spain.,Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid, Avda. Complutense s/n, E-28040 Madrid, Spain
| | - Ana Pérez-Castillo
- Instituto de Investigaciones Biomédicas (CSIC-UAM), C/Arturo Duperier, 4, E-28029 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), C/Valderrebollo 5, E-28031 Madrid, Spain
| | - Eric Del Sastre
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 4, E-28029 Madrid, Spain
| | - Cristina Fernández-Mendívil
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 4, E-28029 Madrid, Spain
| | - Pablo Duarte
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 4, E-28029 Madrid, Spain
| | - Patrycja Michalska
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 4, E-28029 Madrid, Spain
| | - José Lombardía
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 4, E-28029 Madrid, Spain
| | - Sergio Senar
- DrTarget Machine Learning, C/Alejo Carpentier 13, E-28806 Alcalá de Henares, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006 Madrid, Spain.,Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 4, E-28029 Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 4, E-28029 Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa (IIS-IP), C/Diego de León 62, E-28006 Madrid, Spain
| | - María Isabel Rodríguez-Franco
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006 Madrid, Spain
| |
Collapse
|
9
|
Curcumin-Piperlongumine Hybrids with a Multitarget Profile Elicit Neuroprotection in In Vitro Models of Oxidative Stress and Hyperphosphorylation. Antioxidants (Basel) 2021; 11:antiox11010028. [PMID: 35052532 PMCID: PMC8773050 DOI: 10.3390/antiox11010028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Curcumin shows a broad spectrum of activities of relevance in the treatment of Alzheimer’s disease (AD); however, it is poorly absorbed and is also chemically and metabolically unstable, leading to a very low oral bioavailability. A small library of hybrid compounds designed as curcumin analogues and incorporating the key structural fragment of piperlongumine, a natural neuroinflammation inhibitor, were synthesized by a two-step route that combines a three-component reaction between primary amines, β-ketoesters and α-haloesters and a base-promoted acylation with cinnamoyl chlorides. These compounds were predicted to have good oral absorption and CNS permeation, had good scavenging properties in the in vitro DPPH experiment and in a cellular assay based on the oxidation of dichlorofluorescin to a fluorescent species. The compounds showed low toxicity in two cellular models, were potent inductors of the Nrf2-ARE phase II antioxidant response, inhibited PHF6 peptide aggregation, closely related to Tau protein aggregation and were active against the LPS-induced inflammatory response. They also afforded neuroprotection against an oxidative insult induced by inhibition of the mitochondrial respiratory chain with the rotenone-oligomycin A combination and against Tau hyperphosphorylation induced by the phosphatase inhibitor okadaic acid. This multitarget pharmacological profile is highly promising in the development of treatments for AD and provides a good hit structure for future optimization efforts.
Collapse
|
10
|
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: 11] [Impact Index Per Article: 2.8] [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
|
11
|
Martí-Marí O, Martínez-Gualda B, de la Puente-Secades S, Mills A, Quesada E, Abdelnabi R, Sun L, Boonen A, Noppen S, Neyts J, Schols D, Camarasa MJ, Gago F, San-Félix A. Double Arylation of the Indole Side Chain of Tri- and Tetrapodal Tryptophan Derivatives Renders Highly Potent HIV-1 and EV-A71 Entry Inhibitors†. J Med Chem 2021; 64:10027-10046. [PMID: 34229438 PMCID: PMC8389807 DOI: 10.1021/acs.jmedchem.1c00315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
We have recently
described a new generation of potent human immunodeficiency
virus (HIV) and EV-A71 entry inhibitors. The prototypes contain three
or four tryptophan (Trp) residues bearing an isophthalic acid moiety
at the C2 position of each side-chain indole ring. This work is now
extended by both shifting the position of the isophthalic acid to
C7 and synthesizing doubly arylated C2/C7 derivatives. The most potent
derivative (50% effective concentration (EC50) HIV-1, 6
nM; EC50 EV-A71, 40 nM), 33 (AL-518), is a C2/C7 doubly arylated tetrapodal compound. Its superior anti-HIV
potency with respect to the previous C2-arylated prototype is in consonance
with its higher affinity for the viral gp120. 33 (AL-518) showed comparable antiviral activities against X4
and R5 HIV-1 strains and seems to interact with the tip and base of
the gp120 V3 loop. Taken together, these findings support the interest
in 33 (AL-518) as a useful new prototype
for anti-HIV/EV71 drug development.
Collapse
Affiliation(s)
- Olaia Martí-Marí
- Instituto de Química Médica (IQM-CSIC), c/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Belén Martínez-Gualda
- Instituto de Química Médica (IQM-CSIC), c/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | | | - Alberto Mills
- Área de Farmacología, Departamento de Ciencias Biomédicas y Unidad Asociada IQM-UAH, Universidad de Alcalá, E-28805 Alcalá de Henares, Madrid, Spain
| | - Ernesto Quesada
- Instituto de Química Médica (IQM-CSIC), c/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Rana Abdelnabi
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, B-3000 Leuven, Belgium
| | - Liang Sun
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, B-3000 Leuven, Belgium
| | - Arnaud Boonen
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, B-3000 Leuven, Belgium
| | - Sam Noppen
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, B-3000 Leuven, Belgium
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, B-3000 Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, B-3000 Leuven, Belgium
| | - María-José Camarasa
- Instituto de Química Médica (IQM-CSIC), c/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Federico Gago
- Área de Farmacología, Departamento de Ciencias Biomédicas y Unidad Asociada IQM-UAH, Universidad de Alcalá, E-28805 Alcalá de Henares, Madrid, Spain
| | - Ana San-Félix
- Instituto de Química Médica (IQM-CSIC), c/ Juan de la Cierva 3, E-28006 Madrid, Spain
| |
Collapse
|
12
|
Delogu GL, Kumar A, Gatto G, Bustelo F, Saavedra LM, Rodríguez-Franco MI, Laguna R, Viña D. Synthesis and in vitro study of nitro- and methoxy-2-phenylbenzofurans as human monoamine oxidase inhibitors. Bioorg Chem 2021; 107:104616. [PMID: 33444985 DOI: 10.1016/j.bioorg.2020.104616] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/16/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022]
Abstract
A new series of 2-phenylbenzofuran derivatives were designed and synthesized to determine relevant structural features for the MAO inhibitory activity and selectivity. Methoxy substituents were introduced in the 2-phenyl ring, whereas the benzofuran moiety was not substituted or substituted at the positions 5 or 7 with a nitro group. Substitution patterns on both the phenyl ring and the benzofuran moiety determine the affinity for MAO-A or MAO-B. The 2-(3-methoxyphenyl)-5-nitrobenzofuran 9 was the most potent MAO-B inhibitor (IC50 = 0.024 µM) identified in this series, whereas 7-nitro-2-phenylbenzofuran 7 was the most potent MAO-A inhibitor (IC50 = 0.168 µM), both acting as reversible inhibitors. The number and position of the methoxyl groups on the 2-phenyl ring, have an important influence on the inhibitory activity. Molecular docking studies confirmed the experimental results and highlighted the importance of key residues in enzyme inhibition.
Collapse
Affiliation(s)
- Giovanna L Delogu
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Cagliari, Italy.
| | - Amit Kumar
- Department of Electrical and Electronic Engineering, University of Cagliari 09123 Cagliari, Italy
| | - Gianluca Gatto
- Department of Electrical and Electronic Engineering, University of Cagliari 09123 Cagliari, Italy
| | - Fernando Bustelo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Avda Barcelona s/n, Campus Vida 15782, Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Lucía M Saavedra
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), c/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Maria Isabel Rodríguez-Franco
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), c/ Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Reyes Laguna
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Avda Barcelona s/n, Campus Vida 15782, Santiago de Compostela, Spain
| | - Dolores Viña
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Avda Barcelona s/n, Campus Vida 15782, Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
13
|
Jismy B, Guillaumet G, Akssira M, Tikad A, Abarbri M. Efficient microwave-assisted Suzuki–Miyaura cross-coupling reaction of 3-bromo pyrazolo[1,5- a]pyrimidin-5(4 H)-one: towards a new access to 3,5-diarylated 7-(trifluoromethyl)pyrazolo[1,5- a]pyrimidine derivatives. RSC Adv 2021; 11:1287-1302. [PMID: 35747396 PMCID: PMC9134006 DOI: 10.1039/d0ra07959f] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/06/2020] [Indexed: 12/24/2022] Open
Abstract
A convenient and efficient synthetic route to C3-arylated 7-trifluoromethylpyrazolo[1,5-a]pyrimidin-5-one derivatives has been reported starting from 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one through a Suzuki–Miyaura cross-coupling reaction. The arylation (heteroarylation) strategy can be performed using a wide variety of aryl and heteroaryl boronic acids and requiring a tandem catalyst XPhosPdG2/XPhos to avoid the debromination reaction. These optimized conditions were successfully extended to the synthesis of 7-, 8- and 9-arylated pyrimido[1,2-b]indazol-2-ones from their corresponding brominated starting materials. Furthermore, the second C-5 arylation of C3-arylated pyrazolo[1,5-a]pyrimidin-5-ones was achieved under standard Suzuki–Miyaura cross-coupling conditions, after activating the C–O bond of the lactam function with PyBroP, giving access to a small library of 3,5-diarylated 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines in good to excellent yields. The interest of this approach has been highlighted by the synthesis of a known anti-inflammatory agent. Additionally, a preliminary biological evaluation has revealed that a number of derivatives display micromolar IC50 values against monoamine oxidase B, an important target in the field of neurodegenerative disorders. A convenient and efficient synthetic route to C3-arylated 7-trifluoromethylpyrazolo[1,5-a]pyrimidin-5-one derivatives has been reported starting from 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one through a Suzuki–Miyaura cross-coupling reaction.![]()
Collapse
Affiliation(s)
- Badr Jismy
- Laboratoire de Physico-Chimie des Matériaux et des Electrolytes pour l'Energie (PCM2E). EA 6299
- Faculté des Sciences
- 37200 Tours
- France
| | - Gérald Guillaumet
- Institut de Chimie Organique et Analytique (ICOA)
- Université d'Orléans
- UMR CNRS 7311
- France
| | - Mohamed Akssira
- Laboratoire de Chimie Physique et de Chimie Bioorganique
- URAC 22
- 28800 Mohammedia
- Morocco
| | - Abdellatif Tikad
- Laboratoire de Chimie Moléculaire et Substances Naturelles
- Faculté des Sciences
- Département de Chimie
- Université Moulay Ismail
- Meknès 50050
| | - Mohamed Abarbri
- Laboratoire de Physico-Chimie des Matériaux et des Electrolytes pour l'Energie (PCM2E). EA 6299
- Faculté des Sciences
- 37200 Tours
- France
| |
Collapse
|
14
|
Optical control of muscular nicotinic channels with azocuroniums, photoswitchable azobenzenes bearing two N-methyl-N-carbocyclic quaternary ammonium groups. Eur J Med Chem 2020; 200:112403. [DOI: 10.1016/j.ejmech.2020.112403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 12/19/2022]
|
15
|
Ali T, Hao Q, Ullah N, Rahman SU, Shah FA, He K, Zheng C, Li W, Murtaza I, Li Y, Jiang Y, Tan Z, Li S. Melatonin Act as an Antidepressant via Attenuation of Neuroinflammation by Targeting Sirt1/Nrf2/HO-1 Signaling. Front Mol Neurosci 2020; 13:96. [PMID: 32595452 PMCID: PMC7304371 DOI: 10.3389/fnmol.2020.00096] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Physical or psychological stress can cause an immunologic imbalance that disturbs the central nervous system followed by neuroinflammation. The association between inflammation and depression has been widely studied in recent years, though the molecular mechanism is still largely unknown. Thus, targeting the signaling pathways that link stress to neuroinflammation might be a useful strategy against depression. The current study investigated the protective effect of melatonin against lipopolysaccharide (LPS)-induced neuroinflammation and depression. Our results showed that LPS treatment significantly induced depressive-like behavior in mice. Moreover, LPS-treatment enhanced oxidative stress, pro-inflammatory cytokines including TNFα, IL-6, and IL-1β, NF-κB phosphorylation, and glial cell activation markers including GFAP and Iba-1 in the brain of mice. Melatonin treatment significantly abolished the effect of LPS, as indicated by improved depressive-like behaviors, reduced cytokines level, reduced oxidative stress, and normalized LPS-altered Sirt1, Nrf2, and HO-1 expression. However, the melatonin protective effects were reduced after luzindole administration. Collectively, it is concluded that melatonin receptor-dependently protects against LPS-induced depressive-like behaviors via counteracting LPS-induced neuroinflammation.
Collapse
Affiliation(s)
- Tahir Ali
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Hao
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Najeeb Ullah
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.,Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Shafiq Ur Rahman
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.,Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal, Pakistan
| | - Fawad Ali Shah
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.,Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Kaiwu He
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Chengyou Zheng
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Weifen Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Iram Murtaza
- Signal Transduction Lab, Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Yang Li
- Laboratory of Receptor Research, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Shanghai, China
| | - Yuhua Jiang
- Cancer Centre, The Second Hospital of Shandong University, Jinan, China
| | - Zhen Tan
- Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.,Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|