Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Exp Med ; 219(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35201268

RESUMEN

Microglia, the main immunocompetent cells of the brain, regulate neuronal function, but their contribution to cerebral blood flow (CBF) regulation has remained elusive. Here, we identify microglia as important modulators of CBF both under physiological conditions and during hypoperfusion. Microglia establish direct, dynamic purinergic contacts with cells in the neurovascular unit that shape CBF in both mice and humans. Surprisingly, the absence of microglia or blockade of microglial P2Y12 receptor (P2Y12R) substantially impairs neurovascular coupling in mice, which is reiterated by chemogenetically induced microglial dysfunction associated with impaired ATP sensitivity. Hypercapnia induces rapid microglial calcium changes, P2Y12R-mediated formation of perivascular phylopodia, and microglial adenosine production, while depletion of microglia reduces brain pH and impairs hypercapnia-induced vasodilation. Microglial actions modulate vascular cyclic GMP levels but are partially independent of nitric oxide. Finally, microglial dysfunction markedly impairs P2Y12R-mediated cerebrovascular adaptation to common carotid artery occlusion resulting in hypoperfusion. Thus, our data reveal a previously unrecognized role for microglia in CBF regulation, with broad implications for common neurological diseases.


Asunto(s)
Circulación Cerebrovascular/fisiología , Microglía/fisiología , Acoplamiento Neurovascular/fisiología , Receptores Purinérgicos/fisiología , Adulto , Anciano , Animales , Encéfalo/fisiología , Señalización del Calcio/fisiología , Enfermedades de las Arterias Carótidas/fisiopatología , Potenciales Evocados/fisiología , Femenino , Humanos , Hipercapnia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Purinérgicos P2Y12/fisiología , Vasodilatación/fisiología , Vibrisas/inervación
2.
Molecules ; 26(20)2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34684726

RESUMEN

Gliomas are the most common malignant brain tumors in adults, characterized by a high proliferation and invasion. The tumor microenvironment is rich in growth-promoting signals and immunomodulatory pathways, which increase the tumor's aggressiveness. In response to hypoxia and glioma therapy, the amounts of adenosine triphosphate (ATP) and adenosine diphosphate (ADP) strongly increase in the extracellular space, and the purinergic signaling is triggered by nucleotides' interaction in P2 receptors. Several cell types are present in the tumor microenvironment and can facilitate tumor growth. In fact, tumor cells can activate platelets by the ADP-P2Y12 engagement, which plays an essential role in the cancer context, protecting tumors from the immune attack and providing molecules that contribute to the growth and maintenance of a rich environment to sustain the protumor cycle. Besides platelets, the P2Y12 receptor is expressed by some tumors, such as renal carcinoma, colon carcinoma, and gliomas, being related to tumor progression. In this context, this review aims to depict the glioma microenvironment, focusing on the relationship between platelets and tumor malignancy.


Asunto(s)
Glioma/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Receptores Purinérgicos P2Y12/fisiología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Plaquetas/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/fisiopatología , Humanos , Receptores Purinérgicos/metabolismo , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
3.
Acta Pharmacol Sin ; 42(3): 414-421, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32555443

RESUMEN

In the past two decades, purinergic signaling has emerged as a key regulator of hematopoiesis in physiological and pathological conditions. ADP receptor P2y12 is a crucial component of this signaling, but whether it is involved in primitive hematopoiesis remains unknown. To elucidate the function of P2y12 and provide new insights for drug development, we established a zebrafish P2y12 mutant by CRISPR/Cas 9-based genetic modification system, and investigated whether P2y12 acted as an important regulator for primitive hematopoiesis. By using mass spectrometry (MS) combined with RNA sequencing, we showed that absence of P2y12 induced excessive erythropoiesis, evidenced by significantly increased expression of mature erythrocytes marker α-globin (Hbae1 and Hbae3), ß-globin (Hbbe1 and Hbbe3). Expression pattern analysis showed that P2y12 was mainly expressed in red blood cells and endothelial cells of early zebrafish embryos. Further studies revealed that primitive erythroid progenitor marker Gata1 was markedly up-regulated. Remarkably, inhibition of Gata1 by injection of Gata1 morpholino could rescue the erythroid abnormality in P2y12 mutants. The present study demonstrates the essential role of purinergic signaling in differentiation of proerythrocytes during primitive hematopoiesis, and provides potential targets for treatment of blood-related disease and drug development.


Asunto(s)
Factor de Transcripción GATA1/antagonistas & inhibidores , Hematopoyesis/fisiología , Receptores Purinérgicos P2Y12/fisiología , Proteínas de Pez Cebra/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Diferenciación Celular/fisiología , Embrión no Mamífero/fisiología , Endotelio Vascular/fisiología , Eritrocitos/fisiología , Femenino , Factor de Transcripción GATA1/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas de Inactivación de Genes , Hematopoyesis/genética , Masculino , Mutación , Receptores Purinérgicos P2Y12/genética , Regulación hacia Arriba/fisiología , Pez Cebra , Proteínas de Pez Cebra/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 40(6): e166-e179, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32349534

RESUMEN

OBJECTIVE: Recent studies suggest that the P2Y12 (P2Y purinoceptor 12) receptor of vascular smooth muscle cells in atherosclerotic plaques aggravates atherosclerosis, and P2Y12 receptor inhibitors such as CDL (clopidogrel) may effectively treat atherosclerosis. It is imperative to identify an effective biomarker for reflecting the P2Y12 receptor expression on vascular smooth muscle cells in plaques. Approach and Results: We found that there was a positive correlation between the level of circulating sLRP1 (soluble low-density lipoprotein receptor-related protein 1) and the number of LRP1+ α-SMA+ (α-smooth muscle actin), P2Y12+, or P2Y12+ LRP1+ cells in plaques from apoE-/- mice fed a high-fat diet. Furthermore, activation of the P2Y12 receptor increased the expression and shedding of LRP1 in vascular smooth muscle cells by inhibiting cAMP (3'-5'-cyclic adenosine monophosphate)/PKA (protein kinase A)/SREBP-2 (sterol regulatory element binding transcription factor 2). Conversely, genetic knockdown or pharmacological inhibition of the P2Y12 receptor had the opposite effects. Additionally, CDL decreased the number of lesional LRP1+ α-SMA+ cells and the levels of circulating sLRP1 by activating cAMP/PKA/SREBP-2 in apoE-/- mice fed a high-fat diet. CONCLUSIONS: Our study suggests that sLRP1 may be a biomarker that reflects the P2Y12 receptor level in plaques and has the potential to be an indicator for administering P2Y12 receptor inhibitors for patients with atherosclerosis.


Asunto(s)
Biomarcadores/análisis , Expresión Génica , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/análisis , Placa Aterosclerótica/metabolismo , Receptores Purinérgicos P2Y12/genética , Actinas/análisis , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Clopidogrel/farmacología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dieta Alta en Grasa , Técnicas de Silenciamiento del Gen , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/química , Músculo Liso Vascular/metabolismo , Placa Aterosclerótica/química , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2Y12/fisiología , Transducción de Señal , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
5.
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
6.
J Neurosci ; 40(7): 1453-1482, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31896673

RESUMEN

During adult hippocampal neurogenesis, most newborn cells undergo apoptosis and are rapidly phagocytosed by resident microglia to prevent the spillover of intracellular contents. Here, we propose that phagocytosis is not merely passive corpse removal but has an active role in maintaining neurogenesis. First, we found that neurogenesis was disrupted in male and female mice chronically deficient for two phagocytosis pathways: the purinergic receptor P2Y12, and the tyrosine kinases of the TAM family Mer tyrosine kinase (MerTK)/Axl. In contrast, neurogenesis was transiently increased in mice in which MerTK expression was conditionally downregulated. Next, we performed a transcriptomic analysis of the changes induced by phagocytosis in microglia in vitro and identified genes involved in metabolism, chromatin remodeling, and neurogenesis-related functions. Finally, we discovered that the secretome of phagocytic microglia limits the production of new neurons both in vivo and in vitro Our data suggest that microglia act as a sensor of local cell death, modulating the balance between proliferation and survival in the neurogenic niche through the phagocytosis secretome, thereby supporting the long-term maintenance of adult hippocampal neurogenesis.SIGNIFICANCE STATEMENT Microglia are the brain professional phagocytes and, in the adult hippocampal neurogenic niche, they remove newborn cells naturally undergoing apoptosis. Here we show that phagocytosis of apoptotic cells triggers a coordinated transcriptional program that alters their secretome, limiting neurogenesis both in vivo and in vitro In addition, chronic phagocytosis disruption in mice deficient for receptors P2Y12 and MerTK/Axl reduces adult hippocampal neurogenesis. In contrast, inducible MerTK downregulation transiently increases neurogenesis, suggesting that microglial phagocytosis provides a negative feedback loop that is necessary for the long-term maintenance of adult hippocampal neurogenesis. Therefore, we speculate that the effects of promoting engulfment/degradation of cell debris may go beyond merely removing corpses to actively promoting regeneration in development, aging, and neurodegenerative diseases.


Asunto(s)
Hipocampo/citología , Neurogénesis/fisiología , Neuronas/citología , Fagocitosis/fisiología , Animales , Apoptosis , Señalización del Calcio , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Medios de Cultivo Condicionados , Retroalimentación Fisiológica , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Hipocampo/crecimiento & desarrollo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores Purinérgicos P2Y12/fisiología , Transcriptoma , Tirosina Quinasa c-Mer/fisiología
7.
Science ; 367(6477): 528-537, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31831638

RESUMEN

Microglia are the main immune cells in the brain and have roles in brain homeostasis and neurological diseases. Mechanisms underlying microglia-neuron communication remain elusive. Here, we identified an interaction site between neuronal cell bodies and microglial processes in mouse and human brain. Somatic microglia-neuron junctions have a specialized nanoarchitecture optimized for purinergic signaling. Activity of neuronal mitochondria was linked with microglial junction formation, which was induced rapidly in response to neuronal activation and blocked by inhibition of P2Y12 receptors. Brain injury-induced changes at somatic junctions triggered P2Y12 receptor-dependent microglial neuroprotection, regulating neuronal calcium load and functional connectivity. Thus, microglial processes at these junctions could potentially monitor and protect neuronal functions.


Asunto(s)
Lesiones Encefálicas/inmunología , Encéfalo/inmunología , Uniones Intercelulares/inmunología , Microglía/inmunología , Neuronas/inmunología , Receptores Purinérgicos P2Y12/fisiología , Animales , Encéfalo/ultraestructura , Lesiones Encefálicas/patología , Calcio , Comunicación Celular/inmunología , Células HEK293 , Humanos , Ratones , Mitocondrias/inmunología , Canales de Potasio Shab/genética , Canales de Potasio Shab/fisiología , Transducción de Señal
8.
JCI Insight ; 4(16)2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31434806

RESUMEN

Abnormalities in purine availability or purinergic receptor density are commonly seen in patients with lower urinary tract symptoms (LUTS), but the underlying mechanisms relating altered receptor function to LUTS are unknown. Here we provide extensive evidence for the reciprocal interplay of multiple receptors responding to ATP, ADP (adenosine diphosphate), and adenosine, agonists that regulate bladder function significantly. ADP stimulated P2Y12 receptors, causing bladder smooth muscle (BSM) contraction, whereas adenosine signaling through potentially newly defined A2b receptors, actively inhibited BSM purinergic contractility. The modulation of adenylyl cyclase-cAMP signaling via A2b and P2Y12 interaction actively regulated bladder contractility by modulating intracellular calcium levels. KO mice lacking the receptors display diametrically opposed bladder phenotypes, with P2Y12-KO mice exhibiting an underactive bladder (UAB) phenotype with increased bladder capacity and reduced voiding frequency, whereas A2b-KO mice have an overactive bladder (OAB), with decreased capacity and increased voiding frequency. The opposing phenotypes in P2Y12-KO and A2b-KO mice not only resulted from dysregulated BSM contractility, but also from abnormal BSM cell growth. Finally, we demonstrate that i.p. administration of drugs targeting P2Y12 or A2b receptor rescues these abnormal phenotypes in both KO mice. These findings strongly indicate that P2Y12 and A2b receptors are attractive therapeutic targets for human patients with LUTS.


Asunto(s)
Receptor de Adenosina A2B/fisiología , Receptores Purinérgicos P2Y12/fisiología , Vejiga Urinaria/fisiología , Animales , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Contracción Muscular , Músculo Liso/fisiología , Embarazo , Transducción de Señal , Enfermedades de la Vejiga Urinaria/metabolismo
9.
Acta Physiol (Oxf) ; 225(2): e13191, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30257062

RESUMEN

AIM: Therapeutic use of lithium in bipolar disorder is limited by the development of nephrogenic diabetes insipidus (NDI). We reported that pharmacological blockade of P2Y12 receptor (R) with clopidogrel or prasugrel significantly ameliorated lithium-induced NDI in rodents. Using mice genetically lacking P2Y12 -R we evaluated whether the observed amelioration is mediated through P2Y12 -R METHODS: P2ry12-/- mouse line (C57/BL6) was rederived from cryopreserved embryos of the knockout (KO) mice generated by Deltagen Inc. Syngeneic wild type (WT) mice obtained by heterozygous crossing were inbred. Groups of adult WT and KO mice were fed lithium-added (40 mmol LiCl/kg food) or regular diet, and euthanized after 2 or 4 weeks. Twenty-four hour urine samples and terminal blood and kidney samples were analyzed. RESULTS: At both time points, lithium-induced polyuria and decrease in aquaporin-2 (AQP2) protein abundance in the kidney medulla were less marked in KO vs WT mice. Immunofluorescence microscopy revealed that lithium-induced alterations in the cellular disposition of AQP2 protein in the medullary collecting ducts of WT mice were blunted in KO mice. Serum lithium, sodium and osmolality were similar in both genotypes after lithium treatment. After 2 weeks, lithium induced marked increases in urinary excretion of Na, K, and arginine vasopressin in WT mice but not in KO mice. CONCLUSION: Taken together, our data show that similar to pharmacological blockade, deletion of P2Y12 -R significantly ameliorates lithium-induced NDI, without reducing serum lithium levels. Hence, targeting P2Y12 -R with currently available drugs in the market offers a novel and safer method for treating NDI.


Asunto(s)
Diabetes Insípida Nefrogénica/inducido químicamente , Litio/toxicidad , Receptores Purinérgicos P2Y12/fisiología , Animales , Acuaporina 2/metabolismo , Arginina Vasopresina/orina , Diabetes Insípida Nefrogénica/genética , Diabetes Insípida Nefrogénica/metabolismo , Diabetes Insípida Nefrogénica/prevención & control , Dinoprostona/orina , Femenino , Litio/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Natriuresis/efectos de los fármacos , Potasio/orina , Receptores Purinérgicos P2Y12/genética
10.
Drug Discov Today ; 24(1): 325-333, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30291899

RESUMEN

The P2Y12 receptor is one of eight known P2Y receptor subtypes, and belongs to the G-protein-coupled receptor (GPCR) family. The P2Y12 receptor is highly expressed on blood platelets and in the brain. Potent, selective, peripherally acting antagonists for the P2Y12 receptor are used clinically as antithrombotic drugs. Several different scaffolds have been identified as P2Y12 receptor antagonists, including irreversibly acting thienotetrahydropyridines (prodrugs), and reversible competitive antagonists, including adenine nucleotide analogs, piperazinyl-glutamate-quinolines, -pyridines, and -pyrimidines, and anthraquinone derivatives. Here, we provide an overview of the different scaffolds that have been developed as P2Y12 receptor antagonists, some of which have become important therapeutics.


Asunto(s)
Fibrinolíticos/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Nucleótidos de Adenina/farmacología , Descubrimiento de Drogas , Humanos , Agregación Plaquetaria , Profármacos/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Quinolinas/farmacología , Quinonas/farmacología , Receptores Purinérgicos P2Y12/química , Receptores Purinérgicos P2Y12/fisiología
11.
Clin Exp Metastasis ; 35(1-2): 25-35, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29322294

RESUMEN

Activated platelets promote the proliferation and metastatic potential of cancer cells. Platelet activation is largely mediated through ADP engagement of purinergic P2Y12 receptors on platelets. We examined the potential of the reversible P2Y12 inhibitor ticagrelor, an agent used clinically to prevent cardiovascular and cerebrovascular events, to reduce tumor growth and metastasis. In vitro, MCF-7, MDA-MB-468, and MDA-MB-231 human mammary carcinoma cells exhibited decreased interaction with platelets treated with ticagrelor compared to untreated platelets. Prevention of tumor cell-platelet interactions through pretreatment of platelets with ticagrelor did not improve natural killer cell-mediated tumor cell killing of K562 myelogenous leukemia target cells. Additionally, ticagrelor had no effect on proliferation of 4T1 mouse mammary carcinoma cells co-cultured with platelets, or on primary 4T1 tumor growth. In an orthotopic 4T1 breast cancer model, ticagrelor (10 mg/kg), but not clopidogrel (10 mg/kg) or saline, resulted in reduced metastasis and improved survival. Ticagrelor treatment was associated with a marked reduction in tumor cell-platelet aggregates in the lungs at 10, 30 and 60 min post-intravenous inoculation. These findings suggest a role for P2Y12-mediated platelet activation in promoting metastasis, and provide support for the use of ticagrelor in the prevention of breast cancer spread.


Asunto(s)
Plaquetas/efectos de los fármacos , Neoplasias de la Mama/patología , Neoplasias Mamarias Experimentales/patología , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/efectos de los fármacos , Ticagrelor/farmacología , Animales , Plaquetas/inmunología , Plaquetas/metabolismo , Neoplasias de la Mama/inmunología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/inmunología , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Selectina-P/metabolismo , Activación Plaquetaria/fisiología , Receptores Purinérgicos P2Y12/fisiología , Tasa de Supervivencia
12.
Neuron ; 97(2): 299-312.e6, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29290552

RESUMEN

Microglia exhibit two modes of motility: they constantly extend and retract their processes to survey the brain, but they also send out targeted processes to envelop sites of tissue damage. We now show that these motility modes differ mechanistically. We identify the two-pore domain channel THIK-1 as the main K+ channel expressed in microglia in situ. THIK-1 is tonically active, and its activity is potentiated by P2Y12 receptors. Inhibiting THIK-1 function pharmacologically or by gene knockout depolarizes microglia, which decreases microglial ramification and thus reduces surveillance, whereas blocking P2Y12 receptors does not affect membrane potential, ramification, or surveillance. In contrast, process outgrowth to damaged tissue requires P2Y12 receptor activation but is unaffected by blocking THIK-1. Block of THIK-1 function also inhibits release of the pro-inflammatory cytokine interleukin-1ß from activated microglia, consistent with K+ loss being needed for inflammasome assembly. Thus, microglial immune surveillance and cytokine release require THIK-1 channel activity.


Asunto(s)
Interleucina-1beta/fisiología , Microglía/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Adenosina Trifosfato/farmacología , Animales , Movimiento Celular , Polaridad Celular , Forma de la Célula , Extensiones de la Superficie Celular/fisiología , Quimiotaxis/fisiología , Inflamasomas/metabolismo , Potenciales de la Membrana , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Potasio/fisiología , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y12/fisiología , Transcriptoma
13.
Transl Res ; 195: 1-12, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29274308

RESUMEN

It is assumed that platelets in diseased conditions share similar properties to platelets in healthy conditions, although this has never been examined in detail for myocardial infarction (MI). We examined platelets from patients with ST-segment elevation myocardial infarction (STEMI) and non-ST-segment elevation myocardial infarction (NSTEMI) compared with platelets from healthy volunteers to evaluate for differences in platelet phenotype and function. Platelet activation was examined and postreceptor signal transduction pathways were assessed. Platelet-derived plasma biomarkers were evaluated by receiver operator characteristic curve analysis. Maximum platelet activation through the thromboxane receptor was greater in STEMI than in NSTEMI but less through protease-activated receptor 1. Extracellular-signal related-kinase 5 activation, which can activate platelets, was increased in platelets from subjects with STEMI and especially in platelets from patients with NSTEMI. Matrix metalloproteinase 9 (MMP9) protein content and enzymatic activity were several-fold greater in platelets with MI than in control. Mean plasma MMP9 concentration in patients with MI distinguished between STEMI and NSTEMI (area under curve [AUC] 75% [confidence interval (CI) 60-91], P = 0.006) which was superior to troponin T (AUC 66% [CI 48-85, P = 0.08), predicting STEMI with 80% sensitivity (95% CI 56-94), 90% specificity (CI 68-99), 70% AUC (CI 54-86, P < 0.0001), and NSTEMI with 50% sensitivity (CI 27-70), 90% specificity (CI 68-99), 70% AUC (CI 54-86, P = 0.03). Platelets from patients with STEMI and NSTEMI show differences in platelet surface receptor activation and postreceptor signal transduction, suggesting the healthy platelet phenotype in which antiplatelet agents are often evaluated in preclinical studies is different from platelets in patients with MI.


Asunto(s)
Plaquetas/fisiología , Infarto del Miocardio/sangre , Infarto del Miocardio con Elevación del ST/sangre , Anciano , Femenino , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Persona de Mediana Edad , Fenotipo , Receptor PAR-1/fisiología , Receptores Purinérgicos P2Y12/fisiología
14.
Neuroscience ; 326: 31-44, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27058149

RESUMEN

It is known that adenosine 5'-triphosphate (ATP) is released along with the neurotransmitter acetylcholine (ACh) from motor nerve terminals. At mammalian neuromuscular junctions (NMJs), we have previously demonstrated that ATP is able to decrease ACh secretion by activation of P2Y receptors coupled to pertussis toxin-sensitive Gi/o protein. In this group, the receptor subtypes activated by adenine nucleotides are P2Y12 and P2Y13. Here, we investigated, by means of pharmacological and immunohistochemical assays, the P2Y receptor subtype that mediates the modulation of spontaneous and evoked ACh release in mouse phrenic nerve-diaphragm preparations. First, we confirmed that the preferential agonist for P2Y12-13 receptors, 2-methylthioadenosine 5'-diphosphate trisodium salt hydrate (2-MeSADP), reduced MEPP frequency without affecting MEPP amplitude as well as the amplitude and quantal content of end-plate potentials (EPPs). The effect on spontaneous secretion disappeared after the application of the selective P2Y12-13 antagonists AR-C69931MX or 2-methylthioadenosine 5'-monophosphate triethylammonium salt hydrate (2-MeSAMP). 2-MeSADP was more potent than ADP and ATP in reducing MEPP frequency. Then we demonstrated that the selective P2Y13 antagonist MRS-2211 completely prevented the inhibitory effect of 2-MeSADP on MEPP frequency and EPP amplitude, whereas the P2Y12 antagonist MRS-2395 failed to do this. The preferential agonist for P2Y13 receptors inosine 5'-diphosphate sodium salt (IDP) reduced spontaneous and evoked ACh secretion and MRS-2211 abolished IDP-mediated modulation. Immunohistochemical studies confirmed the presence of P2Y13 but not P2Y12 receptors at the end-plate region. Disappearance of P2Y13 receptors after denervation suggests the presynaptic localization of the receptors. We conclude that, at motor nerve terminals, the Gi/o protein-coupled P2Y receptors implicated in presynaptic inhibition of spontaneous and evoked ACh release are of the subtype P2Y13. This study provides new insights into the types of purinergic receptors that contribute to the fine-tuning of cholinergic transmission at mammalian neuromuscular junction.


Asunto(s)
Acetilcolina/metabolismo , Potenciales Postsinápticos Miniatura , Unión Neuromuscular/metabolismo , Receptores Purinérgicos P2/fisiología , Adenosina Difosfato/administración & dosificación , Adenosina Difosfato/análogos & derivados , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Animales , Femenino , Masculino , Ratones , Unión Neuromuscular/efectos de los fármacos , Agonistas del Receptor Purinérgico P2/administración & dosificación , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Receptores Purinérgicos P2Y12/fisiología , Tionucleótidos/administración & dosificación
15.
Proc Natl Acad Sci U S A ; 113(4): 1074-9, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26755608

RESUMEN

Microglia are integral functional elements of the central nervous system, but the contribution of these cells to the structural integrity of the neurovascular unit has not hitherto been assessed. We show here that following blood-brain barrier (BBB) breakdown, P2RY12 (purinergic receptor P2Y, G-protein coupled, 12)-mediated chemotaxis of microglia processes is required for the rapid closure of the BBB. Mice treated with the P2RY12 inhibitor clopidogrel, as well as those in which P2RY12 was genetically ablated, exhibited significantly diminished movement of juxtavascular microglial processes and failed to close laser-induced openings of the BBB. Thus, microglial cells play a previously unrecognized protective role in the maintenance of BBB integrity following cerebrovascular damage. Because clopidogrel antagonizes the platelet P2Y12 receptor, it is widely prescribed for patients with coronary artery and cerebrovascular disease. As such, these observations suggest the need for caution in the postincident continuation of P2RY12-targeted platelet inhibition.


Asunto(s)
Barrera Hematoencefálica , Microglía/fisiología , Receptores Purinérgicos P2Y12/fisiología , Animales , Movimiento Celular , Clopidogrel , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Ticlopidina/análogos & derivados , Ticlopidina/farmacología
16.
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
17.
Biochim Biophys Acta ; 1850(12): 2518-29, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26391841

RESUMEN

BACKGROUND: Two major soluble blood platelet activators are thrombin and ADP. Of these two, only thrombin can induce mitochondrial collapse and programmed cell death leading to phosphatidylserine (PS) exposure required for blood clotting reactions acceleration. Thrombin can also greatly potentiate collagen-induced PS exposure. However, ADP acting through the P2Y12 receptor was shown to increase the PS-exposing (PS+) platelets fraction produced by thrombin or thrombin-plus-collagen via an unknown mechanism. METHODS: We developed a comprehensive multicompartmental computational model of platelet PAR1-and-P2Y12 calcium signal transduction that included cytoplasmic signaling, dense tubular system and mitochondria. To test model predictions, flow cytometry experiments with washed, annexin V-labeled platelets were performed. RESULTS: Stimulation of thrombin receptor PAR1 in the model induced cytoplasmic calcium oscillations, calcium uptake by mitochondria, opening of the permeability transition pore and collapse of the mitochondrial membrane potential. ADP stimulation of P2Y12 led to cAMP decrease that, in turn, caused changes in phospholipase C phosphorylation by protein kinase A, increase in cytoplasmic calcium level and, consequently, PS+ platelet formation. ADP addition before stimulation of PAR1 produced much greater increase of the PS+ fraction because cAMP concentration had time to go down prior to calcium oscillations; this prediction was also tested and confirmed experimentally. CONCLUSION: These results suggest a mechanism of ADP-dependent PS exposure regulation and show a likely mode of action that could be important for the PS exposure regulation in thrombi, where ADP is released before thrombin formation.


Asunto(s)
Plaquetas/citología , Receptor PAR-1/metabolismo , Receptores Purinérgicos P2Y12/fisiología , Transducción de Señal , Adenosina Difosfato/metabolismo , Calcio/metabolismo , Señalización del Calcio , Citosol/metabolismo , Humanos
18.
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
20.
J Neurosci ; 35(6): 2417-22, 2015 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-25673836

RESUMEN

Extracellular calcium concentrations in the brain fluctuate during neuronal activities and may affect the behavior of brain cells. Microglia are highly dynamic immune cells of the brain. However, the effects of extracellular calcium concentrations on microglial dynamics have not been investigated. Here, we addressed this question in mouse brain slices and in vivo using two-photon microscopy. We serendipitously found that extracellular calcium reduction induced microglial processes to converge at distinct sites, a phenomenon we termed microglial process convergence (MPCs). Our studies revealed that MPCs target neuronal dendrites independent of neuronal action potential firing and is mediated by ATP release and microglial P2Y12 receptors. These results indicate that microglia monitor and interact with neurons during conditions of cerebral calcium reduction in the normal and diseased brain.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/fisiología , Dendritas/fisiología , Microglía/fisiología , Neuronas/fisiología , Animales , Espacio Extracelular/fisiología , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Purinas/metabolismo , Receptores Purinérgicos P2Y12/genética , Receptores Purinérgicos P2Y12/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...