Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cell Mol Life Sci ; 81(1): 132, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38472446

RESUMEN

P2Y11 is a G protein-coupled ATP receptor that activates IL-1 receptor (IL-1R) in a cyclic AMP dependent manner. In human macrophages, P2Y11/IL-1R crosstalk with CCL20 as a prime target is controlled by phosphodiesterase 4 (PDE4), which mediates breakdown of cyclic AMP. Here, we used gene expression analysis to identify activation of CXCR4 and CXCR7 as a hallmark of P2Y11 signaling. We found that PDE4 inhibition with rolipram boosts P2Y11/IL-1R-induced upregulation of CXCR7 expression and CCL20 production in an epidermal growth factor receptor dependent manner. Using an astrocytoma cell line, naturally expressing CXCR7 but lacking CXCR4, P2Y11/IL-1R activation effectively induced and CXCR7 agonist TC14012 enhanced CCL20 production even in the absence of PDE4 inhibition. Moreover, CXCR7 depletion by RNA interference suppressed CCL20 production. In macrophages, the simultaneous activation of P2Y11 and CXCR7 by their respective agonists was sufficient to induce CCL20 production with no need of PDE4 inhibition, as CXCR7 activation increased its own and eliminated CXCR4 expression. Finally, analysis of multiple CCL chemokines in the macrophage secretome revealed that CXCR4 inactivation and CXCR7 activation selectively enhanced P2Y11/IL-1R-mediated secretion of CCL20. Altogether, our data establish CXCR7 as an integral component of the P2Y11/IL-1R-initiated signaling cascade and CXCR4-associated PDE4 as a regulatory checkpoint.


Asunto(s)
Receptores CXCR4 , Transducción de Señal , Humanos , Línea Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacología , AMP Cíclico/metabolismo , Macrófagos/metabolismo , Receptores CXCR4/genética , Receptores Purinérgicos/metabolismo
2.
J Cell Biochem ; 120(2): 1783-1793, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30144157

RESUMEN

Extracellular adenosine diphosphate (ADP) mediates a wide range of physiological effects as an extracellular signaling molecule, including platelet aggregation, vascular tone, cell proliferation, and apoptosis by interacting with plasma membrane P2 receptors. However, the effect of ADP on cell proliferation was contradictory. In this study, we found that ADP significantly inhibited cell proliferation of human umbilical vein endothelial cells at high concentrations (50 to 100 µM). Treatment with ADP did not induce cell apoptosis but instead induced cell cycle arrest in the S phase, which may be partly due to the downregulation of cyclin B1. The inhibition of cell proliferation was blocked by suramin, a nonspecific antagonist of the P2 receptors, and high concentrations of ADP significantly upregulated the messenger RNA (mRNA) and protein expression of P2Y11 in endothelial cells. Moreover, the downregulation of P2Y11 by RNA interference reversed the inhibition of cell proliferation. In addition, ADP (100 µM) can induce the formation of cytosolic autophagy in endothelial cells and a rapid phosphorylation of extracellular signal regulated kinase (ERK) 1/2, which is a canonical signal molecule downstream of P2Y receptors, accompanied by a mRNA expression of proinflammatory cytokines such as intercellular adhesion molecule 1 and vascular cell adhesion molecule 1. Taken together, our study excludes a mechanism for extracellular ADP impairing endothelial cells proliferation via P2Y11 receptor by downregulating cyclin B1 and arresting cell cycle at the S phase, besides, ADP induces cell autophagy and mRNA expression of inflammatory cytokines, whether it is mediated by Erk signaling pathways needs further studies to confirm.

3.
IUBMB Life ; 71(10): 1552-1560, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31301116

RESUMEN

Rheumatoid arthritis is a common chronic inflammatory joint disease. Fibroblast-like synoviocytes-mediated inflammation is closely associated with the development of rheumatoid arthritis. In this study, we report that P2Y11 receptor activity is required for cytokine-induced inflammation in primary fibroblast-like synoviocytes (FLS). P2Y11R is fairly expressed in primary FLS isolated from healthy subjects and is elevated to around three- to four-fold in rheumatoid arthritis-derived FLS. The expression of P2Y11R is inducible upon IL-1ß treatment. Blockage of P2Y11R by its antagonist suppresses IL-1ß-induced TNF-α and IL-6 induction and ameliorates oxidative stress as determined by levels of cellular ROS and the oxidative byproduct 4-HNE. Moreover, blockage of P2Y11R by NF340 inhibits IL-1ß-induced matrix metalloproteinase protein expression as indicated by the levels of MMP-1, MMP-3, and MMP-13. Mechanistically, blockage of P2Y11R mitigates IL-1ß-activated NFκB signaling, which was revealed by reduced IκBα phosphorylation, nuclear p65 accumulation, and NFκB promoter activity. Our study provides evidence of a protective mechanism of P2Y11R antagonist NF340 against cytokine-induced inflammation. Therefore, targeting P2Y11R could have potential therapeutic implication in the treatment of RA.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2/genética , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Mediadores de Inflamación/farmacología , Interleucina-1beta/genética , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/genética , Inhibidor NF-kappaB alfa/genética , FN-kappa B/genética , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Receptores Purinérgicos P2/efectos de los fármacos , Sinoviocitos/efectos de los fármacos
5.
Biol Pharm Bull ; 41(10): 1620-1626, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30022772

RESUMEN

External stimuli, such as radiation, induce inflammatory cytokine and chemokine production in skin, but the mechanisms involved are not completely understood. We previously showed that the P2Y11 nucleotide receptor, p38 mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) all participate in interleukin (IL)-6 production induced by γ-irradiation. Here, we focused on the transient receptor potential vanilloid 4 (TRPV4) channel, which is expressed in skin keratinocytes and has been reported to play a role in inflammation. We found that irradiation of human epidermal keratinocytes HaCaT cells with 5 Gy of γ-rays (137Cs: 0.75 Gy/min) induced IL-6 and IL-8 production. HaCaT cells treated with TRPV4 channel agonist GSK1016790A also showed increased IL-6 and IL-8 production. In both cases, IL-6/IL-8 production was not increased at 24 h after stimulation, but was increased at 48 h. ATP was released from cells exposed to γ-irradiation or TRPV4 channel agonist, and the release was suppressed by TRPV4 channel inhibitors. The γ-irradiation-induced increase in IL-6 and IL-8 production was suppressed by apyrase (ecto-nucleotidase), NF157 (selective P2Y11 receptor antagonist) and SB203580 (p38 MAPK inhibitor). GSK1016790A-induced inhibitor of kappa B-alpha (IκBα) decomposition, which causes NF-κB activation was suppressed by NF157 and SB203580, and γ-irradiation-induced IκBα decomposition was suppressed by TRPV4 channel inhibitors. Our results suggest that γ-irradiation of keratinocytes induces ATP release via activation of the TRPV4 channel, and then ATP activates P2Y11 receptor and p38 MAPK-NF-κB signaling, resulting in IL-6/IL-8 production.


Asunto(s)
Adenosina Trifosfato/metabolismo , Rayos gamma , Interleucina-6/biosíntesis , Interleucina-8/biosíntesis , Queratinocitos/metabolismo , Canales Catiónicos TRPV/fisiología , Adenosina Trifosfato/efectos de la radiación , Línea Celular Transformada , Epidermis/metabolismo , Epidermis/efectos de la radiación , Humanos , Interleucina-6/efectos de la radiación , Interleucina-8/efectos de la radiación , Queratinocitos/efectos de la radiación , Canales Catiónicos TRPV/efectos de la radiación
6.
Biol Pharm Bull ; 41(6): 925-936, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29553088

RESUMEN

Skin inflammation is caused by excessive production of cytokines and chemokines in response to an external stimulus, such as radiation, but the mechanisms involved are not completely understood. Here, we report a novel mechanism of γ-irradiation-induced interleukin-6 (IL-6) production mediated by P2Y11 receptors in epidermal cells. After irradiation of HaCaT cells derived from human epidermal keratinocytes with 5 Gy of γ-rays (137Cs: 0.78 Gy/min), IL-6 production was unchanged at 24 h after γ-irradiation, but was increased at 48 h. IL-6 mRNA was increased at 30 h, and IL-6 production was increased at 33 h after irradiation. The production of IL-6 was sustained at least for 4 d after irradiation. P2Y11 receptor antagonist NF157 inhibited IL-6 production in irradiated cells. Treatment with ATP, a ligand of P2Y11 receptor caused IL-6 production within 24 h. ATP-induced IL-6 production was also suppressed by NF157. Extracellular ATP level was increased after irradiation. The p38 mitogen-activated protein kinase (MAPK) and nuclear factor-kappaB (NF-κB) signaling was involved in the production of IL-6 at the downstream of P2Y11 receptor activation. In addition, the cell cycle was arrested at the G2/M phase, and DNA repair foci were not disappeared at 48 h after γ-irradiation. The protein level of histone methylation enzyme G9a, which inhibits IL-6 production, was decreased after γ-irradiation. In conclusion, we suggest that γ-irradiation induces sustained IL-6 production in HaCaT cells from 33 h after irradiation, which is mediated through P2Y11 receptor-p38 MAPK-NF-κB signaling pathway and G9a degradation. This is a novel mechanism of cytokine production in γ-irradiated cells.


Asunto(s)
Rayos gamma , Interleucina-6/metabolismo , Queratinocitos/efectos de la radiación , Receptores Purinérgicos P2/metabolismo , Línea Celular , Daño del ADN , Células Epidérmicas , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Queratinocitos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de la radiación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Mol Cell Biochem ; 431(1-2): 37-44, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28213772

RESUMEN

There is a growing body of evidence pointing to the role of purinergic signaling in the development and progression of various conditions that have inflammation as a common pathogenetic denominator. The aim of the present study was to assess the involvement of P2Y11 purinergic receptors in the regulation of vascular function in aortic segments obtained using an experimental model of acute inflammation, the lipopolysaccharide (LPS, 8 mg/kg, i.p)-treated rats. Twelve hours after LPS administration, thoracic aortas were isolated and used for studies of vascular reactivity in the organ bath and for the measurement of reactive oxygen species (ROS) generation, respectively. LPS treatment significantly increased contractility to phenylephrine and attenuated the endothelium-dependent relaxation of the vascular segments in response to acetylcholine; an increased production of hydrogen peroxide (H2O2) was also recorded. The P2Y11 activator, NF546, decreased the LPS-induced aortic H2O2 release and partially normalized the vasomotor function, namely reduced contractility and improved relaxation. The effect was abolished by co-treatment with the P2Y11 inhibitor, NF340, and also after endothelium denudation. Importantly, NF546 did not elicit an antioxidant effect by acting as a H2O2 scavenger, suggesting that the beneficial outcome of this treatment on the vasculature is the consequence of P2Y11 stimulation. In conclusion, purinergic P2Y11 receptors stimulation improves vascular function and mitigates oxidative stress in the setting of acute systemic inflammation, revealing salutary effects and therapeutic potential in pathologies associated with endothelial dysfunction.


Asunto(s)
Aorta Torácica/metabolismo , Aorta Torácica/fisiopatología , Lipopolisacáridos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Receptores Purinérgicos P2/metabolismo , Vasodilatación/efectos de los fármacos , Enfermedad Aguda , Animales , Aorta Torácica/patología , Difosfonatos/farmacología , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Inflamación/fisiopatología , Naftalenosulfonatos/farmacología , Ratas , Ratas Sprague-Dawley
8.
Adv Exp Med Biol ; 1051: 107-122, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29134605

RESUMEN

The P2Y11 receptor is a G protein-coupled receptor that is stimulated by endogenous purine nucleotides, particularly ATP. Amongst P2Y receptors it has several unique properties; (1) it is the only human P2Y receptor gene that contains an intron in the coding sequence; (2) the gene does not appear to be present in the rodent genome; (3) it couples to stimulation of both phospholipase C and adenylyl cyclase. Its absence in mice and rats, along with a limited range of selective pharmacological tools, has hampered the development of our knowledge and understanding of its properties and functions. Nonetheless, through a combination of careful use of the available tools, suppression of receptor expression using siRNA and genetic screening for SNPs, possible functions of native P2Y11 receptors have been identified in a variety of human cells and tissues. Many are in blood cells involved in inflammatory responses, consistent with extracellular ATP being a damage-associated signalling molecule in the immune system. Thus proposed potential therapeutic applications relate, in the main, to modulation of acute and chronic inflammatory responses.


Asunto(s)
Adenosina Trifosfato/metabolismo , Sistemas de Lectura Abierta , Polimorfismo de Nucleótido Simple , Receptores Purinérgicos P2 , Adenosina Trifosfato/genética , Animales , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Intrones , Ratones , Especificidad de Órganos , Ratas , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Especificidad de la Especie
9.
J Neurochem ; 129(4): 602-13, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24524250

RESUMEN

The P2Y11 nucleotide receptor detects high extracellular ATP concentrations. Mutations of the human P2RY11 gene can play a role in brain autoimmune responses, and the P2Y11 receptor alanine-87-threonine (A87T) polymorphism has been suggested to affect immune-system functions. We investigated receptor functionality of the P2Y11 A87T mutant using HEK293 and 1321N1 astrocytoma cells. In HEK293 cells, the P2Y11 receptor agonist 3'-O-(4-benzoylbenzoyl)adenosine 5'-triphosphate (BzATP) was completely inactive in evoking intracellular calcium release while the potency of ATP was reduced. ATP was also less potent in triggering cAMP generation. However, 1321N1 astrocytoma cells, which lack any endogenous P2Y1 receptors, did not display a reduction. Only when 1321N1 cells were co-transfected with P2Y11 A87T and P2Y1 receptors, the calcium responses to the P2Y11 receptor-specific agonist BzATP were reduced. It is already known that P2Y1 and P2Y11 receptors interact. We thus conclude that the physiological impact of A87T mutation of the P2Y11 receptor derives from detrimental effects on P2Y1 -P2Y11 receptor interaction. We additionally investigated alanine-87-serine and alanine-87-tyrosine P2Y11 receptor mutants. Both mutations rescue the response to BzATP in HEK293 cells, thus ruling out polarity of amino acid-87 to be the molecular basis for altered receptor characteristics. We further found that the P2Y11 A87T receptor shows complete loss of nucleotide-induced internalization in HEK293 cells. Thus, we demonstrate impaired signaling of the P2Y11 A87T-mutated receptors when co-operating with P2Y1 receptors.


Asunto(s)
Sustitución de Aminoácidos , Señalización del Calcio/efectos de los fármacos , Polimorfismo de Nucleótido Simple , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2/genética , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Astrocitoma/patología , Autoinmunidad , Encéfalo/inmunología , Línea Celular Tumoral , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Mapeo de Interacción de Proteínas , Agonistas Purinérgicos/farmacología , Receptores Purinérgicos P2/metabolismo , Relación Estructura-Actividad , Tionucleótidos/farmacología , Transfección
10.
Front Immunol ; 14: 1216580, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37868982

RESUMEN

Since the late 1970s, there has been an alarming increase in the incidence of asthma and its morbidity and mortality. Acute obstruction and inflammation of allergic asthmatic airways are frequently caused by inhalation of exogenous substances such as allergens cross-linking IgE receptors expressed on the surface of the human lung mast cells (HLMC). The degree of constriction of human airways produced by identical amounts of inhaled allergens may vary from day to day and even hour to hour. Endogenous factors in the human mast cell (HMC)'s microenvironment during allergen exposure may markedly modulate the degranulation response. An increase in allergic responsiveness may significantly enhance bronchoconstriction and breathlessness. This review focuses on the role that the ubiquitous endogenous purine nucleotide, extracellular adenosine 5'-triphosphate (ATP), which is a component of the damage-associated molecular patterns, plays in mast cells' physiology. ATP activates P2 purinergic cell-surface receptors (P2R) to trigger signaling cascades resulting in heightened inflammatory responses. ATP is the most potent enhancer of IgE-mediated HLMC degranulation described to date. Current knowledge of ATP as it relates to targeted receptor(s) on HMC along with most recent studies exploring HMC post-receptor activation pathways are discussed. In addition, the reviewed studies may explain why brief, minimal exposures to allergens (e.g., dust, cat, mouse, and grass) can unpredictably lead to intense clinical reactions. Furthermore, potential therapeutic approaches targeting ATP-related enhancement of allergic reactions are presented.


Asunto(s)
Asma , Hipersensibilidad , Receptores Purinérgicos P2 , Humanos , Animales , Ratones , Mastocitos , Transducción de Señal , Adenosina Trifosfato/metabolismo , Asma/metabolismo , Pulmón , Hipersensibilidad/metabolismo , Alérgenos/metabolismo , Receptores Purinérgicos P2/metabolismo
11.
Cancers (Basel) ; 13(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34503103

RESUMEN

ATP released by bone osteocytes is shown to activate purinergic signaling and inhibit the metastasis of breast cancer cells into the bone. However, the underlying molecular mechanism is not well understood. Here, we demonstrate the important roles of the CXCR4 and P2Y11 purinergic receptors in mediating the inhibitory effect of ATP on breast cancer cell migration and bone metastasis. Wound-healing and transwell migration assays showed that non-hydrolysable ATP analogue, ATPγS, inhibited migration of bone-tropic human breast cancer cells in a dose-dependent manner. BzATP, an agonist for P2X7 and an inducer for P2Y11 internalization, had a similar dose-dependent inhibition on cell migration. Both ATPγS and BzATP suppressed the expression of CXCR4, a chemokine receptor known to promote breast cancer bone metastasis, and knocking down CXCR4 expression by siRNA attenuated the inhibitory effect of ATPγS on cancer cell migration. While a P2X7 antagonist A804598 had no effect on the impact of ATPγS on cell migration, antagonizing P2Y11 by NF157 ablated the effect of ATPγS. Moreover, the reduction in P2Y11 expression by siRNA decreased cancer cell migration and abolished the impact of ATPγS on cell migration and CXCR4 expression. Similar to the effect of ATPγS on cell migration, antagonizing P2Y11 inhibited bone-tropic breast cancer cell migration in a dose-dependent manner. An in vivo study using an intratibial bone metastatic model showed that ATPγS inhibited breast cancer growth in the bone. Taken together, these results suggest that ATP inhibits bone-tropic breast cancer cells by down-regulating the P2Y11 purinergic receptor and the down-regulation of CXCR4 expression.

12.
Sleep ; 44(8)2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-33710305

RESUMEN

STUDY OBJECTIVES: Narcolepsy type 1 (NT1) is associated with hypocretin neuron loss. However, there are still unexplained phenotypic NT1 features. We investigated the associations between clinical and sleep phenotypic characteristics, the NT1-associated P2RY11 polymorphism rs2305795, and P2Y11 protein levels in T lymphocytes in patients with NT1, their first-degree relatives and unrelated controls. METHODS: The P2RY11 SNP was genotyped in 100 patients (90/100 H1N1-(Pandemrix)-vaccinated), 119 related and 123 non-related controls. CD4 and CD8 T lymphocyte P2Y11 protein levels were quantified using flow cytometry in 167 patients and relatives. Symptoms and sleep recording parameters were also collected. RESULTS: We found an association between NT1 and the rs2305795 A allele (OR = 2, 95% CI (1.3, 3.0), p = 0.001). T lymphocyte P2Y11 protein levels were significantly lower in patients and relatives homozygous for the rs2305795 risk A allele (CD4: p = 0.012; CD8: p = 0.007). The nocturnal sleep fragmentation index was significantly negatively correlated with patients' P2Y11 protein levels (CD4: p = 0.004; CD8: p = 0.006). Mean MSLT sleep latency, REM-sleep latency, and core clinical symptoms were not associated with P2Y11 protein levels. CONCLUSIONS: We confirmed that the P2RY11 polymorphism rs2305795 is associated with NT1 also in a mainly H1N1-(Pandemrix)-vaccinated cohort. We demonstrated that homozygosity for the A risk allele is associated with lower P2Y11 protein levels. A high level of nocturnal sleep fragmentation was associated with low P2Y11 levels in patients. This suggests that P2Y11 has a previously unknown function in sleep-wake stabilization that affects the severity of NT1.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Narcolepsia , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza , Narcolepsia/genética , Sueño/genética , Privación de Sueño/genética , Linfocitos T
13.
Br J Pharmacol ; 178(7): 1541-1555, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33463722

RESUMEN

BACKGROUND AND PURPOSE: The ATP receptor P2Y11 , which couples to Gq and Gs proteins, senses cell stress and promotes cytoprotective responses. P2Y11 receptors are upregulated during differentiation of M2 macrophages. However, it is unclear whether and how P2Y11 receptors contribute to the anti-inflammatory properties of M2 macrophages. EXPERIMENTAL APPROACH: Transcriptome and secretome profiling of ectopic P2Y11 receptors was used to analyse their signalling and function. Findings were validated in human monocyte-derived M2 macrophages. The suramin analogue NF340 and P2Y11 receptor-knockout cells confirmed that agonist-mediated responses were specific to P2Y11 receptor stimulation. KEY RESULTS: Temporal transcriptome profiling of P2Y11 receptor stimulation showed a strong and tightly controlled response of IL-1 receptors, including activation of the IL-1 receptor target genes, IL6 and IL8. Secretome profiling confirmed the presence of IL-6 and IL-8 proteins and additionally identified soluble tumour necrosis factor receptor 1 and 2 (sTNFR1 and sTNFR2) as targets of P2Y11 receptor activation. Raised levels of intracellular cAMP in M2 macrophages, after inhibition of phosphodiesterases (PDE), especially PDE4, strongly increased P2Y11 receptor-induced release of sTNFR2 through ectodomain shedding mediated by TNF-α converting enzyme (TACE/ADAM17). Both IL-1α and IL-1ß synergistically enhanced P2Y11 receptor- induced IL-6 and IL-8 secretion and release of sTNFR2. During lipopolysaccharide-induced activation of TLR4, which shares the downstream signalling pathway with IL-1 receptors, P2Y11 receptors specifically prevented secretion of TNF-α. CONCLUSIONS AND IMPLICATIONS: Targeting P2Y11 receptors activates IL-1 receptor signalling to promote sTNFR2 release and suppress TLR4 signalling to prevent TNF-α secretion, thus facilitating resolution of inflammation.


Asunto(s)
Receptores Purinérgicos P2 , Antiinflamatorios , Humanos , Lipopolisacáridos/farmacología , Macrófagos , Transducción de Señal , Factor de Necrosis Tumoral alfa
14.
Cells ; 10(5)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34064383

RESUMEN

Macrophages comprise a phenotypically and functionally diverse group of hematopoietic cells. Versatile macrophage subsets engage to ensure maintenance of tissue integrity. To perform tissue stress surveillance, macrophages express many different stress-sensing receptors, including purinergic P2X and P2Y receptors that respond to extracellular nucleotides and their sugar derivatives. Activation of G protein-coupled P2Y receptors can be both pro- and anti-inflammatory. Current examples include the observation that P2Y14 receptor promotes STAT1-mediated inflammation in pro-inflammatory M1 macrophages as well as the demonstration that P2Y11 receptor suppresses the secretion of tumor necrosis factor (TNF)-α and concomitantly promotes the release of soluble TNF receptors from anti-inflammatory M2 macrophages. Here, we review macrophage regulation by P2Y purinergic receptors, both in physiological and disease-associated inflammation. Therapeutic targeting of anti-inflammatory P2Y receptor signaling is desirable to attenuate excessive inflammation in infectious diseases such as COVID-19. Conversely, anti-inflammatory P2Y receptor signaling must be suppressed during cancer therapy to preserve its efficacy.


Asunto(s)
Inflamación/inmunología , Macrófagos/inmunología , Receptores Purinérgicos P2Y/metabolismo , Estrés Fisiológico/inmunología , Animales , COVID-19/sangre , COVID-19/inmunología , Humanos , Vigilancia Inmunológica/efectos de los fármacos , Vigilancia Inmunológica/inmunología , Inflamación/sangre , Inflamación/tratamiento farmacológico , Macrófagos/metabolismo , Ratones , Neoplasias/sangre , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Agonistas del Receptor Purinérgico P2Y/farmacología , Agonistas del Receptor Purinérgico P2Y/uso terapéutico , Antagonistas del Receptor Purinérgico P2Y/farmacología , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Tratamiento Farmacológico de COVID-19
15.
J Cell Physiol ; 223(1): 215-23, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20054824

RESUMEN

Extracellular beta-NAD is known to elevate intracellular levels of calcium ions, inositol 1,4,5-trisphate and cAMP. Recently, beta-NAD was identified as an agonist for P2Y1 and P2Y11 purinergic receptors. Since beta-NAD can be released extracellularly from endothelial cells (EC), we have proposed its involvement in the regulation of EC permeability. Here we show, for the first time, that endothelial integrity can be enhanced in EC endogenously expressing beta-NAD-activated purinergic receptors upon beta-NAD stimulation. Our data demonstrate that extracellular beta-NAD increases the transendothelial electrical resistance (TER) of human pulmonary artery EC (HPAEC) monolayers in a concentration-dependent manner indicating endothelial barrier enhancement. Importantly, beta-NAD significantly attenuated thrombin-induced EC permeability as well as the barrier-compromising effects of Gram-negative and Gram-positive bacterial toxins representing the barrier-protective function of beta-NAD. Immunofluorescence microscopy reveals more pronounced staining of cell-cell junctional protein VE-cadherin at the cellular periphery signifying increased tightness of the cell-cell contacts after beta-NAD stimulation. Interestingly, inhibitory analysis (pharmacological antagonists and receptor sequence specific siRNAs) indicates the participation of both P2Y1 and P2Y11 receptors in beta-NAD-induced TER increase. beta-NAD-treatment attenuates the lipopolysaccharide (LPS)-induced phosphorylation of myosin light chain (MLC) indicating its involvement in barrier protection. Our studies also show the involvement of cAMP-dependent protein kinase A and EPAC1 pathways as well as small GTPase Rac1 in beta-NAD-induced EC barrier enhancement. With these results, we conclude that beta-NAD regulates the pulmonary EC barrier integrity via small GTPase Rac1- and MLCP- dependent signaling pathways.


Asunto(s)
Actinas/metabolismo , Permeabilidad Capilar , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Células Endoteliales/enzimología , Factores de Intercambio de Guanina Nucleótido/metabolismo , NAD/metabolismo , Arteria Pulmonar/enzimología , Proteína de Unión al GTP rac1/metabolismo , Antígenos CD/metabolismo , Proteínas Bacterianas/farmacología , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Uniones Intercelulares/metabolismo , Lipopolisacáridos/farmacología , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1 , Transducción de Señal , Estreptolisinas/farmacología , Trombina/metabolismo , Factores de Tiempo , Proteína de Unión al GTP rac1/genética
16.
Front Immunol ; 11: 549889, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133068

RESUMEN

Intracellular ATP is the universal energy carrier that fuels many cellular processes. However, immune cells can also release a portion of their ATP into the extracellular space. There, ATP activates purinergic receptors that mediate autocrine and paracrine signaling events needed for the initiation, modulation, and termination of cell functions. Mitochondria contribute to these processes by producing ATP that is released. Here, we summarize the synergistic interplay between mitochondria and purinergic signaling that regulates T cell functions. Specifically, we discuss how mitochondria interact with P2X1, P2X4, and P2Y11 receptors to regulate T cell metabolism, cell migration, and antigen recognition. These mitochondrial and purinergic signaling mechanisms are indispensable for host immune defense. However, they also represent an Achilles heel that can render the host susceptible to infections and inflammatory disorders. Hypoxia and mitochondrial dysfunction deflate the purinergic signaling mechanisms that regulate T cells, while inflammation and tissue damage generate excessive systemic ATP levels that distort autocrine purinergic signaling and impair T cell function. An improved understanding of the metabolic and purinergic signaling mechanisms that regulate T cells may lead to novel strategies for the diagnosis and treatment of infectious and inflammatory diseases.


Asunto(s)
Inmunomodulación , Mitocondrias/metabolismo , Receptores Purinérgicos P2X/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Comunicación Celular/inmunología , Ciclo Celular/genética , Ciclo Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Metabolismo Energético/genética , Metabolismo Energético/inmunología , Humanos , Sinapsis Inmunológicas/inmunología , Sinapsis Inmunológicas/metabolismo , Mitocondrias/genética , Transducción de Señal
17.
Artículo en Inglés | MEDLINE | ID: mdl-32528416

RESUMEN

The activation of purinergic receptors by nucleotides and/or nucleosides plays an important role in the control of vascular function, including modulation of vascular smooth muscle excitability, and vascular reactivity. Accordingly, purinergic receptor actions, acting as either ion channels (P2X) or G protein-coupled receptors (GCPRs) (P1, P2Y), target diverse downstream effectors, and substrates to regulate vascular smooth muscle function and vascular reactivity. Both vasorelaxant and vasoconstrictive effects have been shown to be mediated by different purinergic receptors in a vascular bed- and species-specific manner. Purinergic signaling has been shown to play a key role in altering vascular smooth muscle excitability and vascular reactivity following acute and short-term elevations in extracellular glucose (e.g., hyperglycemia). Moreover, there is evidence that vascular smooth muscle excitability and vascular reactivity is severely impaired during diabetes and that this is mediated, at least in part, by activation of purinergic receptors. Thus, purinergic receptors present themselves as important candidates mediating vascular reactivity in hyperglycemia, with potentially important clinical and therapeutic potential. In this review, we provide a narrative summarizing our current understanding of the expression, function, and signaling of purinergic receptors specifically in vascular smooth muscle cells and discuss their role in vascular complications following hyperglycemia and diabetes.


Asunto(s)
Hiperglucemia/complicaciones , Músculo Liso Vascular/patología , Receptores Purinérgicos/metabolismo , Enfermedades Vasculares/patología , Animales , Humanos , Músculo Liso Vascular/metabolismo , Transducción de Señal , Enfermedades Vasculares/etiología , Enfermedades Vasculares/metabolismo
18.
Eur J Pharmacol ; 876: 173060, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32142768

RESUMEN

Chronic inflammation is the hallmark of cardiovascular pathologies with a major role in both disease progression and occurrence of long-term complications. The massive release of ATP during the inflammatory process activates various purinergic receptors, including P2Y11. This receptor is less studied but ubiquitously expressed in all cells relevant for cardiovascular pathology: cardiomyocytes, fibroblasts, endothelial and immune cells. While several studies suggested a potential pro-inflammatory role for P2Y11 receptors, recent literature data are supportive of an anti-inflammatory profile characterized by the immunosuppression of dendritic cells, inhibition of fibroblast proliferation and of cytokines and ATP secretion. Moreover, modulation of its activity appears to mediate the positive inotropic effect of ATP and mitigate endothelial dysfunction, thus rendering this receptor a promising therapeutic target in the cardiovascular disease armamentarium. The aim of the present review is to summarize the current available knowledge on P2Y11-related purinergic signaling in the setting of inflammation and cardio-metabolic diseases.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Enfermedades Metabólicas/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/inmunología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Humanos , Inflamación , Enfermedades Metabólicas/complicaciones , Enfermedades Metabólicas/inmunología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Receptores Purinérgicos P2/genética , Transducción de Señal
19.
Pharmacol Rep ; 71(5): 926-928, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31450027

RESUMEN

BACKGROUND: Narcolepsy with cataplexy is a neurological sleep disorder, which is believed to arise from the autoimmune destruction of hypocretin-producing neurons. The purinergic receptor P2Y11 is associated with narcolepsy in genome-wide association studies, and P2RY11 sequencing has further revealed eight rare missense mutations associated with the disease. Some of these mutations alter the signaling properties of P2Y11, but for some, no functional effects have been discovered so far. METHODS: This study examined the surface expression of the eight narcolepsy-associated P2Y11 mutations using an in vitro surface expression assay. RESULTS: The assay showed excellent discrimination between cells transfected with tagged wild type and the untagged P2Y11 receptor, proving complete specificity towards the 3HA-N-tag used for the assay. Our results show a decreased surface expression of the R307W P2Y11 mutant and a surface expression similar to wild type for the other seven mutants. CONCLUSION: Based on the present findings, alteration in surface expression is not likely to play a role in how P2Y11 influences narcolepsy pathogenesis. This is important because intact surface expression increases the usefulness of P2Y11 as a future drug target.


Asunto(s)
Expresión Génica , Narcolepsia/genética , Receptores Purinérgicos P2/genética , Variación Genética , Células HEK293 , Humanos , Mutación , Neuronas/metabolismo , Orexinas/metabolismo , Transfección
20.
Front Immunol ; 10: 1870, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31447857

RESUMEN

The G protein-coupled P2Y11 receptor is known to sense extracellular ATP during inflammatory and immune responses. The dinucleotide NAD+ has also been proposed to be a P2Y11 receptor ligand but its role is less clear. Here, we have examined for the first time human P2Y11 receptor protein levels and show that the receptor was upregulated during polarization of M2 macrophages. IL-10 reinforced P2Y11 receptor expression during differentiation of M2c macrophages expressing CD163, CD16, and CD274 (PD-L1). Nutlin-3a mediated p53 stabilization further increased P2Y11 receptor, CD16, and PD-L1 expression. AMP-activated kinase (AMPK), which mediates anti-inflammatory effects of IL-10, and nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme of the NAD+ salvage pathway, which is under the control of AMPK, were also required for P2Y11 receptor expression. The P2Y11 receptor agonist ATPγS and NAD+ could independently stimulate the production of IL-8 in M2 macrophages, however, only the ATPγS-induced response was mediated by P2Y11 receptor. Both in a recombinant system and in macrophages, P2Y11 receptor-driven IL-8 production predominantly depended on IkB kinase (IKK), and extracellular signal-regulated kinase (ERK). In conclusion, our data indicate that an AMPK-NAMPT-NAD+ signaling axis promotes P2Y11 receptor expression during M2 polarization of human macrophages in response to IL-10. PD-L1 expressing M2c macrophages that secrete the cancer-promoting chemokine IL-8 in response to P2Y11 receptor stimulation may represent an important target in checkpoint blockade immunotherapy.


Asunto(s)
Citocinas/metabolismo , Macrófagos/metabolismo , Receptores Purinérgicos P2/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA