Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Neurol Sci ; 36(8): 1441-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25805704

RESUMEN

Evidence indicates that the adenosine A2A receptor subtype is of critical importance in stroke. In previous studies, in the model of permanent middle cerebral artery occlusion (pMCAo), the adenosine A2A receptor antagonist, SCH58261, administered soon after ischemia, proved protective against excessive glutamate outflow in the first 4 h after ischemia and against neurological deficit and tissue damage evaluated 24 h after pMCAo. In the present work, we investigated if neuroprotective effect of SCH58261 was maintained 7 days after transient MCAo (tMCAo). SCH58261 (0.01 mg/kg, i.p.), administered twice/day for 7 days, protected from neurological deficit 1 day after tMCAo, but no more after 5 and 7 days. Two days after tMCAo, SCH58261 did not reduce blood cell infiltration, evaluated as HIS-48 positive cells, into ischemic striatal and cortical tissue. Moreover, 7 days after tMCAo, SCH58261 has not protected ischemic areas from damage and has not ameliorated myelin organization into the ischemic striatum. Protection by the A2A receptor antagonist 24 h after ischemia is attributable to reduced excitotoxicity. Seven days after ischemia the early protective effect of the A2A receptor antagonist likely has been overwhelmed by a secondary damage due to blood cell infiltration and neuroinflammation.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/uso terapéutico , Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Ataque Isquémico Transitorio/complicaciones , Pirimidinas/uso terapéutico , Triazoles/uso terapéutico , Análisis de Varianza , Animales , Células Sanguíneas/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Masculino , Glicoproteína Asociada a Mielina/metabolismo , Examen Neurológico , Ratas , Ratas Wistar , Factores de Tiempo
2.
Mediators Inflamm ; 2014: 805198, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25165414

RESUMEN

The extracellular concentration of adenosine in the brain increases dramatically during ischemia. Adenosine A(2A) receptor is expressed in neurons and glial cells and in inflammatory cells (lymphocytes and granulocytes). Recently, adenosine A(2A) receptor emerged as a potential therapeutic attractive target in ischemia. Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia the early massive increase of extracellular glutamate is followed by activation of resident immune cells, that is, microglia, and production or activation of inflammation mediators. Proinflammatory cytokines, which upregulate cell adhesion molecules, exert an important role in promoting recruitment of leukocytes that in turn promote expansion of the inflammatory response in ischemic tissue. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. A(2A) receptors present on central cells and on blood cells account for important effects depending on the time-related evolution of the pathological condition. Evidence suggests that A(2A) receptor antagonists provide early protection via centrally mediated control of excessive excitotoxicity, while A(2A) receptor agonists provide protracted protection by controlling massive blood cell infiltration in the hours and days after ischemia. Focus on inflammatory responses provides for adenosine A(2A) receptor agonists a wide therapeutic time-window of hours and even days after stroke.


Asunto(s)
Isquemia Encefálica/inmunología , Isquemia Encefálica/metabolismo , Receptor de Adenosina A2A/metabolismo , Animales , Citocinas/metabolismo , Humanos
3.
J Neuroinflammation ; 8: 31, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21486435

RESUMEN

BACKGROUND: Permanent functional deficits following spinal cord injury (SCI) arise both from mechanical injury and from secondary tissue reactions involving inflammation. Enhanced release of adenosine and glutamate soon after SCI represents a component in the sequelae that may be responsible for resulting functional deficits. The role of adenosine A2A receptor in central ischemia/trauma is still to be elucidated. In our previous studies we have demonstrated that the adenosine A2A receptor-selective agonist CGS21680, systemically administered after SCI, protects from tissue damage, locomotor dysfunction and different inflammatory readouts. In this work we studied the effect of the adenosine A2A receptor antagonist SCH58261, systemically administered after SCI, on the same parameters. We investigated the hypothesis that the main action mechanism of agonists and antagonists is at peripheral or central sites. METHODS: Spinal trauma was induced by extradural compression of SC exposed via a four-level T5-T8 laminectomy in mouse. Three drug-dosing protocols were utilized: a short-term systemic administration by intraperitoneal injection, a chronic administration via osmotic minipump, and direct injection into the spinal cord. RESULTS: SCH58261, systemically administered (0.01 mg/kg intraperitoneal. 1, 6 and 10 hours after SCI), reduced demyelination and levels of TNF-α, Fas-L, PAR, Bax expression and activation of JNK mitogen-activated protein kinase (MAPK) 24 hours after SCI. Chronic SCH58261 administration, by mini-osmotic pump delivery for 10 days, improved the neurological deficit up to 10 days after SCI. Adenosine A2A receptors are physiologically expressed in the spinal cord by astrocytes, microglia and oligodendrocytes. Soon after SCI (24 hours), these receptors showed enhanced expression in neurons. Both the A2A agonist and antagonist, administered intraperitoneally, reduced expression of the A2A receptor, ruling out the possibility that the neuroprotective effects of the A2A agonist are due to A2A receptor desensitization. When the A2A antagonist and agonist were centrally injected into injured SC, only SCH58261 appeared neuroprotective, while CGS21680 was ineffective. CONCLUSIONS: Our results indicate that the A2A antagonist protects against SCI by acting on centrally located A2A receptors. It is likely that blockade of A2A receptors reduces excitotoxicity. In contrast, neuroprotection afforded by the A2A agonist may be primarily due to peripheral effects.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Antagonistas del Receptor de Adenosina A2/farmacología , Adenosina/análogos & derivados , Fenetilaminas/farmacología , Pirimidinas/farmacología , Traumatismos de la Médula Espinal/prevención & control , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiopatología , Triazoles/farmacología , Adenosina/farmacología , Animales , Inflamación/metabolismo , Inflamación/patología , Bombas de Infusión Implantables , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Médula Espinal/patología , Traumatismos de la Médula Espinal/patología
4.
Brain ; 132(Pt 6): 1480-95, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19359287

RESUMEN

Adenosine is a potent biological mediator, the concentration of which increases dramatically following brain ischaemia. During ischaemia, adenosine is in a concentration range (muM) that stimulates all four adenosine receptor subtypes (A(1), A(2A), A(2B) and A(3)). In recent years, evidence has indicated that the A(2A) receptor subtype is of critical importance in stroke. We have previously shown that 24 h after medial cerebral artery occlusion (MCAo), A(2A) receptors up-regulate on neurons and microglia of ischaemic striatum and cortex and that subchronically administered adenosine A(2A) receptor antagonists protect against brain damage and neurological deficit and reduce activation of p38 mitogen-activated protein kinase (MAPK) in microglial cells. The mechanisms by which A(2A) receptors are noxious during ischaemia still remain elusive. The objective of the present study was to investigate whether the adenosine A(2A) antagonist SCH58261 affects JNK and MEK1/ERK MAPK activation. A further aim was to investigate cell types expressing activated JNK and MEK1/ERK MAPK after ischaemia. We hereby report that the selective adenosine A(2A) receptor antagonist, SCH58261, administered subchronically (0.01 mg/kg i.p) 5 min, 6 and 20 h after MCAo in male Wistar rats, reduced JNK MAPK activation (immunoblot analysis: phospho-JNK54 isoform by 81% and phospho-JNK46 isoform by 60%) in the ischaemic striatum. Twenty-four hours after MCAo, the Olig2 transcription factor of oligodendroglial progenitor cells and mature oligodendrocytes was highly expressed in cell bodies in the ischaemic striatum. Immunofluorescence staining showed that JNK MAPK is maximally expressed in Olig2-stained oligodendrocytes and in a few NeuN stained neurons. Striatal cell fractioning into nuclear and extra-nuclear fractions demonstrated the presence of Olig2 transcription factor and JNK MAPK in both fractions. The A(2A) antagonist reduced striatal Olig 2 transcription factor (immunoblot analysis: by 55%) and prevented myelin disorganization, assessed by myelin-associated glycoprotein staining. Twenty-four hours after MCAo, ERK1/2 MAPK was highly activated in the ischaemic striatum, mostly in microglia, while it was reduced in the ischaemic cortex. The A(2A) antagonist did not affect activation of the ERK1/2 pathway. The efficacy of A(2A) receptor antagonism in reducing activation of JNK MAPK in oligodendrocytes suggests a mechanism of protection consisting of scarring oligodendrocyte inhibitory molecules that can hinder myelin reconstitution and neuron functionality.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Isquemia Encefálica/prevención & control , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Oligodendroglía/efectos de los fármacos , Pirimidinas/uso terapéutico , Triazoles/uso terapéutico , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Isquemia Encefálica/enzimología , Isquemia Encefálica/patología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/enzimología , Evaluación Preclínica de Medicamentos/métodos , Activación Enzimática/efectos de los fármacos , Masculino , Microglía/metabolismo , Actividad Motora/efectos de los fármacos , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/enzimología , Pirimidinas/farmacología , Ratas , Ratas Wistar , Triazoles/farmacología
5.
Prog Neurobiol ; 83(5): 310-31, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18023959

RESUMEN

This review summarizes recent developments that have contributed to understand how adenosine receptors, particularly A2A receptors, modulate brain injury in various animal models of neurological disorders, including Parkinson's disease (PD), stroke, Huntington's disease (HD), multiple sclerosis, Alzheimer's disease (AD) and HIV-associated dementia. It is clear that extracellular adenosine acting at adenosine receptors influences the functional outcome in a broad spectrum of brain injuries, indicating that A2A Rs may modulate some general cellular processes to affect neuronal cells death. Pharmacological, neurochemical and molecular/genetic approaches to the complex actions of A2A receptors in different cellular elements suggest that A2A receptor activation can be detrimental or protective after brain insults, depending on the nature of brain injury and associated pathological conditions. An interesting concept that emerges from these studies is A2A R's ability to fine tune neuronal and glial functions to produce neuroprotective effects. While the data presented here clearly highlight the complexity of using adenosinergic agents therapeutically in PD and other neurodegenerative disorders and point out many areas for further inquiry, they also confirm that adenosine receptor ligands, particularly A2A receptor ligands, have many promising characteristics that encourage the pursuit of their therapeutic potential.


Asunto(s)
Adenosina/metabolismo , Encefalopatías/metabolismo , Encéfalo/metabolismo , Citoprotección/fisiología , Degeneración Nerviosa/metabolismo , Receptor de Adenosina A2A/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Encefalopatías/tratamiento farmacológico , Encefalopatías/genética , Lesión Encefálica Crónica/genética , Lesión Encefálica Crónica/metabolismo , Lesión Encefálica Crónica/fisiopatología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Citoprotección/efectos de los fármacos , Humanos , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Fármacos Neuroprotectores/farmacología
6.
J Neurochem ; 104(2): 479-90, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17953669

RESUMEN

Ischemia, through modulation of adenosine receptors (ARs), may influence adenosine-mediated-cellular responses. In the present study, we investigated the modulation of rat A(2A) receptor expression and functioning, in rat cerebral cortex and striatum, following in vivo focal ischemia (24 h). In cortex, middle cerebral artery occlusion did not induce any alterations in A(2A) receptor binding and functioning. On the contrary, in striatum, a significant decrease in A(2A) ligand affinity, associated with an increase in receptor density, were detected. In striatum, ischemia also induced a significant reduction both in G protein pool and in A(2A) receptor-G protein coupling. On the contrary, A(2A) receptor functional responsiveness, measured as stimulation of adenylyl cyclise, was not affected by ischemia, suggesting receptor up-regulation may represent a compensatory mechanism to maintain receptor functioning during cerebral damage. Immunohistochemical study showed that following 24 h middle cerebral artery occlusion, A(2A) ARs were definitely expressed both on neurons and activated microglia in ischemic striatum and cortex, but were not detected on astrocytes. In the non-ischemic hemisphere and in sham-operated rats A(2A) ARs were barely detected. Modifications of ARs may play a significant role in determining adenosine effects during ischemia and therefore should be taken into account when evaluating time-dependent protective effects of specific A(2A) active compounds.


Asunto(s)
Isquemia Encefálica/fisiopatología , Regulación de la Expresión Génica/fisiología , Receptores de Adenosina A2/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacocinética , Agonistas del Receptor de Adenosina A2 , Animales , Astrocitos/metabolismo , Conducta Animal , Encéfalo/patología , Encéfalo/fisiopatología , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Masculino , Actividad Motora/fisiología , Examen Neurológico , Fenetilaminas/farmacocinética , Ratas , Ratas Wistar , Receptores de Adenosina A2/genética
7.
Front Pharmacol ; 9: 1231, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30420807

RESUMEN

Cerebral ischemia is a multifactorial pathology characterized by different events evolving in time. The acute injury, characterized by excitoxicity, is followed by a secondary brain injury that develops from hours to days after ischemia. Extracellular levels of histamine increase in the ischemic area after focal cerebral ischemia induced by occlusion of the middle cerebral artery (MCAo). The histamine H4 receptor (H4R) is predominantly expressed in cell types of immune system where is involved in the regulation of immunological and inflammatory responses, and in numerous area of the Central Nervous System (CNS) including cortex and striatum. Our aim was to assess the putative neuroprotective effects of the potent and selective H4R antagonist, JNJ7777120 (JNJ), chronically administered (1 mg/kg, i.p., twice/day for 7 days) on damage parameters in a rat model of focal ischemia induced by transient MCAo (tMCAo). Chronic treatment with the H4R antagonist JNJ, significantly protected from the neurological deficit and from body weight loss after tMCAo. Seven days after the ischemic insult, JNJ reduced the volume of the ischemic cortical and striatal damage, the number of activated microglia and astrocytes in the ischemic cortex and striatum and decreased the plasma levels of IL-1ß and TNF-α, while increased the levels of IL-10. Two days after ischemia, JNJ has reduced granulocyte infiltration in the ischemic area. Results demonstrate that the selective antagonist of H4R, JNJ, systemically and chronically administered after ischemia, reduces the ischemic brain damage, improves the neurological deficit and decreases blood pro-inflammatory cytokines, suggesting that H4R is a valuable pharmacological target after focal brain ischemia.

8.
Exp Gerontol ; 96: 46-62, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28606482

RESUMEN

We investigated the quantitative and morphofunctional alterations of neuron-astrocyte-microglia triads in CA3 hippocampus, in comparison to CA1, after 2 Vessel Occlusion (2VO) and the protective effect of dipyridamole. We evaluated 3 experimental groups: sham-operated rats (sham, n=15), 2VO-operated rats treated with vehicle (2VO-vehicle, n=15), and 2VO-operated rats treated with dipyridamole from day 0 to day 7 (2VO-dipyridamole, n=15), 90days after 2VO. We analyzed Stratum Pyramidalis (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of CA3. 1) ectopic neurons increased in SL and SR of 2VO-vehicle, and 2VO-dipyridamole rats; 2) apoptotic neurons increased in SP of 2VO-vehicle rats and dipyridamole reverted this effect; 3) astrocytes increased in SP, SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 4) TNF-α expression increased in astrocytes, blocked by dipyridamole, and in dendrites in SR of 2VO-vehicle rats; 5) total microglia increased in SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 6) triads increased in SR of 2VO-vehicle rats and dipyridamole reverted this effect. Microglia cooperated with astrocytes to phagocytosis of apoptotic neurons and debris, and engulfed ectopic non-fragmented neurons in SL of 2VO-vehicle and 2VO-dipyridamole rats, through a new mechanism called phagoptosis. CA3 showed a better adaptive capacity than CA1 to the ischemic insult, possibly due to the different behaviour of astrocytes and microglial cells. Dipyridamole had neuroprotective effects.


Asunto(s)
Astrocitos/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Dipiridamol/farmacología , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Vasodilatadores/farmacología , Análisis de Varianza , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/fisiopatología , Región CA3 Hipocampal/citología , Región CA3 Hipocampal/efectos de los fármacos , Arteria Carótida Común/cirugía , Estenosis Carotídea/fisiopatología , Masculino , Microscopía Confocal , Fagocitosis/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
9.
J Cereb Blood Flow Metab ; 26(7): 974-82, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16395292

RESUMEN

Adenosine 5'-triphosphate outflow increases after an ischemic insult in the brain and may induce the expression of P2X7 receptors in resting microglia, determining its modification into an activated state. To assess the effects of P2X7 receptor blockade in preventing microglia activation and ameliorating brain damage and neurological impairment, we delivered the P2 unselective antagonist Reactive Blue 2 to rats after middle cerebral artery occlusion. In sham-operated animals, devoid of brain damage, double immunofluorescence verified the absence of P2X7 immunoreactivity on resting microglia, astrocytes, and neurons, identified, respectively, by OX-42, glial fibrillary acid protein, and neuronal nuclei (NeuN) immunoreactivity. After ischemia, vehicle-treated rats showed monolateral sensorimotor deficit and tissue damage in striatum and frontoparietal cortex. Moreover, P2X7 immunoreactivity was de novo expressed on activated microglia in infarcted and surrounding areas, as well as on a reactive form of microglia, resting in shape but P2X7 immunoreactive, present in ipsi- and contralateral cingulate and medial frontal cortex. Reactive Blue 2 improved sensorimotor deficit and restricted the volume of infarction, without preventing the expression of P2X7, but inducing it in the microglia of contralateral frontal and parietal cortex and striatum, which had lost reciprocal connections with the remote infarct area. De novo expression of P2X7 occurred in both activated and reactive microglia, suggesting their differentiated roles in the area of infarct and in remote regions. Reactive Blue 2 reduced ischemic brain damage, likely blocking the function of activated microglia in the infarct area, but in the remote brain regions promoted the expression of P2X7 on reactive microglia, developing defense and reparative processes.


Asunto(s)
Isquemia Encefálica/prevención & control , Infarto Cerebral/prevención & control , Infarto de la Arteria Cerebral Media/fisiopatología , Microglía/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Isquemia Encefálica/etiología , Isquemia Encefálica/fisiopatología , Infarto Cerebral/etiología , Infarto Cerebral/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Microglía/citología , Antagonistas del Receptor Purinérgico P2 , Ratas , Ratas Wistar , Receptores Purinérgicos P2/biosíntesis , Receptores Purinérgicos P2X7 , Triazinas/farmacología
10.
Brain Res ; 1073-1074: 470-80, 2006 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-16443200

RESUMEN

We investigated the protective effect of subchronic treatment of the A2A receptor antagonist, SCH 58261 (0.01 mg/kg, i.p.), administered 5 min, 6 h and 15 h after permanent right middle cerebral artery occlusion (MCAo). Twenty-four hours after ischemia, an extensive pallid area, evaluated by cresyl violet staining, is evident in the vascular territories supplied by the MCA, the striatum and the sensory motor cortex. The pallid area reflects the extent of necrotic neurons. Soon after waking, rats showed a definite contralateral turning behavior which was significantly reduced by SCH 58261 treatment. Twenty-four hours after MCAo, SCH 58261 significantly improved the neurological deficit and reduced ischemic damage in the striatum and cortex. Phospho-p38 mitogen-activated protein kinase (MAPK), evaluated by Western Blot, increased by 500% in the ischemic striatum 24 h after MCAo. SCH 58261 treatment significantly reduced phospho-p38 MAPK by 70%. Microglia was immunostained using the OX-42 antibody. Phospho-p38 MAPK and OX-42-immunoreactive cells are localized in the ventral striatum and frontoparietal cortex. Furthermore, both OX-42 and phospho-p38 MAPK-immunoreactive cells have overlapping morphological features, typical of reactive microglia. SCH 58261 reduced phospho-p38 MAPK immunoreactivity in the striatum and in the cortex without changing the microglial cell morphology. These results indicate that the protective effect of the adenosine antagonist SCH 58261 during ischemia is not due to reduced microglial activation but involves inhibition of phospho-p38 MAPK and suggest that treatment with the A2A antagonist from the first hour to several hours after ischemia may be a useful therapeutic approach in cerebral ischemia.


Asunto(s)
Lesiones Encefálicas/prevención & control , Enfermedades del Sistema Nervioso/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Pirimidinas/administración & dosificación , Triazoles/administración & dosificación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Análisis de Varianza , Animales , Western Blotting/métodos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Lateralidad Funcional/fisiología , Inmunohistoquímica/métodos , Masculino , Microscopía Confocal/métodos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Enfermedades del Sistema Nervioso/etiología , Examen Neurológico/métodos , Ratas , Ratas Wistar
11.
Neuropharmacology ; 104: 105-30, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26581499

RESUMEN

Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia a primary damage due to the early massive increase of extracellular glutamate is followed by activation of resident immune cells, i.e microglia, and production or activation of inflammation mediators. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. Extracellular concentrations of ATP and adenosine in the brain increase dramatically during ischemia in concentrations able to stimulate their respective specific P2 and P1 receptors. Both ATP P2 and adenosine P1 receptor subtypes exert important roles in ischemia. Although adenosine exerts a clear neuroprotective effect through A1 receptors during ischemia, the use of selective A1 agonists is hampered by undesirable peripheral effects. Evidence up to now in literature indicate that A2A receptor antagonists provide protection centrally by reducing excitotoxicity, while agonists at A2A (and possibly also A2B) and A3 receptors provide protection by controlling massive infiltration and neuroinflammation in the hours and days after brain ischemia. Among P2X receptors most evidence indicate that P2X7 receptor contribute to the damage induced by the ischemic insult due to intracellular Ca(2+) loading in central cells and facilitation of glutamate release. Antagonism of P2X7 receptors might represent a new treatment to attenuate brain damage and to promote proliferation and maturation of brain immature resident cells that can promote tissue repair following cerebral ischemia. Among P2Y receptors, antagonists of P2Y12 receptors are of value because of their antiplatelet activity and possibly because of additional anti-inflammatory effects. Moreover strategies that modify adenosine or ATP concentrations at injury sites might be of value to limit damage after ischemia. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.


Asunto(s)
Adenosina Trifosfato/metabolismo , Adenosina/metabolismo , Isquemia Encefálica/metabolismo , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Encefalitis/metabolismo , Humanos , Antagonistas de Receptores Purinérgicos P1/farmacología , Antagonistas del Receptor Purinérgico P2/farmacología , Transducción de Señal
12.
Neurochem Int ; 47(6): 442-8, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16029911

RESUMEN

Interest is growing in the role of adenosine triphosphate (ATP) on P2 receptors during hypoxic/ischemic events in the brain. However, there is no direct evidence of an increase in extracellular ATP levels during cerebral ischemia in vivo. The aim of the present study was to evaluate ATP outflow from the rat striatum by the microdialysis technique associated with focal cerebral ischemia in vivo by intraluminal occlusion of the right middle cerebral artery (MCA). Between 1 and 4h after ischemia, rats showed a clear turning behavior contralateral to the ischemic side. Twenty-four hour after MCA occlusion, ischemic rats had definite neurological deficit and striatal and cortical damage. The ATP concentration (mean+/-S.E.M.) in the striatum of normoxic rats (n = 8) was 3.10+/-0.34 nM. During 220 min after MCA occlusion, the extracellular ATP levels significantly increased two-fold, being 5.90+/-0.61 nM (p < 0.01 versus normoxic level). ATP outflow showed a tendency to increase over time during the 220 min of ischemia. Since extracellular ATP is rapidly metabolized to adenosine, we also assessed ATP outflow in the presence of the ecto-5'-nucleotidase inhibitor, alpha,beta-methylene-adenosine diphosphate (AOPCP, 1 mM) directly perfused into the striatum. The ATP concentration in normoxic rats (n = 8) was increased three-fold in the presence of the ecto-5'-nucleotidase inhibitor (9.57+/-0.26 nM). During 220 min of ischemia, extracellular ATP levels significantly increased 1.3-fold in AOPCP-treated rats (12.62+/-0.65 nM, p < 0.01 versus normoxic level). The present study confirms that ATP is continuously released in the brain and demonstrates for the first time that ATP outflow increases during ischemia in vivo. These results confirm that ATP may be an important mediator in brain ischemia.


Asunto(s)
Adenosina Trifosfato/metabolismo , Isquemia Encefálica/metabolismo , Cuerpo Estriado/metabolismo , Líquido Extracelular/metabolismo , Regulación hacia Arriba/fisiología , 5'-Nucleotidasa/antagonistas & inhibidores , 5'-Nucleotidasa/metabolismo , Adenosina/metabolismo , Animales , Infarto Encefálico/etiología , Infarto Encefálico/metabolismo , Infarto Encefálico/fisiopatología , Isquemia Encefálica/fisiopatología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Microdiálisis , Trastornos del Movimiento/etiología , Trastornos del Movimiento/metabolismo , Trastornos del Movimiento/fisiopatología , Ratas , Ratas Wistar , Receptores Purinérgicos P2/metabolismo , Regulación hacia Arriba/efectos de los fármacos
13.
Front Cell Neurosci ; 9: 155, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25964740

RESUMEN

Differentiation and maturation of oligodendroglial cells are postnatal processes that involve specific morphological changes correlated with the expression of stage-specific surface antigens and functional voltage-gated ion channels. A small fraction of oligodendrocyte progenitor cells (OPCs) generated during development are maintained in an immature and slowly proliferative or quiescent state in the adult central nervous system (CNS) representing an endogenous reservoir of immature cells. Adenosine receptors are expressed by OPCs and a key role of adenosine in oligodendrocyte maturation has been recently recognized. As evaluated on OPC cultures, adenosine, by stimulating A1 receptors, promotes oligodendrocyte maturation and inhibits their proliferation; on the contrary, by stimulating A2A receptors, it inhibits oligodendrocyte maturation. A1 and A2A receptor-mediated effects are related to opposite modifications of outward delayed rectifying membrane K(+) currents (IK) that are involved in the regulation of oligodendrocyte differentiation. Brain A1 and A2A receptors might represent new molecular targets for drugs useful in demyelinating pathologies, such as multiple sclerosis (MS), stroke and brain trauma.

14.
Brain Res ; 979(1-2): 225-9, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12850590

RESUMEN

In the quinolinic acid (QA)-rat model of Huntington's disease (HD), 15 days after QA injection, striatal glutamate, measured by in vivo microdialysis, was unchanged while a significant decrease in adenosine occurred. The decrease in adenosine may depend on QA-induced striatal cell loss. Probe perfusion of the adenosine A(2A) receptor antagonist SCH 58261 significantly increased striatal glutamate outflow, suggesting a potential detrimental effect of A(2A) antagonism at later stages of the neurodegenerative process induced by QA.


Asunto(s)
Cuerpo Estriado/química , Ácido Glutámico/metabolismo , Enfermedad de Huntington/fisiopatología , Antagonistas de Receptores Purinérgicos P1 , Adenosina/análisis , Animales , Cromatografía Líquida de Alta Presión , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Espacio Extracelular/química , Ácido Glutámico/análisis , Ácido Glutámico/efectos de los fármacos , Enfermedad de Huntington/inducido químicamente , Masculino , Microdiálisis , Modelos Animales , Fármacos Neuroprotectores/farmacología , Pirimidinas/farmacología , Ácido Quinolínico/farmacología , Ratas , Ratas Wistar , Receptor de Adenosina A2A , Factores de Tiempo , Triazoles/farmacología
15.
Brain Res ; 959(2): 243-50, 2003 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-12493612

RESUMEN

Adenosine A(2A) receptor antagonists have been proved protective in different ischemia models. In this study we verified if the protective effect of the selective A(2A) antagonist, SCH 58261, could be attributed to the reduction of the excitatory amino acid outflow induced by cerebral focal ischemia. A vertical microdialysis probe was inserted into the striatum of male Wistar rats and, after 24 h, permanent right intraluminal middle cerebral artery occlusion (MCAo) was induced. Soon after waking, rats showed a definite contralateral turning behavior, which persisted up to 7 h after MCAo. During 4 h after MCAo, glutamate, aspartate, GABA, adenosine and taurine outflow increased. SCH 58261 (0.01 mg/kg, i.p.), administered 5 min after MCAo, suppressed turning behavior and significantly reduced the outflow of glutamate, aspartate, GABA and adenosine. At 24 h after MCAo, the rats showed severe sensorimotor deficit and damage in both the striatum and cortex. SCH 58261 significantly reduced cortical damage but did not protect against the sensorimotor deficit. The protective effect of SCH 58261 against turning behavior and increased outflow of excitatory amino acids in the first hours after MCAo suggests the potential utility of selective adenosine A(2A) antagonists when administered in the first hours after ischemia. Furthermore, this study, for the first time, proposes that turning behavior after permanent intraluminal MCAo, be used as a precocious index of neurological deficit and neuronal damage.


Asunto(s)
Isquemia Encefálica/metabolismo , Cuerpo Estriado/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Neurotransmisores/metabolismo , Antagonistas de Receptores Purinérgicos P1 , Pirimidinas/farmacología , Triazoles/farmacología , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Cuerpo Estriado/metabolismo , Masculino , Pirimidinas/uso terapéutico , Ratas , Ratas Wistar , Receptor de Adenosina A2A , Receptores Purinérgicos P1/metabolismo , Triazoles/uso terapéutico
16.
Eur J Pharmacol ; 464(1): 33-8, 2003 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-12600692

RESUMEN

The objective of the work was to study, by in vivo microdialysis, the effect of the adenosine A(2A) receptor antagonist 7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine (SCH 58261) on glutamate outflow in the striata of unilateral 6-hydroxydopamine-infused rats. Two vertical microdialysis probes were implanted bilaterally in both the denervated striatum and in the intact striatum. Glutamate concentrations in the dialysate were determined by high-performance liquid chromatography (HPLC). Infusion of the adenosine A(2A) receptor antagonist SCH 58261 (50 nM), through the microdialysis fiber, significantly increased glutamate outflow from the denervated striatum while it decreased glutamate outflow from the intact striatum. The opposite effects of SCH 58261 on glutamate outflow in the intact and 6-hydroxydopamine-lesioned striatum might be attributed to blockade of striatal adenosine A(2A) receptors located on either striatal indirect output pathways or glutamatergic terminals. These results may be relevant to our understanding of the mechanism of action of adenosine A(2A) receptor antagonists in Parkinson's disease.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Ácido Glutámico/metabolismo , Antagonistas de Receptores Purinérgicos P1 , Pirimidinas/farmacología , Triazoles/farmacología , Animales , Cuerpo Estriado/metabolismo , Desnervación , Dopamina/fisiología , Masculino , Microdiálisis , Oxidopamina/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A2A , Receptores Purinérgicos P1/fisiología , Factores de Tiempo
17.
Neurosci Lett ; 346(1-2): 93-6, 2003 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12850556

RESUMEN

Adenosine present in human brain glioma extracellular spaces is a marker of astrocyte purine metabolism. In this study, we evaluated adenosine levels in the extracellular fluid of 21 human gliomas of high-grade malignancy using brain microdialysis techniques coupled to high-performance liquid chromatography. The adenosine concentration (mean+/-SEM) within the control tissue was 2.99+/-0.37 microM and in the tumour tissue 1.56+/-0.46 microM. The reduction was statistically significant. It is concluded that the adenosine concentrations reached in the tumour tissue are sufficient to stimulate all adenosine receptor subtypes, suppress local anti-tumour immune responses and affect glial and endothelial cell proliferation.


Asunto(s)
Adenosina/metabolismo , Neoplasias Encefálicas/metabolismo , Espacio Extracelular/metabolismo , Glioma/metabolismo , Microdiálisis/métodos , Adulto , Anciano , Neoplasias Encefálicas/cirugía , Cromatografía Líquida de Alta Presión/métodos , Femenino , Glioma/cirugía , Humanos , Periodo Intraoperatorio/métodos , Masculino , Persona de Mediana Edad
18.
Int Rev Neurobiol ; 119: 309-48, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25175971

RESUMEN

Ischemic stroke is a complex pathology characterized by a sequence of events that evolve over time and space. It is the second leading cause of death and the main cause of adult long-term disability in developed countries. At the moment, there is no promising pharmacotherapy for acute ischemic stroke. Adenosine receptors (A1, A2A, A2B, A3) are important targets for therapeutic implementation in the treatment of stroke because extracellular adenosine concentrations increase dramatically soon after ischemia. Adenosine receptors located both on central nervous system cells and on immune blood cells exert important roles during ischemia. The neuroprotective role of adenosine through A1 receptor subtype during ischemia is accepted, but the use of selective A1 agonists is hampered by undesirable side effects such as sedation, bradycardia, and hypotension. Recently, the A2A receptor subtype emerged as a potential therapeutic attractive target in ischemia. Evidence suggests that A2A receptor has dual role: in a first phase of ischemia, it potentiates excitotoxicity, while hours and days after ischemia, A2A receptors on immune blood cells potentiate cell adhesion mechanisms and infiltration in the ischemic parenchyma. Consistently, the use of A2A receptor agonists/antagonists (administered at doses that do not modify blood pressure and heart rate) should be carefully evaluated in function of time after ischemia. Although much is still to be known about the role of A2B and A3 receptor subtypes in brain ischemia, most consistent information indicates their role in regulation of immunosuppression and inflammation.


Asunto(s)
Isquemia Encefálica/metabolismo , Receptores Purinérgicos P1/uso terapéutico , Animales , Isquemia Encefálica/tratamiento farmacológico , Humanos , Purinérgicos/farmacología , Purinérgicos/uso terapéutico
19.
Brain Res ; 1551: 59-72, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24457041

RESUMEN

Evidence indicate that adenosine A2A receptor subtype is of critical importance in stroke. An overexpression of A2A adenosine receptors occurs at central level on neurons and microglia of ischemic striatum and cortex after focal ischemia. Adenosine A2A receptor subtype is localized not only at central level but also peripherally on blood cells, where it is known to exert antiinflammatory effect. Purpose of the present work was to investigate the putative neuroprotective effect of the adenosine A2A receptor agonist CGS21680 in a rat model of transient medial cerebral artery occlusion (MCAo). Transient cerebral ischemia was induced by 1h occlusion of MCA. CGS21680 (0.01 and 0.1mg/kg, i.p.) was administered starting 4h after ischemia according to a chronic protocol (twice/day for 7 days). CGS21680, at the dose of 0.1mg/kg transiently increased heart frequency but did not modify blood pressure. At the dose of 0.01mg/kg the drug did not modify either heart frequency or blood pressure. Following transient MCAo, CGS21680 at both doses protected from neurological deficit from the first day up to 7 days thereafter. At this time, it has reduced microgliosis, astrogliosis and improved myelin organization in the striatum and cytoarchitecture of the ischemic cortex and striatum. Two days after transient MCAo, CGS21680 has reduced the number of infiltrated granulocytes into the ischemic tissue. Data indicate that CGS21680 systemically administered is protective by immunosuppressive effects.


Asunto(s)
Agonistas del Receptor de Adenosina A2/uso terapéutico , Adenosina/análogos & derivados , Isquemia Encefálica/tratamiento farmacológico , Corteza Cerebral/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Fenetilaminas/uso terapéutico , Adenosina/uso terapéutico , Animales , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Corteza Cerebral/patología , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Gliosis/tratamiento farmacológico , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Wistar , Pérdida de Peso/efectos de los fármacos
20.
Front Aging Neurosci ; 6: 322, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25505884

RESUMEN

Chronic cerebral hypoperfusion during aging may cause progressive neurodegeneration as ischemic conditions persist. Proper functioning of the interplay between neurons and glia is fundamental for the functional organization of the brain. The aim of our research was to study the pathophysiological mechanisms, and particularly the derangement of the interplay between neurons and astrocytes-microglia with the formation of "triads," in a model of chronic cerebral hypoperfusion induced by the two-vessel occlusion (2VO) in adult Wistar rats (n = 15). The protective effect of dipyridamole given during the early phases after 2VO (4 mg/kg/day i.v., the first 7 days after 2VO) was verified (n = 15). Sham-operated rats (n = 15) were used as controls. Immunofluorescent triple staining of neurons (NeuN), astrocytes (GFAP), and microglia (IBA1) was performed 90 days after 2VO. We found significantly higher amount of "ectopic" neurons, neuronal debris and apoptotic neurons in CA1 Str. Radiatum and Str. Pyramidale of 2VO rats. In CA1 Str. Radiatum of 2VO rats the amount of astrocytes (cells/mm(2)) did not increase. In some instances several astrocytes surrounded ectopic neurons and formed a "micro scar" around them. Astrocyte branches could infiltrate the cell body of ectopic neurons, and, together with activated microglia cells formed the "triads." In the triad, significantly more numerous in CA1 Str. Radiatum of 2VO than in sham rats, astrocytes and microglia cooperated in the phagocytosis of ectopic neurons. These events might be common mechanisms underlying many neurodegenerative processes. The frequency to which they appear might depend upon, or might be the cause of, the burden and severity of neurodegeneration. Dypiridamole significantly reverted all the above described events. The protective effect of chronic administration of dipyridamole might be a consequence of its vasodilatory, antioxidant and anti-inflammatory role during the early phases after 2VO.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA