1
|
Liu ZK, Liu Y, Hu XQ, Zou YQ. Decarboxylative Aminomethylation of Indole-3-carboxylic Acids via Strain Release-Driven Ring Opening of 1,2-Oxazetidines. Org Lett 2024; 26:8934-8938. [PMID: 39382327 DOI: 10.1021/acs.orglett.4c03426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
A copper-catalyzed decarboxylative aminomethylation of indole-3-carboxylic acids with 1,2-oxazetidines has been developed, enabling the rapid synthesis of structurally diverse 3-aminomethylindoles in good to excellent yields. Remarkably, an unprecedented decarboxylative aminomethylation/cyclization cascade was further achieved by a combination of copper and iron salts to construct complex γ-carbolines with high efficiency. It is worth noting that one of the obtained products proved to be a good dual-emissive luminogen, exhibiting both aggregation-caused quenching and aggregation-induced emission.
Collapse
Affiliation(s)
- Zi-Kui Liu
- Department of Otolaryngology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Yun Liu
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education and Hubei Key Laboratory of Catalysis and Materials Science, School of Chemistry and Materials Science, South-Central Minzu University, Wuhan 430074, China
| | - Xiao-Qiang Hu
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education and Hubei Key Laboratory of Catalysis and Materials Science, School of Chemistry and Materials Science, South-Central Minzu University, Wuhan 430074, China
| | - You-Quan Zou
- Department of Otolaryngology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
2
|
Song G, Li B, Yang Z, Lin H, Cheng J, Huang Y, Xing C, Lv F, Bai H, Wang S. Regulation of Cell Membrane Potential through Supramolecular System for Activating Calcium Ion Channels. J Am Chem Soc 2024; 146:25383-25393. [PMID: 39196894 DOI: 10.1021/jacs.4c10710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
The regulation of the cell membrane potential plays a crucial role in governing the transmembrane transport of various ions and cellular life processes. However, in situ and on-demand modulation of cell membrane potential for ion channel regulation is challenging. Herein, we have constructed a supramolecular assembly system based on water-soluble cationic oligo(phenylenevinylene) (OPV) and cucurbit[7]uril (CB[7]). The controllable disassembly of OPV/4CB[7] combined with the subsequent click reaction provides a step-by-step adjustable surface positive potential. These processes can be employed in situ on the plasma membrane to modulate the membrane potential on-demand for precisely controlling the activation of the transient receptor potential vanilloid 1 (TRPV1) ion channel and up-regulating exogenous calcium-responsive gene expression. Compared with typical optogenetics, electrogenetics, and mechanogenetics, our strategy provides a perspective supramolecular genetics toolbox for the regulation of membrane potential and downstream intracellular gene regulation events.
Collapse
Affiliation(s)
- Gang Song
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Boying Li
- School of Chemical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Zhiwen Yang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongrui Lin
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Junjie Cheng
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yiming Huang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Chengfen Xing
- School of Chemical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Haotian Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
3
|
Di Pierdomenico J, Gallego-Ortega A, Norte-Muñoz M, Vidal-Villegas B, Bravo I, Boluda-Ruiz M, Bernal-Garro JM, Fernandez-Bueno I, Pastor-Jimeno JC, Villegas-Pérez MP, Avilés-Trigueros M, de Los Ríos C, Vidal-Sanz M. Evaluation of the neuroprotective efficacy of the gramine derivative ITH12657 against NMDA-induced excitotoxicity in the rat retina. Front Neuroanat 2024; 18:1335176. [PMID: 38415017 PMCID: PMC10898249 DOI: 10.3389/fnana.2024.1335176] [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: 11/08/2023] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
Purpose The aim of this study was to investigate, the neuroprotective effects of a new Gramine derivative named: ITH12657, in a model of retinal excitotoxicity induced by intravitreal injection of NMDA. Methods Adult Sprague Dawley rats received an intravitreal injection of 100 mM NMDA in their left eye and were treated daily with subcutaneous injections of ITH12657 or vehicle. The best dose-response, therapeutic window study, and optimal treatment duration of ITH12657 were studied. Based on the best survival of Brn3a + RGCs obtained from the above-mentioned studies, the protective effects of ITH12657 were studied in vivo (retinal thickness and full-field Electroretinography), and ex vivo by quantifying the surviving population of Brn3a + RGCs, αRGCs and their subtypes α-ONsRGCs, α-ONtRGCs, and α-OFFRGCs. Results Administration of 10 mg/kg ITH12657, starting 12 h before NMDA injection and dispensed for 3 days, resulted in the best significant protection of Brn3a + RGCs against NMDA-induced excitotoxicity. In vivo, ITH12657-treated rats showed significant preservation of retinal thickness and functional protection against NMDA-induced retinal excitotoxicity. Ex vivo results showed that ITH12657 afforded a significant protection against NMDA-induced excitotoxicity for the populations of Brn3a + RGC, αRGC, and αONs-RGC, but not for the population of αOFF-RGC, while the population of α-ONtRGC was fully resistant to NMDA-induced excitotoxicity. Conclusion Subcutaneous administration of ITH12657 at 10 mg/kg, initiated 12 h before NMDA-induced retinal injury and continued for 3 days, resulted in the best protection of Brn3a + RGCs, αRGC, and αONs-RGC against excitotoxicity-induced RGC death. The population of αOFF-RGCs was extremely sensitive while α-ONtRGCs were fully resistant to NMDA-induced excitotoxicity.
Collapse
Affiliation(s)
| | | | - María Norte-Muñoz
- Departamento de Oftalmología, Universidad de Murcia e IMIB-Arrixaca, Murcia, Spain
| | | | - Isaac Bravo
- Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - María Boluda-Ruiz
- Departamento de Oftalmología, Universidad de Murcia e IMIB-Arrixaca, Murcia, Spain
| | | | - Iván Fernandez-Bueno
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, Valladolid, Spain
| | - Jose Carlos Pastor-Jimeno
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, Valladolid, Spain
| | | | | | - Cristobal de Los Ríos
- Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Universidad de Murcia e IMIB-Arrixaca, Murcia, Spain
| |
Collapse
|
4
|
Perez Garcia G, Bicak M, Buros J, Haure-Mirande JV, Perez GM, Otero-Pagan A, Gama Sosa MA, De Gasperi R, Sano M, Gage FH, Barlow C, Dudley JT, Glicksberg BS, Wang Y, Readhead B, Ehrlich ME, Elder GA, Gandy S. Beneficial effects of physical exercise and an orally active mGluR2/3 antagonist pro-drug on neurogenesis and behavior in an Alzheimer's amyloidosis model. FRONTIERS IN DEMENTIA 2023; 2:1198006. [PMID: 39081972 PMCID: PMC11285632 DOI: 10.3389/frdem.2023.1198006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/31/2023] [Indexed: 08/02/2024]
Abstract
Background Modulation of physical activity represents an important intervention that may delay, slow, or prevent mild cognitive impairment (MCI) or dementia due to Alzheimer's disease (AD). One mechanism proposed to underlie the beneficial effect of physical exercise (PE) involves the apparent stimulation of adult hippocampal neurogenesis (AHN). BCI-838 is a pro-drug whose active metabolite BCI-632 is a negative allosteric modulator at group II metabotropic glutamate receptors (mGluR2/3). We previously demonstrated that administration of BCI-838 to a mouse model of brain accumulation of oligomeric AβE22Q (APP E693Q = "Dutch APP") reduced learning behavior impairment and anxiety, both of which are associated with the phenotype of Dutch APP mice. Methods 3-month-old mice were administered BCI-838 and/or physical exercise for 1 month and then tested in novel object recognition, neurogenesis, and RNAseq. Results Here we show that (i) administration of BCI-838 and a combination of BCI-838 and PE enhanced AHN in a 4-month old mouse model of AD amyloid pathology (APP KM670/671NL /PSEN1 Δexon9= APP/PS1), (ii) administration of BCI-838 alone or with PE led to stimulation of AHN and improvement in recognition memory, (iii) the hippocampal dentate gyrus transcriptome of APP/PS1 mice following BCI-838 treatment showed up-regulation of brain-derived neurotrophic factor (BDNF), PIK3C2A of the PI3K-mTOR pathway, and metabotropic glutamate receptors, and down-regulation of EIF5A involved in modulation of mTOR activity by ketamine, and (iv) validation by qPCR of an association between increased BDNF levels and BCI-838 treatment. Conclusion Our study points to BCI-838 as a safe and orally active compound capable of mimicking the beneficial effect of PE on AHN and recognition memory in a mouse model of AD amyloid pathology.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Mesude Bicak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jacqueline Buros
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Gissel M. Perez
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Alena Otero-Pagan
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Miguel A. Gama Sosa
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rita De Gasperi
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mary Sano
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- BrainCells, Inc., La Jolla, CA, United States
| | - Carrolee Barlow
- BrainCells, Inc., La Jolla, CA, United States
- E-Scape Bio, South San Francisco, CA, United States
| | - Joel T. Dudley
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Benjamin S. Glicksberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yanzhuang Wang
- Department of Developmental and Cell Biology, University of Michigan, Ann Arbor, MI, United States
| | - Benjamin Readhead
- Arizona State University-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, United States
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Gregory A. Elder
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, United States
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Cognitive Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
5
|
Zhang J, Jia Q, Li N, Gu L, Dan W, Dai J. Recent Developments of Gramine: Chemistry and Biological Activity. Molecules 2023; 28:5695. [PMID: 37570664 PMCID: PMC10419902 DOI: 10.3390/molecules28155695] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The natural alkaloid gramine has attracted significant attention in both academic and industrial circles because of its potential and diverse biological activities, including antiviral, antibacterial, antifungal, anti-inflammatory and antitumor activities; application in therapy for Alzheimer's disease; serotonin-receptor-related activity; insecticidal activity; and application as an algicide. In this review, we focus on the research advances that have been made for gramine-based molecules since their discovery, providing key information on their extraction and separation, chemical synthesis and diverse biological activities. Data regarding their mechanisms of action are also presented. This comprehensive and critical review will serve as a guide for developing more drug candidates based on gramine skeletons.
Collapse
Affiliation(s)
- Jiaoyue Zhang
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, China; (J.Z.); (Q.J.)
| | - Qitao Jia
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, China; (J.Z.); (Q.J.)
| | - Na Li
- Instrumental Analysis Center, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Liqiang Gu
- School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China;
| | - Wenjia Dan
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, China; (J.Z.); (Q.J.)
| | - Jiangkun Dai
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, China; (J.Z.); (Q.J.)
| |
Collapse
|
6
|
Bravo I, Viejo L, de Los Ríos C, García-Frutos EM, Darder M. Cellulose/pectin-based materials incorporating Laponite-indole derivative hybrid for oral administration and controlled delivery of the neuroprotective drug. Int J Biol Macromol 2023; 234:123765. [PMID: 36812973 DOI: 10.1016/j.ijbiomac.2023.123765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
Bionanocomposite materials based on clays have been designed for oral administration and controlled release of a neuroprotective drug derivative of 5-methylindole, which had featured an innovative pharmacological mechanism for the treatment of neurodegenerative diseases such as Alzheimer's. This drug was adsorbed in the commercially available Laponite® XLG (Lap). X-ray diffractograms confirmed its intercalation in the interlayer region of the clay. The loaded drug was 62.3 meq/100 g Lap, close to the cation exchange capacity of Lap. Per se toxicity studies and neuroprotective experiments versus the neurotoxin okadaic acid, a potent and selective inhibitor of protein phosphatase 2A (PP2A), confirmed that the clay-intercalated drug did not exert toxicity in cell cultures and provided neuroprotection. Release tests of the hybrid material performed in media mimicking the gastrointestinal tract indicated a drug release in acid medium close to 25 %. The hybrid was encapsulated in a micro/nanocellulose matrix and processed as microbeads, with pectin coating for additional protection, to minimize release under acidic conditions. Alternatively, low density materials based on a microcellulose/pectin matrix were evaluated as orodispersible foams showing fast disintegration times, sufficient mechanical resistance for handling, and release profiles in simulated media that confirmed a controlled release of the encapsulated neuroprotective drug.
Collapse
Affiliation(s)
- Isaac Bravo
- Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid 28049, Spain; Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid 28006, Spain; Instituto Fundación Teófilo Hernando (IFTH), Madrid 28029, Spain
| | - Lucía Viejo
- Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid 28006, Spain; Instituto Fundación Teófilo Hernando (IFTH), Madrid 28029, Spain
| | - Cristóbal de Los Ríos
- Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid 28006, Spain; Instituto Fundación Teófilo Hernando (IFTH), Madrid 28029, Spain
| | - Eva M García-Frutos
- Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid 28049, Spain.
| | - Margarita Darder
- Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid 28049, Spain.
| |
Collapse
|
7
|
Arribas RL, Viejo L, Bravo I, Martínez M, Ramos E, Romero A, García-Frutos EM, Janssens V, Montiel C, de Los Ríos C. C-glycosides analogues of the okadaic acid central fragment exert neuroprotection via restoration of PP2A-phosphatase activity: A rational design of potential drugs for Alzheimer's disease targeting tauopathies. Eur J Med Chem 2023; 251:115245. [PMID: 36905916 DOI: 10.1016/j.ejmech.2023.115245] [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: 10/31/2022] [Revised: 01/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Protein phosphatase 2A (PP2A) is an important Ser/Thr phosphatase that participates in the regulation of multiple cellular processes. This implies that any deficient activity of PP2A is the responsible of severe pathologies. For instance, one of the main histopathological features of Alzheimer's disease is neurofibrillary tangles, which are mainly comprised by hyperphosphorylated forms of tau protein. This altered rate of tau phosphorylation has been correlated with PP2A depression AD patients. With the goal of preventing PP2A inactivation in neurodegeneration scenarios, we have aimed to design, synthesize and evaluate new ligands of PP2A capable of preventing its inhibition. To achieve this goal, the new PP2A ligands present structural similarities with the central fragment C19-C27 of the well-established PP2A inhibitor okadaic acid (OA). Indeed, this central moiety of OA does not exert inhibitory actions. Hence, these compounds lack PP2A-inhibiting structural motifs but, in contrast, compete with PP2A inhibitors, thus recovering phosphatase activity. Proving this hypothesis, most compounds showed a good neuroprotective profile in neurodegeneration models related to PP2A impairment, highlighting derivative 10, named ITH12711, as the most promising one. This compound (1) restored in vitro and cellular PP2A catalytic activity, measured on a phospho-peptide substrate and by western-blot analyses, (2) proved good brain penetration measured by PAMPA, and (3) prevented LPS-induced memory impairment of mice in the object recognition test. Thus, the promising outcomes of the compound 10 validate our rational approach to design new PP2A-activating drugs based on OA central fragment.
Collapse
Affiliation(s)
- Raquel L Arribas
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Spain
| | - Lucía Viejo
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006, Madrid, Spain
| | - Isaac Bravo
- Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006, Madrid, Spain; Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, 28049, Madrid, Spain
| | - Minerva Martínez
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Eva Ramos
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain
| | - Alejandro Romero
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain
| | - Eva M García-Frutos
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, 28049, Madrid, Spain; Universidad de Alcalá, Departamento de Química Orgánica y Química Inorgánica, Ctra. Madrid-Barcelona Km.33,600, 28871, Alcalá de Henares, Madrid, Spain
| | - Veerle Janssens
- Department of Cellular & Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, B-3000, Leuven, Belgium; LBI (KU Leuven Brain Institute), B-3000, Leuven, Belgium
| | - Carmen Montiel
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Cristóbal de Los Ríos
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Spain; Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006, Madrid, Spain.
| |
Collapse
|
8
|
de los Ríos C, Viejo L, Carretero VJ, Juárez NH, Cruz-Martins N, Hernández-Guijo JM. Promising Molecular Targets in Pharmacological Therapy for Neuronal Damage in Brain Injury. Antioxidants (Basel) 2023; 12:118. [PMID: 36670980 PMCID: PMC9854812 DOI: 10.3390/antiox12010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
The complex etiopathogenesis of brain injury associated with neurodegeneration has sparked a lot of studies in the last century. These clinical situations are incurable, and the currently available therapies merely act on symptoms or slow down the course of the diseases. Effective methods are being sought with an intent to modify the disease, directly acting on the properly studied targets, as well as to contribute to the development of effective therapeutic strategies, opening the possibility of refocusing on drug development for disease management. In this sense, this review discusses the available evidence for mitochondrial dysfunction induced by Ca2+ miscommunication in neurons, as well as how targeting phosphorylation events may be used to modulate protein phosphatase 2A (PP2A) activity in the treatment of neuronal damage. Ca2+ tends to be the catalyst for mitochondrial dysfunction, contributing to the synaptic deficiency seen in brain injury. Additionally, emerging data have shown that PP2A-activating drugs (PADs) suppress inflammatory responses by inhibiting different signaling pathways, indicating that PADs may be beneficial for the management of neuronal damage. In addition, a few bioactive compounds have also triggered the activation of PP2A-targeted drugs for this treatment, and clinical studies will help in the authentication of these compounds. If the safety profiles of PADs are proven to be satisfactory, there is a case to be made for starting clinical studies in the setting of neurological diseases as quickly as possible.
Collapse
Affiliation(s)
- Cristóbal de los Ríos
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, University Rey Juan Carlos, Avda. Atenas s/n, 28922 Alcorcón, Spain
| | - Lucía Viejo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Victoria Jiménez Carretero
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natalia Hernández Juárez
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natália Cruz-Martins
- Faculty of Medicine, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Advanced Training in Health Sciences and Technologies, Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Ramón y Cajal Institute for Health Research, IRYCIS, Hospital Ramón y Cajal, Ctra. de Colmenar Viejo, Km. 9,100, 28029 Madrid, Spain
| |
Collapse
|
9
|
Targeting neuronal calcium channels and GSK3β for Alzheimer's disease with naturally-inspired Diels-Alder adducts. Bioorg Chem 2022; 129:106152. [PMID: 36155094 DOI: 10.1016/j.bioorg.2022.106152] [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: 07/26/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/22/2022]
Abstract
The complexity of neurodegenerative diseases, among which Alzheimer's disease plays a pivotal role, poses one of the tough therapeutic challenges of present time. In this perspective, a multitarget approach appears as a promising strategy to simultaneously interfere with different defective pathways. In this paper, a structural simplification plan was performed on our previously reported multipotent polycyclic compounds, in order to obtain a simpler pharmacophoric central core with improved pharmacokinetic properties, while maintaining the modulating activity on neuronal calcium channels and glycogen synthase kinase 3-beta (GSK-3β), as validated targets to combat Alzheimer's disease. The molecular pruning approach applied here led to tetrahydroisoindole-dione (1), tetrahydromethanoisoindole-dione (2) and tetrahydroepoxyisoindole-dione (3) structures, easily affordable by Diels-Alder cycloaddition. Preliminary data indicated structure 3 as the most appropriate, thus a SAR study was performed by introducing different substituents, selected on the basis of the commercial availability of the furan derivatives required for the synthetic procedure. The results indicated compound 10 as a promising, structurally atypical, safe and BBB-penetrating Cav modulator, inhibiting both L- and N-calcium channels, likely responsible for the Ca2+ overload observed in Alzheimer's disease. In a multitarget perspective, compound 11 appeared as an effective prototype, endowed with improved Cav inhibitory activity, with respect to the reference drug nifedipine, and encouraging modulating activity on GSK-3β.
Collapse
|
10
|
Won E, Na KS, Kim YK. Associations between Melatonin, Neuroinflammation, and Brain Alterations in Depression. Int J Mol Sci 2021; 23:ijms23010305. [PMID: 35008730 PMCID: PMC8745430 DOI: 10.3390/ijms23010305] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/21/2021] [Accepted: 12/26/2021] [Indexed: 12/14/2022] Open
Abstract
Pro-inflammatory systemic conditions that can cause neuroinflammation and subsequent alterations in brain regions involved in emotional regulation have been suggested as an underlying mechanism for the pathophysiology of major depressive disorder (MDD). A prominent feature of MDD is disruption of circadian rhythms, of which melatonin is considered a key moderator, and alterations in the melatonin system have been implicated in MDD. Melatonin is involved in immune system regulation and has been shown to possess anti-inflammatory properties in inflammatory conditions, through both immunological and non-immunological actions. Melatonin has been suggested as a highly cytoprotective and neuroprotective substance and shown to stimulate all stages of neuroplasticity in animal models. The ability of melatonin to suppress inflammatory responses through immunological and non-immunological actions, thus influencing neuroinflammation and neurotoxicity, along with subsequent alterations in brain regions that are implicated in depression, can be demonstrated by the antidepressant-like effects of melatonin. Further studies that investigate the associations between melatonin, immune markers, and alterations in the brain structure and function in patients with depression could identify potential MDD biomarkers.
Collapse
Affiliation(s)
- Eunsoo Won
- Department of Psychiatry, Chaum, Seoul 06062, Korea;
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea
| | - Kyoung-Sae Na
- Department of Psychiatry, Gachon University Gil Medical Center, Incheon 21565, Korea;
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
- Correspondence:
| |
Collapse
|
11
|
Synthesis and Biological Assessment of 4,1-Benzothiazepines with Neuroprotective Activity on the Ca 2+ Overload for the Treatment of Neurodegenerative Diseases and Stroke. Molecules 2021; 26:molecules26154473. [PMID: 34361628 PMCID: PMC8347512 DOI: 10.3390/molecules26154473] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
In excitable cells, mitochondria play a key role in the regulation of the cytosolic Ca2+ levels. A dysregulation of the mitochondrial Ca2+ buffering machinery derives in serious pathologies, where neurodegenerative diseases highlight. Since the mitochondrial Na+/Ca2+ exchanger (NCLX) is the principal efflux pathway of Ca2+ to the cytosol, drugs capable of blocking NCLX have been proposed to act as neuroprotectants in neuronal damage scenarios exacerbated by Ca2+ overload. In our search of optimized NCLX blockers with augmented drug-likeness, we herein describe the synthesis and pharmacological characterization of new benzothiazepines analogues to the first-in-class NCLX blocker CGP37157 and its further derivative ITH12575, synthesized by our research group. As a result, we found two new compounds with an increased neuroprotective activity, neuronal Ca2+ regulatory activity and improved drug-likeness and pharmacokinetic properties, such as clog p or brain permeability, measured by PAMPA experiments.
Collapse
|
12
|
Huang Y, Wang J, Liu J, Shi D, Li X, Wang M, Lu T, Jiang B, Xia C, Lin H, Xu Y, Li J. Rapid Repurposing of Novel Combination Drugs for the Treatment of Heart Failure via a Computationally Guided Network Screening Approach. J Chem Inf Model 2021; 62:5223-5232. [PMID: 34151561 DOI: 10.1021/acs.jcim.1c00132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Combination drugs, characterized by high efficacy and few side effects, have received extensive attention from pharmaceutical companies and researchers for the treatment of complex diseases such as heart failure (HF). Traditional combination drug discovery depends on large-scale high-throughput experimental approaches that are time-consuming and costly. Herein we developed a novel, rapid, and potentially universal computer-guided combination drug-network-screening approach based on a set of databases and web services that are easy for individuals to obtain and operate, and we discovered for the first time that the menthol-allethrin combination screened by this approach exhibited a significant synergistic cardioprotective effect in vitro. Further mechanistic studies indicated that allethrin and menthol could synergistically block calcium channels. Allethrin bound to the central cavity of the voltage-dependent L-type calcium channel subunit alpha-1S (CACNA1S) lead to a conformational change in an allosteric site of CACNA1S, thereby enhancing the binding of menthol to this allosteric site. In summary, we reported a potentially universal computational approach to combination drug screening that has been used to discover a new combination of menthol-allethrin against HF in vitro, providing a new synergistic mechanism and prospective agent for HF treatment.
Collapse
Affiliation(s)
- Yunyuan Huang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jiqun Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jiaqi Liu
- Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Donglei Shi
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Xiaokang Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Manjiong Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Taotao Lu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Bei Jiang
- College of Pharmacy and Chemistry, Dali University, 5 Xue Ren Road, Dali 671000, China
| | - Conglong Xia
- College of Pharmacy and Chemistry, Dali University, 5 Xue Ren Road, Dali 671000, China
| | - Houwen Lin
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200127, China
| | - Yixiang Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China.,College of Pharmacy and Chemistry, Dali University, 5 Xue Ren Road, Dali 671000, China.,Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
13
|
The structural simplification of lysergic acid as a natural lead for synthesizing novel anti-Alzheimer agents. Bioorg Med Chem Lett 2021; 47:128205. [PMID: 34139326 DOI: 10.1016/j.bmcl.2021.128205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/06/2021] [Accepted: 06/11/2021] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, projected to be the second leading cause of mortality by 2040. AD is characterized by a progressive impairment of memory leading to dementia and loss of ability to carry out daily functions. In addition to the deficiency of acetylcholine release in synapse, there are other mechanisms explaining the etiology of the disease. The most disputing ones are associated with the accumulation of damaged proteins β-amyloid (Aβ) and hyperphosphorylated tau outside and inside neurons, respectively. Lysergic acid derivatives have been shown to possess promising anti-Alzheimer effect. Moreover, lysergic acid structure encompasses the general structural requirements for acetylcholinesterase inhibition. In this study, sixteen analogues, derived from lysergic acid structure, were synthesized. Heck and Mannich reactions were carried out to 4-bromo indole nucleus to generate potentially active analogues. Some of them were subsequently cyclized by nitromethane and zinc reduction procedures. Some of these compounds showed neuroprotective and anti-inflammatory effects stronger than the currently used anti-Alzheimer drug; donepezil. Some of the synthesized com-pounds showed a noticeable acetylcholinesterase inhibition. Twelve molecular targets attributed with AD etiology were tested versus the synthesized compounds by in silico modeling. Docking scores of modeling were plotted against in vitro activity of the compounds. The one afforded the strongest positive correlation was ULK-1 which has a significant role in autophagy.
Collapse
|
14
|
Multitarget Therapeutic Strategies for Alzheimer's Disease: Review on Emerging Target Combinations. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5120230. [PMID: 32714977 PMCID: PMC7354643 DOI: 10.1155/2020/5120230] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases represent nowadays one of the major health problems. Despite the efforts made to unveil the mechanism leading to neurodegeneration, it is still not entirely clear what triggers this phenomenon and what allows its progression. Nevertheless, it is accepted that neurodegeneration is a consequence of several detrimental processes, such as protein aggregation, oxidative stress, and neuroinflammation, finally resulting in the loss of neuronal functions. Starting from these evidences, there has been a wide search for novel agents able to address more than a single event at the same time, the so-called multitarget-directed ligands (MTDLs). These compounds originated from the combination of different pharmacophoric elements which endowed them with the ability to interfere with different enzymatic and/or receptor systems, or to exert neuroprotective effects by modulating proteins and metal homeostasis. MTDLs have been the focus of the latest strategies to discover a new treatment for Alzheimer's disease (AD), which is considered the most common form of dementia characterized by neurodegeneration and cognitive dysfunctions. This review is aimed at collecting the latest and most interesting target combinations for the treatment of AD, with a detailed discussion on new agents with favorable in vitro properties and on optimized structures that have already been assessed in vivo in animal models of dementia.
Collapse
|
15
|
Arribas RL, Bordas A, Domènech Omella J, Cedillo JL, Janssens V, Montiel C, de Los Ríos C. An okadaic acid fragment analogue prevents nicotine-induced resistance to cisplatin by recovering PP2A activity in non-small cell lung cancer cells. Bioorg Chem 2020; 100:103874. [PMID: 32361056 DOI: 10.1016/j.bioorg.2020.103874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022]
Abstract
We herein report the design, synthesis, and functional impact of an okadaic acid (OA) small analogue, ITH12680, which restores the activity of phosphoprotein phosphatase 2A (PP2A), whose deficient activity has been implicated in nicotine-mediated tumor progression and chemoresistance in non-small cell lung cancer (NSCLC). For its design, we paid attention to the structure of the PP2A-OA complex, where the C16-C38 OA fragment confers PP2A affinity and selectivity, but it is not involved in the inhibitory effect. Confirming this hypothesis, PP2A activity was not inhibited by ITH12680. By contrast, the compound partially restored OA-exerted PP2A inhibition in vitro. Moreover, flow cytometry and immunoblotting experiments revealed that ITH12680 reversed nicotine-induced cisplatin resistance in NSCLC cells, as it prevented nicotine-induced reduction of Bax expression and inhibited nicotine-mediated activation of cell survival and proliferation kinases, Akt and ERK1/2. Our findings suggest that the rescue of nicotine-inhibited PP2A activity could diminish the resistance to cisplatin treatment observed in NSCLC patients who continue smoking.
Collapse
Affiliation(s)
- Raquel L Arribas
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Anna Bordas
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Judit Domènech Omella
- Department of Cellular & Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Herestraat 49, B-3000 Leuven, & LKI (Leuven Cancer Institute), Belgium
| | - Jose Luis Cedillo
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Veerle Janssens
- Department of Cellular & Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Herestraat 49, B-3000 Leuven, & LKI (Leuven Cancer Institute), Belgium
| | - Carmen Montiel
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain.
| | - Cristóbal de Los Ríos
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain; Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006 Madrid, Spain.
| |
Collapse
|
16
|
Liu L, Labani N, Cecon E, Jockers R. Melatonin Target Proteins: Too Many or Not Enough? Front Endocrinol (Lausanne) 2019; 10:791. [PMID: 31803142 PMCID: PMC6872631 DOI: 10.3389/fendo.2019.00791] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022] Open
Abstract
The neurohormone N-acetyl-5-methoxytryptamine, better known as melatonin, is a tryptophan derivative with a wide range of biological effects that is present in many organisms. These effects are believed to rely either on the chemical properties of melatonin itself as scavenger of free radicals or on the binding of melatonin to protein targets. More than 15 proteins, including receptors (MT1, MT2, Mel1c, CAND2, ROR, VDR), enzymes (QR2, MMP-9, pepsin, PP2A, PR-10 proteins), pores (mtPTP), transporters (PEPT1/2, Glut1), and other proteins (HBS, CaM, tubulin, calreticuline), have been suggested to interact with melatonin at sub-nanomolar to millimolar melatonin concentrations. In this review we assemble for the first time the available information on proposed melatonin targets and discuss them in a comprehensive manner to evaluate the robustness of these findings in terms of methodology, physiological relevance, and independent replication.
Collapse
Affiliation(s)
- Lei Liu
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Nedjma Labani
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Erika Cecon
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Ralf Jockers
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| |
Collapse
|
17
|
Polycyclic maleimide-based derivatives as first dual modulators of neuronal calcium channels and GSK-3β for Alzheimer's disease treatment. Eur J Med Chem 2019; 163:394-402. [DOI: 10.1016/j.ejmech.2018.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/30/2018] [Accepted: 12/02/2018] [Indexed: 01/06/2023]
|
18
|
Chikazawa M, Sato R. Identification of Functional Food Factors as β 2-Adrenergic Receptor Agonists and Their Potential Roles in Skeletal Muscle. J Nutr Sci Vitaminol (Tokyo) 2018; 64:68-74. [PMID: 29491275 DOI: 10.3177/jnsv.64.68] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Maintaining skeletal muscle functions by controlling muscle metabolism is of utmost importance. β2-Adrenergic receptor (β2-AR), which is expressed in skeletal muscle, is a member of the G-protein-coupled receptor family that plays a critical role in the maintenance of muscle mass. In the present study, using luciferase reporter assays in β2-AR-expressing HEK293 cells, we discovered several food factors that exhibited agonistic activity at mouse or human β2-AR. Osthole, gramine, and hordenine were identified as both mouse and human β2-AR agonists, whereas berberine was identified as a mouse β2-AR agonist only. Additionally, intramuscular injection of gramine or hordenine in mice facilitated gene expression of several cAMP response element binding protein targets, which is thought to result in increased skeletal muscle protein synthesis. This study provides evidence that several food factors might exert potential health effects on skeletal muscle by enhancing cAMP signaling through the activation of β2-AR.
Collapse
Affiliation(s)
- Miho Chikazawa
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo
| | - Ryuichiro Sato
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo.,Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo.,AMED-CREST, Japan Agency for Medical Research and Development
| |
Collapse
|
19
|
Lajarín-Cuesta R, Arribas RL, Nanclares C, García-Frutos EM, Gandía L, de los Ríos C. Design and synthesis of multipotent 3-aminomethylindoles and 7-azaindoles with enhanced protein phosphatase 2A-activating profile and neuroprotection. Eur J Med Chem 2018; 157:294-309. [DOI: 10.1016/j.ejmech.2018.07.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 02/02/2023]
|
20
|
Gandini A, Bartolini M, Tedesco D, Martinez-Gonzalez L, Roca C, Campillo NE, Zaldivar-Diez J, Perez C, Zuccheri G, Miti A, Feoli A, Castellano S, Petralla S, Monti B, Rossi M, Moda F, Legname G, Martinez A, Bolognesi ML. Tau-Centric Multitarget Approach for Alzheimer’s Disease: Development of First-in-Class Dual Glycogen Synthase Kinase 3β and Tau-Aggregation Inhibitors. J Med Chem 2018; 61:7640-7656. [DOI: 10.1021/acs.jmedchem.8b00610] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Annachiara Gandini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136 Trieste, Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Daniele Tedesco
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | | | - Carlos Roca
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Nuria E. Campillo
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Josefa Zaldivar-Diez
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Concepción Perez
- Instituto de Quimica Medica, CSIC, Calle Juan de la Cierva 3, 28006 Madrid, Spain
| | - Giampaolo Zuccheri
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- S3 Center of the Institute of Nanosciences, Italian National Research Council (CNR), I-41125 Modena, Italy
| | - Andrea Miti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- S3 Center of the Institute of Nanosciences, Italian National Research Council (CNR), I-41125 Modena, Italy
| | - Alessandra Feoli
- EpigeneticMedChemLab, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano, Italy
| | - Sabrina Castellano
- EpigeneticMedChemLab, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Martina Rossi
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136 Trieste, Italy
| | - Fabio Moda
- Fondazione IRCCS Istituto Neurologico Carlo Besta, via Celoria 11, I-20133 Milan, Italy
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136 Trieste, Italy
| | - Ana Martinez
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| |
Collapse
|
21
|
Arribas RL, Romero A, Egea J, de los Ríos C. Modulation of serine/threonine phosphatases by melatonin: therapeutic approaches in neurodegenerative diseases. Br J Pharmacol 2018; 175:3220-3229. [PMID: 29781146 PMCID: PMC6057903 DOI: 10.1111/bph.14365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/27/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022] Open
Abstract
Melatonin is an endogenous hormone produced by the pineal gland as well as many other tissues and organs. The natural decline in melatonin levels with ageing contributes significantly to the development of neurodegenerative disorders. Neurodegenerative diseases share common mechanisms of toxicity such as proteinopathy, mitochondrial dysfunction, metal dyshomeostasis, oxidative stress, neuroinflammation and an imbalance in the phosphorylation/dephosphorylation ratio. Several reports have proved the usefulness of melatonin in counteracting the events that lead to a neurodegenerative scenario. In this review, we have focused on the fact that melatonin could rectify the altered phosphorylation/dephosphorylation rate found in some neurodegenerative diseases by influencing the activity of phosphoprotein phosphatases. We analyse whether melatonin offers any protective activity towards these enzymes through a direct interaction. LINKED ARTICLES: This article is part of a themed section on Recent Developments in Research of Melatonin and its Potential Therapeutic Applications. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.16/issuetoc.
Collapse
Affiliation(s)
- Raquel L Arribas
- Instituto‐Fundación Teófilo Hernando, Departamento de Farmacología y TerapéuticaUniversidad Autónoma de MadridMadridSpain
| | - Alejandro Romero
- Department of Pharmacology & Toxicology, Faculty of Veterinary MedicineComplutense University of MadridMadridSpain
| | - Javier Egea
- Instituto‐Fundación Teófilo Hernando, Departamento de Farmacología y TerapéuticaUniversidad Autónoma de MadridMadridSpain
- Molecular Neuroinflammation and Neuronal Plasticity Laboratory, Research UnitHospital Universitario Santa CristinaMadridSpain
- Instituto de Investigación SanitariaHospital Universitario de la PrincesaMadridSpain
| | - Cristóbal de los Ríos
- Instituto‐Fundación Teófilo Hernando, Departamento de Farmacología y TerapéuticaUniversidad Autónoma de MadridMadridSpain
- Instituto de Investigación SanitariaHospital Universitario de la PrincesaMadridSpain
| |
Collapse
|
22
|
Gonzalez D, Arribas RL, Viejo L, Lajarin-Cuesta R, de los Rios C. Substituent effect of N-benzylated gramine derivatives that prevent the PP2A inhibition and dissipate the neuronal Ca2+ overload, as a multitarget strategy for the treatment of Alzheimer’s disease. Bioorg Med Chem 2018; 26:2551-2560. [DOI: 10.1016/j.bmc.2018.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/06/2018] [Accepted: 04/07/2018] [Indexed: 11/28/2022]
|
23
|
Borowiecki P, Justyniak I, Ochal Z. Lipase-catalyzed kinetic resolution approach toward enantiomerically enriched 1-(β-hydroxypropyl)indoles. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.tetasy.2017.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
24
|
Ramos E, Patiño P, Reiter RJ, Gil-Martín E, Marco-Contelles J, Parada E, de Los Rios C, Romero A, Egea J. Ischemic brain injury: New insights on the protective role of melatonin. Free Radic Biol Med 2017; 104:32-53. [PMID: 28065781 DOI: 10.1016/j.freeradbiomed.2017.01.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 12/20/2016] [Accepted: 01/04/2017] [Indexed: 12/15/2022]
Abstract
Stroke represents one of the most common causes of brain's vulnerability for many millions of people worldwide. The plethora of physiopathological events associated with brain ischemia are regulate through multiple signaling pathways leading to the activation of oxidative stress process, Ca2+ dyshomeostasis, mitochondrial dysfunction, proinflammatory mediators, excitotoxicity and/or programmed neuronal cell death. Understanding this cascade of molecular events is mandatory in order to develop new therapeutic strategies for stroke. In this review article, we have highlighted the pleiotropic effects of melatonin to counteract the multiple processes of the ischemic cascade. Additionally, experimental evidence supports its actions to ameliorate ischemic long-term behavioural and neuronal deficits, preserving the functional integrity of the blood-brain barrier, inducing neurogenesis and cell proliferation through receptor-dependent mechanism, as well as improving synaptic transmission. Consequently, the synthesis of melatonin derivatives designed as new multitarget-directed products has focused a great interest in this area. This latter has been reinforced by the low cost of melatonin and its reduced toxicity. Furthermore, its spectrum of usages seems to be wide and with the potential for improving human health. Nevertheless, the molecular and cellular mechanisms underlying melatonin´s actions need to be further exploration and accordingly, new clinical studies should be conducted in human patients with ischemic brain pathologies.
Collapse
Affiliation(s)
- Eva Ramos
- Department of Toxicology & Pharmacology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Paloma Patiño
- Paediatric Unit, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Russel J Reiter
- Department of Cellular and Structural Biology. University of Texas Health Science Center at San Antonio, USA
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, Vigo, Spain
| | - José Marco-Contelles
- Medicinal Chemistry Laboratory, Institute of General Organic Chemistry (CSIC), Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Esther Parada
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain; Instituto de I+D del Medicamento Teófilo Hernando (ITH), Facultad de Medicina, Universidad Autónoma de Madrid, Spain
| | - Cristobal de Los Rios
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain; Instituto de I+D del Medicamento Teófilo Hernando (ITH), Facultad de Medicina, Universidad Autónoma de Madrid, Spain
| | - Alejandro Romero
- Department of Toxicology & Pharmacology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Javier Egea
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain; Instituto de I+D del Medicamento Teófilo Hernando (ITH), Facultad de Medicina, Universidad Autónoma de Madrid, Spain.
| |
Collapse
|