1
|
Humpert S, Schneider D, Bier D, Schulze A, Neumaier F, Neumaier B, Holschbach M. 8-Bicycloalkyl-CPFPX derivatives as potent and selective tools for in vivo imaging of the A 1 adenosine receptor. Eur J Med Chem 2024; 271:116380. [PMID: 38615410 DOI: 10.1016/j.ejmech.2024.116380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/16/2024]
Abstract
Imaging of the A1 adenosine receptor (A1R) by positron emission tomography (PET) with 8-cyclopentyl-3-(3-[18F]fluoropropyl)-1-propyl-xanthine ([18F]CPFPX) has been widely used in preclinical and clinical studies. However, this radioligand suffers from rapid peripheral metabolism and subsequent accumulation of radiometabolites in the vascular compartment. In the present work, we prepared four derivatives of CPFPX by replacement of the cyclopentyl group with norbornane moieties. These derivatives were evaluated by competition binding studies, microsomal stability assays and LC-MS analysis of microsomal metabolites. In addition, the 18F-labeled isotopologue of 8-(1-norbornyl)-3-(3-fluoropropyl)-1-propylxanthine (1-NBX) as the most promising candidate was prepared by radiofluorination of the corresponding tosylate precursor and the resulting radioligand ([18F]1-NBX) was evaluated by permeability assays with Caco-2 cells and in vitro autoradiography in rat brain slices. Our results demonstrate that 1-NBX exhibits significantly improved A1R affinity and selectivity when compared to CPFPX and that it does not give rise to lipophilic metabolites expected to cross the blood-brain-barrier in microsomal assays. Furthermore, [18F]1-NBX showed a high passive permeability (Pc = 6.9 ± 2.9 × 10-5 cm/s) and in vitro autoradiography with this radioligand resulted in a distribution pattern matching A1R expression in the brain. Moreover, a low degree of non-specific binding (5%) was observed. Taken together, these findings identify [18F]1-NBX as a promising candidate for further preclinical evaluation as potential PET tracer for A1R imaging.
Collapse
Affiliation(s)
- Swen Humpert
- Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428, Jülich, Germany
| | - Daniela Schneider
- Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428, Jülich, Germany
| | - Dirk Bier
- Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428, Jülich, Germany
| | - Annette Schulze
- Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428, Jülich, Germany
| | - Felix Neumaier
- Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428, Jülich, Germany; Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Bernd Neumaier
- Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428, Jülich, Germany; Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany; Max Planck Institute for Metabolism Research, Gleueler Straße 50, 50931, Cologne, Germany.
| | - Marcus Holschbach
- Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428, Jülich, Germany
| |
Collapse
|
2
|
Perfilova VN, Muzyko EA, Taran AS, Shevchenko AA, Naumenko LV. Problems and prospects for finding new pharmacological agents among adenosine receptor agonists, antagonists, or their allosteric modulators for the treatment of cardiovascular diseases. BIOMEDITSINSKAIA KHIMIIA 2023; 69:353-370. [PMID: 38153051 DOI: 10.18097/pbmc20236906353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
A1-adenosine receptors (A1AR) are widely distributed in the human body and mediate many different effects. They are abundantly present in the cardiovascular system, where they control angiogenesis, vascular tone, heart rate, and conduction. This makes the cardiovascular system A1AR an attractive target for the treatment of cardiovascular diseases (CVD). The review summarizes the literature data on the structure and functioning of A1AR, and analyzes their involvement in the formation of myocardial hypertrophy, ischemia-reperfusion damage, various types of heart rhythm disorders, chronic heart failure, and arterial hypertension. Special attention is paid to the role of some allosteric regulators of A1AR as potential agents for the CVD treatment.
Collapse
Affiliation(s)
- V N Perfilova
- Volgograd State Medical University, Volgograd, Russia; Volgograd Medical Research Center, Volgograd, Russia
| | - E A Muzyko
- Volgograd State Medical University, Volgograd, Russia
| | - A S Taran
- Volgograd State Medical University, Volgograd, Russia
| | | | - L V Naumenko
- Volgograd State Medical University, Volgograd, Russia
| |
Collapse
|
3
|
Wang L, Graziano B, Encalada N, Fernandez-Abascal J, Kaplan DH, Bianchi L. Glial regulators of ions and solutes required for specific chemosensory functions in Caenorhabditis elegans. iScience 2022; 25:105684. [PMID: 36567707 PMCID: PMC9772852 DOI: 10.1016/j.isci.2022.105684] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/11/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Glia and accessory cells regulate the microenvironment around neurons and primary sensory cells. However, the impact of specific glial regulators of ions and solutes on functionally diverse primary cells is poorly understood. Here, we systemically investigate the requirement of ion channels and transporters enriched in Caenorhabditis elegans Amsh glia for the function of chemosensory neurons. Although Amsh glia ablated worms show reduced function of ASH, AWC, AWA, and ASE neurons, we show that the loss of glial enriched ion channels and transporters impacts these neurons differently, with nociceptor ASH being the most affected. Furthermore, our analysis underscores the importance of K+, Cl-, and nucleoside homeostasis in the Amphid sensory organ and uncovers the contribution of glial genes implicated in neurological disorders. Our findings build a unique fingerprint of each glial enriched ion channel and transporter and may provide insights into the function of supporting cells of mammalian sensory organs.
Collapse
Affiliation(s)
- Lei Wang
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Rm 5133 Rosenstiel Building, 1600 NW 10 Avenue, Miami, FL33136, USA
| | - Bianca Graziano
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Rm 5133 Rosenstiel Building, 1600 NW 10 Avenue, Miami, FL33136, USA
| | - Nicole Encalada
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Rm 5133 Rosenstiel Building, 1600 NW 10 Avenue, Miami, FL33136, USA
| | - Jesus Fernandez-Abascal
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Rm 5133 Rosenstiel Building, 1600 NW 10 Avenue, Miami, FL33136, USA
| | - Daryn H. Kaplan
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Rm 5133 Rosenstiel Building, 1600 NW 10 Avenue, Miami, FL33136, USA
| | - Laura Bianchi
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Rm 5133 Rosenstiel Building, 1600 NW 10 Avenue, Miami, FL33136, USA
| |
Collapse
|
4
|
Catarzi D, Varano F, Vigiani E, Calenda S, Melani F, Varani K, Vincenzi F, Pasquini S, Mennini N, Nerli G, Dal Ben D, Volpini R, Colotta V. 4-Heteroaryl Substituted Amino-3,5-Dicyanopyridines as New Adenosine Receptor Ligands: Novel Insights on Structure-Activity Relationships and Perspectives. Pharmaceuticals (Basel) 2022; 15:ph15040478. [PMID: 35455475 PMCID: PMC9024521 DOI: 10.3390/ph15040478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/09/2022] [Indexed: 12/04/2022] Open
Abstract
A new set of amino-3,5-dicyanopyridines was synthesized and biologically evaluated at the adenosine receptors (ARs). This chemical class is particularly versatile, as small structural modifications can influence not only affinity and selectivity, but also the pharmacological profile. Thus, in order to deepen the structure–activity relationships (SARs) of this series, different substituents were evaluated at the diverse positions on the dicyanopyridine scaffold. In general, the herein reported compounds show nanomolar binding affinity and interact better with both the human (h) A1 and A2A ARs than with the other subtypes. Docking studies at hAR structure were performed to rationalize the observed affinity data. Of interest are compounds 1 and 5, which can be considered as pan ligands as binding all the ARs with comparable nanomolar binding affinity (A1AR: 1, Ki = 9.63 nM; 5, Ki = 2.50 nM; A2AAR: 1, Ki = 21 nM; 5, Ki = 24 nM; A3AR: 1, Ki = 52 nM; 5, Ki = 25 nM; A2BAR: 1, EC50 = 1.4 nM; 5, EC50 = 1.12 nM). Moreover, these compounds showed a partial agonist profile at all the ARs. This combined AR partial agonist activity could lead us to hypothesize a potential effect in the repair process of damaged tissue that would be beneficial in both wound healing and remodeling.
Collapse
Affiliation(s)
- Daniela Catarzi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via Ugo Schiff, 6, 50019 Sesto Fiorentino, Italy; (F.V.); (E.V.); (S.C.); (F.M.); (V.C.)
- Correspondence:
| | - Flavia Varano
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via Ugo Schiff, 6, 50019 Sesto Fiorentino, Italy; (F.V.); (E.V.); (S.C.); (F.M.); (V.C.)
| | - Erica Vigiani
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via Ugo Schiff, 6, 50019 Sesto Fiorentino, Italy; (F.V.); (E.V.); (S.C.); (F.M.); (V.C.)
| | - Sara Calenda
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via Ugo Schiff, 6, 50019 Sesto Fiorentino, Italy; (F.V.); (E.V.); (S.C.); (F.M.); (V.C.)
| | - Fabrizio Melani
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via Ugo Schiff, 6, 50019 Sesto Fiorentino, Italy; (F.V.); (E.V.); (S.C.); (F.M.); (V.C.)
| | - Katia Varani
- Dipartimento di Medicina Traslazionale, Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (K.V.); (F.V.); (S.P.)
| | - Fabrizio Vincenzi
- Dipartimento di Medicina Traslazionale, Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (K.V.); (F.V.); (S.P.)
| | - Silvia Pasquini
- Dipartimento di Medicina Traslazionale, Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (K.V.); (F.V.); (S.P.)
| | - Natascia Mennini
- Dipartimento di Chimica Ugo Schiff, Università degli Studi di Firenze, Via della Lastruccia, 3, 50019 Sesto Fiorentino, Italy; (N.M.); (G.N.)
| | - Giulia Nerli
- Dipartimento di Chimica Ugo Schiff, Università degli Studi di Firenze, Via della Lastruccia, 3, 50019 Sesto Fiorentino, Italy; (N.M.); (G.N.)
| | - Diego Dal Ben
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università degli Studi di Camerino, Via S.Agostino 1, 62032 Camerino, Italy; (D.D.B.); (R.V.)
| | - Rosaria Volpini
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università degli Studi di Camerino, Via S.Agostino 1, 62032 Camerino, Italy; (D.D.B.); (R.V.)
| | - Vittoria Colotta
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via Ugo Schiff, 6, 50019 Sesto Fiorentino, Italy; (F.V.); (E.V.); (S.C.); (F.M.); (V.C.)
| |
Collapse
|
5
|
Larrick JW, Larrick JW, Mendelsohn AR. Response to Hypoxia in Cognitive Decline. Rejuvenation Res 2021; 24:319-324. [PMID: 34314252 DOI: 10.1089/rej.2021.0051] [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] [Indexed: 12/20/2022] Open
Abstract
Inflammaging, the increase of proinflammatory processes with increasing age, has multiple mechanisms from increasing numbers of senescent cells secreting cytokines to changes in metabolic processes. Alterations of oxygen metabolism with aging, especially decreased levels of O2 with age resulting from endocrine and cardiovascular dysfunction as well as desensitization of cellular response to hypoxia, may exacerbate inflammaging, which in turn creates further oxygen metabolic dysfunction. During aging, decline in levels of erythrocyte 2,3-bisphosphoglycerate (2,3-BPG), BPG mutase, and adenosine A2B receptor, a key adenosine signaling receptor that can augment 2,3-BPG expression, may fail to protect sensitive brain tissue from subtly reduced O2 levels, in turn resulting in increased numbers of activated microglia and secretion of proinflammatory cytokines, ultimately promoting inflammaging and senescence of endothelial cells. Interventions to restore O2 levels directly or via increasing 2,3-BPG may help promote cognitive health in old age, but significant work to quantify the degree of reduced O2 during aging in mammals, and especially humans, needs to be pursued.
Collapse
Affiliation(s)
- James W Larrick
- Panorama Research Institute and Regenerative Sciences Institute, Sunnyvale, California, USA
| | - Jasmine W Larrick
- Division of Pulmonary, Critical Care and Immunology, University of California, San Francisco, San Francisco, California, USA
| | - Andrew R Mendelsohn
- Panorama Research Institute and Regenerative Sciences Institute, Sunnyvale, California, USA
| |
Collapse
|
6
|
Zarrinmayeh H, Territo PR. Purinergic Receptors of the Central Nervous System: Biology, PET Ligands, and Their Applications. Mol Imaging 2021; 19:1536012120927609. [PMID: 32539522 PMCID: PMC7297484 DOI: 10.1177/1536012120927609] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purinergic receptors play important roles in central nervous system (CNS). These receptors are involved in cellular neuroinflammatory responses that regulate functions of neurons, microglial and astrocytes. Based on their endogenous ligands, purinergic receptors are classified into P1 or adenosine, P2X and P2Y receptors. During brain injury or under pathological conditions, rapid diffusion of extracellular adenosine triphosphate (ATP) or uridine triphosphate (UTP) from the damaged cells, promote microglial activation that result in the changes in expression of several of these receptors in the brain. Imaging of the purinergic receptors with selective Positron Emission Tomography (PET) radioligands has advanced our understanding of the functional roles of some of these receptors in healthy and diseased brains. In this review, we have accumulated a list of currently available PET radioligands of the purinergic receptors that are used to elucidate the receptor functions and participations in CNS disorders. We have also reviewed receptors lacking radiotracer, laying the foundation for future discoveries of novel PET radioligands to reveal these receptors roles in CNS disorders.
Collapse
Affiliation(s)
- Hamideh Zarrinmayeh
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul R Territo
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
7
|
Qiang Q, Manalo JM, Sun H, Zhang Y, Song A, Wen AQ, Wen YE, Chen C, Liu H, Cui Y, Nemkov T, Reisz JA, Edwards III G, Perreira FA, Kellems RE, Soto C, D’Alessandro A, Xia Y. Erythrocyte adenosine A2B receptor prevents cognitive and auditory dysfunction by promoting hypoxic and metabolic reprogramming. PLoS Biol 2021; 19:e3001239. [PMID: 34138843 PMCID: PMC8211187 DOI: 10.1371/journal.pbio.3001239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
Hypoxia drives aging and promotes age-related cognition and hearing functional decline. Despite the role of erythrocytes in oxygen (O2) transport, their role in the onset of aging and age-related cognitive decline and hearing loss (HL) remains undetermined. Recent studies revealed that signaling through the erythrocyte adenosine A2B receptor (ADORA2B) promotes O2 release to counteract hypoxia at high altitude. However, nothing is known about a role for erythrocyte ADORA2B in age-related functional decline. Here, we report that loss of murine erythrocyte-specific ADORA2B (eAdora2b-/-) accelerates early onset of age-related impairments in spatial learning, memory, and hearing ability. eAdora2b-/- mice display the early aging-like cellular and molecular features including the proliferation and activation of microglia and macrophages, elevation of pro-inflammatory cytokines, and attenuation of hypoxia-induced glycolytic gene expression to counteract hypoxia in the hippocampus (HIP), cortex, or cochlea. Hypoxia sufficiently accelerates early onset of cognitive and cochlear functional decline and inflammatory response in eAdora2b-/- mice. Mechanistically, erythrocyte ADORA2B-mediated activation of AMP-activated protein kinase (AMPK) and bisphosphoglycerate mutase (BPGM) promotes hypoxic and metabolic reprogramming to enhance production of 2,3-bisphosphoglycerate (2,3-BPG), an erythrocyte-specific metabolite triggering O2 delivery. Significantly, this finding led us to further discover that murine erythroblast ADORA2B and BPGM mRNA levels and erythrocyte BPGM activity are reduced during normal aging. Overall, we determined that erythrocyte ADORA2B-BPGM axis is a key component for anti-aging and anti-age-related functional decline.
Collapse
Affiliation(s)
- Qingfen Qiang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Jeanne M. Manalo
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Hong Sun
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yujin Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Anren Song
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Alexander Q. Wen
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of California at San Diego, La Jolla, California, United States of America
| | - Y. Edward Wen
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas Southwestern Medical School, Dallas, Texas, United States of America
| | - Changhan Chen
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Hong Liu
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Cui
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - George Edwards III
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Fred A. Perreira
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rodney E. Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Claudio Soto
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| |
Collapse
|
8
|
Kundu D, Dubey VK. Purines and Pyrimidines: Metabolism, Function and Potential as Therapeutic Options in Neurodegenerative Diseases. Curr Protein Pept Sci 2021; 22:170-189. [PMID: 33292151 DOI: 10.2174/1389203721999201208200605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/01/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
Various neurodegenerative disorders have various molecular origins but some common molecular mechanisms. In the current scenario, there are very few treatment regimens present for advanced neurodegenerative diseases. In this context, there is an urgent need for alternate options in the form of natural compounds with an ameliorating effect on patients. There have been individual scattered experiments trying to identify potential values of various intracellular metabolites. Purines and Pyrimidines, which are vital molecules governing various aspects of cellular biochemical reactions, have been long sought as crucial candidates for the same, but there are still many questions that go unanswered. Some critical functions of these molecules associated with neuromodulation activities have been identified. They are also known to play a role in foetal neurodevelopment, but there is a lacuna in understanding their mechanisms. In this review, we have tried to assemble and identify the importance of purines and pyrimidines, connecting them with the prevalence of neurodegenerative diseases. The leading cause of this class of diseases is protein misfolding and the formation of amyloids. A direct correlation between loss of balance in cellular homeostasis and amyloidosis is yet an unexplored area. This review aims at bringing the current literature available under one umbrella serving as a foundation for further extensive research in this field of drug development in neurodegenerative diseases.
Collapse
Affiliation(s)
- Debanjan Kundu
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, UP - 221005, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, UP - 221005, India
| |
Collapse
|
9
|
Marucci G, Ben DD, Lambertucci C, Navia AM, Spinaci A, Volpini R, Buccioni M. Combined Therapy of A 1AR Agonists and A 2AAR Antagonists in Neuroinflammation. Molecules 2021; 26:1188. [PMID: 33672225 PMCID: PMC7926490 DOI: 10.3390/molecules26041188] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's, Parkinson's, and multiple sclerosis are neurodegenerative diseases related by neuronal degeneration and death in specific areas of the central nervous system. These pathologies are associated with neuroinflammation, which is involved in disease progression, and halting this process represents a potential therapeutic strategy. Evidence suggests that microglia function is regulated by A1 and A2A adenosine receptors (AR), which are considered as neuroprotective and neurodegenerative receptors, respectively. The manuscript's aim is to elucidate the role of these receptors in neuroinflammation modulation through potent and selective A1AR agonists (N6-cyclopentyl-2'- or 3'-deoxyadenosine substituted or unsubstituted in 2 position) and A2AAR antagonists (9-ethyl-adenine substituted in 8 and/or in 2 position), synthesized in house, using N13 microglial cells. In addition, the combined therapy of A1AR agonists and A2AAR antagonists to modulate neuroinflammation was evaluated. Results showed that A1AR agonists were able, to varying degrees, to prevent the inflammatory effect induced by cytokine cocktail (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and interferon (IFN)-γ), while A2AAR antagonists showed a good ability to counteract neuroinflammation. Moreover, the effect achieved by combining the two most effective compounds (1 and 6) in doses previously found to be non-effective was greater than the treatment effect of each of the two compounds used separately at maximal dose.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michela Buccioni
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, 62032 Camerino, MC, Italy; (G.M.); (D.D.B.); (C.L.); (A.M.N.); (A.S.); (R.V.)
| |
Collapse
|
10
|
Effendi WI, Nagano T, Kobayashi K, Nishimura Y. Focusing on Adenosine Receptors as a Potential Targeted Therapy in Human Diseases. Cells 2020; 9:E785. [PMID: 32213945 PMCID: PMC7140859 DOI: 10.3390/cells9030785] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Adenosine is involved in a range of physiological and pathological effects through membrane-bound receptors linked to G proteins. There are four subtypes of adenosine receptors, described as A1AR, A2AAR, A2BAR, and A3AR, which are the center of cAMP signal pathway-based drug development. Several types of agonists, partial agonists or antagonists, and allosteric substances have been synthesized from these receptors as new therapeutic drug candidates. Research efforts surrounding A1AR and A2AAR are perhaps the most enticing because of their concentration and affinity; however, as a consequence of distressing conditions, both A2BAR and A3AR levels might accumulate. This review focuses on the biological features of each adenosine receptor as the basis of ligand production and describes clinical studies of adenosine receptor-associated pharmaceuticals in human diseases.
Collapse
Affiliation(s)
- Wiwin Is Effendi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| |
Collapse
|
11
|
Schneider D, Oskamp A, Holschbach M, Neumaier B, Bier D, Bauer A. Influence of binding affinity and blood plasma level on cerebral pharmacokinetics and PET imaging characteristics of two novel xanthine PET radioligands for the A1 adenosine receptor. Nucl Med Biol 2020; 82-83:1-8. [DOI: 10.1016/j.nucmedbio.2019.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/16/2019] [Accepted: 12/01/2019] [Indexed: 10/25/2022]
|
12
|
Pereira-Figueiredo D, Brito R, Araújo DSM, Nascimento AA, Lyra ESB, Cheibub AMSS, Pereira Netto AD, Ventura ALM, Paes-de-Carvalho R, Calaza KC. Caffeine exposure ameliorates acute ischemic cell death in avian developing retina. Purinergic Signal 2020; 16:41-59. [PMID: 32078115 PMCID: PMC7166236 DOI: 10.1007/s11302-020-09687-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022] Open
Abstract
In infants, the main cause of blindness is retinopathy of prematurity that stems in a hypoxic-ischemic condition. Caffeine is a psychoactive compound that at low to moderate concentrations, selectively inhibits adenosine A1 and A2A receptors. Caffeine exerts beneficial effects in central nervous system of adult animal models and humans, whereas it seems to have malefic effect on the developing tissue. We observed that 48-h exposure (during synaptogenesis) to a moderate dose of caffeine (30 mg/kg of egg) activated pro-survival signaling pathways, including ERK, CREB, and Akt phosphorylation, alongside BDNF production, and reduced retinal cell death promoted by oxygen glucose deprivation in the chick retina. Blockade of TrkB receptors and inhibition of CREB prevented caffeine protection effect. Similar signaling pathways were described in previously reported data concerning chemical preconditioning mechanism triggered by NMDA receptors activation, with low concentrations of agonist. In agreement to these data, caffeine increased NMDA receptor activity. Caffeine decreased the levels of the chloride co-transporter KCC2 and delayed the developmental shift on GABAA receptor response from depolarizing to hyperpolarizing. These results suggest that the caffeine-induced delaying in depolarizing effect of GABA could be facilitating NMDA receptor activity. DPCPX, an A1 adenosine receptor antagonist, but not A2A receptor inhibitor, mimicked the effect of caffeine, suggesting that the effect of caffeine occurs through A1 receptor blockade. In summary, an in vivo caffeine exposure could increase the resistance of the retina to ischemia-induced cell death, by triggering survival pathways involving CREB phosphorylation and BDNF production/TrkB activation.
Collapse
Affiliation(s)
- D. Pereira-Figueiredo
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
| | - R. Brito
- Cellular Signaling and Metabolic Modulation Laboratory, Cellular and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, RJ Brazil
| | - D. S. M. Araújo
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - A. A. Nascimento
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
| | - E. S. B. Lyra
- Fundamental and Applied Analytical Chemistry Laboratory (LAQAFA), Department of Analytical Chemistry, Chemistry Institute, Fluminense Federal University, Niterói, RJ Brazil
| | - A. M. S. S. Cheibub
- Fundamental and Applied Analytical Chemistry Laboratory (LAQAFA), Department of Analytical Chemistry, Chemistry Institute, Fluminense Federal University, Niterói, RJ Brazil
| | - A. D. Pereira Netto
- Fundamental and Applied Analytical Chemistry Laboratory (LAQAFA), Department of Analytical Chemistry, Chemistry Institute, Fluminense Federal University, Niterói, RJ Brazil
| | - A. L. M. Ventura
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
- Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, Rio de Janeiro Brazil
| | - R. Paes-de-Carvalho
- Laboratory of Cellular Neurobiology, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
- Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, Rio de Janeiro Brazil
| | - K. C. Calaza
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
- Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, Rio de Janeiro Brazil
| |
Collapse
|
13
|
Alves ACDB, Bristot VJDO, Limana MD, Speck AE, Barros LSD, Solano AF, Aguiar AS. Role of Adenosine A 2A Receptors in the Central Fatigue of Neurodegenerative Diseases. J Caffeine Adenosine Res 2019. [DOI: 10.1089/caff.2019.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Ana Cristina de Bem Alves
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | | | - Mirieli Denardi Limana
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | - Ana Elisa Speck
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | - Leonardo Soares de Barros
- LABOX—Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, UFSC—Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Alexandre Francisco Solano
- LABOX—Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, UFSC—Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Aderbal S. Aguiar
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| |
Collapse
|
14
|
Blaise JH, Park JE, Bellas NJ, Gitchell TM, Phan V. Caffeine consumption disrupts hippocampal long-term potentiation in freely behaving rats. Physiol Rep 2019; 6. [PMID: 29512310 PMCID: PMC5840440 DOI: 10.14814/phy2.13632] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/25/2018] [Accepted: 01/31/2018] [Indexed: 12/24/2022] Open
Abstract
Caffeine, one of the most commonly consumed psychoactive substances in the world, has long been known to alter neurological functions, such as alertness, attention, and memory. Despite caffeine's popularity, systematic investigations of its effects on synaptic plasticity in the brain are still lacking. Here we used a freely behaving rodent model of long-term potentiation (LTP), a frequently studied form of synaptic plasticity, to assess the effects of caffeine consumption on hippocampal plasticity. LTP, which is a persistent increase in the strength of synaptic connections between neurons, is a cellular mechanism widely considered to underlie the processes of learning and memory. A group of 10-week-old Sprague-Dawley rats were administered caffeine (1 g/L) in their drinking water 3 weeks prior to collection of electrophysiological data. Another group of age-matched animals received tap water and served as controls. Stimulating and recording electrodes were chronically implanted in the perforant pathway (PP) and dentate gyrus (DG) region of the hippocampus, respectively, to permit stable electrophysiological recordings of synaptic transmission at this synapse. Population spike amplitude (PSA) measures of LTP induction and duration were acquired in vivo while animals were freely behaving using a well-established electrophysiological recording protocol. Results indicate caffeine-treated rats (n = 9) had a significantly (P < 0.05) reduced level of LTP induction compared with controls (n = 10). More studies are needed to identify the exact mechanism through which caffeine alters LTP induction in this freely behaving model of synaptic plasticity.
Collapse
Affiliation(s)
- J Harry Blaise
- Interdisciplinary Science Program, Trinity College, Hartford, Connecticut.,Neuroscience Program, Trinity College, Hartford, Connecticut.,Engineering Department, Trinity College, Hartford, Connecticut
| | - Jee E Park
- Interdisciplinary Science Program, Trinity College, Hartford, Connecticut.,Biology Department, Trinity College, Hartford, Connecticut
| | - Nicholas J Bellas
- Interdisciplinary Science Program, Trinity College, Hartford, Connecticut.,Chemistry Department, Trinity College, Hartford, Connecticut
| | - Thomas M Gitchell
- Interdisciplinary Science Program, Trinity College, Hartford, Connecticut.,Neuroscience Program, Trinity College, Hartford, Connecticut
| | - Vy Phan
- Interdisciplinary Science Program, Trinity College, Hartford, Connecticut.,Neuroscience Program, Trinity College, Hartford, Connecticut
| |
Collapse
|
15
|
Hughes B, Herron CE. Cannabidiol Reverses Deficits in Hippocampal LTP in a Model of Alzheimer's Disease. Neurochem Res 2018; 44:703-713. [PMID: 29574668 DOI: 10.1007/s11064-018-2513-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 01/12/2023]
Abstract
Here we demonstrate for the first time that cannabidiol (CBD) acts to protect synaptic plasticity in an in vitro model of Alzheimer's disease (AD). The non-psycho active component of Cannabis sativa, CBD has previously been shown to protect against the neurotoxic effects of beta amyloid peptide (Aβ) in cell culture and cognitive behavioural models of neurodegeneration. Hippocampal long-term potentiation (LTP) is an activity dependent increase in synaptic efficacy often used to study cellular mechanisms related to memory. Here we show that acute application of soluble oligomeric beta amyloid peptide (Aβ1-42) associated with AD, attenuates LTP in the CA1 region of hippocampal slices from C57Bl/6 mice. Application of CBD alone did not alter LTP, however pre-treatment of slices with CBD rescued the Aβ1-42 mediated deficit in LTP. We found that the neuroprotective effects of CBD were not reversed by WAY100635, ZM241385 or AM251, demonstrating a lack of involvement of 5HT1A, adenosine (A2A) or Cannabinoid type 1 (CB1) receptors respectively. However in the presence of the PPARγ antagonist GW9662 the neuroprotective effect of CBD was prevented. Our data suggests that this major component of Cannabis sativa, which lacks psychoactivity may have therapeutic potential for the treatment of AD.
Collapse
Affiliation(s)
- Blathnaid Hughes
- School of Biomolecular and Biomedical Sciences, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Caroline E Herron
- School of Biomolecular and Biomedical Sciences, Conway Institute, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
16
|
Impact of Coffee and Cacao Purine Metabolites on Neuroplasticity and Neurodegenerative Disease. Neurochem Res 2018; 44:214-227. [PMID: 29417473 DOI: 10.1007/s11064-018-2492-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/31/2018] [Accepted: 02/02/2018] [Indexed: 12/20/2022]
Abstract
Increasing evidence suggests that regular consumption of coffee, tea and dark chocolate (cacao) can promote brain health and may reduce the risk of age-related neurodegenerative disorders. However, the complex array of phytochemicals in coffee and cacao beans and tea leaves has hindered a clear understanding of the component(s) that affect neuronal plasticity and resilience. One class of phytochemicals present in relatively high amounts in coffee, tea and cacao are methylxanthines. Among such methylxanthines, caffeine has been the most widely studied and has clear effects on neuronal network activity, promotes sustained cognitive performance and can protect neurons against dysfunction and death in animal models of stroke, Alzheimer's disease and Parkinson's disease. Caffeine's mechanism of action relies on antagonism of various subclasses of adenosine receptors. Downstream xanthine metabolites, such as theobromine and theophylline, may also contribute to the beneficial effects of coffee, tea and cacao on brain health.
Collapse
|
17
|
Zhang MQ, Li R, Wang YQ, Huang ZL. Neural Plasticity Is Involved in Physiological Sleep, Depressive Sleep Disturbances, and Antidepressant Treatments. Neural Plast 2017; 2017:5870735. [PMID: 29181202 PMCID: PMC5664320 DOI: 10.1155/2017/5870735] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/27/2017] [Accepted: 07/13/2017] [Indexed: 12/28/2022] Open
Abstract
Depression, which is characterized by a pervasive and persistent low mood and anhedonia, greatly impacts patients, their families, and society. The associated and recurring sleep disturbances further reduce patient's quality of life. However, therapeutic sleep deprivation has been regarded as a rapid and robust antidepressant treatment for several decades, which suggests a complicated role of sleep in development of depression. Changes in neural plasticity are observed during physiological sleep, therapeutic sleep deprivation, and depression. This correlation might help us to understand better the mechanism underlying development of depression and the role of sleep. In this review, we first introduce the structure of sleep and the facilitated neural plasticity caused by physiological sleep. Then, we introduce sleep disturbances and changes in plasticity in patients with depression. Finally, the effects and mechanisms of antidepressants and therapeutic sleep deprivation on neural plasticity are discussed.
Collapse
Affiliation(s)
- Meng-Qi Zhang
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Rui Li
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yi-Qun Wang
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Zhi-Li Huang
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
18
|
Adenosine A1 and A2A Receptors in the Brain: Current Research and Their Role in Neurodegeneration. Molecules 2017; 22:molecules22040676. [PMID: 28441750 PMCID: PMC6154612 DOI: 10.3390/molecules22040676] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022] Open
Abstract
The inhibitory adenosine A1 receptor (A1R) and excitatory A2A receptor (A2AR) are predominantly expressed in the brain. Whereas the A2AR has been implicated in normal aging and enhancing neurotoxicity in multiple neurodegenerative diseases, the inhibitory A1R has traditionally been ascribed to have a neuroprotective function in various brain insults. This review provides a summary of the emerging role of prolonged A1R signaling and its potential cross-talk with A2AR in the cellular basis for increased neurotoxicity in neurodegenerative disorders. This A1R signaling enhances A2AR-mediated neurodegeneration, and provides a platform for future development of neuroprotective agents in stroke, Parkinson’s disease and epilepsy.
Collapse
|
19
|
Picchioni D, Reith RM, Nadel JL, Smith CB. Sleep, plasticity and the pathophysiology of neurodevelopmental disorders: the potential roles of protein synthesis and other cellular processes. Brain Sci 2014; 4:150-201. [PMID: 24839550 PMCID: PMC4020186 DOI: 10.3390/brainsci4010150] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/26/2014] [Accepted: 03/07/2014] [Indexed: 12/28/2022] Open
Abstract
Sleep is important for neural plasticity, and plasticity underlies sleep-dependent memory consolidation. It is widely appreciated that protein synthesis plays an essential role in neural plasticity. Studies of sleep-dependent memory and sleep-dependent plasticity have begun to examine alterations in these functions in populations with neurological and psychiatric disorders. Such an approach acknowledges that disordered sleep may have functional consequences during wakefulness. Although neurodevelopmental disorders are not considered to be sleep disorders per se, recent data has revealed that sleep abnormalities are among the most prevalent and common symptoms and may contribute to the progression of these disorders. The main goal of this review is to highlight the role of disordered sleep in the pathology of neurodevelopmental disorders and to examine some potential mechanisms by which sleep-dependent plasticity may be altered. We will also briefly attempt to extend the same logic to the other end of the developmental spectrum and describe a potential role of disordered sleep in the pathology of neurodegenerative diseases. We conclude by discussing ongoing studies that might provide a more integrative approach to the study of sleep, plasticity, and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Dante Picchioni
- Behavioral Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; E-Mail:
- Advanced MRI Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| | - R. Michelle Reith
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| | - Jeffrey L. Nadel
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| | - Carolyn B. Smith
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| |
Collapse
|
20
|
Nagpure BV, Bian JS. Hydrogen sulfide inhibits A2A adenosine receptor agonist induced β-amyloid production in SH-SY5Y neuroblastoma cells via a cAMP dependent pathway. PLoS One 2014; 9:e88508. [PMID: 24523906 PMCID: PMC3921165 DOI: 10.1371/journal.pone.0088508] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 01/11/2014] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of senile dementia in today's society. Its debilitating symptoms are manifested by disturbances in many important brain functions, which are influenced by adenosine. Hence, adenosinergic system is considered as a potential therapeutic target in AD treatment. In the present study, we found that sodium hydrosulfide (NaHS, an H2S donor, 100 µM) attenuated HENECA (a selective A2A receptor agonist, 10-200 nM) induced β-amyloid (1-42) (Aβ42) production in SH-SY5Y cells. NaHS also interfered with HENECA-stimulated production and post-translational modification of amyloid precursor protein (APP) by inhibiting its maturation. Measurement of the C-terminal APP fragments generated from its enzymatic cleavage by β-site amyloid precursor protein cleaving enzyme 1 (BACE1) showed that NaHS did not have any significant effect on β-secretase activity. However, the direct measurements of HENECA-elevated γ-secretase activity and mRNA expressions of presenilins suggested that the suppression of Aβ42 production in NaHS pretreated cells was mediated by inhibiting γ-secretase. NaHS induced reductions were accompanied by similar decreases in intracellular cAMP levels and phosphorylation of cAMP responsive element binding protein (CREB). NaHS significantly reduced the elevated cAMP and Aβ42 production caused by forskolin (an adenylyl cyclase, AC agonist) alone or forskolin in combination with IBMX (a phosphodiesterase inhibitor), but had no effect on those caused by IBMX alone. Moreover, pretreatment with NaHS significantly attenuated HENECA-elevated AC activity and mRNA expressions of various AC isoforms. These data suggest that NaHS may preferentially suppress AC activity when it was stimulated. In conclusion, H2S attenuated HENECA induced Aβ42 production in SH-SY5Y neuroblastoma cells through inhibiting γ-secretase via a cAMP dependent pathway.
Collapse
Affiliation(s)
- Bhushan Vijay Nagpure
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- * E-mail:
| |
Collapse
|
21
|
Sheth S, Brito R, Mukherjea D, Rybak LP, Ramkumar V. Adenosine receptors: expression, function and regulation. Int J Mol Sci 2014; 15:2024-52. [PMID: 24477263 PMCID: PMC3958836 DOI: 10.3390/ijms15022024] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
Adenosine receptors (ARs) comprise a group of G protein-coupled receptors (GPCR) which mediate the physiological actions of adenosine. To date, four AR subtypes have been cloned and identified in different tissues. These receptors have distinct localization, signal transduction pathways and different means of regulation upon exposure to agonists. This review will describe the biochemical characteristics and signaling cascade associated with each receptor and provide insight into how these receptors are regulated in response to agonists. A key property of some of these receptors is their ability to serve as sensors of cellular oxidative stress, which is transmitted by transcription factors, such as nuclear factor (NF)-κB, to regulate the expression of ARs. Recent observations of oligomerization of these receptors into homo- and heterodimers will be discussed. In addition, the importance of these receptors in the regulation of normal and pathological processes such as sleep, the development of cancers and in protection against hearing loss will be examined.
Collapse
Affiliation(s)
- Sandeep Sheth
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Rafael Brito
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Debashree Mukherjea
- Department of Surgery (Otolaryngology), Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Leonard P Rybak
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Vickram Ramkumar
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| |
Collapse
|
22
|
Brito R, Pereira MR, Paes-de-Carvalho R, Calaza KDC. Expression of A1 adenosine receptors in the developing avian retina: in vivo modulation by A(2A) receptors and endogenous adenosine. J Neurochem 2012; 123:239-49. [PMID: 22862679 DOI: 10.1111/j.1471-4159.2012.07909.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 07/30/2012] [Accepted: 08/01/2012] [Indexed: 12/13/2022]
Abstract
Little is known about the mechanisms that regulate the expression of adenosine receptors during CNS development. We demonstrate here that retinas from chick embryos injected in ovo with selective adenosine receptor ligands show changes in A1 receptor expression after 48 h. Exposure to A1 agonist N⁶-cyclohexyladenosine (CHA) or antagonist 8-Cyclopentyl-1, 3-dipropylxanthine (DPCPX) reduced or increased, respectively, A1 receptor protein and [³H]DPCPX binding, but together, CHA+DPCPX had no effect. Interestingly, treatment with A(2A) agonist 3-[4-[2-[[6-amino-9-[(2R,3R,4S,5S)-5-(ethylcarbamoyl)-3,4-dihydroxy-oxolan-2-yl]purin-2-yl]amino] ethyl]phenyl] propanoic acid (CGS21680) increased A1 receptor protein and [³H]DPCPX binding, and reduced A(2A) receptors. The A(2A) antagonists 7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-trizolo[1,5-c] pyrimidine (SCH58261) and 4-(2-[7-amino-2-[2-furyl][1,2,4]triazolo[2,3-a][1,3,5]triazo-5-yl-amino]ethyl)phenol (ZM241385) had opposite effects on A1 receptor expression. Exposure to CGS21680 + CHA did not change A1 receptor levels, whereas CHA + ZM241385 or CGS21680 + DPCPX had no synergic effect. The blockade of adenosine transporter with S-(4-nitrobenzyl)-6-thioinosine (NBMPR) also reduced [³H]DPCPX binding, an effect blocked by DPCPX, but not enhanced by ZM241385. [³H]DPCPX binding kinetics showed that treatment with CHA reduced and CGS21680 increased the Bmax, but did not affect Kd values. CHA, DPCPX, CGS21680, and ZM241385 had no effect on A1 receptor mRNA. These data demonstrated an in vivo regulation of A1 receptor expression by endogenous adenosine or long-term treatment with A1 and A(2A) receptors modulators.
Collapse
Affiliation(s)
- Rafael Brito
- Neurobiology of the Retina Laboratory, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niteroi, RJ, Brazil
| | | | | | | |
Collapse
|
23
|
Working memory and the homeostatic control of brain adenosine by adenosine kinase. Neuroscience 2012; 213:81-92. [PMID: 22521820 DOI: 10.1016/j.neuroscience.2012.03.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 03/12/2012] [Accepted: 03/13/2012] [Indexed: 12/16/2022]
Abstract
The neuromodulator adenosine maintains brain homeostasis and regulates complex behaviour via activation of inhibitory and excitatory adenosine receptors (ARs) in a brain region-specific manner. AR antagonists such as caffeine have been shown to ameliorate cognitive impairments in animal disease models but their effects on learning and memory in normal animals are equivocal. An alternative approach to reduce AR activation is to lower the extracellular tone of adenosine, which can be achieved by up-regulating adenosine kinase (ADK), the key enzyme of metabolic adenosine clearance. However, mice that globally over-express an Adk transgene ('Adk-tg' mice) were devoid of a caffeine-like pro-cognitive profile; they instead exhibited severe spatial memory deficits. This may be mechanistically linked to cortical/hippocampal N-methyl-d-aspartate receptor (NMDAR) hypofunction because the motor response to acute MK-801 was also potentiated in Adk-tg mice. Here, we evaluated the extent to which the behavioural phenotypes of Adk-tg mice might be modifiable by up-regulating adenosine levels in the cortex/hippocampus. To this end, we investigated mutant 'fb-Adk-def' mice in which ADK expression was specifically reduced in the telencephalon leading to a selective increase in cortical/hippocampal adenosine, while the rest of the brain remained as adenosine-deficient as in Adk-tg mice. The fb-Adk-def mice showed an even greater impairment in spatial working memory and a more pronounced motor response to NMDAR blockade than Adk-tg mice. These outcomes suggest that maintenance of cortical/hippocampal adenosine homeostasis is essential for effective spatial memory and deviation in either direction is detrimental with increased expression seemingly more disruptive than decreased expression.
Collapse
|
24
|
Burnstock G, Krügel U, Abbracchio MP, Illes P. Purinergic signalling: from normal behaviour to pathological brain function. Prog Neurobiol 2011; 95:229-74. [PMID: 21907261 DOI: 10.1016/j.pneurobio.2011.08.006] [Citation(s) in RCA: 315] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 08/12/2011] [Accepted: 08/15/2011] [Indexed: 02/07/2023]
Abstract
Purinergic neurotransmission, involving release of ATP as an efferent neurotransmitter was first proposed in 1972. Later, ATP was recognised as a cotransmitter in peripheral nerves and more recently as a cotransmitter with glutamate, noradrenaline, GABA, acetylcholine and dopamine in the CNS. Both ATP, together with some of its enzymatic breakdown products (ADP and adenosine) and uracil nucleotides are now recognised to act via P2X ion channels and P1 and P2Y G protein-coupled receptors, which are widely expressed in the brain. They mediate both fast signalling in neurotransmission and neuromodulation and long-term (trophic) signalling in cell proliferation, differentiation and death. Purinergic signalling is prominent in neurone-glial cell interactions. In this review we discuss first the evidence implicating purinergic signalling in normal behaviour, including learning and memory, sleep and arousal, locomotor activity and exploration, feeding behaviour and mood and motivation. Then we turn to the involvement of P1 and P2 receptors in pathological brain function; firstly in trauma, ischemia and stroke, then in neurodegenerative diseases, including Alzheimer's, Parkinson's and Huntington's, as well as multiple sclerosis and amyotrophic lateral sclerosis. Finally, the role of purinergic signalling in neuropsychiatric diseases (including schizophrenia), epilepsy, migraine, cognitive impairment and neuropathic pain will be considered.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK.
| | | | | | | |
Collapse
|
25
|
Costenla AR, Diógenes MJ, Canas PM, Rodrigues RJ, Nogueira C, Maroco J, Agostinho PM, Ribeiro JA, Cunha RA, de Mendonça A. Enhanced role of adenosine A2A receptors in the modulation of LTP in the rat hippocampus upon ageing. Eur J Neurosci 2011; 34:12-21. [DOI: 10.1111/j.1460-9568.2011.07719.x] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Ferguson AL, Stone TW. Glutamate-induced depression of EPSP-spike coupling in rat hippocampal CA1 neurons and modulation by adenosine receptors. Eur J Neurosci 2010; 31:1208-18. [PMID: 20345917 DOI: 10.1111/j.1460-9568.2010.07157.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The presence of high concentrations of glutamate in the extracellular fluid following brain trauma or ischaemia may contribute substantially to subsequent impairments of neuronal function. In this study, glutamate was applied to hippocampal slices for several minutes, producing over-depolarization, which was reflected in an initial loss of evoked population potential size in the CA1 region. Orthodromic population spikes recovered only partially over the following 60 min, whereas antidromic spikes and excitatory postsynaptic potentials (EPSPs) showed greater recovery, implying a change in EPSP-spike coupling (E-S coupling), which was confirmed by intracellular recording from CA1 pyramidal cells. The recovery of EPSPs was enhanced further by dizocilpine, suggesting that the long-lasting glutamate-induced change in E-S coupling involves NMDA receptors. This was supported by experiments showing that when isolated NMDA-receptor-mediated EPSPs were studied in isolation, there was only partial recovery following glutamate, unlike the composite EPSPs. The recovery of orthodromic population spikes and NMDA-receptor-mediated EPSPs following glutamate was enhanced by the adenosine A1 receptor blocker DPCPX, the A2A receptor antagonist SCH58261 or adenosine deaminase, associated with a loss of restoration to normal of the glutamate-induced E-S depression. The results indicate that the long-lasting depression of neuronal excitability following recovery from glutamate is associated with a depression of E-S coupling. This effect is partly dependent on activation of NMDA receptors, which modify adenosine release or the sensitivity of adenosine receptors. The results may have implications for the use of A1 and A2A receptor ligands as cognitive enhancers or neuroprotectants.
Collapse
Affiliation(s)
- Alexandra L Ferguson
- Neuroscience and Molecular Pharmacology, Faculty of Biomedical & Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | | |
Collapse
|
27
|
Fontinha BM, Diógenes MJ, Ribeiro JA, Sebastião AM. Enhancement of long-term potentiation by brain-derived neurotrophic factor requires adenosine A2A receptor activation by endogenous adenosine. Neuropharmacology 2008; 54:924-33. [PMID: 18384819 DOI: 10.1016/j.neuropharm.2008.01.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 01/08/2008] [Accepted: 01/30/2008] [Indexed: 10/22/2022]
Abstract
The excitatory action of brain-derived neurotrophic factor (BDNF) on synaptic transmission is triggered by adenosine A2A receptor activation. Since high-frequency neuronal firing, such as that inducing long-term potentiation (LTP), favours both A2A receptor activation and BDNF effects on transmission, we now evaluated the influence of adenosine on the facilitatory action of BDNF upon CA1 hippocampal LTP. theta-Burst stimulation of the pyramidal inputs induced a significant and persistent increase in field EPSP slopes, and this potentiation was augmented in the presence of BDNF (20 ng/ml), an action prevented by the inhibitor of Trk receptor autophosphorylation, K252a (200 nM). Removal of endogenous extracellular adenosine with adenosine deaminase (ADA, 1 U/ml), as well as the antagonism of adenosine A2A receptors with SCH58261 (100 nM), prevented the excitatory action of BDNF upon LTP. In an adenosine depleted background (with ADA), activation of adenosine A2A receptors (with 10nM CGS21680) restored the facilitatory effect of BDNF on LTP; this was fully prevented by the protein kinase A inhibitor, H-89 (1 microM) and mimicked by the adenylate cyclase activator, forskolin (10 microM). In similar experiments, activation of adenosine inhibitory A1 receptors (with 5 nM CPA) did not affect the facilitatory effect of BDNF. In conclusion, the facilitatory action of BDNF upon hippocampal LTP is critically dependent on the presence of extracellular adenosine and A2A receptor activation through a cAMP/PKA-dependent mechanism. Since extracellular adenosine accumulates upon high-frequency neuronal firing, the present results reveal a key process to allow the influence of BDNF upon synaptic plasticity.
Collapse
Affiliation(s)
- B M Fontinha
- Institute of Pharmacology and Neurosciences, Faculty of Medicine and Unit of Neurosciences, Institute of Molecular Medicine, University of Lisbon, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | | | | | | |
Collapse
|
28
|
Chen JF, Sonsalla PK, Pedata F, Melani A, Domenici MR, Popoli P, Geiger J, Lopes LV, de Mendonça A. Adenosine A2A receptors and brain injury: Broad spectrum of neuroprotection, multifaceted actions and “fine tuning” modulation. Prog Neurobiol 2007; 83:310-31. [DOI: 10.1016/j.pneurobio.2007.09.002] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 08/10/2007] [Accepted: 09/21/2007] [Indexed: 10/22/2022]
|
29
|
Cerqueira V, de Mendonça A, Minez A, Dias AR, de Carvalho M. Does caffeine modify corticomotor excitability? Neurophysiol Clin 2006; 36:219-26. [PMID: 17095411 DOI: 10.1016/j.neucli.2006.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2006] [Accepted: 06/29/2006] [Indexed: 11/30/2022] Open
Abstract
AIMS To test the influence of caffeine on the lower and upper motor neuron excitability. METHODS In Experiment A, 18 healthy subjects received 200 mg of caffeine or placebo, in a randomized, double-blind, placebo-controlled design protocol. Mean F-waves amplitude, amplitude of the motor response evoked by magnetic stimulation (MEP), MEP duration, cortical silent period (CSP), central conduction time, and cortical threshold were evaluated. In Experiment B, 6 healthy controls received 400 mg of caffeine, the peripheral silent period (PSP) and CSP were evaluated. CSP was recorded bilaterally in biceps brachii (intensity 10% above threshold) and abductor digiti minimi (ADM) (intensity at 10% and 50% above threshold). Muscle contraction was above 50% of the maximum force in both experiments. Latencies were defined by a technician who was not aware of this investigation. Serum caffeine level was evaluated. RESULTS In Experiment A, only the CSP, recorded in both ADM with intensity at 10% above threshold showed a significant change after caffeine (decrease of 17.1+/-34.0 ms, about 12% reduction). In Experiment B, PSP did not change, but CSP tested with intensities 10% above threshold was significantly decreased by 20.8+/-34.4 ms in ADM and 13.5+/-13.8 ms in biceps (about 13 and 16%, respectively). Serum caffeine level clearly increased after consumption but no correlation could be found between these levels and CSP reduction. CONCLUSIONS In our investigation, caffeine elicited a consistent decrease of the CSP, suggesting that caffeine increases cortical neuronal excitability.
Collapse
Affiliation(s)
- V Cerqueira
- Dementia Group, Institute of Molecular Medicine, Faculty of Medicine of Lisbon and Hospital de Santa Maria, Portugal
| | | | | | | | | |
Collapse
|
30
|
da Silva BM, de Mendonça A, Ribeiro JA. Long-term depression is not modulated by ATP receptors in the rat CA1 hippocampal region. Neurosci Lett 2005; 383:345-9. [PMID: 15882930 DOI: 10.1016/j.neulet.2005.04.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2005] [Revised: 04/18/2005] [Accepted: 04/19/2005] [Indexed: 11/23/2022]
Abstract
ATP is an important extracellular messenger in the CNS. In the hippocampus, a brain structure relevant for learning and memory processes, it acts both as a modulator and as a mediator of synaptic transmission, with implications for synaptic plasticity phenomena. Recent evidence suggests that ATP modulates activity-dependent long-term potentiation (LTP) of Schaffer collateral-CA1 synapses. However, it remains unclear if ATP also modulates LTP counterpart's phenomenon, long-term depression (LTD), in the rat hippocampus. This study investigated the effect of ATP analogues on homosynaptic LTD, induced by low-frequency stimulation of the Schaffer collaterals (1 Hz; 900 pulses) in the CA1 region of young rat hippocampal slices. The metabolically stable ATP analogues beta,gamma-ImATP (20 microM), a P2 receptor agonist, and alpha,beta-MeATP (20 microM), a preferential P2X(1,3) receptor agonist, did not modify LTD (LTD values of 14.7+/-0.5% and 14.1+/-3% for aCSF controls and of 15.1+/-4% and 19.0+/-5.2% for beta,gamma-ImATP and alpha,beta-MeATP, respectively). The ATP analogue beta,gamma-ImATP (20 microM) did not modify LTD also in the presence of the adenosine A1 receptor antagonist DPCPX (50 nM) (21.5+/-4.2% for DPCPX only and of 23.8+/-8.9% for DPCPX plus beta,gamma-ImATP). Finally, the preferential P2X(1,3) receptor antagonist NF023 (10 microM) had also no effect on LTD (18.6+/-5.2% for aCSF and of 18.7+/-5.2% for NF023). The present results suggest that ATP does not modulate activity-dependent homosynaptic LTD in the rat CA1 hippocampal region by activating P2 receptors.
Collapse
Affiliation(s)
- Bruno M da Silva
- Laboratory of Neurosciences, Institute of Molecular Medicine and Faculty of Medicine of Lisbon, Lisbon, Portugal.
| | | | | |
Collapse
|
31
|
Bhattacharya P, Leonard JT, Roy K. Exploring 3D-QSAR of thiazole and thiadiazole derivatives as potent and selective human adenosine A3 receptor antagonists+. J Mol Model 2005; 11:516-24. [PMID: 15928917 DOI: 10.1007/s00894-005-0273-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Accepted: 01/18/2005] [Indexed: 11/26/2022]
Abstract
Binding affinity data [Bioorg Med Chem (2004) 12:613-623] of thiazole and thiadiazole derivatives (n = 30) for the human adenosine A3 receptor subtype have been subjected to 3D-QSAR (Quantitative structure-activity relationships) analyses by molecular shape analysis (MSA) and molecular field analysis (MFA) techniques using Cerius2 Version 4.8. In the case of the MSA, the major steps were (1) generation of conformers and energy minimization; (2) hypothesizing an active conformer (global minimum of the most active compound); (3) selecting a candidate shape-reference compound (based on the active conformation); (4) performing pairwise molecular superimposition using the maximum common subgroup (MCSG) method; (5) measuring molecular shape commonality using MSA descriptors; (6) determining other molecular features by calculating spatial, electronic and conformational parameters; (7) selection of conformers; (8) generation of QSAR equations by genetic function algorithm (GFA) or stepwise regression. The best 3D-QSAR equation (MSA) obtained from GFA technique shows 70.0% predicted variance (leave-one-out) and 77.7% explained variance. This equation shows the importance of Jurs descriptors (atomic charge weighted positive surface area, relative negative charge and relative positive charge surface area), partial moment of inertia, energy of the most stable conformer and the ratio of common overlap steric volume to volume of individual molecules. In the case of stepwise regression, the best relation showed 46.1% predicted variance and 72.3% explained variance. In the case of MFA, the major steps were (1) generating conformers and energy minimization; (2) matching atoms using a maximum common substructure (MCS) search and aligning molecules using the default options; (3) setting MFA preferences (rectangular grid with 2 A step size, charges by the Gasteiger algorithm, H+ and CH3 as probes); (4) creating the field; (5) analysis by the Genetic partial least squares (G/PLS) method. The equation obtained was of excellent statistical quality: 96.1% explained variance and 71.6% predicted variance. Statistically reliable 3D-QSAR models obtained from this study suggest that these techniques could be useful to design potent A3 receptor antagonists.
Collapse
Affiliation(s)
- Prosenjit Bhattacharya
- Drug Theoretics and Cheminformatics Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700 032, India
| | | | | |
Collapse
|
32
|
Almeida T, Rodrigues RJ, de Mendonça A, Ribeiro JA, Cunha RA. Purinergic P2 receptors trigger adenosine release leading to adenosine A2A receptor activation and facilitation of long-term potentiation in rat hippocampal slices. Neuroscience 2004; 122:111-21. [PMID: 14596853 DOI: 10.1016/s0306-4522(03)00523-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Electrophysiological recordings were used to investigate the effects of ATP analogues on theta-burst-induced long-term potentiation (LTP) in rat hippocampal slices. alpha,beta-Methylene ATP (alpha,beta-MeATP; 20 microM) decreased LTP from 36+/-9% to 17+/-5%, an effect prevented by adenosine A(1) receptor blockade in accordance with the localised catabolism of ATP analogues into adenosine, leading to adenosine A(1) receptor activation. Thus, to probe the role of extracellular ATP, all experiments were performed with the A(1) receptor selective antagonist, 1,3-dipropyl-8-cyclopentylxanthine (50 nM). In these conditions, alpha,beta-MeATP or 5'-adenylylimido-diphosphate (beta,gamma-ImATP; 20 microM) facilitated LTP by 120%, an effect prevented by the P2 receptor antagonists, pyridoxalphosphate-6-azophenyl-2'-4'-disulphonic acid (PPADS; 20 microM) or suramin (75 microM), as well as by the P2X(1/3)-selective antagonist 8-(benzamido)naphthalene-1,3,5-trisulfonate (10 microM). The facilitations of LTP by either alpha,beta-MeATP or beta,gamma-ImATP (20 microM) were also prevented by both 4-(2-[7-amino-2-(2-furyl(1,2,4)-triazolo(2,3a)-(1,3,5)triazin-5-yl-amino]ethyl)phenol (50 nM) or 7-2(-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c] pyrimidine (50 nM), antagonists of facilitatory adenosine A(2A) receptors, were occluded by the A(2A) receptor agonist, CGS 21680 (10 nM) and were prevented by the protein kinase C inhibitor, chelerythrine (6 microM) and unaffected by the protein kinase A inhibitor, H89 (1 microM). Furthermore, beta,gamma-ImATP (20 microM) enhanced [(3)H]adenosine outflow from rat hippocampal slices by nearly 150%, an effect prevented by PPADS (20 microM) or suramin (75 microM). The adenosine transport inhibitors, nitrobenzylthioinosine (5 microM) and dipyridamole (10 microM) also prevented beta,gamma-ImATP (20 microM)-induced [(3)H]adenosine outflow and facilitation of LTP. These results suggest that ATP analogues facilitate LTP through P2 receptor activation that mainly triggers adenosine release leading to the activation of adenosine A(2A) receptors.
Collapse
Affiliation(s)
- T Almeida
- Laboratory of Neurosciences, Faculty of Medicine of Lisbon, University of Coimbra, Coimbra, Portugal
| | | | | | | | | |
Collapse
|
33
|
Ribeiro JA, Sebastião AM, de Mendonça A. Adenosine receptors in the nervous system: pathophysiological implications. Prog Neurobiol 2002; 68:377-92. [PMID: 12576292 DOI: 10.1016/s0301-0082(02)00155-7] [Citation(s) in RCA: 374] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adenosine is a ubiquitous homeostatic substance released from most cells, including neurones and glia. Once in the extracellular space, adenosine modifies cell functioning by operating G-protein-coupled receptors (GPCR; A(1), A(2A), A(2B), A(3)) that can inhibit (A(1)) or enhance (A(2)) neuronal communication. Interactions between adenosine receptors and other G-protein-coupled receptors, ionotropic receptors and receptors for neurotrophins also occur, and this might contribute to a fine-tuning of neuronal function. Manipulations of adenosine receptors influence sleep and arousal, cognition and memory, neuronal damage and degeneration, as well as neuronal maturation. These actions might have therapeutic implications for neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, as well as for other neurological situations such as epilepsy, idiopathic pain or even drug addition. Peripheral side effects associated with adenosine receptor agonists limit their usefulness in therapeutics; in contrast, adenosine receptor antagonists appear to have less side effects as it is the case of the well-known non-selective antagonists theophylline (present in tea) or caffeine (abundant in coffee and tea), and their emerging beneficial actions in Parkinson's disease and Alzheimer's disease are encouraging. A(1) receptor antagonism may also be useful to enhance cognition and facilitate arousal, as well as in the periphery when deficits of neurotransmitter release occur (e.g. myasthenic syndromes). Enhancement of extracellular adenosine levels through drugs that influence its metabolism might prove useful approaches in situations such as neuropathic pain, where enhanced activation of inhibitory adenosine A(1) receptors is beneficial. One might then consider adenosine as a fine-tuning modulator of neuronal activity, which via subtle effects causes harmonic actions on neuronal activity. Whenever this homeostasis is disrupted, pathology may be installed and selective receptor antagonism or agonism required.
Collapse
Affiliation(s)
- J A Ribeiro
- Laboratory of Neurosciences, Faculty of Medicine, Institute for Molecular Medicine, University of Lisbon, Lisbon, Portugal.
| | | | | |
Collapse
|