Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Purinergic Signal ; 17(4): 681-691, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34351588

RESUMEN

The ability of cardiac adipose-derived stem cells (cADSC) to differentiate into multiple cell types has opened new perspectives in cardiac cell-based regenerative therapies. P2Y nucleotide receptors have already been described as regulators of adipogenic differentiation of cADSC and bone marrow-derived stem cells. In this study, we defined UTP as a regulator of cADSC endothelial differentiation. A daily UTP stimulation of cADSC during endothelial predifferentiation increased their capacity to form an endothelial network in matrigel. Additionally, pro-angiogenic UTP target genes such as epiregulin and hyaluronan synthase-1 were identified in predifferentiated cADSC by RNA sequencing experiments. Their regulation by UTP was confirmed by qPCR and ELISA experiments. We then evaluated the capacity of UTP-treated predifferentiated cADSC to increase post-ischemic revascularization in mice subjected to left anterior descending artery ligation. Predifferentiated cADSC treated or not with UTP were injected in the periphery of the infarcted zone, 3 days after ligation. We observed a significant increase of capillary density 14 and 30 days after UTP-treated predifferentiated cADSC injection, correlated with a reduction of cardiac fibrosis. This revascularization increase was not observed after injection of UTP-treated cADSC deficient for UTP and ATP nucleotide receptor P2Y2. The present study highlights the P2Y2 receptor as a regulator of cADSC endothelial differentiation and as a potential target for the therapeutic use of cADSC in post-ischemic heart revascularization.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Multipotentes/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Uridina Trifosfato/farmacología , Animales , Epirregulina/genética , Epirregulina/metabolismo , Ratones , Ratones Noqueados , Células Madre Multipotentes/metabolismo , Receptores Purinérgicos P2Y2/genética , Receptores Purinérgicos P2Y2/metabolismo
2.
J Biol Chem ; 291(30): 15841-52, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27231349

RESUMEN

The study of the mechanisms leading to cardiac hypertrophy is essential to better understand cardiac development and regeneration. Pathological conditions such as ischemia or pressure overload can induce a release of extracellular nucleotides within the heart. We recently investigated the potential role of nucleotide P2Y receptors in cardiac development. We showed that adult P2Y4-null mice displayed microcardia resulting from defective cardiac angiogenesis. Here we show that loss of another P2Y subtype called P2Y6, a UDP receptor, was associated with a macrocardia phenotype and amplified pathological cardiac hypertrophy. Cardiomyocyte proliferation and size were increased in vivo in hearts of P2Y6-null neonates, resulting in enhanced postnatal heart growth. We then observed that loss of P2Y6 receptor enhanced pathological cardiac hypertrophy induced after isoproterenol injection. We identified an inhibitory effect of UDP on in vitro isoproterenol-induced cardiomyocyte hyperplasia and hypertrophy. The present study identifies mouse P2Y6 receptor as a regulator of cardiac development and cardiomyocyte function. P2Y6 receptor could constitute a therapeutic target to regulate cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/patología , Hiperplasia , Isoproterenol/efectos adversos , Isoproterenol/farmacología , Masculino , Ratones , Ratones Noqueados , Isquemia Miocárdica/inducido químicamente , Isquemia Miocárdica/genética , Isquemia Miocárdica/patología , Miocitos Cardíacos/patología , Receptores Purinérgicos P2/genética
3.
J Immunol ; 194(4): 1874-81, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595790

RESUMEN

Nucleotides are released in the heart under pathological conditions, but little is known about their contribution to cardiac inflammation. The present study defines the P2Y4 nucleotide receptor, expressed on cardiac microvascular endothelial cells and involved in postnatal heart development, as an important regulator of the inflammatory response to cardiac ischemia. P2Y4-null mice displayed smaller infarcts in the left descending artery ligation model, as well as reduced neutrophil infiltration and fibrosis. Gene profiling identified inter alia endothelin-1 (ET-1) as one of the target genes of P2Y4 in ischemic heart. The reduced level of ET-1 was correlated with reduction of microvascular hyperpermeability, neutrophil infiltration, and endothelial adhesion molecule expression, and it could be explained by the decreased number of endothelial cells in P2Y4-null mice. Expression analysis of metalloproteinases and their tissue inhibitors in ischemic heart revealed reduced expression of matrix metalloproteinase (MMP)-9, reported to be potentially regulated by ET-1, and MMP-8, considered as neutrophil collagenase, as well as reduction of tissue inhibitor of MMP-1 and tissue inhibitor of MMP-4 in P2Y4-null mice. Reduction of cardiac permeability and neutrophil infiltration was also observed in P2Y4-null mice in LPS-induced inflammation model. Protection against infarction resulting from loss of P2Y4 brings new therapeutic perspectives for cardiac ischemia and remodeling.


Asunto(s)
Endotelina-1/biosíntesis , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Receptores Purinérgicos P2/deficiencia , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunohistoquímica , Ratones , Ratones Noqueados , Infarto del Miocardio/fisiopatología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
4.
Crit Care Med ; 41(12): e411-22, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23963133

RESUMEN

OBJECTIVE: As adenosine monophosphate (AMP)-activated protein kinase both controls cytoskeleton organization in endothelial cells and exerts anti-inflammatory effects, we here postulated that it could influence vascular permeability and inflammation, thereby counteracting cardiac wall edema during sepsis. DESIGN: Controlled animal study. SETTINGS: University research laboratory. SUBJECTS: C57BL/6J, α1AMPK, and α1AMPK mice. INTERVENTION: Sepsis was triggered in vivo using a sublethal injection of lipopolysaccharide (O55B5, 10 mg/kg), inducing systolic left ventricular dysfunction. Left ventricular function, edema, vascular permeability, and inflammation were assessed in vivo in both wild-type mice (α1AMPK) and α1AMP-activated protein kinase-deficient mice (α1AMPK). The 5-aminoimidazole-4-carboxamide riboside served to study the impact of AMP-activated protein kinase activation on vascular permeability in vivo. The integrity of endothelial cell monolayers was also examined in vitro after lipopolysaccharide challenge in the presence of aminoimidazole-4-carboxamide riboside and/or after α1AMP-activated protein kinase silencing. MEASUREMENTS AND MAIN RESULTS: α1AMP-activated protein kinase deficiency dramatically impaired tolerance to lipopolysaccharide challenge. Indeed, α1AMPK exhibited heightened cardiac vascular permeability after lipopolysaccharide challenge compared with α1AMPK. Consequently, an increase in left ventricular mass corresponding to exaggerated wall edema occurred in α1AMPK, without any further decrease in systolic function. Mechanistically, the lipopolysaccharide-induced α1AMPK cardiac phenotype could not be attributed to major changes in the systemic inflammatory response but was due to an increased disruption of interendothelial tight junctions. Accordingly, AMP-activated protein kinase activation by aminoimidazole-4-carboxamide riboside counteracted lipopolysaccharide-induced hyperpermeability in wild-type mice in vivo as well as in endothelial cells in vitro. This effect was associated with a potent protection of zonula occludens-1 linear border pattern in endothelial cells. CONCLUSIONS: Our results demonstrate for the first time the involvement of a signaling pathway in the control of left ventricular wall edema during sepsis. AMP-activated protein kinase exerts a protective action through the preservation of interendothelial tight junctions. Interestingly, exaggerated left ventricular wall edema was not coupled with aggravated systolic dysfunction. However, it could contribute to diastolic dysfunction in patients with sepsis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Permeabilidad Capilar , Edema/etiología , Endotoxemia/complicaciones , Endotoxemia/enzimología , Cardiopatías/etiología , Inflamación/etiología , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Colorantes/farmacocinética , Citocinas/sangre , Ecocardiografía , Edema/diagnóstico , Edema/fisiopatología , Células Endoteliales/efectos de los fármacos , Endotoxemia/inducido químicamente , Azul de Evans/farmacocinética , Silenciador del Gen , Cardiopatías/diagnóstico , Cardiopatías/fisiopatología , Ventrículos Cardíacos/fisiopatología , Humanos , Inflamación/sangre , Lipopolisacáridos/farmacología , Pulmón/enzimología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/metabolismo , Ribonucleósidos/farmacología , Uniones Estrechas/efectos de los fármacos
5.
Angiogenesis ; 15(3): 349-60, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22437266

RESUMEN

Communication between endothelial cells and cardiomyocytes is critical for cardiac development and regeneration. However the mechanisms involved in these endothelial-cardiomyocyte interactions remain poorly understood. Nucleotides are released within the heart, especially under ischemia or pressure overload. The function of P2Y nucleotide receptors in cardiac development has never been investigated. Here we show that adult P2Y(4)-null mice display microcardia. P2Y(4) nucleotide receptor is expressed in cardiac endothelial cells but not in cardiomyocytes. Loss of P2Y(4) in cardiac endothelial cells strongly inhibits their growth, migration and PDGF-B secretion in response to UTP. Proliferation of microvessels and cardiomyocytes is reduced in P2Y(4)-null hearts early after birth, resulting in reduced heart growth. Our study uncovers mouse P2Y(4) receptor as an essential regulator of cardiac endothelial cell function, and illustrates the involvement of endothelial-cardiomyocyte interactions in post-natal heart development. We also detected P2Y(4) expression in human cardiac microvessels. P2Y(4) receptor could constitute a therapeutic target to regulate cardiac remodelling and post-ischemic revascularisation.


Asunto(s)
Corazón/crecimiento & desarrollo , Receptores Purinérgicos P2/fisiología , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Neovascularización Fisiológica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Am J Physiol Heart Circ Physiol ; 303(7): H835-43, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22865387

RESUMEN

Nucleotides released within the heart under pathological conditions can be involved in cardioprotection or cardiac fibrosis through the activation purinergic P2Y(2) and P2Y(6) receptors, respectively. We previously demonstrated that adult P2Y(4)-null mice display a microcardia phenotype related to a cardiac angiogenic defect. To evaluate the functional consequences of this defect, we performed here a combination of cardiac monitoring and exercise tests. We investigated the exercise capacity of P2Y(4) wild-type and P2Y(4)-null mice in forced swimming and running tests. Analysis of their stress, locomotion, and resignation was realized in open field, black and white box, and tail suspension experiments. Exercise-induced cardiac hypertrophy was evaluated after repeated and prolonged exercise in P2Y(4) wild-type and P2Y(4)-null hearts. We showed that P2Y(4)-null mice have a lower exercise capacity in both swimming and treadmill tests. This was not related to decreased motivation or increased stress, since open field, white and black box, and mouse tail suspension tests gave comparable results in P2Y(4) wild-type and P2Y(4)-null mice. Heart rate and blood pressure rose normally in P2Y(4)-null swimming mice equipped with a telemetric implant. On the contrary, we observed a delayed recovery of postexercise blood pressure after exercise in P2Y(4)-null mice. The heart rate increment in response to catecholamines was also similar in P2Y(4) wild-type and P2Y(4)-null implanted mice, which is consistent with a similar level of cardiac ß-receptor expression. Interestingly, the heart of P2Y(4)-null mice displayed a reduced sympathetic innervation associated with a decreased norepinephrine level. We also demonstrated that exercise-induced cardiac hypertrophy was lower in P2Y(4)-null mice after repeated and prolonged exercise. This was associated with a lower increase in cardiomyocyte size and microvessel density. In conclusion, besides its role in cardiac development, P2Y(4) receptor could constitute an important regulator of acute and chronic response to exercise.


Asunto(s)
Cardiomegalia Inducida por el Ejercicio/genética , Cardiomegalia/prevención & control , Tolerancia al Ejercicio/genética , Eliminación de Gen , Corazón/fisiopatología , Miocardio/metabolismo , Receptores Purinérgicos P2/deficiencia , Natación , Fibras Adrenérgicas/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Conducta Animal , Presión Sanguínea/genética , Monitoreo Ambulatorio de la Presión Arterial , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Catecolaminas/metabolismo , Modelos Animales de Enfermedad , Dobutamina/farmacología , Prueba de Esfuerzo , Tolerancia al Ejercicio/efectos de los fármacos , Genotipo , Corazón/inervación , Frecuencia Cardíaca/genética , Hipotermia/genética , Hipotermia/metabolismo , Hipotermia/fisiopatología , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Fenotipo , Receptores Adrenérgicos beta/efectos de los fármacos , Receptores Adrenérgicos beta/metabolismo , Receptores Purinérgicos P2/genética , Recuperación de la Función , Telemetría , Factores de Tiempo
7.
Blood ; 116(17): 3219-26, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20651071

RESUMEN

ATP, which has an important proinflammatory action as danger signal, induces the semimaturation of dendritic cells (DCs) that can be associated with immune tolerance. We identified epidermal growth factor receptor ligands as target genes of ATPγS, a slowly hydrolyzed ATP derivative, by a gene profiling approach in DCs. Amphiregulin was the most highly up-regulated gene in response to ATPγS. Human monocyte-derived DCs and mouse bone marrow-derived DCs released amphiregulin (AREG) after purinergic receptor activation, with a contribution of P2Y(11) and A(2B) receptor, respectively. Supernatants of LPS+ATPγS-stimulated DCs induced smooth muscle cell and Lewis Lung Carcinoma (LLC) cell growth in vitro. The coinjection of LPS+ATPγS-stimulated DCs or their supernatants with LLC cells increased tumor weight in mice compared with LPS-treated DCs. The preincubation of LPS+ATPγS-treated DC supernatants with an anti-AREG blocking antibody inhibited their positive effect on smooth muscle cell density and tumor growth. The present study demonstrates for the first time that DCs can be a source of AREG. ATP released from tumor cells might exert a tumorigenic action by stimulating the secretion of AREG from DCs. Antagonists of purinergic receptors expressed on DCs and anti-AREG blocking antibodies could have a therapeutic potential as antitumor agents.


Asunto(s)
Adenosina Trifosfato/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Células Dendríticas/inmunología , Células Dendríticas/patología , Glicoproteínas/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Adenosina Trifosfato/análogos & derivados , Anfirregulina , Animales , Células de la Médula Ósea/citología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Proliferación Celular , Células Dendríticas/metabolismo , Familia de Proteínas EGF , Factor de Crecimiento Epidérmico/genética , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Miocitos del Músculo Liso/citología , Regulación hacia Arriba
8.
J Immunol ; 185(6): 3702-7, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20720203

RESUMEN

ATP has been defined as a key mediator of asthma. In this study, we evaluated lung inflammation in mice deficient for the P2Y(2) purinergic receptor. We observed that eosinophil accumulation, a distinctive feature of lung allergic inflammation, was defective in OVA-treated P2Y(2)-deficient mice compared with OVA-treated wild type animals. Interestingly, the upregulation of VCAM-1 was lower on lung endothelial cells of OVA-treated P2Y(2)(-/-) mice compared with OVA-treated wild type animals. Adhesion assays demonstrated that the action of UTP on leukocyte adhesion through the regulation of endothelial VCAM-1 was abolished in P2Y(2)-deficient lung endothelial cells. Additionally, the level of soluble VCAM-1, reported as an inducer of eosinophil chemotaxis, was strongly reduced in the bronchoalveolar lavage fluid (BALF) of P2Y(2)-deficient mice. In contrast, we observed comparable infiltration of macrophages and neutrophils in the BALF of LPS-aerosolized P2Y(2)(+/+) and P2Y(2)(-/-) mice. This difference could be related to the much lower level of ATP in the BALF of LPS-treated mice compared with OVA-treated mice. Our data define P2Y(2) as a regulator of membrane and soluble forms of VCAM-1 and eosinophil accumulation during lung inflammation.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Membrana Celular/inmunología , Movimiento Celular/inmunología , Eosinófilos/inmunología , Pulmón/inmunología , Receptores Purinérgicos P2Y2/fisiología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/patología , Modelos Animales de Enfermedad , Eosinófilos/patología , Lipopolisacáridos/toxicidad , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/administración & dosificación , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/sangre , Isoformas de Proteínas/fisiología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología , Receptores Purinérgicos P2Y2/biosíntesis , Receptores Purinérgicos P2Y2/deficiencia , Solubilidad , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/sangre
9.
Front Pharmacol ; 13: 906173, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784739

RESUMEN

Adipose tissue is a source of stem cells with a high potential of differentiation for cell-based regenerative therapies. We previously identified mouse P2Y2, an ATP and UTP nucleotide receptor, as a regulator of adipogenic and endothelial differentiation of cardiac adipose-derived stem cells (cADSC). We investigated here the potential involvement of P2Y2 receptor in the cardioprotective action of undifferentiated cADSC transplantation in mouse ischemic heart. Transplantation of cADSC was realized in the periphery of the infarcted zone of ischemic heart, 3 days after left anterior descending artery ligation. A strong reduction of collagen stained area was observed 14 days after cADSC injection, compared to PBS injection. Interestingly, loss of P2Y2 expression totally inhibits the ability of transplanted cADSC to reduce cardiac fibrosis. A detailed gene ontology enrichment analysis was realized by comparing RNA-sequencing data obtained for UTP-treated wild type cASDC and UTP-treated P2Y2-null cASDC. We identified UTP target genes linked to extracellular matrix organization such as matrix metalloproteinases and various collagen types, UTP target genes related to macrophage chemotaxis and differentiation into pro-fibrotic foam cells, and a significant number of UTP target genes linked to angiogenesis regulation. More particularly, we showed that UTP regulated the secretion of CCL5, CXCL5, and CCL12 chemokines and serum amyloid apolipoprotein 3, in the supernatants of UTP-treated cADSC. Interestingly, CCL5 is reported as a key factor in post-infarction heart failure and in the reparative and angiogenic action of transplanted ADSC on ischemic tissue. We investigated then if a UTP-pretreatment of cADSC amplifies their effect on cardiac revascularization in mouse ischemic heart. Transplantation of cADSC was able to increase peri-infarct capillary density, 14 days after their injection. This beneficial effect on cardiac revascularization was enhanced by a UTP-pretreatment of cADSC before their transplantation, and not observed using P2Y2-null cADSC. Our data support that the efficacy of transplanted cADSC can be regulated by the release of inflammatory mediators such as extracellular nucleotides in the ischemic site. The present study highlights the P2Y2 receptor as a regulator of cADSC cardioprotective action, and as a potential target for the therapeutic use of undifferentiated cADSC in post-ischemic cardiac ischemia.

10.
Front Immunol ; 13: 1061958, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36389796

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2022.1006934.].

11.
Front Immunol ; 13: 1006934, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36248854

RESUMEN

A better understanding of the immune function of pericardial adipose tissue is essential to adapt treatments after myocardial infarction. We showed previously that inactivation of mouse P2Y4 nucleotide receptor induces adiponectin overexpression and protection against myocardial infarction. We investigated here the inflammatory state of pericardial adipose tissue in ischemic P2Y4-deficient mice. We demonstrated that P2Y4-deficient mice displayed adipocyte beiging with increased PD-L1 expression and a higher number of regulatory leukocytes in their pericardial adipose tissue after left anterior descending artery ligation, compared to wild type mice. Effectively, a higher level of anti-inflammatory M2c macrophages and regulatory T cells was observed in pericardial adipose tissue of P2Y4 KO mice and correlated with reduced post-ischemic expansion of fat-associated lymphoid clusters. Interestingly, the anti-inflammatory effects observed in P2Y4 KO mice, were no more observed in P2Y4/adiponectin double KO ischemic mice. Finally, the reduction of T cell infiltration and cardiac fibrosis observed in P2Y4-deficient heart was lost after injection of anti-PD-L1 blocking antibody in ischemic mice. The present study defines P2Y4 as a regulator of PD-L1 and adiponectin, and as a potential target for anti-inflammatory therapies to improve myocardial infarction outcome. The combined effect of P2Y4 loss on adipocyte beiging and regulatory leukocyte increase highlights this nucleotide receptor as an important player in post-ischemic cardiac response.


Asunto(s)
Adiponectina , Antígeno B7-H1/metabolismo , Infarto del Miocardio , Receptores Purinérgicos P2/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , Nucleótidos
12.
Front Pharmacol ; 13: 1049696, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36532779

RESUMEN

Human P2Y4 is a UTP receptor, while in mice it is activated by both ATP and UTP. P2Y4 knockout (KO) in mice protects against myocardial infarction and is characterized by increased adiponectin secretion by adipocytes, and decreased cardiac inflammation and permeability under ischemic conditions. The relevance of these data has, however, not been explored to date in humans. In a population study comprising 50 patients with coronary artery disease (CAD) and 50 age-matched control individuals, we analyzed P2RY4 mutations and their potential association with CAD severity and fasting plasma parameters. Among the mutations identified, we focused our attention on a coding region polymorphism (rs3745601) that results in replacement of the asparagine at residue 178 with threonine (N178T) located in the second extracellular loop of the P2Y4 receptor. The N178T variant is a loss-of-function mutation of the human P2Y4 receptor and is encountered less frequently in coronary patients than in control individuals. In coronary patients, carriers of the N178T variant had significantly reduced jeopardy and Gensini cardiac severity scores, as well as lower resting heart rates and plasma levels of N-terminal pro-brain natriuretic peptide (NT-proBNP). Regarding fasting plasma parameters, the N178T variant was associated with a lower concentration of glucose. Accordingly, P2Y4 KO mice had significantly improved glucose tolerance and insulin sensitivity compared with their WT littermate controls. The improvement of insulin sensitivity resulting from lack of the P2Y4 receptor was no longer observed in the absence of adiponectin. The present study identifies a frequent loss-of-function P2Y4 variant associated with less severe coronary artery atherosclerosis and lower fasting plasma glucose in coronary patients. The role of the P2Y4 receptor in glucose homeostasis was confirmed in mouse. P2Y4 antagonists could thus have therapeutic applications in the treatment of myocardial infarction and type 2 diabetes.

13.
Nat Med ; 9(7): 936-43, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12796773

RESUMEN

Therapeutic angiogenesis is likely to require the administration of factors that complement each other. Activation of the receptor tyrosine kinase (RTK) Flk1 by vascular endothelial growth factor (VEGF) is crucial, but molecular interactions of other factors with VEGF and Flk1 have been studied to a limited extent. Here we report that placental growth factor (PGF, also known as PlGF) regulates inter- and intramolecular cross talk between the VEGF RTKs Flt1 and Flk1. Activation of Flt1 by PGF resulted in intermolecular transphosphorylation of Flk1, thereby amplifying VEGF-driven angiogenesis through Flk1. Even though VEGF and PGF both bind Flt1, PGF uniquely stimulated the phosphorylation of specific Flt1 tyrosine residues and the expression of distinct downstream target genes. Furthermore, the VEGF/PGF heterodimer activated intramolecular VEGF receptor cross talk through formation of Flk1/Flt1 heterodimers. The inter- and intramolecular VEGF receptor cross talk is likely to have therapeutic implications, as treatment with VEGF/PGF heterodimer or a combination of VEGF plus PGF increased ischemic myocardial angiogenesis in a mouse model that was refractory to VEGF alone.


Asunto(s)
Proteínas Gestacionales/metabolismo , Receptor Cross-Talk/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Dimerización , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Activación Enzimática , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/metabolismo , Linfocinas/farmacología , Ratones , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Factor de Crecimiento Placentario , Proteínas Gestacionales/farmacología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteínas Virales/metabolismo , Proteínas Virales/farmacología
14.
Biochem Pharmacol ; 187: 114347, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33232731

RESUMEN

The family of P2Y nucleotide receptors is composed of eight members differentiated by their pharmacology and their coupling to specific G-proteins and transduction mechanisms. The laboratory studying these nucleotide receptors at IRIBHM institute (Free University of Brussels) has participated actively in their cloning. We used classical cloning by homology strategies relying on polymerase chain reactions with degenerate primers or on DNA libraries screening with P2Y receptors-related primers or probes, respectively. We identified and characterised four of the eight human P2Y receptors cloned so far: P2Y4, P2Y6, P2Y11 and P2Y13 receptors. These human receptors displayed specific features in terms of pharmacology such as affinity for pyrimidine nucleotides for P2Y4 and P2Y6 receptors and differential G-protein coupling. Their specific and restricted tissue distribution compared to ubiquitous P2Y1 and P2Y2 receptors led us to study their physiological role in chosen cell systems or using mice deficient for these P2Y subtypes. These studies revealed over the years that the P2Y11 receptor was able to confer tolerogenic and tumorigenic properties to human dendritic cells and that P2Y4 and P2Y6 receptors were involved in mouse heart post-natal development and cardioprotection. P2Y receptors and their identified target genes could constitute therapeutic targets to regulate cardiac hypertrophy and regeneration. The multiple roles of P2Y receptors identified in the ischemic heart and cardiac adipose tissue could have multiple innovative clinical applications and present a major interest in the field of cardiovascular diseases. P2Y receptors can induce cardioprotection by the regulation of cardiac inflammation and the modulation of the volume and composition of cardiac adipose tissue. These findings might lead to the pre-clinical validation of P2Y receptors as new targets for the treatment of myocardial ischemia.


Asunto(s)
Clonación Molecular/métodos , Receptores Purinérgicos P2/fisiología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Cardiopatías/tratamiento farmacológico , Cardiopatías/fisiopatología , Humanos , Agonistas del Receptor Purinérgico P2/administración & dosificación , Antagonistas del Receptor Purinérgico P2/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
J Pharmacol Exp Ther ; 332(1): 238-47, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19815812

RESUMEN

The G protein-coupled P2Y(11) receptor is involved in immune system modulation. In-depth physiological evaluation is hampered, however, by a lack of selective and potent ligands. By screening a library of sulfonic and phosphonic acid derivatives at P2Y(11) receptors recombinantly expressed in human 1321N1 astrocytoma cells (calcium and cAMP assays), the selective non-nucleotide P2Y(11) agonist NF546 [4,4'-(carbonylbis(imino-3,1-phenylene-carbonylimino-3,1-(4-methyl-phenylene)carbonylimino))-bis(1,3-xylene-alpha,alpha'-diphosphonic acid) tetrasodium salt] was identified. NF546 had a pEC(50) of 6.27 and is relatively selective for P2Y(11) over P2Y(1), P2Y(2), P2Y(4), P2Y(6), P2Y(12), P2X(1), P2X(2), and P2X(2)-X(3). Adenosine-5'-O-(3-thio)triphosphate (ATPgammaS), a nonhydrolyzable analog of the physiological P2Y(11) agonist ATP, and NF546 use a common binding site as suggested by molecular modeling studies and their competitive behavior toward the nanomolar potency antagonist NF340 [4,4'-(carbonylbis(imino-3,1-(4-methyl-phenylene)carbonylimino))bis(naphthalene-2,6-disulfonic acid) tetrasodium salt] in Schild analysis. The pA(2) of NF340 was 8.02 against ATPgammaS and 8.04 against NF546 (calcium assays). NF546 was further tested for P2Y(11)-mediated effects in monocyte-derived dendritic cells. Similarly to ATPgammaS, NF546 led to thrombospondin-1 secretion and inhibition of lipopolysaccharide-stimulated interleukin-12 release, whereas NF340 inhibited these effects. Further, for the first time, it was shown that ATPgammaS or NF546 stimulation promotes interleukin 8 (IL-8) release from dendritic cells, which could be inhibited by NF340. In conclusion, we have described the first selective, non-nucleotide agonist NF546 for P2Y(11) receptors in both recombinant and physiological expression systems and could show a P2Y(11)-stimulated IL-8 release, further supporting the immunomodulatory role of P2Y(11) receptors.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Difosfonatos/farmacología , Interleucina-8/metabolismo , Naftalenosulfonatos/farmacología , Agonistas del Receptor Purinérgico P2 , Calcio/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Clonación Molecular , AMP Cíclico/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Unión Proteica , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X , Proteínas Recombinantes , Transfección
16.
Stem Cells Dev ; 29(2): 100-109, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31829837

RESUMEN

The formation of pericardial adipose tissue (PAT) and its regulatory function in cardiac inflammation are not well understood. We investigated the potential role of the ubiquitous ATP/UTP nucleotide receptor P2Y2 in the PAT by using P2Y2-null mice. We observed that P2Y2-null mice displayed a lower mass of PAT and a reduced density of its fat-associated lymphoid clusters (FALCs) and, more particularly, B cells. Loss of P2Y2 receptor in pericardial preadipocytes decreased their adipogenic differentiation and maturation abilities in vitro. Gene profiling identified P2Y2 target genes in PAT linked to immunomodulation. These data led to the identification of an increase of M2c anti-inflammatory macrophages correlated with increased apoptosis of B lymphocytes in P2Y2-null pericardial fat. In addition, follicular helper T cells, which contribute to B cell expansion in germinal centers, were dramatically decreased. The effect of P2Y2 loss was also investigated after ischemia-mediated expansion of FALCs in a model of myocardial infarct. Loss of P2Y2 led to reduced expansion of B and neutrophil populations in these clusters, whereas density of M2c anti-inflammatory macrophages was increased. Our study defines the P2Y2 nucleotide receptor as a regulator of the formation and inflammatory status of pericardial fat. The P2Y2 receptor could represent a therapeutic target in the regulation of PAT function before and during cardiac ischemia.


Asunto(s)
Tejido Adiposo/metabolismo , Grasas/metabolismo , Linfocitos/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Células Madre/metabolismo , Adipogénesis/genética , Tejido Adiposo/citología , Animales , Linfocitos B/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pericardio/citología , Pericardio/metabolismo , Receptores Purinérgicos P2Y2/genética
17.
Eur J Pharmacol ; 566(1-3): 167-71, 2007 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-17481603

RESUMEN

Extracellular nucleotides regulate ion transport, mucociliary clearance as well as inflammatory properties of the airway epithelium by acting on P2 receptors. Cyclooxygenase-2 (COX-2) is a key enzyme involved in the synthesis of prostaglandins during inflammation. In this study, using calcium imaging, DNA microarray experiments, real-time Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR) and prostaglandin E2 (PGE2) measurement, we show for the first time that ATP, UTP or INS365 compound (P2Y2 receptor agonists) up-regulate COX-2 expression by approximately 3-fold and enhance the release of PGE2 in human A549 airway epithelial cells. Our data suggest that P2Y receptors may represent putative targets in airway inflammatory diseases.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Dinoprostona/biosíntesis , Células Epiteliales/efectos de los fármacos , Nucleótidos/farmacología , Calcio/metabolismo , Línea Celular , Ciclooxigenasa 2/genética , Células Epiteliales/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Alveolos Pulmonares/citología , Agonistas del Receptor Purinérgico P2 , ARN Mensajero/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y2 , Regulación hacia Arriba
18.
Stem Cells Dev ; 26(5): 363-373, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27855539

RESUMEN

Cardiac adipose tissue-derived stem cells (cASCs) have the ability to differentiate into multiple cell lineages giving them a high potential for use in regenerative medicine. Cardiac fat tissue still raises many unsolved questions related to its formation and features. P2Y nucleotide receptors have already been described as regulators of differentiation of bone-marrow derived stem cells, but remain poorly investigated in cASCs. We defined, in this study, the P2Y4 nucleotide receptor as a negative regulator of cardiac fat formation and cASC adipogenic differentiation. Higher expression of P2Y4 receptor in cardiac fat tissue was observed compared to other adipose tissues. P2Y4-null mice displayed a higher mass of cardiac adipose tissue specifically. We therefore examined the role of P2Y4 receptor in cASC adipogenic differentiation. An inhibitory effect of uridine 5'-triphosphate (UTP), ligand of P2Y4, was observed on the maturation state of differentiated cASCs, and on the expression of adipogenesis-linked genes and adiponectin, a cardioprotective adipokine. Higher adiponectin secretion by P2Y4-null adipocytes could be linked with cardioprotection previously observed in the heart of P2Y4-null ischemic mice. We realized here left anterior descending artery ligation on simple and double-knockout mice for P2Y4 and adiponectin. No cardioprotective effect of P2Y4 loss was observed in the absence of adiponectin secretion. In addition, P2Y4 loss was correlated with higher expression of UCP-1 (uncoupling protein-1) and CD137, two markers of brown/beige cardiac adipocytes. Our data highlight the P2Y4 receptor as an inhibitor of cardiac fat formation and cASC adipogenic differentiation, and as a potential therapeutic target in the regulation of cardioprotective function of cardiac fat.


Asunto(s)
Tejido Adiposo/citología , Adiposidad , Diferenciación Celular , Miocardio/citología , Receptores Purinérgicos P2/metabolismo , Células Madre/citología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Adiponectina/metabolismo , Adiposidad/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Cardiotónicos/farmacología , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Femenino , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Uridina Trifosfato/farmacología
19.
J Invest Dermatol ; 126(5): 943-4, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16619009

RESUMEN

In this issue, Seiffert et al. show that adenosine triphosphate enhances the production of inflammatory mediators by human dermal endothelial cells. However, a growing literature shows that adenosine triphosphate exerts anti-inflammatory effects, partly by inducing a semi-maturation of dendritic cells associated with immune tolerance. These discrepancies can be reconciled knowing that extracellular nucleotides constitute danger signals that are released in response to both external aggression (chemical, microbial) and excess inflammation.


Asunto(s)
Adenosina Trifosfato/fisiología , Células Endoteliales/inmunología , Mediadores de Inflamación/metabolismo , Inflamación/etiología , Receptores Purinérgicos/fisiología , Células Dendríticas/fisiología , Células Endoteliales/metabolismo , Humanos , Inflamación/prevención & control , Transducción de Señal
20.
FEBS Lett ; 580(3): 747-54, 2006 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-16413542

RESUMEN

Extracellular ATP is known to affect the maturation of monocyte-derived dendritic cells mainly by regulation of cytokines and costimulatory molecules. The present study describes the inhibition of MCP-1 (CCL2) and MIP-1alpha (CCL3) release by human monocyte-derived dendritic cells in response to adenine nucleotides. Our pharmacological data support the involvement of P2Y11 and P2Y1 purinergic receptors in the downregulation of these major monocyte recruiters. Migration assays have demonstrated that supernatants of dendritic cells treated with adenine nucleotides or anti-MCP-1/MIP-1alpha blocking antibodies display a strongly reduced capacity to attract monocytes and immature dendritic cells.


Asunto(s)
Nucleótidos de Adenina/farmacología , Quimiocina CCL2/inmunología , Quimiotaxis/efectos de los fármacos , Células Dendríticas/inmunología , Proteínas Inflamatorias de Macrófagos/inmunología , Monocitos/inmunología , Nucleótidos de Adenina/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Quimiotaxis/inmunología , Células Dendríticas/citología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Monocitos/citología , Receptores Purinérgicos P2/inmunología , Receptores Purinérgicos P2Y1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA