Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 41(4): 594-612, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33303678

RESUMEN

Spontaneous bursts of electrical activity in the developing auditory system arise within the cochlea before hearing onset and propagate through future sound-processing circuits of the brain to promote maturation of auditory neurons. Studies in isolated cochleae revealed that this intrinsically generated activity is initiated by ATP release from inner supporting cells (ISCs), resulting in activation of purinergic autoreceptors, K+ efflux, and subsequent depolarization of inner hair cells. However, it is unknown when this activity emerges or whether different mechanisms induce activity during distinct stages of development. Here we show that spontaneous electrical activity in mouse cochlea from both sexes emerges within ISCs during the late embryonic period, preceding the onset of spontaneous correlated activity in inner hair cells and spiral ganglion neurons, which begins at birth and follows a base to apex developmental gradient. At all developmental ages, pharmacological inhibition of P2Y1 purinergic receptors dramatically reduced spontaneous activity in these three cell types. Moreover, in vivo imaging within the inferior colliculus revealed that auditory neurons within future isofrequency zones exhibit coordinated neural activity at birth. The frequency of these discrete bursts increased progressively during the postnatal prehearing period yet remained dependent on P2RY1. Analysis of mice with disrupted cholinergic signaling in the cochlea indicate that this efferent input modulates, rather than initiates, spontaneous activity before hearing onset. Thus, the auditory system uses a consistent mechanism involving ATP release from ISCs and activation of P2RY1 autoreceptors to elicit coordinated excitation of neurons that will process similar frequencies of sound.SIGNIFICANCE STATEMENT In developing sensory systems, groups of neurons that will process information from similar sensory space exhibit highly correlated electrical activity that is critical for proper maturation and circuit refinement. Defining the period when this activity is present, the mechanisms responsible and the features of this activity are crucial for understanding how spontaneous activity influences circuit development. We show that, from birth to hearing onset, the auditory system relies on a consistent mechanism to elicit correlate firing of neurons that will process similar frequencies of sound. Targeted disruption of this activity will increase our understanding of how these early circuits mature and may provide insight into processes responsible for developmental disorders of the auditory system.


Asunto(s)
Vías Auditivas/crecimiento & desarrollo , Vías Auditivas/fisiología , Receptores Purinérgicos/fisiología , Adenosina Trifosfato/metabolismo , Animales , Señalización del Calcio/fisiología , Cóclea/crecimiento & desarrollo , Cóclea/fisiología , Femenino , Células Ciliadas Auditivas/fisiología , Células Ciliadas Auditivas Internas/fisiología , Colículos Inferiores/fisiología , Células Laberínticas de Soporte/fisiología , Masculino , Ratones , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/fisiología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/fisiología , Retina/fisiología , Ganglio Espiral de la Cóclea/fisiología
2.
J Neurosci ; 40(7): 1373-1388, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31896671

RESUMEN

Microglia exhibit multiple, phenotype-dependent motility patterns often triggered by purinergic stimuli. However, little data exist on motility of human microglia in pathological situations. Here we examine motility of microglia stained with a fluorescent lectin in tissue slices from female and male epileptic patients diagnosed with mesial temporal lobe epilepsy or cortical glioma (peritumoral cortex). Microglial shape varied from ramified to amoeboid cells predominantly in regions of high neuronal loss or closer to a tumor. Live imaging revealed unstimulated or purine-induced microglial motilities, including surveillance movements, membrane ruffling, and process extension or retraction. At different concentrations, ADP triggered opposing motilities. Low doses triggered process extension. It was suppressed by P2Y12 receptor antagonists, which also reduced process length and surveillance movements. Higher purine doses caused process retraction and membrane ruffling, which were blocked by joint application of P2Y1 and P2Y13 receptor antagonists. Purinergic effects on motility were similar for all microglia tested. Both amoeboid and ramified cells from mesial temporal lobe epilepsy or peritumoral cortex tissue expressed P2Y12 receptors. A minority of microglia expressed the adenosine A2A receptor, which has been linked with process withdrawal of rodent cells. Laser-mediated tissue damage let us test the functional significance of these effects. Moderate damage induced microglial process extension, which was blocked by P2Y12 receptor antagonists. Overall, the purine-induced motility of human microglia in epileptic tissue is similar to that of rodent microglia in that the P2Y12 receptor initiates process extension. It differs in that retraction is triggered by joint activation of P2Y1/P2Y13 receptors.SIGNIFICANCE STATEMENT Microglial cells are brain-resident immune cells with multiple functions in healthy or diseased brains. These diverse functions are associated with distinct phenotypes, including different microglial shapes. In the rodent, purinergic signaling is associated with changes in cell shape, such as process extension toward tissue damage. However, there are little data on living human microglia, especially in diseased states. We developed a reliable technique to stain microglia from epileptic and glioma patients to examine responses to purines. Low-intensity purinergic stimuli induced process extension, as in rodents. In contrast, high-intensity stimuli triggered a process withdrawal mediated by both P2Y1 and P2Y13 receptors. P2Y1/P2Y13 receptor activation has not previously been linked to microglial morphological changes.


Asunto(s)
Epilepsia del Lóbulo Temporal/fisiopatología , Glioma/fisiopatología , Microglía/fisiología , Receptores Purinérgicos P2Y12/fisiología , Receptores Purinérgicos P2Y1/fisiología , Receptores Purinérgicos P2/fisiología , Neoplasias Supratentoriales/fisiopatología , Adenosina Difosfato/farmacología , Adulto , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Forma de la Célula/efectos de los fármacos , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/fisiología , Extensiones de la Superficie Celular/ultraestructura , Epilepsia del Lóbulo Temporal/etiología , Epilepsia del Lóbulo Temporal/patología , Femenino , Glioma/patología , Humanos , Microscopía Intravital , Masculino , Microglía/efectos de los fármacos , Microglía/ultraestructura , Persona de Mediana Edad , Lectinas de Plantas , Agonistas Purinérgicos/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Neoplasias Supratentoriales/patología , Esclerosis Tuberosa/complicaciones
3.
Cereb Cortex ; 30(3): 1272-1290, 2020 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-31407772

RESUMEN

To understand the pathogenesis of specific neuronal circuit dysfunction in Alzheimer's disease (AD), we investigated the fate of three subclasses of "modulatory interneurons" in hippocampal CA1 using the AppNL-F/NL-F knock-in mouse model of AD. Cholecystokinin- and somatostatin-expressing interneurons were aberrantly hyperactive preceding the presence of the typical AD hallmarks: neuroinflammation and amyloid-ß (Aß) accumulation. These interneurons showed an age-dependent vulnerability to Aß penetration and a reduction in density and coexpression of the inhibitory neurotransmitter GABA synthesis enzyme, glutamic acid decarboxylase 67 (GAD67), suggesting a loss in their inhibitory function. However, calretinin (CR) interneurons-specialized to govern only inhibition, showed resilience to Aß accumulation, preservation of structure, and displayed synaptic hyperinhibition, despite the lack of inhibitory control of CA1 excitatory pyramidal cells from midstages of the disease. This aberrant inhibitory homeostasis observed in CA1 CR cells and pyramidal cells was "normalized" by blocking P2Y1 purinoreceptors, which were "upregulated" and strongly expressed in CR cells and astrocytes in AppNL-F/NL-F mice in the later stages of AD. In summary, AD-associated cell-type selective destruction of inhibitory interneurons and disrupted inhibitory homeostasis rectified by modulation of the upregulated purinoreceptor system may serve as a novel therapeutic strategy to normalize selective dysfunctional synaptic homeostasis during pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Región CA1 Hipocampal/fisiopatología , Calbindina 2/fisiología , Interneuronas/fisiología , Inhibición Neural , Receptores Purinérgicos P2Y1/fisiología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Interneuronas/patología , Masculino , Ratones Endogámicos C57BL , Regulación hacia Arriba
4.
Neuroscience ; 423: 98-108, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31689490

RESUMEN

Non-synaptic transmission is pervasive throughout the nervous system. It appears especially prevalent in peripheral ganglia, where non-synaptic interactions between neighboring cell bodies have been described in both physiological and pathological conditions, a phenomenon referred to as cross-depolarization (CD) and thought to play a role in sensory processing and chronic pain. CD has been proposed to be mediated by a chemical agent, but its identity has remained elusive. Here, we report that in the rat dorsal root ganglion (DRG), the P2Y1 purinergic receptor (P2RY1) plays an important role in regulating CD. The effect of P2RY1 is cell-type specific: pharmacological blockade of P2RY1 inhibited CD in A-type neurons while enhancing it in C-type neurons. In the nodose ganglion of the vagus, CD requires extracellular calcium in a large percentage of cells. In contrast, we show that in the DRG extracellular calcium appears to play no major role, pointing to a mechanistic difference between the two peripheral ganglia. Furthermore, we show that DRG glial cells also play a cell-type specific role in CD regulation. Fluorocitrate-induced glial inactivation had no effect on A-cells but enhanced CD in C-cells. These findings shed light on the mechanism of CD in the DRG and pave the way for further analysis of non-synaptic neuronal communication in sensory ganglia.


Asunto(s)
Comunicación Celular/fisiología , Ganglios Espinales/fisiología , Neuronas/fisiología , Receptores Purinérgicos P2Y1/fisiología , 2-Amino-5-fosfonovalerato/farmacología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Potenciales de Acción/fisiología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Bencenosulfonatos/farmacología , Calcio/fisiología , Citratos/farmacología , Estimulación Eléctrica , Masculino , Inhibición Neural/fisiología , Neuroglía/efectos de los fármacos , Neuroglía/fisiología , Neuronas/efectos de los fármacos , Ganglio Nudoso/fisiología , Piperazinas/farmacología , Ratas , Receptores Purinérgicos P2Y1/efectos de los fármacos
5.
J Neurosci ; 39(27): 5377-5392, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31048325

RESUMEN

Extracellular ATP activates inflammatory responses to tissue injury. It is also implicated in establishing lasting network hyperexcitability in the brain by acting upon independent receptor systems. Whereas the fast-acting P2X channels have well-established roles driving neuroinflammation and increasing hyperexcitability, the slower-acting metabotropic P2Y receptors have received much less attention. Recent studies of P2Y1 receptor function in seizures and epilepsy have produced contradictory results, suggesting that the role of this receptor during seizure pathology may be highly sensitive to context. Here, by using male mice, we demonstrate that the metabotropic P2Y1 receptor mediates either proconvulsive or anticonvulsive responses, dependent on the time point of activation in relation to the induction of status epilepticus. P2Y1 deficiency or a P2Y1 antagonist (MRS2500) administered before a chemoconvulsant, exacerbates epileptiform activity, whereas a P2Y1 agonist (MRS2365) administered at this time point is anticonvulsant. When these drugs are administered after the onset of status epilepticus, however, their effect on seizure severity is reversed, with the antagonist now anticonvulsant and the agonist proconvulsant. This result was consistent across two different mouse models of status epilepticus (intra-amygdala kainic acid and intraperitoneal pilocarpine). Pharmacologic P2Y1 blockade during status epilepticus reduces also associated brain damage, delays the development of epilepsy and, when applied during epilepsy, suppresses spontaneous seizures, in mice. Our data show a context-specific role for P2Y1 during seizure pathology and demonstrate that blocking P2Y1 after status epilepticus and during epilepsy has potent anticonvulsive effects, suggesting that P2Y1 may be a novel candidate for the treatment of drug-refractory status epilepticus and epilepsy.SIGNIFICANCE STATEMENT This is the first study to fully characterize the contribution of a metabotropic purinergic P2Y receptor during acute seizures and epilepsy. The findings suggest that targeting P2Y1 may offer a potential novel treatment strategy for drug-refractory status epilepticus and epilepsy. Our data demonstrate a context-specific role of P2Y1 activation during seizures, switching from a proconvulsive to an anticonvulsive role depending on physiopathological context. Thus, our study provides a possible explanation for seemingly conflicting results obtained between studies of different brain diseases where P2Y1 targeting has been proposed as a potential treatment strategy and highlights that the timing of pharmacological interventions is of critical importance to the understanding of how receptors contribute to the generation of seizures and the development of epilepsy.


Asunto(s)
Encéfalo/fisiopatología , Epilepsia/fisiopatología , Receptores Purinérgicos P2Y1/fisiología , Estado Epiléptico/fisiopatología , Adenosina Difosfato/administración & dosificación , Adenosina Difosfato/análogos & derivados , Animales , Encéfalo/efectos de los fármacos , Nucleótidos de Desoxiadenina/administración & dosificación , Modelos Animales de Enfermedad , Electroencefalografía , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Agonistas del Receptor Purinérgico P2Y/administración & dosificación , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Receptores Purinérgicos P2Y1/genética
6.
J Cereb Blood Flow Metab ; 39(11): 2144-2156, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30334687

RESUMEN

Extracellular ATP, which is released from damaged cells after ischemia, activates P2 receptors. P2Y1 receptors (P2Y1R) have received considerable attention, especially in astrocytes, because their activation plays a central role in the regulation of neuron-to-glia communication. However, the functions or even existence of P2Y1R in microglia remain unknown, despite the fact that many microglial P2 receptors are involved in several brain diseases. Herein, we demonstrate the presence and functional capability of microglial P2Y1R to provide neuroprotective effects following ischemic stress. Cerebral ischemia resulted in increased microglial P2Y1R expression. The number of injured hippocampal neurons was significantly higher in P2Y1 R knockout (KO) mice than wildtype mice after forebrain ischemia. Propidium iodide (PI) uptake, a marker for dying cells, was significantly higher in P2Y1R KO hippocampal slices compared with wildtype hippocampal slices at 48 h after 40-min oxygen-glucose deprivation (OGD). Furthermore, increased PI uptake following OGD was rescued by ectopic overexpression of P2Y1R in microglia. In summary, these data suggest that microglial P2Y1R mediate neuroprotective effects against ischemic stress and OGD insult.


Asunto(s)
Microglía/química , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Receptores Purinérgicos P2Y1/fisiología , Animales , Isquemia Encefálica , Muerte Celular/efectos de los fármacos , Glucosa/deficiencia , Hipocampo/metabolismo , Hipocampo/patología , Hipoxia , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Receptores Purinérgicos P2Y1/análisis
7.
Brain Res Bull ; 151: 74-83, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30593879

RESUMEN

Nucleotides can contribute to the survival of different glial and neuronal models at the nervous system via activation of purinergic P2X and P2Y receptors. Their activation counteracts different proapoptotic events, such as excitotoxicity, mitochondrial impairment, oxidative stress and DNA damage, which concur to elicit cell loss in different processes of neurodegeneration and brain injury. Thus, it is frequent to find that different neuroprotective mediators converge in the activation of the same intracellular survival pathways to protect cells from death. The present review focuses on the role of P2Y1 and P2Y13 metabotropic receptors, and P2X7 ionotropic receptors to regulate the balance between survival and apoptosis. In particular, we analyze the intracellular pathways involved in the signaling of these nucleotide receptors to elicit survival, including calcium/PLC, PI3K/Akt/GSK3, MAPK cascades, and the expression of antioxidant and antiapoptotic genes. This review emphasizes the novel contribution of nucleotide receptors to maintain cell homeostasis through the regulation of MAP kinases and phosphatases. Unraveling the different roles found for nucleotide receptors in different models and cellular contexts may be crucial to delineate future therapeutic applications based on targeting nucleotide receptors for neuroprotection.


Asunto(s)
Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Neuroglía/metabolismo , Neuronas/metabolismo , Neuroprotección/fisiología , Fármacos Neuroprotectores/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Receptores Purinérgicos P2/fisiología , Receptores Purinérgicos P2X/metabolismo , Receptores Purinérgicos P2X/fisiología , Receptores Purinérgicos P2X7/fisiología , Receptores Purinérgicos P2Y/metabolismo , Receptores Purinérgicos P2Y/fisiología , Receptores Purinérgicos P2Y1/fisiología , Transducción de Señal
8.
J Neurosci ; 38(6): 1383-1395, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29305530

RESUMEN

Fine processes of astrocytes enwrap synapses and are well positioned to sense neuronal information via synaptic transmission. In rodents, astrocyte processes sense synaptic transmission via Gq-protein coupled receptors (GqPCR), including the P2Y1 receptor (P2Y1R), to generate Ca2+ signals. Astrocytes display numerous spontaneous microdomain Ca2+ signals; however, it is not clear whether such signals are due to local synaptic transmission and/or in what timeframe astrocytes sense local synaptic transmission. To ask whether GqPCRs mediate microdomain Ca2+ signals, we engineered mice (both sexes) to specifically overexpress P2Y1Rs in astrocytes, and we visualized Ca2+ signals via a genetically encoded Ca2+ indicator, GCaMP6f, in astrocytes from adult mice. Astrocytes overexpressing P2Y1Rs showed significantly larger Ca2+ signals in response to exogenously applied ligand and to repetitive electrical stimulation of axons compared with controls. However, we found no evidence of increased microdomain Ca2+ signals. Instead, Ca2+ waves appeared and propagated to occupy areas that were up to 80-fold larger than microdomain Ca2+ signals. These Ca2+ waves accounted for only 2% of total Ca2+ events, but they were 1.9-fold larger and 2.9-fold longer in duration than microdomain Ca2+ signals at processes. Ca2+ waves did not require action potentials for their generation and occurred in a probenecid-sensitive manner, indicating that the endogenous ligand for P2Y1R is elevated independently of synaptic transmission. Our data suggest that spontaneous microdomain Ca2+ signals occur independently of P2Y1R activation and that astrocytes may not encode neuronal information in response to synaptic transmission at a point source of neurotransmitter release.SIGNIFICANCE STATEMENT Astrocytes are thought to enwrap synapses with their processes to receive neuronal information via Gq-protein coupled receptors (GqPCRs). Astrocyte processes display numerous microdomain Ca2+ signals that occur spontaneously. To determine whether GqPCRs play a role in microdomain Ca2+ signals and the timeframe in which astrocytes sense neuronal information, we engineered mice whose astrocytes specifically overexpress the P2Y1 receptor, a major GqPCR in astrocytes. We found that overexpression of P2Y1 receptors in astrocytes did not increase microdomain Ca2+ signals in astrocyte processes but caused Ca2+ wavelike signals. Our data indicate that spontaneous microdomain Ca2+ signals do not require activation of P2Y1 receptors.


Asunto(s)
Astrocitos/fisiología , Señalización del Calcio/fisiología , Receptores Purinérgicos P2Y1/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Señalización del Calcio/efectos de los fármacos , Femenino , Hipocampo/fisiología , Masculino , Ratones , Ratones Transgénicos , Probenecid/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/genética , Sinapsis/fisiología
9.
J Physiol ; 596(15): 3245-3269, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28678385

RESUMEN

KEY POINTS: The ventilatory response to reduced oxygen (hypoxia) is biphasic, comprising an initial increase in ventilation followed by a secondary depression. Our findings indicate that, during hypoxia, astrocytes in the pre-Bötzinger complex (preBötC), a critical site of inspiratory rhythm generation, release a gliotransmitter that acts via P2Y1 receptors to stimulate ventilation and reduce the secondary depression. In vitro analyses reveal that ATP excitation of the preBötC involves P2Y1 receptor-mediated release of Ca2+ from intracellular stores. By identifying a role for gliotransmission and the sites, P2 receptor subtype, and signalling mechanisms via which ATP modulates breathing during hypoxia, these data advance our understanding of the mechanisms underlying the hypoxic ventilatory response and highlight the significance of purinergic signalling and gliotransmission in homeostatic control. Clinically, these findings are relevant to conditions in which hypoxia and respiratory depression are implicated, including apnoea of prematurity, sleep disordered breathing and congestive heart failure. ABSTRACT: The hypoxic ventilatory response (HVR) is biphasic, consisting of a phase I increase in ventilation followed by a secondary depression (to a steady-state phase II) that can be life-threatening in premature infants who suffer from frequent apnoeas and respiratory depression. ATP released in the ventrolateral medulla oblongata during hypoxia attenuates the secondary depression. We explored a working hypothesis that vesicular release of ATP by astrocytes in the pre-Bötzinger Complex (preBötC) inspiratory rhythm-generating network acts via P2Y1 receptors to mediate this effect. Blockade of vesicular exocytosis in preBötC astrocytes bilaterally (using an adenoviral vector to specifically express tetanus toxin light chain in astrocytes) reduced the HVR in anaesthetized rats, indicating that exocytotic release of a gliotransmitter within the preBötC contributes to the hypoxia-induced increases in ventilation. Unilateral blockade of P2Y1 receptors in the preBötC via local antagonist injection enhanced the secondary respiratory depression, suggesting that a significant component of the phase II increase in ventilation is mediated by ATP acting at P2Y1 receptors. In vitro responses of the preBötC inspiratory network, preBötC inspiratory neurons and cultured preBötC glia to purinergic agents demonstrated that the P2Y1 receptor-mediated increase in fictive inspiratory frequency involves Ca2+ recruitment from intracellular stores leading to increases in intracellular Ca2+ ([Ca2+ ]i ) in inspiratory neurons and glia. These data suggest that ATP is released by preBötC astrocytes during hypoxia and acts via P2Y1 receptors on inspiratory neurons (and/or glia) to evoke Ca2+ release from intracellular stores and an increase in ventilation that counteracts the hypoxic respiratory depression.


Asunto(s)
Adenosina Trifosfato/fisiología , Astrocitos/fisiología , Hipoxia/fisiopatología , Bulbo Raquídeo/fisiología , Receptores Purinérgicos P2Y1/fisiología , Animales , Calcio/fisiología , Masculino , Ventilación Pulmonar , Ratas Sprague-Dawley
10.
Brain Res Bull ; 130: 165-172, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28153540

RESUMEN

Although interleukin-1ß (IL-1ß) is a prototypical pro-inflammatory cytokine, the specific mechanisms underlying the role of its cognate receptor, the interleukin-1 receptor (IL-1R) in peripheral sensitization remain to be investigated. Since emerging evidence in the literature indicates that IL-1ß can modulate membrane-bound receptors, we decided to examine the involvement of P2Y1 receptor (P2Y1R) in IL-1ß induced pain and the potential interaction of P2Y1Rs and IL-1Rs in both naïve and carrageenan injected rats. Intraplantar (i.pl) injection of IL-1ß dose-dependently produced mechanical and thermal hypersensitivity in naïve rats. Pre-treatment with IL-1ra (i.pl, 30 and 100ng), an endogenous IL-1R antagonist, prevented the IL-1ß induced mechanical and thermal hypersensitivity. Pre-treatment with MRS2500 (i.pl, 1 and 3nmol), a specific P2Y1R antagonist, dose-dependently reduced IL-1ß induced thermal hypersensitivity, but did not affect the development of mechanical hypersensitivity. Conversely coadministration of MRS2500 (i.pl, 0.1nmol, sub-effective dose) together with IL-1ra (10nmol, sub-effective dose) significantly reduced IL-1ß induced thermal, but not mechanical hypersensitivity. We next used immunohistochemistry to demonstrate that P2Y1 and IL-1 type I receptors co-localize predominantly in small diameter neurons in the dorsal root ganglion. We also performed experiments to examine the interaction of P2Y1Rs and IL-1Rs under the inflammatory conditions induced by 2% carrageenan. Intraplantar coadministration of MRS2500 (3nmol, sub-effective dose) and IL-1ra (30ng, sub-effective dose) significantly reduced inflammatory thermal, but not mechanical, hypersensitivity. These data indicate the involvement of P2Y1Rs in IL-1ß mediated pain in both naive and carrageenan injected rats. There is a positive interaction between peripheral P2Y1Rs and IL-1Rs in both IL-1ß and carrageenan-induced thermal hypersensitivity.


Asunto(s)
Hiperalgesia/fisiopatología , Interleucina-1beta/fisiología , Receptores de Interleucina-1/fisiología , Receptores Purinérgicos P2Y1/fisiología , Animales , Carragenina/administración & dosificación , Hiperalgesia/inducido químicamente , Interleucina-1beta/administración & dosificación , Masculino , Umbral del Dolor , Ratas Sprague-Dawley
11.
J Physiol ; 595(3): 713-738, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506492

RESUMEN

KEY POINTS: Neuroinflammation associated with CNS insults leads to neuronal hyperexcitability, which may culminate in epileptiform discharges. Application of the endotoxin lipopolysaccharide (LPS) to brain tissue initiates a neuroinflammatory cascade, providing an experimental model to study the mechanisms of neuroinflammatory neuronal hyperexcitability. Here we show that LPS application to hippocampal slices markedly enhances the excitability of CA1 pyramidal cells by inhibiting a specific potassium current, the M-current, generated by KV 7/M channels, which controls the excitability of almost every neuron in the CNS. The LPS-induced M-current inhibition is triggered by sequential activation of microglia, astrocytes and pyramidal cells, mediated by metabotropic purinergic and glutamatergic transmission, leading to blockade of KV 7/M channels by calcium released from intracellular stores. The identification of the downstream molecular target of neuroinflammation, namely the KV 7/M channel, potentially has far reaching implications for the understanding and treatment of many acute and chronic brain disorders. ABSTRACT: Acute brain insults and many chronic brain diseases manifest an innate inflammatory response. The hallmark of this response is glia activation, which promotes repair of damaged tissue, but also induces structural and functional changes that may lead to an increase in neuronal excitability. We have investigated the mechanisms involved in the modulation of neuronal activity by acute inflammation. Initiating inflammatory responses in hippocampal tissue rapidly led to neuronal depolarization and repetitive firing even in the absence of active synaptic transmission. This action was mediated by a complex metabotropic purinergic and glutamatergic glia-to-neuron signalling cascade, leading to the blockade of neuronal KV 7/M channels by Ca2+ released from internal stores. These channels generate the low voltage-activating, non-inactivating M-type K+ current (M-current) that controls intrinsic neuronal excitability, and its inhibition was the predominant cause of the inflammation-induced hyperexcitability. Our discovery that the ubiquitous KV 7/M channels are the downstream target of the inflammation-induced cascade, has far reaching implications for the understanding and treatment of many acute and chronic brain disorders.


Asunto(s)
Canales de Potasio KCNQ/fisiología , Lipopolisacáridos/farmacología , Células Piramidales/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Región CA1 Hipocampal/citología , Calcio/fisiología , Masculino , Células Piramidales/fisiología , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Receptores Purinérgicos P2Y1/fisiología
12.
J Theor Biol ; 408: 127-136, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27521526

RESUMEN

A novel platelet-derived growth factor receptor alpha-positive cell (PDGFRα(+)) has recently been identified as part of the purinergic inhibitory neural control mechanism in the gastrointestinal (GI) tract. The mechanism through which PDGFRα(+) cells mediate GI muscle relaxation has been found to be associated with the purine receptors P2Y1 and apamin-sensitive SK3 channels that are highly expressed in these cells. This study aims to develop a mechanistic model elucidating a proposed mechanism through which PDGFRα(+) cells contribute to purinergic inhibitory neuromuscular transmission. In accordance with recent experimental findings, the model describes how the binding of neurotransmitters, released from enteric neurons, triggers the release of Ca(2+) from the endoplasmic reticulum in the PDGFRα(+) cells, and how this subsequently leads to large amplitude transient outward currents, which in turn hyperpolarize the cell. The model has been validated against experimental recordings and good agreement was found under normal and pharmacologically-altered conditions. This model demonstrates the feasibility of the proposed mechanism and provides a basis for understanding the mechanism underlying purinergic control of colonic motility.


Asunto(s)
Tracto Gastrointestinal/citología , Modelos Biológicos , Relajación Muscular/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Calcio/metabolismo , Humanos , Neurotransmisores/metabolismo , Receptores Purinérgicos P2Y1/fisiología
13.
BMC Pharmacol Toxicol ; 17(1): 29, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27384918

RESUMEN

BACKGROUND: All hematopoietic cells express P2 receptors, however pharmacological characteristics such as expression and affinity in granulocytes are unknown. METHODS: Pharmacological characteristics of P2 receptors were evaluated by Ca(2+) measurements using Fura-2 fluorophore. P2 receptors expression were analyzed by flow cytometry and RT-PCR. P2 interaction were shown by coimmunoprecipitation, western blotting and FRET. RESULTS: Granulocytes were responsive to P2Y agonists, whereas P2X agonists were ineffective. Ca(2+) increase, elicited by ADP and UTP was dependent on intracellular stocks and sensitive to G-coupled receptor inhibition. Moreover, MRS2179, a specific antagonist of the P2Y1 receptor, abolished ADP response. Interestingly, ADP and UTP exhibited full heterologous desensitization, suggesting that these agonists interact with the same receptor. The heteromeric association between P2Y1 receptor and the P2Y2 and P2Y4 receptors was shown by immunoprecipitation and FRET analysis. CONCLUSION: Clear evidence of heteromeric association of P2Y receptors was found during the evaluation of P2 receptors present in mice granulocytes, which could impact in the classical pharmacology of P2Y receptors in granulocytes.


Asunto(s)
Granulocitos/fisiología , Receptores Purinérgicos P2Y1/fisiología , Receptores Purinérgicos P2Y2/fisiología , Receptores Purinérgicos P2/fisiología , Animales , Femenino , Citometría de Flujo , Granulocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Unión Proteica/fisiología , Agonistas Purinérgicos/farmacología , Receptores Purinérgicos P2/química , Receptores Purinérgicos P2Y1/química , Receptores Purinérgicos P2Y2/química , Células Madre/efectos de los fármacos , Células Madre/fisiología
14.
Neurochem Res ; 41(7): 1784-96, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27038933

RESUMEN

Retinal glial (Müller) cells possess an endogenous purinergic signal transduction cascade which normally prevents cellular swelling in osmotic stress. The cascade can be activated by osmotic or glutamate receptor-dependent ATP release. We determined whether activation of this cascade is altered in Müller cells of transgenic rats that suffer from a slow photoreceptor degeneration due to the expression of a truncated human cilia gene polycystin-2 (CMV-PKD21/703 HA). Age-matched Sprague-Dawley rats served as control. Retinal slices were superfused with a hypoosmotic solution (60 % osmolarity). Müller cells in retinas of PKD21/703 rats swelled immediately in hypoosmotic stress; this was not observed in control retinas. Pharmacological blockade of P2Y1 or adenosine A1 receptors induced osmotic swelling of Müller cells from control rats. The swelling induced by the P2Y1 receptor antagonist was mediated by induction of oxidative-nitrosative stress, mitochondrial dysfunction, production of inflammatory lipid mediators, and a sodium influx from the extracellular space. Exogenous VEGF or glutamate prevented the hypoosmotic swelling of Müller cells from PKD21/703 rats; this effect was mediated by activation of the purinergic signaling cascade. In neuroretinas of PKD21/703 rats, the gene expression levels of P2Y1 and A1 receptors, pannexin-1, connexin 45, NTPDases 1 and 2, and various subtypes of nucleoside transporters are elevated compared to control. The data may suggest that the osmotic swelling of Müller cells from PKD21/703 rats is caused by an abrogation of the osmotic ATP release while the glutamate-induced ATP release is functional. In the normal retina, ATP release and autocrine P2Y1 receptor activation serve to inhibit the induction of oxidative-nitrosative stress, mitochondrial dysfunction, and production of inflammatory lipid mediators, which otherwise will induce a sodium influx and cytotoxic Müller cell swelling under anisoosmotic conditions. Purinergic receptors may represent a target for the protection of retinal glial cells from mitochondrial oxidative stress.


Asunto(s)
Tamaño de la Célula , Células Ependimogliales/metabolismo , Receptor de Adenosina A1/fisiología , Receptores Purinérgicos P2Y1/fisiología , Retina/metabolismo , Canales Catiónicos TRPP/biosíntesis , Animales , Células Ependimogliales/patología , Regulación de la Expresión Génica , Humanos , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Retina/patología , Canales Catiónicos TRPP/genética
15.
Yakugaku Zasshi ; 135(12): 1335-40, 2015.
Artículo en Japonés | MEDLINE | ID: mdl-26632148

RESUMEN

Adenosine and its precursors, ATP and ADP, exert various physiological effects via binding to purinergic receptors. We previously used co-immunoprecipitation, bioluminescence resonance energy transfer (BRET) and immunoelectron microscopy to demonstrate the hetero-oligomerization of purinergic receptor subtypes. Furthermore, pharmacological studies found significant changes in receptor-mediated signaling in human embryonic kidney (HEK) 293T cells co-transfected with these receptors. These findings suggest that heterodimers of purinergic receptors may have distinct pharmacological profiles, possibly due to dimerization-induced conformational changes, further suggesting that hetero-dimerization may be employed to "fine-tune" purinergic receptor signaling. Adenosine A(2A) receptor (A(2A)R), P2Y1 receptor (P2Y1R) and P2Y12 receptor (P2Y12R) are predominantly expressed on human platelets. ADP activates human platelets by stimulating both P2Y1R and P2Y12R, which act sequentially and in concert to achieve complete platelet aggregation. In contrast, adenosine stimulates Gs-coupled A(2A)R, followed by activativation of adenylate cyclase, leading to an increase in intracellular cAMP levels, which potently inhibits platelet activation. We examined the hetero-oligomerization and functional interactions of A(2A)R, P2Y1R, and P2Y12R. In HEK293T cells triply expressing all three receptors, hetero-oligomerization was observed among the three receptors. Additionally, P2Y1R agonist-evoked Ca(2+) signaling was significantly inhibited by co-treatment with an A(2A)R antagonist in HEK293T cells. In human platelets, we identified endogenous A(2A)R/P2Y1R and A(2A)R/P2Y12R heterodimers. We also observed functional Ca(2+)-signaling-related cross-talk similar to those found in HEK293T cells, and found that they appeared to affect platelet shape. These results collectively suggest that intermolecular signal transduction and specific conformational changes occur among components of the hetero-oligomers formed by these three receptors.


Asunto(s)
Plaquetas/química , Plaquetas/citología , Receptores Purinérgicos/fisiología , Adenosina Difosfato/fisiología , Dimerización , Humanos , Agregación Plaquetaria/fisiología , Receptor Cross-Talk/fisiología , Receptor de Adenosina A2A/fisiología , Receptores Purinérgicos/análisis , Receptores Purinérgicos P2Y1/fisiología , Receptores Purinérgicos P2Y12/fisiología
16.
Br J Pharmacol ; 172(21): 5199-210, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26282717

RESUMEN

BACKGROUND AND PURPOSE: Stimulation of soluble guanylyl cyclase (sGC) is a valuable therapeutic strategy for the treatment of several cardiovascular diseases. The sGC stimulator riociguat has been approved for the treatment of two forms of pulmonary hypertension. Platelets contain large amounts of sGC and play a key role in the regulation of haemostasis. Therefore, we investigated the effects of riociguat on platelet function. EXPERIMENTAL APPROACH: The effect of riociguat treatment on human platelet activation and aggregation was investigated. The sGC-specific effects of riociguat were determined by comparing wild-type and platelet-specific sGC-knockout mice. KEY RESULTS: Riociguat induced cGMP synthesis and subsequent PKG activation in human platelets, suggesting that the inhibitory effects are mediated by cGMP signalling. This finding was confirmed when sGC-knockout platelets were not inhibited by riociguat. In washed human platelets, 100 nM riociguat reduced ADP-induced GPIIb/IIIa activation, while a 10-fold higher concentration was required to reduce convulxin-stimulated GPIIb/IIIa activation. Riociguat inhibited ADP-induced platelet shape change and aggregation, while ATP-induced shape change remained unaffected. However, in PRP and whole blood, 50-100 µM riociguat was required to inhibit platelet activation and aggregation. Riociguat in combination with iloprost significantly inhibited platelet aggregation, even in whole blood. CONCLUSIONS AND IMPLICATIONS: Riociguat inhibits platelet activation in whole blood only at concentrations above 50 µM, while the plasma concentrations in riociguat-treated patients are 150 to 500 nM. This finding indicates that riociguat treatment does not affect platelet function in patients. Nevertheless, the possibility that riociguat acts synergistically with iloprost to inhibit platelet activation should be considered.


Asunto(s)
Sangre , Guanilato Ciclasa/metabolismo , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Activación Enzimática , Humanos , Iloprost/farmacología , Ratones , Ratones Noqueados , Agregación Plaquetaria/fisiología , Receptores Purinérgicos P2Y1/efectos de los fármacos , Receptores Purinérgicos P2Y1/fisiología , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2Y12/fisiología , Guanilil Ciclasa Soluble
17.
J Mol Neurosci ; 57(3): 446-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26036470

RESUMEN

ATP is co-stored and co-released with acetylcholine (ACh) at the pre-synaptic vesicles in vertebrate neuromuscular junction (nmj). Several lines of studies demonstrated that binding of ATP to its corresponding P2Y1 and P2Y2 receptors in the muscle regulated post-synaptic gene expressions. To further support the notion that P2Y receptors are playing indispensable role in formation of post-synaptic specifications at the nmj, the knock-out mice of P2Y1 receptor (P2Y1R (-/-)) were employed here for analyses. In P2Y1R (-/-) mice, the expression of P2Y2 receptor in muscle was reduced by over 50 %, as compared to P2Y1R (+/+) mice. In parallel, the expression of acetylcholinesterase (AChE) in muscle was markedly decreased. In the analysis of the expression of anchoring subunits of AChE in P2Y1R (-/-) mice, the proline-rich membrane anchor (PRiMA) subunit was reduced by 60 %; while the collagen tail (ColQ) subunit was reduced by 50 %. AChE molecular forms in the muscle were not changed, except the amount of enzyme was reduced. Immuno-staining of P2Y1R (-/-) mice nmj, both AChE and AChR were still co-localized at the nmj, and the staining was diminished. Taken together our data demonstrated that P2Y1 receptor regulated the nmj gene expression.


Asunto(s)
Acetilcolinesterasa/biosíntesis , Regulación de la Expresión Génica/fisiología , Unión Neuromuscular/metabolismo , Receptores Purinérgicos P2Y1/deficiencia , Receptores Purinérgicos P2Y2/biosíntesis , Acetilcolinesterasa/química , Acetilcolinesterasa/genética , Adenosina Trifosfato/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Subunidades de Proteína , Receptores Colinérgicos/metabolismo , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y1/fisiología , Receptores Purinérgicos P2Y2/genética
18.
Pharmacol Biochem Behav ; 128: 23-32, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25449358

RESUMEN

Metabotropic P2Y receptors subfamily consists of eight functional mammalian receptors. Specifically, P2Y1, P2Y6 and P2Y11 receptors have been described in the sensory nervous system, but their participation, at peripheral level, in behavioral pain models is scarcely understood. This study assessed the role of peripheral P2Y1, P2Y6 and P2Y11 receptors in formalin-induced inflammatory pain. Ipsilateral, but not contralateral peripheral pre-treatment with the endogenous P2Y1 (ADP, 100-1000nmol/paw), P2Y6 (UDP, 180-300nmol/paw) and P2Y11 (ATP, 100-1000nmol/paw), or selective P2Y1 (MRS2365, 0.1-10nmol/paw), P2Y6 (PSB0474, 0.1-0.10pmol/paw) and P2Y11 (NF546, 0.3-3nmol/paw) receptor agonists increased 0.5% formalin-induced flinching behavior. Concordantly, peripheral pre-treatment with the selective P2Y1 (MRS2500, 0.01-10pmol/paw), P2Y6 (MRS2578, 3-30nmol/paw) and P2Y11 (NF340, 1-10nmol/paw) receptor antagonists significantly decreased 1% formalin-induced flinching behavior. Furthermore, the pronociceptive effect of ADP (100nmol/paw) or MRS2365 (10nmol/paw), UDP (300nmol/paw) or PSB0474 (10pmol/paw) and ATP (1000nmol/paw) or NF546 (3nmol/paw) was blocked by the selective P2Y1 (MRS2500, 0.01nmol/paw), P2Y6 (MRS2578, 3nmol/paw), and P2Y11 (NF340, 1nmol/paw) receptor antagonists, respectively. Western blot analysis confirmed the presence of P2Y1 (66kDa), P2Y6 (36kDa) and P2Y11 (75kDa) receptors in dorsal root ganglia (DRG) and sciatic nerve. Results suggest that peripheral activation of P2Y1, P2Y6 and P2Y11 receptors plays a pronociceptive role in formalin-induced pain.


Asunto(s)
Dolor Nociceptivo/fisiopatología , Receptores Purinérgicos P2Y1/fisiología , Receptores Purinérgicos P2/fisiología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Femenino , Formaldehído/toxicidad , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiopatología , Inflamación/fisiopatología , Isotiocianatos/farmacología , Nocicepción/efectos de los fármacos , Nocicepción/fisiología , Dolor Nociceptivo/inducido químicamente , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Agonistas del Receptor Purinérgico P2/farmacología , Antagonistas del Receptor Purinérgico P2/farmacología , Ratas , Ratas Wistar , Nervio Ciático/efectos de los fármacos , Nervio Ciático/fisiopatología , Tiourea/análogos & derivados , Tiourea/farmacología , Uridina Difosfato/análogos & derivados , Uridina Difosfato/metabolismo , Uridina Difosfato/farmacología
19.
Eur J Pharmacol ; 740: 442-54, 2014 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-24998877

RESUMEN

The purine receptor involved in inhibitory responses in the gastrointestinal tract has been recently identified. P2Y1 receptor activation mediates the fast component of the inhibitory junction potential (IJPf) and the non-nitrergic relaxation. The aim of the present work has been to investigate which purinergic agonist better mimics endogenous responses. We used different agonist and antagonist of P2 receptors. Contractility and microelectrode experiments were used to compare the effects of exogenously added purines and electrical field stimulation (EFS)-induced nerve mediated effects in rat and human colonic strips. In rat colon, the IJPf and EFS-induced inhibition of contractions were concentration-dependently inhibited by the P2Y1 antagonist MRS2500 but not by iso-PPADS or NF023 (P2X antagonists) up to 1 µM. In samples from human colon, EFS-induced inhibition of contractions was inhibited by either MRS2500 or apamin (1 µM) but not by iso-PPADS. In both species, α,ß-meATP, a stable analog of ATP, caused inhibition of spontaneous contractions. α,ß-meATP effect was concentration-dependent (EC50: 2.7 µM rat, 4.4 µM human) and was antagonized by either MRS2500 or apamin but unaffected by P2X antagonists. ATP, ADP, ß-NAD and ADP-ribose inhibited spontaneous contractions but did not show the same sensitivity profile to purine receptor antagonists as EFS-induced inhibition of contractions. The effect of α,ß-meATP is due to P2Y1 receptor activation leading the opening of sKca channels. Accordingly, α,ß-meATP mimics the endogenous purinergic mediator. In contrast, exogenously added putative neurotransmitters do not exactly mimic the endogenous mediator. Quick degradation by ecto-nuclease or different distribution of receptors (junctionally vs extrajunctionally) might explain these results.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Colon/efectos de los fármacos , Agonistas Purinérgicos/farmacología , Antagonistas del Receptor Purinérgico P2/farmacología , Receptores Purinérgicos P2Y1/fisiología , Adenosina Trifosfato/farmacología , Anciano , Animales , Apamina/farmacología , Colon/fisiología , Nucleótidos de Desoxiadenina/farmacología , Estimulación Eléctrica , Femenino , Humanos , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiología , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Ratas Sprague-Dawley , Suramina/análogos & derivados , Suramina/farmacología
20.
J Physiol ; 592(14): 3089-111, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24879869

RESUMEN

PreBötzinger complex inspiratory rhythm-generating networks are excited by metabotropic purinergic receptor subtype 1 (P2Y1R) activation. Despite this, and the fact that inspiratory MNs express P2Y1Rs, the role of P2Y1Rs in modulating motor output is not known for any MN pool. We used rhythmically active brainstem-spinal cord and medullary slice preparations from neonatal rats to investigate the effects of P2Y1R signalling on inspiratory output of phrenic and XII MNs that innervate diaphragm and airway muscles, respectively. MRS2365 (P2Y1R agonist, 0.1 mm) potentiated XII inspiratory burst amplitude by 60 ± 9%; 10-fold higher concentrations potentiated C4 burst amplitude by 25 ± 7%. In whole-cell voltage-clamped XII MNs, MRS2365 evoked small inward currents and potentiated spontaneous EPSCs and inspiratory synaptic currents, but these effects were absent in TTX at resting membrane potential. Voltage ramps revealed a persistent inward current (PIC) that was attenuated by: flufenamic acid (FFA), a blocker of the Ca(2+)-dependent non-selective cation current ICAN; high intracellular concentrations of BAPTA, which buffers Ca(2+) increases necessary for activation of ICAN; and 9-phenanthrol, a selective blocker of TRPM4 channels (candidate for ICAN). Real-time PCR analysis of mRNA extracted from XII punches and laser-microdissected XII MNs revealed the transcript for TRPM4. MRS2365 potentiated the PIC and this potentiation was blocked by FFA, which also blocked the MRS2365 potentiation of glutamate currents. These data suggest that XII MNs are more sensitive to P2Y1R modulation than phrenic MNs and that the P2Y1R potentiation of inspiratory output occurs in part via potentiation of TRPM4-mediated ICAN, which amplifies inspiratory inputs.


Asunto(s)
Nervio Hipogloso/fisiología , Neuronas Motoras/fisiología , Nervio Frénico/fisiología , Receptores Purinérgicos P2Y1/fisiología , Animales , Animales Recién Nacidos , Tronco Encefálico/fisiología , Técnicas In Vitro , Inhalación/fisiología , Ratas Sprague-Dawley , Ratas Wistar , Médula Espinal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA