1
|
Heit BS, Chu A, McRay A, Richmond JE, Heckman CJ, Larson J. Interference with glutamate antiporter system x c - enables post-hypoxic long-term potentiation in hippocampus. Exp Physiol 2024; 109:1572-1592. [PMID: 39153228 PMCID: PMC11363115 DOI: 10.1113/ep092045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 08/19/2024]
Abstract
Our group previously showed that genetic or pharmacological inhibition of the cystine/glutamate antiporter, system xc -, mitigates excitotoxicity after anoxia by increasing latency to anoxic depolarization, thus attenuating the ischaemic core. Hypoxia, however, which prevails in the ischaemic penumbra, is a condition where neurotransmission is altered, but excitotoxicity is not triggered. The present study employed mild hypoxia to further probe ischaemia-induced changes in neuronal responsiveness from wild-type and xCT KO (xCT-/-) mice. Synaptic transmission was monitored in hippocampal slices from both genotypes before, during and after a hypoxic episode. Although wild-type and xCT-/- slices showed equal suppression of synaptic transmission during hypoxia, mutant slices exhibited a persistent potentiation upon re-oxygenation, an effect we termed 'post-hypoxic long-term potentiation (LTP)'. Blocking synaptic suppression during hypoxia by antagonizing adenosine A1 receptors did not preclude post-hypoxic LTP. Further examination of the induction and expression mechanisms of this plasticity revealed that post-hypoxic LTP was driven by NMDA receptor activation, as well as increased calcium influx, with no change in paired-pulse facilitation. Hence, the observed phenomenon engaged similar mechanisms as classical LTP. This was a remarkable finding as theta-burst stimulation-induced LTP was equivalent between genotypes. Importantly, post-hypoxic LTP was generated in wild-type slices pretreated with system xc - inhibitor, S-4-carboxyphenylglycine, thereby confirming the antiporter's role in this phenomenon. Collectively, these data indicate that system xc - interference enables neuroplasticity in response to mild hypoxia, and, together with its regulation of cellular damage in the ischaemic core, suggest a role for the antiporter in post-ischaemic recovery of the penumbra.
Collapse
Affiliation(s)
- Bradley S. Heit
- Department of Neuroscience and Department of Biomedical EngineeringNorthwestern UniversityChicagoIllinoisUSA
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alex Chu
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alyssa McRay
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Janet E. Richmond
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Charles J. Heckman
- Department of Neuroscience and Department of Biomedical EngineeringNorthwestern UniversityChicagoIllinoisUSA
| | - John Larson
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
2
|
Zironi I, Aicardi G. Hypoxia Depresses Synaptic Transmission in the Primary Motor Cortex of the Infant Rat-Role of Adenosine A 1 Receptors and Nitric Oxide. Biomedicines 2022; 10:2875. [PMID: 36359395 PMCID: PMC9687150 DOI: 10.3390/biomedicines10112875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 09/08/2024] Open
Abstract
The acute and long-term consequences of perinatal asphyxia have been extensively investigated, but only a few studies have focused on postnatal asphyxia. In particular, electrophysiological changes induced in the motor cortex by postnatal asphyxia have not been examined so far, despite the critical involvement of this cortical area in epilepsy. In this study, we exposed primary motor cortex slices obtained from infant rats in an age window (16-18 day-old) characterized by high incidence of hypoxia-induced seizures associated with epileptiform motor behavior to 10 min of hypoxia. Extracellular field potentials evoked by horizontal pathway stimulation were recorded in layers II/III of the primary motor cortex before, during, and after the hypoxic event. The results show that hypoxia reversibly depressed glutamatergic synaptic transmission and neuronal excitability. Data obtained in the presence of specific blockers suggest that synaptic depression was mediated by adenosine acting on pre-synaptic A1 receptors to decrease glutamate release, and by a nitric oxide (NO)/cGMP postsynaptic pathway. These effects are neuroprotective because they limit energy failure. The present findings may be helpful in the preclinical search for therapeutic strategies aimed at preventing acute and long-term neurological consequences of postnatal asphyxia.
Collapse
Affiliation(s)
- Isabella Zironi
- Department of Physics and Astronomy, University of Bologna, 40127 Bologna, Italy
| | - Giorgio Aicardi
- Department for Life Quality Studies, University of Bologna, 40127 Bologna, Italy
| |
Collapse
|
3
|
The Role of the Adenosine System on Emotional and Cognitive Disturbances Induced by Ethanol Binge Drinking in the Immature Brain and the Beneficial Effects of Caffeine. Pharmaceuticals (Basel) 2022; 15:ph15111323. [DOI: 10.3390/ph15111323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Binge drinking intake is the most common pattern of ethanol consumption by adolescents, which elicits emotional disturbances, mainly anxiety and depressive symptoms, as well as cognitive alterations. Ethanol exposure may act on the adenosine neuromodulation system by increasing adenosine levels, consequently increasing the activation of adenosine receptors in the brain. The adenosine modulation system is involved in the control of mood and memory behavior. However, there is a gap in the knowledge about the exact mechanisms related to ethanol exposure’s hazardous effects on the immature brain (i.e., during adolescence) and the role of the adenosine system thereupon. The present review attempts to provide a comprehensive picture of the role of the adenosinergic system on emotional and cognitive disturbances induced by ethanol during adolescence, exploring the potential benefits of caffeine administration in view of its action as a non-selective antagonist of adenosine receptors.
Collapse
|
4
|
Schubert C, Schulz K, Träger S, Plath AL, Omriouate A, Rosenkranz SC, Morellini F, Friese MA, Hirnet D. Neuronal Adenosine A1 Receptor is Critical for Olfactory Function but Unable to Attenuate Olfactory Dysfunction in Neuroinflammation. Front Cell Neurosci 2022; 16:912030. [PMID: 35846561 PMCID: PMC9279574 DOI: 10.3389/fncel.2022.912030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Adenine nucleotides, such as adenosine triphosphate (ATP), adenosine diphosphate (ADP), as well as the nucleoside adenosine are important modulators of neuronal function by engaging P1 and P2 purinergic receptors. In mitral cells, signaling of the G protein-coupled P1 receptor adenosine 1 receptor (A1R) affects the olfactory sensory pathway by regulating high voltage-activated calcium channels and two-pore domain potassium (K2P) channels. The inflammation of the central nervous system (CNS) impairs the olfactory function and gives rise to large amounts of extracellular ATP and adenosine, which act as pro-inflammatory and anti-inflammatory mediators, respectively. However, it is unclear whether neuronal A1R in the olfactory bulb modulates the sensory function and how this is impacted by inflammation. Here, we show that signaling via neuronal A1R is important for the physiological olfactory function, while it cannot counteract inflammation-induced hyperexcitability and olfactory deficit. Using neuron-specific A1R-deficient mice in patch-clamp recordings, we found that adenosine modulates spontaneous dendro-dendritic signaling in mitral and granule cells via A1R. Furthermore, neuronal A1R deficiency resulted in olfactory dysfunction in two separate olfactory tests. In mice with experimental autoimmune encephalomyelitis (EAE), we detected immune cell infiltration and microglia activation in the olfactory bulb as well as hyperexcitability of mitral cells and olfactory dysfunction. However, neuron-specific A1R activity was unable to attenuate glutamate excitotoxicity in the primary olfactory bulb neurons in vitro or EAE-induced olfactory dysfunction and disease severity in vivo. Together, we demonstrate that A1R modulates the dendro-dendritic inhibition (DDI) at the site of mitral and granule cells and impacts the processing of the olfactory sensory information, while A1R activity was unable to counteract inflammation-induced hyperexcitability.
Collapse
Affiliation(s)
- Charlotte Schubert
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Schulz
- Division of Neurophysiology, Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Simone Träger
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna-Lena Plath
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Asina Omriouate
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C. Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabio Morellini
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Manuel A. Friese,
| | - Daniela Hirnet
- Division of Neurophysiology, Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
- Daniela Hirnet,
| |
Collapse
|
5
|
Spanoghe J, Larsen LE, Craey E, Manzella S, Van Dycke A, Boon P, Raedt R. The Signaling Pathways Involved in the Anticonvulsive Effects of the Adenosine A 1 Receptor. Int J Mol Sci 2020; 22:ijms22010320. [PMID: 33396826 PMCID: PMC7794785 DOI: 10.3390/ijms22010320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 12/20/2022] Open
Abstract
Adenosine acts as an endogenous anticonvulsant and seizure terminator in the brain. Many of its anticonvulsive effects are mediated through the activation of the adenosine A1 receptor, a G protein-coupled receptor with a wide array of targets. Activating A1 receptors is an effective approach to suppress seizures. This review gives an overview of the neuronal targets of the adenosine A1 receptor focusing in particular on signaling pathways resulting in neuronal inhibition. These include direct interactions of G protein subunits, the adenyl cyclase pathway and the phospholipase C pathway, which all mediate neuronal hyperpolarization and suppression of synaptic transmission. Additionally, the contribution of the guanyl cyclase and mitogen-activated protein kinase cascades to the seizure-suppressing effects of A1 receptor activation are discussed. This review ends with the cautionary note that chronic activation of the A1 receptor might have detrimental effects, which will need to be avoided when pursuing A1 receptor-based epilepsy therapies.
Collapse
Affiliation(s)
- Jeroen Spanoghe
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Lars E. Larsen
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Erine Craey
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Simona Manzella
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Annelies Van Dycke
- Department of Neurology, General Hospital Sint-Jan Bruges, 8000 Bruges, Belgium;
| | - Paul Boon
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Robrecht Raedt
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
- Correspondence:
| |
Collapse
|
6
|
Gomes JI, Farinha-Ferreira M, Rei N, Gonçalves-Ribeiro J, Ribeiro JA, Sebastião AM, Vaz SH. Of adenosine and the blues: The adenosinergic system in the pathophysiology and treatment of major depressive disorder. Pharmacol Res 2020; 163:105363. [PMID: 33285234 DOI: 10.1016/j.phrs.2020.105363] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022]
Abstract
Major depressive disorder (MDD) is the foremost cause of global disability, being responsible for enormous personal, societal, and economical costs. Importantly, existing pharmacological treatments for MDD are partially or totally ineffective in a large segment of patients. As such, the search for novel antidepressant drug targets, anchored on a clear understanding of the etiological and pathophysiological mechanisms underpinning MDD, becomes of the utmost importance. The adenosinergic system, a highly conserved neuromodulatory system, appears as a promising novel target, given both its regulatory actions over many MDD-affected systems and processes. With this goal in mind, we herein review the evidence concerning the role of adenosine as a potential player in pathophysiology and treatment of MDD, combining data from both human and animal studies. Altogether, evidence supports the assertions that the adenosinergic system is altered in both MDD patients and animal models, and that drugs targeting this system have considerable potential as putative antidepressants. Furthermore, evidence also suggests that modifications in adenosine signaling may have a key role in the effects of several pharmacological and non-pharmacological antidepressant treatments with demonstrated efficacy, such as electroconvulsive shock, sleep deprivation, and deep brain stimulation. Lastly, it becomes clear from the available literature that there is yet much to study regarding the role of the adenosinergic system in the pathophysiology and treatment of MDD, and we suggest several avenues of research that are likely to prove fruitful.
Collapse
Affiliation(s)
- Joana I Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Farinha-Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
7
|
Lutte AH, Majolo JH, Da Silva RS. Inhibition of ecto-5'-nucleotidase and adenosine deaminase is able to reverse long-term behavioural effects of early ethanol exposure in zebrafish (Danio rerio). Sci Rep 2020; 10:17809. [PMID: 33082435 PMCID: PMC7576130 DOI: 10.1038/s41598-020-74832-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
The behavioural impacts of prenatal exposure to ethanol include a lower IQ, learning problems, anxiety and conduct disorders. Several components of the neurochemical network could contribute to the long-lasting effects of ethanol embryonic exposure. Adenosine is an important neuromodulator, that has been indicated to be affected by acute and chronic exposure to ethanol. Here, embryos of zebrafish exposed to 1% ethanol during the developmental stages of gastrula/segmentation or pharyngula exhibited anxiolytic effect, increased aggressiveness, and decreased social interaction. The exposure during pharyngula stage was able to affect all behavioural parameters analysed at 3 months-post fertilization (mpf), while the treatment during gastrula stage affected the anxiety and social interaction parameters. The aggressiveness was the only behavioural effect of early ethanol exposure that lasted to 12 mpf. The use of a specific inhibitor of adenosine production, the inhibitor of ecto-5′-nucleotidase (AMPCP/150 mg/kg), and the specific inhibitor of adenosine degradation, the inhibitor of adenosine deaminase, EHNA (100 mg/kg) did not affect the effects over anxiety. However, AMPCP at 3 mpf, but not EHNA, reversed aggressive parameters. AMPCP also recovered the social interaction parameter at 3 mpf in animals treated in both stages, while EHNA recovered this parameter just in those animals treated with ethanol during the gastrula stage. These results suggest that long-lasting behavioural effects of ethanol can be modulated by intervention on ecto-5′-nucleotidase and adenosine deaminase activities.
Collapse
Affiliation(s)
- Aline Haab Lutte
- Laboratório de Neuroquímica E Psicofarmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Avenida Ipiranga, 6681, Porto Alegre, RS, 90619-900, Brazil
| | - Julia Huppes Majolo
- Laboratório de Neuroquímica E Psicofarmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Avenida Ipiranga, 6681, Porto Alegre, RS, 90619-900, Brazil
| | - Rosane Souza Da Silva
- Laboratório de Neuroquímica E Psicofarmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Avenida Ipiranga, 6681, Porto Alegre, RS, 90619-900, Brazil.
| |
Collapse
|
8
|
Martins RS, Rombo DM, Gonçalves-Ribeiro J, Meneses C, Borges-Martins VPP, Ribeiro JA, Vaz SH, Kubrusly RCC, Sebastião AM. Caffeine has a dual influence on NMDA receptor-mediated glutamatergic transmission at the hippocampus. Purinergic Signal 2020; 16:503-518. [PMID: 33025424 DOI: 10.1007/s11302-020-09724-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
Caffeine, a stimulant largely consumed around the world, is a non-selective adenosine receptor antagonist, and therefore caffeine actions at synapses usually, but not always, mirror those of adenosine. Importantly, different adenosine receptors with opposing regulatory actions co-exist at synapses. Through both inhibitory and excitatory high-affinity receptors (A1R and A2R, respectively), adenosine affects NMDA receptor (NMDAR) function at the hippocampus, but surprisingly, there is a lack of knowledge on the effects of caffeine upon this ionotropic glutamatergic receptor deeply involved in both positive (plasticity) and negative (excitotoxicity) synaptic actions. We thus aimed to elucidate the effects of caffeine upon NMDAR-mediated excitatory post-synaptic currents (NMDAR-EPSCs), and its implications upon neuronal Ca2+ homeostasis. We found that caffeine (30-200 μM) facilitates NMDAR-EPSCs on pyramidal CA1 neurons from Balbc/ByJ male mice, an action mimicked, as well as occluded, by 1,3-dipropyl-cyclopentylxantine (DPCPX, 50 nM), thus likely mediated by blockade of inhibitory A1Rs. This action of caffeine cannot be attributed to a pre-synaptic facilitation of transmission because caffeine even increased paired-pulse facilitation of NMDA-EPSCs, indicative of an inhibition of neurotransmitter release. Adenosine A2ARs are involved in this likely pre-synaptic action since the effect of caffeine was mimicked by the A2AR antagonist, SCH58261 (50 nM). Furthermore, caffeine increased the frequency of Ca2+ transients in neuronal cell culture, an action mimicked by the A1R antagonist, DPCPX, and prevented by NMDAR blockade with AP5 (50 μM). Altogether, these results show for the first time an influence of caffeine on NMDA receptor activity at the hippocampus, with impact in neuronal Ca2+ homeostasis.
Collapse
Affiliation(s)
- Robertta S Martins
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Pós-Graduação em Neurociências, Universidade Federal Fluminense, Niterói, Brazil
| | - Diogo M Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Carlos Meneses
- Área Departamental de Engenharia de Electrónica e Telecomunicações e de Computadores, Instituto Superior de Engenharia de Lisboa, Lisbon, Portugal
| | - Vladimir P P Borges-Martins
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Pós-Graduação em Neurociências, Universidade Federal Fluminense, Niterói, Brazil
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Regina C C Kubrusly
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Pós-Graduação em Neurociências, Universidade Federal Fluminense, Niterói, Brazil
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal. .,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
9
|
Pliássova A, Henriques M, Silva HB, Agostinho P, Cunha RA, Ferreira SG. Control of NMDA Receptor-Mediated Currents by Adenosine A1 and A2A Receptors Within the Basolateral Amygdala. J Caffeine Adenosine Res 2020. [DOI: 10.1089/caff.2019.0024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Anna Pliássova
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Margarida Henriques
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Henrique B. Silva
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Paula Agostinho
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Rodrigo A. Cunha
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Samira G. Ferreira
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
10
|
Tescarollo FC, Rombo DM, DeLiberto LK, Fedele DE, Alharfoush E, Tomé ÂR, Cunha RA, Sebastião AM, Boison D. Role of Adenosine in Epilepsy and Seizures. J Caffeine Adenosine Res 2020; 10:45-60. [PMID: 32566903 PMCID: PMC7301316 DOI: 10.1089/caff.2019.0022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adenosine is an endogenous anticonvulsant and neuroprotectant of the brain. Seizure activity produces large quantities of adenosine, and it is this seizure-induced adenosine surge that normally stops a seizure. However, within the context of epilepsy, adenosine plays a wide spectrum of different roles. It not only controls seizures (ictogenesis), but also plays a major role in processes that turn a normal brain into an epileptic brain (epileptogenesis). It is involved in the control of abnormal synaptic plasticity and neurodegeneration and plays a major role in the expression of comorbid symptoms and complications of epilepsy, such as sudden unexpected death in epilepsy (SUDEP). Given the important role of adenosine in epilepsy, therapeutic strategies are in development with the goal to utilize adenosine augmentation not only for the suppression of seizures but also for disease modification and epilepsy prevention, as well as strategies to block adenosine A2A receptor overfunction associated with neurodegeneration. This review provides a comprehensive overview of the role of adenosine in epilepsy.
Collapse
Affiliation(s)
- Fabio C. Tescarollo
- Deptartment of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Diogo M. Rombo
- Faculty of Medicine, Institute of Pharmacology and Neurosciences, Lisbon, Portugal
- Institute of Molecular Medicine, University of Lisbon, Lisbon, Portugal
| | - Lindsay K. DeLiberto
- Deptartment of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Denise E. Fedele
- Deptartment of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Enmar Alharfoush
- Deptartment of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Ângelo R. Tomé
- Faculty of Science and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Rodrigo A. Cunha
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana M. Sebastião
- Faculty of Medicine, Institute of Pharmacology and Neurosciences, Lisbon, Portugal
- Institute of Molecular Medicine, University of Lisbon, Lisbon, Portugal
| | - Detlev Boison
- Deptartment of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
- Department of Neurosurgery, New Jersey Medical School, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
11
|
Reklow RJ, Alvares TS, Zhang Y, Miranda Tapia AP, Biancardi V, Katzell AK, Frangos SM, Hansen MA, Toohey AW, Cass CE, Young JD, Pagliardini S, Boison D, Funk GD. The Purinome and the preBötzinger Complex - A Ménage of Unexplored Mechanisms That May Modulate/Shape the Hypoxic Ventilatory Response. Front Cell Neurosci 2019; 13:365. [PMID: 31496935 PMCID: PMC6712068 DOI: 10.3389/fncel.2019.00365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
Exploration of purinergic signaling in brainstem homeostatic control processes is challenging the traditional view that the biphasic hypoxic ventilatory response, which comprises a rapid initial increase in breathing followed by a slower secondary depression, reflects the interaction between peripheral chemoreceptor-mediated excitation and central inhibition. While controversial, accumulating evidence supports that in addition to peripheral excitation, interactions between central excitatory and inhibitory purinergic mechanisms shape this key homeostatic reflex. The objective of this review is to present our working model of how purinergic signaling modulates the glutamatergic inspiratory synapse in the preBötzinger Complex (key site of inspiratory rhythm generation) to shape the hypoxic ventilatory response. It is based on the perspective that has emerged from decades of analysis of glutamatergic synapses in the hippocampus, where the actions of extracellular ATP are determined by a complex signaling system, the purinome. The purinome involves not only the actions of ATP and adenosine at P2 and P1 receptors, respectively, but diverse families of enzymes and transporters that collectively determine the rate of ATP degradation, adenosine accumulation and adenosine clearance. We summarize current knowledge of the roles played by these different purinergic elements in the hypoxic ventilatory response, often drawing on examples from other brain regions, and look ahead to many unanswered questions and remaining challenges.
Collapse
Affiliation(s)
- Robert J. Reklow
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tucaaue S. Alvares
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yong Zhang
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ana P. Miranda Tapia
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Vivian Biancardi
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexis K. Katzell
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Sara M. Frangos
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Megan A. Hansen
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexander W. Toohey
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Carol E. Cass
- Professor Emerita, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - James D. Young
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Silvia Pagliardini
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School and New Jersey Medical School, Rutgers University, New Brunswick, NJ, United States
| | - Gregory D. Funk
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
12
|
Kawamura M, Ruskin DN, Masino SA. Adenosine A 1 receptor-mediated protection of mouse hippocampal synaptic transmission against oxygen and/or glucose deprivation: a comparative study. J Neurophysiol 2019; 122:721-728. [PMID: 31242045 DOI: 10.1152/jn.00813.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adenosine receptors are widely expressed in the brain, and adenosine is a key bioactive substance for neuroprotection. In this article, we clarify systematically the role of adenosine A1 receptors during a range of timescales and conditions when a significant amount of adenosine is released. Using acute hippocampal slices obtained from mice that were wild type or null mutant for the adenosine A1 receptor, we quantified and characterized the impact of varying durations of experimental ischemia, hypoxia, and hypoglycemia on synaptic transmission in the CA1 subregion. In normal tissue, these three stressors rapidly and markedly reduced synaptic transmission, and only treatment of sufficient duration led to incomplete recovery. In contrast, inactivation of adenosine A1 receptors delayed and/or lessened the reduction in synaptic transmission during all three stressors and reduced the magnitude of the recovery significantly. We reproduced the responses to hypoxia and hypoglycemia by applying an adenosine A1 receptor antagonist, validating the clear effects of genetic receptor inactivation on synaptic transmission. We found activation of adenosine A1 receptor inhibited hippocampal synaptic transmission during the acute phase of ischemia, hypoxia, or hypoglycemia and caused the recovery from synaptic impairment after these three stressors using genetic mutant. These studies quantify the neuroprotective role of the adenosine A1 receptor during a variety of metabolic stresses within the same recording system.NEW & NOTEWORTHY Deprivation of oxygen and/or glucose causes a rapid adenosine A1 receptor-mediated decrease in synaptic transmission in mouse hippocampus. We quantified adenosine A1 receptor-mediated inhibition during and synaptic recovery after ischemia, hypoxia, and hypoglycemia of varying durations using a genetic mutant and confirmed these findings using pharmacology. Overall, using the same recording conditions, we found the acute response and the neuroprotective ability of the adenosine A1 receptor depended on the type and duration of deprivation event.
Collapse
Affiliation(s)
- Masahito Kawamura
- Department of Pharmacology, Jikei University School of Medicine, Tokyo, Japan
| | - David N Ruskin
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, Connecticut
| | - Susan A Masino
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, Connecticut
| |
Collapse
|
13
|
Lanigan SM, O'Connor JJ. Prolyl hydroxylase domain inhibitors: can multiple mechanisms be an opportunity for ischemic stroke? Neuropharmacology 2018; 148:117-130. [PMID: 30578795 DOI: 10.1016/j.neuropharm.2018.12.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022]
Abstract
Stroke and cerebrovascular disease are now the fifth most common cause of death behind other diseases such as heart, cancer and respiratory disease and accounts for approximately 40-50 fatalities per 100,000 people each year in the United States. Currently the only therapy for acute stroke, is intravenous administration of tissue plasminogen activator which was approved in 1996 by the FDA. Surprisingly no new treatments have come on the market since, although endovascular mechanical thrombectomy is showing promising results in trials. Recently focus has shifted towards a preventative therapy rather than trying to reverse or limit the amount of damage occurring following stroke onset. During one of the components of ischemia, hypoxia, a number of physiological changes occur within neurons which include the stabilization of hypoxia-inducible factors. The activity of these proteins is regulated by O2, Fe2+, 2-OG and ascorbate-dependant hydroxylases which contain prolyl-4-hydroxylase domains (PHDs). PHD inhibitors are capable of pharmacologically activating the body's own endogenous adaptive response to low levels of oxygen and have therefore become an attractive therapeutic target for treating ischemia. They have been widely used in the periphery and have been shown to have a preconditioning and protective effect against a later and more severe ischemic insult. Currently there are a number of these agents in phase 1, 2 and 3 clinical trials for the treatment of anemia. In this review we assess the neuroprotective effects of PHD inhibitors, including dimethyloxalylglycine and deferoxamine and suggest that not all of their effects in the CNS are HIF-dependent. Unravelling new roles and a better understanding of the function of PHD inhibitors in the CNS may be of great benefit especially when investigating their use in the treatment of stroke and other ischemic diseases.
Collapse
Affiliation(s)
- Sinead M Lanigan
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - John J O'Connor
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
14
|
Minic Z, O'Leary DS, Goshgarian HG, Scislo TJ. Colocalization of A 2a but not A 1 adenosine receptors with GABA-ergic neurons in cardiopulmonary chemoreflex network in the caudal nucleus of the solitary tract. Physiol Rep 2018; 6:e13913. [PMID: 30467998 PMCID: PMC6250926 DOI: 10.14814/phy2.13913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/24/2018] [Accepted: 10/12/2018] [Indexed: 12/03/2022] Open
Abstract
Adenosine operating in the nucleus of the solitary tract (NTS) may inhibit or facilitate neurotransmitter release from nerve terminals and directly inhibit or facilitate central neurons via A1 and A2a pre- and postsynaptic receptors, respectively. However, adenosine A2a receptors, may also activate GABA-ergic neurons/terminals which in turn inhibit glutamatergic transmission in the NTS network. Our previous studies showed that adenosine operating via both A1 (inhibitor) and A2a (activator) receptors powerfully inhibits the cardiopulmonary chemoreflex (CCR) at the level of the caudal NTS. A1 receptors most likely inhibit glutamate release in the CCR network, whereas A2a receptors facilitate NTS GABA-ergic mechanisms which in turn inhibit CCR glutamatergic transmission. Therefore, we hypothesized that A2a receptors are located on NTS GABA-ergic neurons/terminals whereas A1 receptors may be located on NTS glutamatergic neurons/terminals. We investigated this hypothesis using double immunofluorescent staining for A2a or A1 adenosine receptors and GABA synthesizing enzyme, GAD67, in 30 μm thick, floating, medullary rat sections. We found that A2a adenosine receptors are localized within the GABA-ergic cells in the caudal NTS, whereas A1 adenosine receptors are absent from these neurons. Instead, A1 receptors were located on non-GABA-ergic (likely glutamatergic) neurons/terminals in the caudal NTS. These data support our functional findings and the hypothesis that adenosine A2a, but not A1 receptors are located on GABA-ergic neurons.
Collapse
Affiliation(s)
- Zeljka Minic
- Department of PhysiologyWayne State University School of MedicineDetroitMichigan
- Department of Anatomy and Cell BiologyWayne State University School of MedicineDetroitMichigan
- Department of Emergency MedicineWayne State University School of MedicineDetroitMichigan
- Cardiovascular Research InstituteWayne State University School of MedicineDetroitMichigan
| | - Donal S. O'Leary
- Department of PhysiologyWayne State University School of MedicineDetroitMichigan
- Cardiovascular Research InstituteWayne State University School of MedicineDetroitMichigan
| | - Harry G. Goshgarian
- Department of Anatomy and Cell BiologyWayne State University School of MedicineDetroitMichigan
| | - Tadeusz J. Scislo
- Department of PhysiologyWayne State University School of MedicineDetroitMichigan
- Cardiovascular Research InstituteWayne State University School of MedicineDetroitMichigan
| |
Collapse
|
15
|
Mouro FM, Rombo DM, Dias RB, Ribeiro JA, Sebastião AM. Adenosine A 2A receptors facilitate synaptic NMDA currents in CA1 pyramidal neurons. Br J Pharmacol 2018; 175:4386-4397. [PMID: 30220081 PMCID: PMC6240125 DOI: 10.1111/bph.14497] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/18/2018] [Accepted: 08/21/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE NMDA receptors play a key role in both synaptic plasticity and neurodegeneration. Adenosine is an endogenous neuromodulator and through membrane receptors of the A2A subtype can influence both synaptic plasticity and neuronal death. The present work was designed to evaluate the influence of adenosine A2A receptors upon NMDA receptor activity in CA1 hippocampal neurons. We discriminated between modulation of synaptic versus extrasynaptic receptors, since extrasynaptic NMDA receptors are mostly associated with neurodegeneration while synaptic NMDA receptors are linked to plasticity phenomena. EXPERIMENTAL APPROACH Whole-cell patch-clamp recordings were obtained to evaluate NMDA receptor actions on CA1 pyramidal neurons of young adult (5-10 weeks) male Wistar rat hippocampus. KEY RESULTS Activation of A2A receptors with CGS 21680 (30 nM) consistently facilitated chemically-evoked NMDA receptor-currents (NMDA-PSCs) and afferent-evoked NMDA-currents (NMDA-EPSCs), an action prevented by an A2A receptor antagonist (SCH58261, 100 nM) and a PKA inhibitor, H-89 (1 μM). These actions did not reflect facilitation in glutamate release since there was no change in NMDA-EPSCs paired pulse ratio. A2A receptor actions were lost in the presence of an open-channel NMDA receptor blocker, MK-801 (10 μM), but persisted in the presence of memantine, at a concentration (10 μM) known to preferentially block extrasynaptic NMDA receptors. CONCLUSION AND IMPLICATIONS These results show that A2A receptors exert a positive postsynaptic modulatory effect over synaptic, but not extrasynaptic, NMDA receptors in CA1 neurons and, therefore, under non-pathological conditions may contribute to shift the dual role of NMDA receptors towards enhancement of synaptic plasticity.
Collapse
Affiliation(s)
- Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Diogo M Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Raquel B Dias
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
16
|
Nehlig A. Interindividual Differences in Caffeine Metabolism and Factors Driving Caffeine Consumption. Pharmacol Rev 2018; 70:384-411. [PMID: 29514871 DOI: 10.1124/pr.117.014407] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Most individuals adjust their caffeine intake according to the objective and subjective effects induced by the methylxanthine. However, to reach the desired effects, the quantity of caffeine consumed varies largely among individuals. It has been known for decades that the metabolism, clearance, and pharmacokinetics of caffeine is affected by many factors such as age, sex and hormones, liver disease, obesity, smoking, and diet. Caffeine also interacts with many medications. All these factors will be reviewed in the present document and discussed in light of the most recent data concerning the genetic variability affecting caffeine levels and effects at the pharmacokinetic and pharmacodynamic levels that both critically drive the level of caffeine consumption. The pharmacokinetics of caffeine are highly variable among individuals due to a polymorphism at the level of the CYP1A2 isoform of cytochrome P450, which metabolizes 95% of the caffeine ingested. Moreover there is a polymorphism at the level of another critical enzyme, N-acetyltransferase 2. At the pharmacodynamic level, there are several polymorphisms at the main brain target of caffeine, the adenosine A2A receptor or ADORA2. Genetic studies, including genome-wide association studies, identified several loci critically involved in caffeine consumption and its consequences on sleep, anxiety, and potentially in neurodegenerative and psychiatric diseases. We start reaching a better picture on how a multiplicity of biologic mechanisms seems to drive the levels of caffeine consumption, although much more knowledge is still required to understand caffeine consumption and effects on body functions.
Collapse
Affiliation(s)
- Astrid Nehlig
- INSERM U 1129, Pediatric Neurology, Necker-Enfants Malades Hospital, University of Paris Descartes, Inserm U1129, Paris, France
| |
Collapse
|
17
|
Kerkhofs A, Xavier AC, da Silva BS, Canas PM, Idema S, Baayen JC, Ferreira SG, Cunha RA, Mansvelder HD. Caffeine Controls Glutamatergic Synaptic Transmission and Pyramidal Neuron Excitability in Human Neocortex. Front Pharmacol 2018; 8:899. [PMID: 29354052 PMCID: PMC5758559 DOI: 10.3389/fphar.2017.00899] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 11/27/2017] [Indexed: 12/13/2022] Open
Abstract
Caffeine is the most widely used psychoactive drug, bolstering attention and normalizing mood and cognition, all functions involving cerebral cortical circuits. Whereas studies in rodents showed that caffeine acts through the antagonism of inhibitory A1 adenosine receptors (A1R), neither the role of A1R nor the impact of caffeine on human cortical neurons is known. We here provide the first characterization of the impact of realistic concentrations of caffeine experienced by moderate coffee drinkers (50 μM) on excitability of pyramidal neurons and excitatory synaptic transmission in the human temporal cortex. Moderate concentrations of caffeine disinhibited several of the inhibitory A1R-mediated effects of adenosine, similar to previous observations in the rodent brain. Thus, caffeine restored the adenosine-induced decrease of both intrinsic membrane excitability and excitatory synaptic transmission in the human pyramidal neurons through antagonism of post-synaptic A1R. Indeed, the A1R-mediated effects of endogenous adenosine were more efficient to inhibit synaptic transmission than neuronal excitability. This was associated with a distinct affinity of caffeine for synaptic versus extra-synaptic human cortical A1R, probably resulting from a different molecular organization of A1R in human cortical synapses. These findings constitute the first neurophysiological description of the impact of caffeine on pyramidal neuron excitability and excitatory synaptic transmission in the human temporal cortex, providing adequate ground for the effects of caffeine on cognition in humans.
Collapse
Affiliation(s)
- Amber Kerkhofs
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana C Xavier
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Beatriz S da Silva
- Portuguese National Institute of Legal Medicine and Forensic Sciences, Coimbra, Portugal
| | - Paula M Canas
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Sander Idema
- Department of Neurosurgery, Neuroscience Amsterdam, VU University Medical Center Amsterdam, Amsterdam, Netherlands
| | - Johannes C Baayen
- Department of Neurosurgery, Neuroscience Amsterdam, VU University Medical Center Amsterdam, Amsterdam, Netherlands
| | - Samira G Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Rodrigo A Cunha
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
18
|
Purinergic system in psychiatric diseases. Mol Psychiatry 2018; 23:94-106. [PMID: 28948971 DOI: 10.1038/mp.2017.188] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/15/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
Psychiatric disorders are debilitating diseases, affecting >80 million people worldwide. There are no causal cures for psychiatric disorders and available therapies only treat the symptoms. The etiology of psychiatric disorders is unknown, although it has been speculated to be a combination of environmental, stress and genetic factors. One of the neurotransmitter systems implicated in the biology of psychiatric disorders is the purinergic system. In this review, we performed a comprehensive search of the literature about the role and function of the purinergic system in the development and predisposition to psychiatric disorders, with a focus on depression, schizophrenia, bipolar disorder, autism, anxiety and attention deficit/hyperactivity disorder. We also describe how therapeutics used for psychiatric disorders act on the purinergic system.
Collapse
|
19
|
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: 134] [Impact Index Per Article: 16.8] [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
|
20
|
Rombo DM, Ribeiro JA, Sebastião AM. Hippocampal GABAergic transmission: a new target for adenosine control of excitability. J Neurochem 2016; 139:1056-1070. [PMID: 27778347 DOI: 10.1111/jnc.13872] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 01/01/2023]
Abstract
Physiological network functioning in the hippocampus is dependent on a balance between glutamatergic cell excitability and the activity of diverse local circuit neurons that release the inhibitory neurotransmitter γ-aminobutyric acid (GABA). Tuners of neuronal communication such as adenosine, an endogenous modulator of synapses, control hippocampal network operations by regulating excitability. Evidence has been recently accumulating on the influence of adenosine on different aspects of GABAergic transmission to shape hippocampal function. This review addresses how adenosine, through its high-affinity A1 (A1 R) and A2A receptors (A2A R), interferes with different GABA-mediated forms of inhibition in the hippocampus to regulate neuronal excitability. Adenosine-mediated modulation of phasic/tonic inhibitory transmission, of GABA transport mechanisms and its interference with other modulatory systems are discussed together with the putative implications for neuronal function in physiological and pathological conditions. This article is part of a mini review series: 'Synaptic Function and Dysfunction in Brain Diseases'.
Collapse
Affiliation(s)
- Diogo M Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
21
|
Alhaider IA, Alkadhi KA. Caffeine treatment prevents rapid eye movement sleep deprivation-induced impairment of late-phase long-term potentiation in the dentate gyrus. Eur J Neurosci 2016; 42:2843-50. [PMID: 26449851 DOI: 10.1111/ejn.13092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
Abstract
The CA1 and dentate gyrus (DG) are physically and functionally closely related areas of the hippocampus, but they differ in various respects, including their reactions to different insults. The purpose of this study was to determine the protective effects of chronic caffeine treatment on late-phase long-term potentiation (L-LTP) and its signalling cascade in the DG area of the hippocampus of rapid eye movement sleep-deprived rats. Rats were chronically treated with caffeine (300 mg/L drinking water) for 4 weeks, after which they were sleep-deprived for 24 h. L-LTP was induced in in anaesthetized rats, and extracellular field potentials from the DG area were recorded in vivo. The levels of L-LTP-related signalling proteins were assessed by western blot analysis. Sleep deprivation markedly reduced L-LTP magnitude, and basal levels of total cAMP response element-binding protein (CREB), phosphorylated CREB (P-CREB), and calcium/calmodulin kinase IV (CaMKIV). Chronic caffeine treatment prevented the reductions in the basal levels of P-CREB, total CREB and CaMKIV in sleep-deprived rats. Furthermore, caffeine prevented post-L-LTP sleep deprivation-induced downregulation of P-CREB and brain-derived neurotrophic factor in the DG. The current findings show that caffeine treatment prevents acute sleep deprivation-induced deficits in brain function.
Collapse
Affiliation(s)
- Ibrahim A Alhaider
- Department of Pharmacological and Pharmaceutical Sciences, 521E Science and Research 2 Bldg, College of Pharmacy, University of Houston, Houston, TX 77204-5037, USA
| | - Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, 521E Science and Research 2 Bldg, College of Pharmacy, University of Houston, Houston, TX 77204-5037, USA
| |
Collapse
|
22
|
Cunha RA. How does adenosine control neuronal dysfunction and neurodegeneration? J Neurochem 2016; 139:1019-1055. [PMID: 27365148 DOI: 10.1111/jnc.13724] [Citation(s) in RCA: 335] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/23/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022]
Abstract
The adenosine modulation system mostly operates through inhibitory A1 (A1 R) and facilitatory A2A receptors (A2A R) in the brain. The activity-dependent release of adenosine acts as a brake of excitatory transmission through A1 R, which are enriched in glutamatergic terminals. Adenosine sharpens salience of information encoding in neuronal circuits: high-frequency stimulation triggers ATP release in the 'activated' synapse, which is locally converted by ecto-nucleotidases into adenosine to selectively activate A2A R; A2A R switch off A1 R and CB1 receptors, bolster glutamate release and NMDA receptors to assist increasing synaptic plasticity in the 'activated' synapse; the parallel engagement of the astrocytic syncytium releases adenosine further inhibiting neighboring synapses, thus sharpening the encoded plastic change. Brain insults trigger a large outflow of adenosine and ATP, as a danger signal. A1 R are a hurdle for damage initiation, but they desensitize upon prolonged activation. However, if the insult is near-threshold and/or of short-duration, A1 R trigger preconditioning, which may limit the spread of damage. Brain insults also up-regulate A2A R, probably to bolster adaptive changes, but this heightens brain damage since A2A R blockade affords neuroprotection in models of epilepsy, depression, Alzheimer's, or Parkinson's disease. This initially involves a control of synaptotoxicity by neuronal A2A R, whereas astrocytic and microglia A2A R might control the spread of damage. The A2A R signaling mechanisms are largely unknown since A2A R are pleiotropic, coupling to different G proteins and non-canonical pathways to control the viability of glutamatergic synapses, neuroinflammation, mitochondria function, and cytoskeleton dynamics. Thus, simultaneously bolstering A1 R preconditioning and preventing excessive A2A R function might afford maximal neuroprotection. The main physiological role of the adenosine modulation system is to sharp the salience of information encoding through a combined action of adenosine A2A receptors (A2A R) in the synapse undergoing an alteration of synaptic efficiency with an increased inhibitory action of A1 R in all surrounding synapses. Brain insults trigger an up-regulation of A2A R in an attempt to bolster adaptive plasticity together with adenosine release and A1 R desensitization; this favors synaptotocity (increased A2A R) and decreases the hurdle to undergo degeneration (decreased A1 R). Maximal neuroprotection is expected to result from a combined A2A R blockade and increased A1 R activation. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
23
|
Alkadhi KA, Alhaider IA. Caffeine and REM sleep deprivation: Effect on basal levels of signaling molecules in area CA1. Mol Cell Neurosci 2016; 71:125-31. [DOI: 10.1016/j.mcn.2015.12.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 12/16/2015] [Accepted: 12/31/2015] [Indexed: 01/19/2023] Open
|
24
|
The Effects of Hypoxia and Inflammation on Synaptic Signaling in the CNS. Brain Sci 2016; 6:brainsci6010006. [PMID: 26901230 PMCID: PMC4810176 DOI: 10.3390/brainsci6010006] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/21/2016] [Accepted: 02/02/2016] [Indexed: 12/16/2022] Open
Abstract
Normal brain function is highly dependent on oxygen and nutrient supply and when the demand for oxygen exceeds its supply, hypoxia is induced. Acute episodes of hypoxia may cause a depression in synaptic activity in many brain regions, whilst prolonged exposure to hypoxia leads to neuronal cell loss and death. Acute inadequate oxygen supply may cause anaerobic metabolism and increased respiration in an attempt to increase oxygen intake whilst chronic hypoxia may give rise to angiogenesis and erythropoiesis in order to promote oxygen delivery to peripheral tissues. The effects of hypoxia on neuronal tissue are exacerbated by the release of many inflammatory agents from glia and neuronal cells. Cytokines, such as TNF-α, and IL-1β are known to be released during the early stages of hypoxia, causing either local or systemic inflammation, which can result in cell death. Another growing body of evidence suggests that inflammation can result in neuroprotection, such as preconditioning to cerebral ischemia, causing ischemic tolerance. In the following review we discuss the effects of acute and chronic hypoxia and the release of pro-inflammatory cytokines on synaptic transmission and plasticity in the central nervous system. Specifically we discuss the effects of the pro-inflammatory agent TNF-α during a hypoxic event.
Collapse
|
25
|
Purine nucleosides in neuroregeneration and neuroprotection. Neuropharmacology 2015; 104:226-42. [PMID: 26577017 DOI: 10.1016/j.neuropharm.2015.11.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 12/20/2022]
Abstract
In the present review, we stress the importance of the purine nucleosides, adenosine and guanosine, in protecting the nervous system, both centrally and peripherally, via activation of their receptors and intracellular signalling mechanisms. A most novel part of the review focus on the mechanisms of neuronal regeneration that are targeted by nucleosides, including a recently identified action of adenosine on axonal growth and microtubule dynamics. Discussion on the role of the purine nucleosides transversally with the most established neurotrophic factors, e.g. brain derived neurotrophic factor (BDNF), glial derived neurotrophic factor (GDNF), is also focused considering the intimate relationship between some adenosine receptors, as is the case of the A2A receptors, and receptors for neurotrophins. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
|
26
|
Köles L, Kató E, Hanuska A, Zádori ZS, Al-Khrasani M, Zelles T, Rubini P, Illes P. Modulation of excitatory neurotransmission by neuronal/glial signalling molecules: interplay between purinergic and glutamatergic systems. Purinergic Signal 2015; 12:1-24. [PMID: 26542977 DOI: 10.1007/s11302-015-9480-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/26/2015] [Indexed: 12/29/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter of the central nervous system (CNS), released both from neurons and glial cells. Acting via ionotropic (NMDA, AMPA, kainate) and metabotropic glutamate receptors, it is critically involved in essential regulatory functions. Disturbances of glutamatergic neurotransmission can be detected in cognitive and neurodegenerative disorders. This paper summarizes the present knowledge on the modulation of glutamate-mediated responses in the CNS. Emphasis will be put on NMDA receptor channels, which are essential executive and integrative elements of the glutamatergic system. This receptor is crucial for proper functioning of neuronal circuits; its hypofunction or overactivation can result in neuronal disturbances and neurotoxicity. Somewhat surprisingly, NMDA receptors are not widely targeted by pharmacotherapy in clinics; their robust activation or inhibition seems to be desirable only in exceptional cases. However, their fine-tuning might provide a promising manipulation to optimize the activity of the glutamatergic system and to restore proper CNS function. This orchestration utilizes several neuromodulators. Besides the classical ones such as dopamine, novel candidates emerged in the last two decades. The purinergic system is a promising possibility to optimize the activity of the glutamatergic system. It exerts not only direct and indirect influences on NMDA receptors but, by modulating glutamatergic transmission, also plays an important role in glia-neuron communication. These purinergic functions will be illustrated mostly by depicting the modulatory role of the purinergic system on glutamatergic transmission in the prefrontal cortex, a CNS area important for attention, memory and learning.
Collapse
Affiliation(s)
- László Köles
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary.
| | - Erzsébet Kató
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Adrienn Hanuska
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Zoltán S Zádori
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Patrizia Rubini
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany
| | - Peter Illes
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany.
| |
Collapse
|
27
|
Krügel U. Purinergic receptors in psychiatric disorders. Neuropharmacology 2015; 104:212-25. [PMID: 26518371 DOI: 10.1016/j.neuropharm.2015.10.032] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/23/2015] [Accepted: 10/23/2015] [Indexed: 02/07/2023]
Abstract
Psychiatric disorders describe different mental or behavioral patterns, causing suffering or poor coping of ordinary life with manifold presentations. Multifactorial processes can contribute to their development and progression. Purinergic neurotransmission and neuromodulation in the brain have attracted increasing therapeutic interest in the field of psychiatry. Purine nucleotides and nucleosides are well recognized as signaling molecules mediating cell to cell communication. The actions of ATP are mediated by ionotropic P2X and metabotropic P2Y receptor subfamilies, whilst the actions of adenosine are mediated by P1 (A1 or A2) adenosine receptors. Purinergic mechanisms and specific receptor subtypes have been shown to be linked to the regulation of many aspects of behavior and mood and to dysregulation in pathological processes of brain function. In this review the recent knowledge on the role of purinergic receptors in the two most frequent psychiatric diseases, major depression and schizophrenia, as well as on related animal models is summarized. At present the most promising data for therapeutic strategies derive from investigations of the adenosine system emphasizing a unique function of A2A receptors at neurons and astrocytes in these disorders. Among the P2 receptor family, in particular P2X7 and P2Y1 receptors were related to disturbances in major depression and schizophrenia, respectively. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Ute Krügel
- Rudolf Boehm Institute of Pharmacology and Toxicology, Universität Leipzig, Härtelstrasse 16-18, 04107 Leipzig, Germany.
| |
Collapse
|
28
|
Di Angelantonio S, Bertollini C, Piccinin S, Rosito M, Trettel F, Pagani F, Limatola C, Ragozzino D. Basal adenosine modulates the functional properties of AMPA receptors in mouse hippocampal neurons through the activation of A1R A2AR and A3R. Front Cell Neurosci 2015; 9:409. [PMID: 26528137 PMCID: PMC4601258 DOI: 10.3389/fncel.2015.00409] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/25/2015] [Indexed: 11/30/2022] Open
Abstract
Adenosine is a widespread neuromodulator within the CNS and its extracellular level is increased during hypoxia or intense synaptic activity, modulating pre- and postsynaptic sites. We studied the neuromodulatory action of adenosine on glutamatergic currents in the hippocampus, showing that activation of multiple adenosine receptors (ARs) by basal adenosine impacts postsynaptic site. Specifically, the stimulation of both A1R and A3R reduces AMPA currents, while A2AR has an opposite potentiating effect. The effect of ARs stimulation on glutamatergic currents in hippocampal cultures was investigated using pharmacological and genetic approaches. A3R inhibition by MRS1523 increased GluR1-Ser845 phosphorylation and potentiated AMPA current amplitude, increasing the apparent affinity for the agonist. A similar effect was observed blocking A1R with DPCPX or by genetic deletion of either A3R or A1R. Conversely, impairment of A2AR reduced AMPA currents, and decreased agonist sensitivity. Consistently, in hippocampal slices, ARs activation by AR agonist NECA modulated glutamatergic current amplitude evoked by AMPA application or afferent fiber stimulation. Opposite effects of AR subtypes stimulation are likely associated to changes in GluR1 phosphorylation and represent a novel mechanism of physiological modulation of glutamatergic transmission by adenosine, likely acting in normal conditions in the brain, depending on the level of extracellular adenosine and the distribution of AR subtypes.
Collapse
Affiliation(s)
- Silvia Di Angelantonio
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy ; Center for Life Nanoscience, Istituto Italiano di Tecnologia Rome, Italy
| | - Cristina Bertollini
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Sonia Piccinin
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Maria Rosito
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Flavia Trettel
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Francesca Pagani
- Center for Life Nanoscience, Istituto Italiano di Tecnologia Rome, Italy
| | - Cristina Limatola
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy ; Neuromed, Istituto di Ricovero e Cura a Carattere Scientifico Pozzilli, Italy
| | - Davide Ragozzino
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy ; Neuromed, Istituto di Ricovero e Cura a Carattere Scientifico Pozzilli, Italy
| |
Collapse
|
29
|
Novel Therapeutic GPCRs for Psychiatric Disorders. Int J Mol Sci 2015; 16:14109-21. [PMID: 26101869 PMCID: PMC4490542 DOI: 10.3390/ijms160614109] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 05/25/2015] [Accepted: 06/09/2015] [Indexed: 02/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the most common targets of the neuropharmacological drugs in the central nervous system (CNS). GPCRs are activated by manifold neurotransmitters, and their activation in turn evokes slow synaptic transmission. They are deeply involved in multiple neurological and psychiatric disorders such as Parkinson’s disease and schizophrenia. In the brain, the striatum is strongly innervated by the ventral tegmental area (VTA) and plays a central role in manifestation of psychiatric disorders. Recently, anatomical and comprehensive transcriptome analysis of the non-odorant GPCR superfamily revealed that the orphan GPCRs GPR88, GPR6, and GPR52, as well as dopamine D1 and D2 receptors and the adenosine A2a receptor, are the most highly enriched in the rodent striatum. Genetically engineered animal models and molecular biological studies have suggested that these striatally enriched GPCRs have a potential to be therapeutic psychiatric receptors. This review summarizes the current understanding of the therapeutic GPCR candidates for psychiatric disorders.
Collapse
|
30
|
Rombo DM, Dias RB, Duarte ST, Ribeiro JA, Lamsa KP, Sebastião AM. Adenosine A1Receptor Suppresses Tonic GABAAReceptor Currents in Hippocampal Pyramidal Cells and in a Defined Subpopulation of Interneurons. Cereb Cortex 2014; 26:1081-95. [DOI: 10.1093/cercor/bhu288] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
31
|
Sebastião AM, Ribeiro JA. Neuromodulation and metamodulation by adenosine: Impact and subtleties upon synaptic plasticity regulation. Brain Res 2014; 1621:102-13. [PMID: 25446444 DOI: 10.1016/j.brainres.2014.11.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/30/2014] [Accepted: 11/05/2014] [Indexed: 01/06/2023]
Abstract
Synaptic plasticity mechanisms, i.e. the sequence of events that underlies persistent changes in synaptic strength as a consequence of transient alteration in neuronal firing, are greatly influenced by the 'chemical atmosphere' of the synapses, that is to say by the presence of molecules at the synaptic cleft able to fine-tune the activity of other molecules more directly related to plasticity. One of those fine tuners is adenosine, known for a long time as an ubiquitous neuromodulator and metamodulator and recognized early as influencing synaptic plasticity. In this review we will refer to the mechanisms that adenosine can use to affect plasticity, emphasizing aspects of the neurobiology of adenosine relevant to its ability to control synaptic functioning. This article is part of a Special Issue entitled Brain and Memory.
Collapse
Affiliation(s)
- Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Unidade de Neurociências, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal.
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Unidade de Neurociências, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
32
|
Prolonged adenosine A1 receptor activation in hypoxia and pial vessel disruption focal cortical ischemia facilitates clathrin-mediated AMPA receptor endocytosis and long-lasting synaptic inhibition in rat hippocampal CA3-CA1 synapses: differential regulation of GluA2 and GluA1 subunits by p38 MAPK and JNK. J Neurosci 2014; 34:9621-43. [PMID: 25031403 DOI: 10.1523/jneurosci.3991-13.2014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Activation of presynaptic adenosine A1 receptors (A1Rs) causes substantial synaptic depression during hypoxia/cerebral ischemia, but postsynaptic actions of A1Rs are less clear. We found that A1Rs and GluA2-containing AMPA receptors (AMPARs) form stable protein complexes from hippocampal brain homogenates and cultured hippocampal neurons from Sprague Dawley rats. In contrast, adenosine A2A receptors (A2ARs) did not coprecipitate or colocalize with GluA2-containing AMPARs. Prolonged stimulation of A1Rs with the agonist N(6)-cyclopentyladenosine (CPA) caused adenosine-induced persistent synaptic depression (APSD) in hippocampal brain slices, and APSD levels were blunted by inhibiting clathrin-mediated endocytosis of GluA2 subunits with the Tat-GluA2-3Y peptide. Using biotinylation and membrane fractionation assays, prolonged CPA incubation showed significant depletion of GluA2/GluA1 surface expression from hippocampal brain slices and cultured neurons. Tat-GluA2-3Y peptide or dynamin inhibitor Dynasore prevented CPA-induced GluA2/GluA1 internalization. Confocal imaging analysis confirmed that functional A1Rs, but not A2ARs, are required for clathrin-mediated AMPAR endocytosis in hippocampal neurons. Pharmacological inhibitors or shRNA knockdown of p38 MAPK and JNK prevented A1R-mediated internalization of GluA2 but not GluA1 subunits. Tat-GluA2-3Y peptide or A1R antagonist 8-cyclopentyl-1,3-dipropylxanthine also prevented hypoxia-mediated GluA2/GluA1 internalization. Finally, in a pial vessel disruption cortical stroke model, a unilateral cortical lesion compared with sham surgery reduced hippocampal GluA2, GluA1, and A1R surface expression and also caused synaptic depression in hippocampal slices that was consistent with AMPAR downregulation and decreased probability of transmitter release. Together, these results indicate a previously unknown mechanism for A1R-induced persistent synaptic depression involving clathrin-mediated GluA2 and GluA1 internalization that leads to hippocampal neurodegeneration after hypoxia/cerebral ischemia.
Collapse
|
33
|
Ferrante A, Martire A, Pepponi R, Varani K, Vincenzi F, Ferraro L, Beggiato S, Tebano MT, Popoli P. Expression, pharmacology and functional activity of adenosine A1 receptors in genetic models of Huntington's disease. Neurobiol Dis 2014; 71:193-204. [PMID: 25132555 DOI: 10.1016/j.nbd.2014.08.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/15/2014] [Accepted: 08/06/2014] [Indexed: 12/20/2022] Open
Abstract
Adenosine A1 receptor (A1R) stimulation exerts beneficial effects in response to various insults to the brain and, although it was found neuroprotective in a lesional model of Huntington's disease (HD), the features of this receptor in genetic models of HD have never been explored. In the present study we characterized the expression, affinity and functional effects of A1Rs in R6/2 mice (the most widely used transgenic model of HD) and in a cellular model of HD. Binding studies revealed that the density of A1Rs was significantly reduced in the cortex and the striatum of R6/2 mice compared to age-matched wild-type (WT), while receptor affinity was unchanged. The selective A1R agonist cyclopentyladenosine (CPA, 300nM) was significantly more effective in reducing synaptic transmission in corticostriatal slices from symptomatic R6/2 than in age-matched WT mice. Such an effect was due to a stronger inhibition of glutamate release from the pre-synaptic terminal. The different functional activities of A1Rs in HD mice were associated also to a different intracellular signaling pathway involved in the synaptic effect of CPA. In fact, while the PKA pathway was involved in both genotypes, p38 MAPK inhibitor SB203580 partially prevented synaptic effects of CPA in R6/2, but not in WT, mice; moreover, CPA differently modulated the phosphorylation status of p38 in the two genotypes. In vitro studies confirmed a different behavior of A1Rs in HD: CPA (100 nM for 5h) modulated cell viability in STHdh(Q111/Q111) (mhttHD cells), without affecting the viability of STHdh(Q7/Q7) (wthtt cells). This effect was prevented by the application of SB203580. Our results demonstrate that in the presence of the HD mutation A1Rs undergo profound changes in terms of expression, pharmacology and functional activity. These changes have to be taken in due account when considering A1Rs as a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Antonella Ferrante
- Istituto Superiore di Sanità, Department of Therapeutic Research and Medicines Evaluation, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Alberto Martire
- Istituto Superiore di Sanità, Department of Therapeutic Research and Medicines Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - Rita Pepponi
- Istituto Superiore di Sanità, Department of Therapeutic Research and Medicines Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - Katia Varani
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Fabrizio Vincenzi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy
| | - Sarah Beggiato
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Maria Teresa Tebano
- Istituto Superiore di Sanità, Department of Therapeutic Research and Medicines Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - Patrizia Popoli
- Istituto Superiore di Sanità, Department of Therapeutic Research and Medicines Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
34
|
Giust D, Da Ros T, Martín M, Albasanz JL. [60]Fullerene derivative modulates adenosine and metabotropic glutamate receptors gene expression: a possible protective effect against hypoxia. J Nanobiotechnology 2014; 12:27. [PMID: 25123848 PMCID: PMC4143579 DOI: 10.1186/s12951-014-0027-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 07/23/2014] [Indexed: 01/26/2023] Open
Abstract
Background Glutamate, the main excitatory neurotransmitter, is involved in learning and memory processes but at higher concentration results excitotoxic causing degeneration and neuronal death. Adenosine is a nucleoside that exhibit neuroprotective effects by modulating of glutamate release. Hypoxic and related oxidative conditions, in which adenosine and metabotropic glutamate receptors are involved, have been demonstrated to contribute to neurodegenerative processes occurring in certain human pathologies. Results Human neuroblastoma cells (SH-SY5Y) were used to evaluate the long time (24, 48 and 72 hours) effects of a [60]fullerene hydrosoluble derivative (t3ss) as potential inhibitor of hypoxic insult. Low oxygen concentration (5% O2) caused cell death, which was avoided by t3ss exposure in a concentration dependent manner. In addition, gene expression analysis by real time PCR of adenosine A1, A2A and A2B and metabotropic glutamate 1 and 5 receptors revealed that t3ss significantly increased A1 and mGlu1 expression in hypoxic conditions. Moreover, t3ss prevented the hypoxia-induced increase in A2A mRNA expression. Conclusions As t3ss causes overexpression of adenosine A1 and metabotropic glutamate receptors which have been shown to be neuroprotective, our results point to a radical scavenger protective effect of t3ss through the enhancement of these neuroprotective receptors expression. Therefore, the utility of these nanoparticles as therapeutic target to avoid degeneration and cell death of neurodegenerative diseases is suggested.
Collapse
|
35
|
Perígolo-Vicente R, Ritt K, Gonçalves-de-Albuquerque CF, Castro-Faria-Neto HC, Paes-de-Carvalho R, Giestal-de-Araujo E. IL-6, A1 and A2aR: a crosstalk that modulates BDNF and induces neuroprotection. Biochem Biophys Res Commun 2014; 449:477-82. [PMID: 24845382 DOI: 10.1016/j.bbrc.2014.05.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/08/2014] [Indexed: 11/15/2022]
Abstract
Several diseases are related to retinal ganglion cell death, such as glaucoma, diabetes and other retinopathies. Many studies have attempted to identify factors that could increase neuroprotection after axotomy of these cells. Interleukin-6 has been shown to be able to increase the survival and regeneration of retinal ganglion cells (RGC) in mixed culture as well as in vivo. In this work we show that the trophic effect of IL-6 is mediated by adenosine receptor (A2aR) activation and also by the presence of extracellular BDNF. We also show that there is a complex cross-talk between IL-6, BDNF, the Adenosine A1 and A2a receptors that results in neuroprotection of retinal ganglion cells.
Collapse
Affiliation(s)
- Rafael Perígolo-Vicente
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil; Blizard Institute - Queen Mary, University of London, 4 Newark St, London, City of London, Greater London E1 2AT, United Kingdom.
| | - Karen Ritt
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Departamento de Fisiologia e Farmacodinâmica, Av. Brasil, n° 4365, Manguinhos, CEP: 21045-900 Rio de Janeiro, RJ, Brazil
| | - Hugo Caire Castro-Faria-Neto
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Departamento de Fisiologia e Farmacodinâmica, Av. Brasil, n° 4365, Manguinhos, CEP: 21045-900 Rio de Janeiro, RJ, Brazil
| | - Roberto Paes-de-Carvalho
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil
| | - Elizabeth Giestal-de-Araujo
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil
| |
Collapse
|
36
|
Rial D, Lara DR, Cunha RA. The Adenosine Neuromodulation System in Schizophrenia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 119:395-449. [DOI: 10.1016/b978-0-12-801022-8.00016-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
37
|
Corcoran A, O'Connor JJ. Hypoxia-inducible factor signalling mechanisms in the central nervous system. Acta Physiol (Oxf) 2013; 208:298-310. [PMID: 23692777 DOI: 10.1111/apha.12117] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/01/2013] [Accepted: 05/16/2013] [Indexed: 12/22/2022]
Abstract
In the CNS, neurones are highly sensitive to the availability of oxygen. In conditions where oxygen availability is decreased, neuronal function can be altered, leading to injury and cell death. Hypoxia has been implicated in a number of central nervous system pathologies including stroke, head trauma and neurodegenerative diseases. Cellular responses to oxygen deprivation are complex and result in activation of short- and long-term mechanisms to conserve energy and protect cells. Failure of synaptic transmission can be observed within minutes following this hypoxia. The acute effects of hypoxia on synaptic transmission are primarily mediated by altering ion fluxes across membranes, pre-synaptic effects of adenosine and other actions at glutamatergic receptors. A more long-term feature of the response of neurones to hypoxia is the activation of transcription factors such as hypoxia-inducible factor. The activation of hypoxia-inducible factor is governed by a family of dioxygenases called hypoxia-inducible factor prolyl 4 hydroxylases (PHDs). Under hypoxic conditions, PHD activity is inhibited, thereby allowing hypoxia-inducible factor to accumulate and translocate to the nucleus, where it binds to the hypoxia-responsive element sequences of target gene promoters. Inhibition of PHD activity stabilizes hypoxia-inducible factor and other proteins thus acting as a neuroprotective agent. This review will focus on the response of neuronal cells to hypoxia-inducible factor and its targets, including the prolyl hydroxylases. We also present evidence for acute effects of PHD inhibition on synaptic transmission and plasticity in the hippocampus.
Collapse
Affiliation(s)
- A. Corcoran
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute of Biomolecular and Biomedical Research; UniversityCollege Dublin; Dublin; Ireland
| | - J. J. O'Connor
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute of Biomolecular and Biomedical Research; UniversityCollege Dublin; Dublin; Ireland
| |
Collapse
|
38
|
Gori MB, Girardi E. 3-Mercaptopropionic acid-induced repetitive seizures increase GluN2A expression in rat hippocampus: a potential neuroprotective role of cyclopentyladenosine. Cell Mol Neurobiol 2013; 33:803-13. [PMID: 23748434 PMCID: PMC11497892 DOI: 10.1007/s10571-013-9947-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 05/28/2013] [Indexed: 11/25/2022]
Abstract
The N-methyl-D-aspartate receptor (NMDAR) is involved in synaptic plasticity, learning, memory, and neurological diseases like epilepsy and it is the major mediator of excitotoxicity. Functional NMDARs in the mature brain are heteromeric complexes composed of different subunits: GluN1 and GluN2. There are four different GluN2 subunits (A-D) and each of them critically determines the pharmacological and electrophysiological properties of NMDARs. GluN1 is ubiquitously expressed in the central nervous system while the highest GluN2A expression is in the hippocampus. Adenosine, an endogenous anticonvulsant, is a neuromodulator with a critical role in the regulation of neuronal activity, mediating its effect on specific receptors, among which adenosine A1 receptor is highly expressed in the hippocampus. In the present work hippocampal GluN2A expression after the convulsant drug 3-mercaptopropionic acid (MP) induced seizures and the effect of cyclopentyladenosine (CPA) given alone or prior to MP (CPA + MP) in an acute or repetitive experimental model was studied. CPA administered to rats for one or 4 days increases seizure threshold induced by MP. After one administration of MP, no significant difference in GluN2A expression was observed in CPA and CPA + MP by Western blot, although immunohistochemistry revealed an increase in CA2/3 area. However, repetitive MP administration during 4 days showed a significant increase of GluN2A expression, and the repetitive administration of CPA 30 min prior to MP caused a significant decrease of GluN2A expression with respect to MP treatment, returning to control levels. These results show that GluN2A subunit is involved in repetitive MP-induced seizures, while CPA administration displays a protective effect against it.
Collapse
Affiliation(s)
- María Belén Gori
- Laboratorio de Epilepsia Experimental y Excitoxicidad, Instituto de Biología Celular y Neurociencia “Prof Eduardo De Robertis”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, C1121 ABG Buenos Aires, Argentina
| | - Elena Girardi
- Laboratorio de Epilepsia Experimental y Excitoxicidad, Instituto de Biología Celular y Neurociencia “Prof Eduardo De Robertis”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, C1121 ABG Buenos Aires, Argentina
| |
Collapse
|
39
|
Abstract
Protection against neuronal damage is a major objective of current research in areas such as stroke medicine, Alzheimer's disease and other neurodegenerative conditions. Adenosine receptors are important modulators of cell survival, and thus agents targeting these receptors could be valuable therapeutic agents. Agonists at A(1) receptors and antagonists at A(2A) receptors are known to protect acutely against neuronal damage caused by toxins or ischemia-reperfusion, and these compounds can also protect against the cell damage inflicted by reactive oxygen species. Even endogenous adenosine may be neuroprotective, since its levels rise substantially in association with a period of ischemia-reperfusion. Unfortunately, there is growing evidence that the efficacy of adenosine receptor activation can be reduced by the concomitant activation of glutamate receptors responding to N-methyl-D-aspartate (NMDA), probably acting via the release of nitric oxide. Such problems will need to be resolved before adenosine receptor agonists can proceed far as neuroprotective agents. The use of receptor antagonists may prove a more valuable approach.
Collapse
Affiliation(s)
- Trevor W Stone
- Institute of Biomedical & Life Sciences, West Medical Building, University of Glasgow, Glasgow G12 8QQ, Scotland, UK.
| |
Collapse
|
40
|
Alkadhi K, Zagaar M, Alhaider I, Salim S, Aleisa A. Neurobiological consequences of sleep deprivation. Curr Neuropharmacol 2013; 11:231-249. [PMID: 24179461 PMCID: PMC3648777 DOI: 10.2174/1570159x11311030001] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 01/15/2013] [Accepted: 02/02/2013] [Indexed: 01/30/2023] Open
Abstract
Although the physiological function of sleep is not completely understood, it is well documented that it contributes significantly to the process of learning and memory. Ample evidence suggests that adequate sleep is essential for fostering connections among neuronal networks for memory consolidation in the hippocampus. Sleep deprivation studies are extremely valuable in understanding why we sleep and what are the consequences of sleep loss. Experimental sleep deprivation in animals allows us to gain insight into the mechanism of sleep at levels not possible to study in human subjects. Many useful approaches have been utilized to evaluate the effect of sleep loss on cognitive function, each with relative advantages and disadvantages. In this review we discuss sleep and the detrimental effects of sleep deprivation mostly in experimental animals. The negative effects of sleep deprivation on various aspects of brain function including learning and memory, synaptic plasticity and the state of cognition-related signaling molecules are discussed.
Collapse
Affiliation(s)
- Karim Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Texas, USA
| | - Munder Zagaar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Texas, USA
| | - Ibrahim Alhaider
- College of Clinical Pharmacy, King Faisal University, Al-Hofuf, Kingdom of Saudi Arabia
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Texas, USA
| | - Abdulaziz Aleisa
- Department of Pharmacology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
41
|
Dias RB, Rombo DM, Ribeiro JA, Henley JM, Sebastião AM. Adenosine: setting the stage for plasticity. Trends Neurosci 2013; 36:248-57. [PMID: 23332692 DOI: 10.1016/j.tins.2012.12.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 10/09/2012] [Accepted: 12/14/2012] [Indexed: 12/19/2022]
Abstract
It is widely accepted that Hebbian forms of plasticity mediate selective modifications in synaptic strength underlying information encoding in response to experience and circuit formation or refinement throughout development. Several complementary forms of homeostatic plasticity coordinate to keep Hebbian plasticity in check, frequently through the actions of conserved regulatory molecules. Recent evidence suggests that this may be the case for adenosine, which is ubiquitous in the brain and is released by both neurons and glial cells via constitutive and activity-dependent mechanisms. Through A1 and A2A receptor activation, adenosine modulates neuronal homeostasis and tunes the ability of synapses to undergo and/or sustain plasticity. Here, we review how adenosine equilibrates neuronal activity and sets the stage for synaptic plasticity.
Collapse
Affiliation(s)
- Raquel B Dias
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | | | | | | | | |
Collapse
|
42
|
IL-6 treatment increases the survival of retinal ganglion cells in vitro: The role of adenosine A1 receptor. Biochem Biophys Res Commun 2013; 430:512-8. [DOI: 10.1016/j.bbrc.2012.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 12/03/2012] [Indexed: 11/23/2022]
|
43
|
Lucena GMRS, Matheus FC, Ferreira VM, Tessele PB, Azevedo MS, Cechinel-Filho V, Prediger RD. Effects of Ethanolic Extract and Naphthoquinones Obtained from the Bulbs ofCipura paludosaon Short-Term and Long-Term Memory: Involvement of Adenosine A1and A2AReceptors. Basic Clin Pharmacol Toxicol 2012; 112:229-35. [DOI: 10.1111/bcpt.12022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 09/26/2012] [Indexed: 11/27/2022]
Affiliation(s)
| | - Filipe C. Matheus
- Departamento de Farmacologia, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina (UFSC); Florianópolis; SC; Brazil
| | - Vania M. Ferreira
- Faculdade de Ciências da Saúde; Universidade de Brasília (UnB); Brasília; DF; Brazil
| | - Priscila B. Tessele
- Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Curso de Farmácia-CCS, Universidade do Vale do Itajaí (UNIVALI); Itajaí; SC; Brazil
| | - Mariangela S. Azevedo
- Departamento de Química, Laboratórios de Fitoquímica; Produtos Naturais e Meio Ambiente, Universidade Federal de Rondônia (Labfito-UNIR); Porto Velho; RO; Brazil
| | - Valdir Cechinel-Filho
- Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Curso de Farmácia-CCS, Universidade do Vale do Itajaí (UNIVALI); Itajaí; SC; Brazil
| | - Rui D. Prediger
- Departamento de Farmacologia, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina (UFSC); Florianópolis; SC; Brazil
| |
Collapse
|
44
|
Savio LEB, Vuaden FC, Rosemberg DB, Bogo MR, Bonan CD, Wyse ATS. Long-term proline exposure alters nucleotide catabolism and ectonucleotidase gene expression in zebrafish brain. Metab Brain Dis 2012; 27:541-9. [PMID: 22669495 DOI: 10.1007/s11011-012-9321-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 05/22/2012] [Indexed: 12/20/2022]
Abstract
Hyperprolinemia is an inherited disorder of proline metabolism and hyperprolinemic patients can present neurological manifestations, such as seizures cognitive dysfunctions, and psychotic disorders. However, the underlying mechanisms of these symptoms are still unclear. Since adenine nucleotides play crucial roles in neurotransmission and neuromodulation, we evaluated the in vivo and in vitro effects of proline on ectonucleotidase activities and gene expression in zebrafish brain. For the in vivo studies, animals were exposed at two proline concentrations (1.5 and 3.0 mM) during 1 h or 7 days (short- or long-term treatments, respectively). For the in vitro assays, different proline concentrations (ranging from 3.0 to 1000 μM) were tested. Short-term proline exposure did not promote significant changes on the ectonucleotidase activities and gene expression. Long-term proline exposure significantly increased ATP catabolism in both concentrations tested (14 % and 22 %, respectively), whereas ADP and AMP hydrolysis were increased only at 3.0 mM proline (21 % and 17 %, respectively) when compared to control. Moreover, the relative gene expression of enpd3 increased in both treated groups after long-term proline, whereas enptd1 increased only at 3.0 mM proline. Proline in vitro did not promote significant changes on ectonucleotidase activities. Altogether, these data indicate that the enzymes responsible for the control of extracellular nucleotides levels might be altered after proline exposure in zebrafish, contributing to better understand the pathophysiology of this disease. Moreover, such findings might facilitate the use of the zebrafish as a complementary vertebrate model for studying inborn errors of amino acid metabolism.
Collapse
Affiliation(s)
- Luiz Eduardo Baggio Savio
- Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, 90035-003, Porto Alegre, RS, Brazil
| | | | | | | | | | | |
Collapse
|
45
|
Diógenes MJ, Neves-Tomé R, Fucile S, Martinello K, Scianni M, Theofilas P, Lopatár J, Ribeiro JA, Maggi L, Frenguelli BG, Limatola C, Boison D, Sebastião AM. Homeostatic control of synaptic activity by endogenous adenosine is mediated by adenosine kinase. Cereb Cortex 2012; 24:67-80. [PMID: 22997174 DOI: 10.1093/cercor/bhs284] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Extracellular adenosine, a key regulator of neuronal excitability, is metabolized by astrocyte-based enzyme adenosine kinase (ADK). We hypothesized that ADK might be an upstream regulator of adenosine-based homeostatic brain functions by simultaneously affecting several downstream pathways. We therefore studied the relationship between ADK expression, levels of extracellular adenosine, synaptic transmission, intrinsic excitability, and brain-derived neurotrophic factor (BDNF)-dependent synaptic actions in transgenic mice underexpressing or overexpressing ADK. We demonstrate that ADK: 1) Critically influences the basal tone of adenosine, evaluated by microelectrode adenosine biosensors, and its release following stimulation; 2) determines the degree of tonic adenosine-dependent synaptic inhibition, which correlates with differential plasticity at hippocampal synapses with low release probability; 3) modulates the age-dependent effects of BDNF on hippocampal synaptic transmission, an action dependent upon co-activation of adenosine A2A receptors; and 4) influences GABAA receptor-mediated currents in CA3 pyramidal neurons. We conclude that ADK provides important upstream regulation of adenosine-based homeostatic function of the brain and that this mechanism is necessary and permissive to synaptic actions of adenosine acting on multiple pathways. These mechanistic studies support previous therapeutic studies and implicate ADK as a promising therapeutic target for upstream control of multiple neuronal signaling pathways crucial for a variety of neurological disorders.
Collapse
|
46
|
Genetically controlled upregulation of adenosine A(1) receptor expression enhances the survival of primary cortical neurons. Mol Neurobiol 2012; 46:535-44. [PMID: 22899189 DOI: 10.1007/s12035-012-8321-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 08/02/2012] [Indexed: 12/20/2022]
Abstract
Adenosine has a key endogenous neuroprotective role in the brain, predominantly mediated by the adenosine A(1) receptor (A(1)R). This has been mainly explored using pharmacological tools and/or receptor knockout mice strains. It has long been suggested that the neuroprotective effects of A(1)R are increased following receptor upregulation, thus attenuating neuronal damage in pathological conditions. We have previously shown that the neuroprotective and neuromodulatory actions of the cytokines IL-6 and oncostatin M are mediated by induction of neuronal A(1)R expression. In order to investigate the direct effects of A(1)R upregulation in neurons, we have generated a tetracycline-regulated expression system with a bidirectional promoter, directing the simultaneous expression of the mouse A(1)R and GFP/mCherry reporter genes. In a first step, we tested the efficacy of the system in transiently transfected human embryonic kidney 293 cells. In addition, we confirmed the functional integrity of the expressed A(1)R by whole-cell patch clamp recordings. We demonstrated that A(1)R-transfected primary neurons show enhanced survival against N-methyl-D-aspartate-induced excitotoxicity. Pretreatment with an A(1)R-selective agonist additionally strongly decreased neuronal cell death, while an A(1)R antagonist completely abolished the neuroprotective effects of A(1)R upregulation. The presented data provide for the first time direct evidence that the upregulation of A(1)R enhances neuronal survival.
Collapse
|
47
|
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: 318] [Impact Index Per Article: 22.7] [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
|
48
|
Butler TR, Prendergast MA. Neuroadaptations in adenosine receptor signaling following long-term ethanol exposure and withdrawal. Alcohol Clin Exp Res 2011; 36:4-13. [PMID: 21762181 DOI: 10.1111/j.1530-0277.2011.01586.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ethanol affects the function of neurotransmitter systems, resulting in neuroadaptations that alter neural excitability. Adenosine is one such receptor system that is changed by ethanol exposure. The current review is focused on the A(1) and the A(2A) receptor subtypes in the context of ethanol-related neuroadaptations and ethanol withdrawal because these subtypes (i) are activated by basal levels of adenosine, (ii) have been most well-studied for their role in neuroprotection and ethanol-related phenomena, and (iii) are the primary site of action for caffeine in the brain, a substance commonly ingested with ethanol. It is clear that alterations in adenosinergic signaling mediate many of the effects of acute ethanol administration, particularly with regard to motor function and sedation. Further, prolonged ethanol exposure has been shown to produce adaptations in the cell surface expression or function of both A(1) and the A(2A) receptor subtypes, effects that likely promote neuronal excitability during ethanol withdrawal. As a whole, these findings demonstrate a significant role for ethanol-induced adaptations in adenosine receptor signaling that likely influence neuronal function, viability, and relapse to ethanol intake following abstinence.
Collapse
Affiliation(s)
- Tracy R Butler
- Department of Psychology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, USA.
| | | |
Collapse
|
49
|
Alhaider IA, Aleisa AM, Tran TT, Alkadhi KA. Sleep deprivation prevents stimulation-induced increases of levels of P-CREB and BDNF: protection by caffeine. Mol Cell Neurosci 2011; 46:742-51. [PMID: 21338685 DOI: 10.1016/j.mcn.2011.02.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 02/02/2011] [Accepted: 02/11/2011] [Indexed: 01/12/2023] Open
Abstract
It is well known that caffeine and sleep deprivation have opposing effects on learning and memory; therefore, this study was undertaken to determine the effects of chronic (4wks) caffeine treatment (0.3g/l in drinking water) on long-term memory deficit associated with 24h sleep deprivation. Animals were sleep deprived using the modified multiple platform method. The results showed that chronic caffeine treatment prevented the impairment of long-term memory as measured by performance in the radial arm water maze task and normalized L-LTP in area CA1 of the hippocampi of sleep-deprived anesthetized rats. Sleep deprivation prevents the high frequency stimulation-induced increases in the levels of phosphorylated-cAMP response element binding protein (P-CREB) and brain-derived neurotrophic factor (BDNF) seen during the expression of late phase long-term potentiation (L-LTP). However, chronic caffeine treatment prevented the effect of sleep-deprivation on the stimulated levels of P-CREB and BDNF. The results suggest that chronic caffeine treatment may protect the sleep-deprived brain probably by preserving the levels of P-CREB and BDNF.
Collapse
Affiliation(s)
- Ibrahim A Alhaider
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | | | | | | |
Collapse
|
50
|
Gomes CV, Kaster MP, Tomé AR, Agostinho PM, Cunha RA. Adenosine receptors and brain diseases: neuroprotection and neurodegeneration. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1808:1380-99. [PMID: 21145878 DOI: 10.1016/j.bbamem.2010.12.001] [Citation(s) in RCA: 303] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 11/30/2010] [Accepted: 12/01/2010] [Indexed: 02/06/2023]
Abstract
Adenosine acts in parallel as a neuromodulator and as a homeostatic modulator in the central nervous system. Its neuromodulatory role relies on a balanced activation of inhibitory A(1) receptors (A1R) and facilitatory A(2A) receptors (A2AR), mostly controlling excitatory glutamatergic synapses: A1R impose a tonic brake on excitatory transmission, whereas A2AR are selectively engaged to promote synaptic plasticity phenomena. This neuromodulatory role of adenosine is strikingly similar to the role of adenosine in the control of brain disorders; thus, A1R mostly act as a hurdle that needs to be overcame to begin neurodegeneration and, accordingly, A1R only effectively control neurodegeneration if activated in the temporal vicinity of brain insults; in contrast, the blockade of A2AR alleviates the long-term burden of brain disorders in different neurodegenerative conditions such as ischemia, epilepsy, Parkinson's or Alzheimer's disease and also seem to afford benefits in some psychiatric conditions. In spite of this qualitative agreement between neuromodulation and neuroprotection by A1R and A2AR, it is still unclear if the role of A1R and A2AR in the control of neuroprotection is mostly due to the control of glutamatergic transmission, or if it is instead due to the different homeostatic roles of these receptors related with the control of metabolism, of neuron-glia communication, of neuroinflammation, of neurogenesis or of the control of action of growth factors. In spite of this current mechanistic uncertainty, it seems evident that targeting adenosine receptors might indeed constitute a novel strategy to control the demise of different neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Catarina V Gomes
- Center for Neurosciences of Coimbra, University of Coimbra, Coimbra, Portugal
| | | | | | | | | |
Collapse
|