Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Cell Biol Toxicol ; 40(1): 36, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771396

ABSTRACT

Purinergic receptor P2Y11, a G protein-coupled receptor that is stimulated by extracellular ATP, has been demonstrated to be related to the chemotaxis of granulocytes, apoptosis of neutrophils, and secretion of cytokines in vitro. P2Y11 mutations were associated with narcolepsy. However, little is known about the roles of P2RY11 in the occurrence of narcolepsy and inflammatory response in vivo. In this study, we generated a zebrafish P2Y11 mutant using CRISPR/Cas9 genome editing and demonstrated that the P2Y11 mutant replicated the narcolepsy-like features including reduced HCRT expression and excessive daytime sleepiness, suggesting that P2Y11 is essential for HCRT expression. Furthermore, we accessed the cytokine expression in the mutant and revealed that the P2RY11 mutation disrupted the systemic inflammatory balance by reducing il4, il10 and tgfb, and increasing il6, tnfa, and il1b. In addition, the P2RY11-deficient larvae with caudal fin injuries exhibited significantly slower migration and less recruitment of neutrophils and macrophages at damaged site, and lower expression of anti-inflammatory cytokines during tissue damage. All these findings highlight the vital roles of P2RY11 in maintaining HCRT production and secreting anti-inflammatory cytokines in the native environment, and suggested that P2RY11-deficient zebrafish can serve as a reliable and unique model to further explore narcolepsy and inflammatory-related diseases with impaired neutrophil and macrophage responses.


Subject(s)
Cytokines , Inflammation , Macrophages , Neutrophils , Zebrafish Proteins , Zebrafish , Animals , Neutrophils/metabolism , Neutrophils/immunology , Macrophages/metabolism , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Cytokines/metabolism , Mutation/genetics , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2/deficiency
2.
JCI Insight ; 9(8)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38470490

ABSTRACT

Excessive lipolysis in white adipose tissue (WAT) leads to insulin resistance (IR) and ectopic fat accumulation in insulin-sensitive tissues. However, the impact of Gi-coupled receptors in restraining adipocyte lipolysis through inhibition of cAMP production remained poorly elucidated. Given that the Gi-coupled P2Y13 receptor (P2Y13-R) is a purinergic receptor expressed in WAT, we investigated its role in adipocyte lipolysis and its effect on IR and metabolic dysfunction-associated steatotic liver disease (MASLD). In humans, mRNA expression of P2Y13-R in WAT was negatively correlated to adipocyte lipolysis. In mice, adipocytes lacking P2Y13-R displayed higher intracellular cAMP levels, indicating impaired Gi signaling. Consistently, the absence of P2Y13-R was linked to increased lipolysis in adipocytes and WAT explants via hormone-sensitive lipase activation. Metabolic studies indicated that mice lacking P2Y13-R showed a greater susceptibility to diet-induced IR, systemic inflammation, and MASLD compared with their wild-type counterparts. Assays conducted on precision-cut liver slices exposed to WAT conditioned medium and on liver-specific P2Y13-R-knockdown mice suggested that P2Y13-R activity in WAT protects from hepatic steatosis, independently of liver P2Y13-R expression. In conclusion, our findings support the idea that targeting adipose P2Y13-R activity may represent a pharmacological strategy to prevent obesity-associated disorders, including type 2 diabetes and MASLD.


Subject(s)
Adipocytes , Adipose Tissue, White , Fatty Liver , Insulin Resistance , Lipolysis , Receptors, Purinergic P2 , Animals , Female , Humans , Male , Mice , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue, White/metabolism , Fatty Liver/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/deficiency
3.
J Neuroinflammation ; 18(1): 225, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34635136

ABSTRACT

Inflammation may contribute to multiple brain pathologies. One cause of inflammation is lipopolysaccharide/endotoxin (LPS), the levels of which are elevated in blood and/or brain during bacterial infections, gut dysfunction and neurodegenerative diseases, such as Parkinson's disease. How inflammation causes neuronal loss is unclear, but one potential mechanism is microglial phagocytosis of neurons, which is dependent on the microglial P2Y6 receptor. We investigated here whether the P2Y6 receptor was required for inflammatory neuronal loss. Intraperitoneal injection of LPS on 4 successive days resulted in specific loss of dopaminergic neurons (measured as cells staining with tyrosine hydroxylase or NeuN) in the substantia nigra of wild-type mice, but no neuronal loss in cortex or hippocampus. This supports the hypothesis that neuronal loss in Parkinson's disease may be driven by peripheral LPS. By contrast, there was no LPS-induced neuronal loss in P2Y6 receptor knockout mice. In vitro, LPS-induced microglial phagocytosis of cells was prevented by inhibition of the P2Y6 receptor, and LPS-induced neuronal loss was reduced in mixed glial-neuronal cultures from P2Y6 receptor knockout mice. This supports the hypothesis that microglial phagocytosis contributes to inflammatory neuronal loss, and can be prevented by blocking the P2Y6 receptor, suggesting that P2Y6 receptor antagonists might be used to prevent inflammatory neuronal loss in Parkinson's disease and other brain pathologies involving inflammatory neuronal loss.


Subject(s)
Lipopolysaccharides/toxicity , Neurons/metabolism , Neurons/pathology , Receptors, Purinergic P2/deficiency , Substantia Nigra/metabolism , Substantia Nigra/pathology , Animals , Cell Line, Transformed , Cells, Cultured , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Organ Culture Techniques , PC12 Cells , Rats , Substantia Nigra/drug effects
4.
J Immunol ; 207(6): 1555-1565, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34426542

ABSTRACT

NK cells are critical innate immune cells that target the tumor cells and cancer-initiating cells and clear viruses by producing cytokines and cytotoxic granules. However, the role of the purinergic receptor P2Y6 in the NK cells remains largely unknown. In this study, we discovered that the expression of P2Y6 was decreased upon the activation of the NK cells. Moreover, in the P2Y6-deficient mice, we found that the deficiency of P2Y6 promoted the development of the NK precursor cells into immature NK and mature NK cells. We also found that the P2Y6 deficiency increased, but the P2Y6 receptor agonist UDP or UDP analog 5-OMe-UDP decreased the production of IFN-γ in the activated NK cells. Furthermore, we demonstrated that the P2Y6-deficient NK cells exhibited stronger cytotoxicity in vitro and antimetastatic effects in vivo. Mechanistically, P2Y6 deletion promoted the expression of T-bet (encoded by Tbx21), with or without the stimulation of IL-15. In the absence of P2Y6, the levels of phospho-serine/threonine kinase and pS6 in the NK cells were significantly increased upon the stimulation of IL-15. Collectively, we demonstrated that the P2Y6 receptor acted as a negative regulator of the NK cell function and inhibited the maturation and antitumor activities of the NK cells. Therefore, inhibition of the P2Y6 receptor increases the antitumor activities of the NK cells, which may aid in the design of innovative strategies to improve NK cell-based cancer therapy.


Subject(s)
Cell Differentiation/genetics , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Melanoma/pathology , Receptors, Purinergic P2/deficiency , Signal Transduction/genetics , Skin Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Gene Knockout Techniques , Humans , Interleukin-15/pharmacology , Lung Neoplasms/genetics , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera/immunology , Receptors, Purinergic P2/genetics , Signal Transduction/drug effects , T-Box Domain Proteins/metabolism , Tumor Burden/genetics , Tumor Burden/immunology
6.
J Mol Cell Biol ; 11(5): 395-407, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30137373

ABSTRACT

Among the most important sensors of extracellular danger signals, purinergic receptors have been demonstrated to play crucial roles in host defense against infection. However, the function of P2 receptors in viral infection has been little explored. Here we demonstrated that P2Y13 and its ligand ADP play an important role in protecting hosts from viral infections. First, we demonstrate that P2Y13, as a typical interferon-stimulated gene, is induced together with extracellular ADP during viral infection. Most importantly, extracellular ADP restricts the replication of different kinds of viruses, including vesicular stomatitis virus, Newcastle disease virus, herpes simplex virus 1, and murine leukemia virus. This kind of protection is dependent on P2Y13 but not P2Y1 or P2Y12, which are also considered as receptors for ADP. Furthermore, cyclic adenosine monophosphate and EPAC1 are downregulated by extracellular ADP through the P2Y13-coupled Gi alpha subunit. Accordingly, inhibition or deletion of EPAC1 significantly eliminates ADP/P2Y13-mediated antiviral activities. Taken together, our results show that P2Y13 and ADP play pivotal roles in the clearance of invaded virus and have the potential as antiviral targets.


Subject(s)
Cyclic AMP/metabolism , Gene Expression/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Interferons/pharmacology , Receptors, Purinergic P2/metabolism , Adenosine Diphosphate/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Rhabdoviridae Infections/mortality , Rhabdoviridae Infections/pathology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/veterinary , Signal Transduction/drug effects , Survival Rate , Vesiculovirus/genetics , Vesiculovirus/pathogenicity , Virus Replication/drug effects
7.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978804

ABSTRACT

Glaucoma is an optic neuropathy characterized by progressive degeneration of retinal ganglion cells (RGCs) and visual loss. Although one of the highest risk factors for glaucoma is elevated intraocular pressure (IOP) and reduction in IOP is the only proven treatment, the mechanism of IOP regulation is poorly understood. We report that the P2Y6 receptor is critical for lowering IOP and that ablation of the P2Y6 gene in mice (P2Y6KO) results in hypertensive glaucoma-like optic neuropathy. Topically applied uridine diphosphate, an endogenous selective agonist for the P2Y6 receptor, decreases IOP. The P2Y6 receptor was expressed in nonpigmented epithelial cells of the ciliary body and controlled aqueous humor dynamics. P2Y6KO mice exhibited sustained elevation of IOP, age-dependent damage to the optic nerve, thinning of ganglion cell plus inner plexiform layers, and a reduction of RGC numbers. These changes in P2Y6KO mice were attenuated by an IOP lowering agent. Consistent with RGC damage, visual functions were impaired in middle-aged P2Y6KO mice. We also found that expression and function of P2Y6 receptors in WT mice were significantly reduced by aging, another important risk factor for glaucoma. In summary, our data show that dysfunctional purinergic signaling causes IOP dysregulation, resulting in glaucomatous optic neuropathy.


Subject(s)
Glaucoma/complications , Optic Nerve Diseases/etiology , Receptors, Purinergic P2/physiology , Aging/pathology , Aging/physiology , Animals , Aqueous Humor/metabolism , Ciliary Body/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Glaucoma/drug therapy , Glaucoma/metabolism , Glaucoma/pathology , Intraocular Pressure/drug effects , Intraocular Pressure/physiology , Mice, Knockout , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/pathology , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Retinal Ganglion Cells/pathology , Signal Transduction/physiology , Uridine Diphosphate/administration & dosage , Uridine Diphosphate/pharmacology , Uridine Diphosphate/therapeutic use
8.
Sci Rep ; 7(1): 771, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28396595

ABSTRACT

The role of the P2Y6 receptor in bladder function has recently attracted a great deal of attention in lower urinary tract research. We conducted this study to determine contributions of the P2Y6 receptor in lower urinary tract function of normal phenotypes by comparing P2Y6-deficient mice and wild-type mice. In in vivo experiments, P2Y6-deficient mice had more frequent micturition with smaller bladder capacity compared to wild-type mice; however, there was no difference between these groups in bladder-filling pressure/volume relationships during cystometry under decerebrate, unanaesthetized conditions. Analysis of in vivo bladder contraction revealed significant difference between the 2 groups, with P2Y6-deficient mice presenting markedly shorter bladder contraction duration but no difference in peak contraction pressure. However, analysis of in vitro experiments showed no P2Y6 involvements in contraction and relaxation of bladder muscle strips and in ATP release by mechanical stimulation of primary-cultured urothelial cells. These results suggest that the P2Y6 receptor in the central nervous system, dorsal root ganglion, or both is involved in inhibition of bladder afferent signalling or sensitivity in the pontine micturition centre and that the receptor in the detrusor may be implicated in facilitation to sustain bladder contraction force.


Subject(s)
Muscle Contraction/genetics , Receptors, Purinergic P2/deficiency , Urinary Bladder/physiopathology , Urination , Adenosine Triphosphate/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Electric Stimulation , Gene Expression , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiopathology , RNA, Messenger/genetics , Receptors, Purinergic P2/genetics , Reflex , Urinary Bladder/metabolism
9.
Arterioscler Thromb Vasc Biol ; 36(8): 1598-606, 2016 08.
Article in English | MEDLINE | ID: mdl-27255725

ABSTRACT

OBJECTIVE: Myogenic tone (MT) of resistance arteries ensures autoregulation of blood flow in organs and relies on the intrinsic property of smooth muscle to contract in response to stretch. Nucleotides released by mechanical strain on cells are responsible for pleiotropic vascular effects, including vasoconstriction. Here, we evaluated the contribution of extracellular nucleotides to MT. APPROACH AND RESULTS: We measured MT and the associated pathway in mouse mesenteric resistance arteries using arteriography for small arteries and molecular biology. Of the P2 receptors in mouse mesenteric resistance arteries, mRNA expression of P2X1 and P2Y6 was dominant. P2Y6 fully sustained UDP/UTP-induced contraction (abrogated in P2ry6(-/-) arteries). Preventing nucleotide hydrolysis with the ectonucleotidase inhibitor ARL67156 enhanced pressure-induced MT by 20%, whereas P2Y6 receptor blockade blunted MT in mouse mesenteric resistance arteries and human subcutaneous arteries. Despite normal hemodynamic parameters, P2ry6(-/-) mice were protected against MT elevation in myocardial infarction-induced heart failure. Although both P2Y6 and P2Y2 receptors contributed to calcium mobilization, P2Y6 activation was mandatory for RhoA-GTP binding, myosin light chain, P42-P44, and c-Jun N-terminal kinase phosphorylation in arterial smooth muscle cells. In accordance with the opening of a nucleotide conduit in pressurized arteries, MT was altered by hemichannel pharmacological inhibitors and impaired in Cx43(+/-) and P2rx7(-/-) mesenteric resistance arteries. CONCLUSIONS: Signaling through P2 nucleotide receptors contributes to MT. This mechanism encompasses the release of nucleotides coupled to specific autocrine/paracrine activation of the uracil nucleotide P2Y6 receptor and may contribute to impaired tissue perfusion in cardiovascular diseases.


Subject(s)
Arterioles/metabolism , Mesentery/blood supply , Receptors, Purinergic P2/metabolism , Vasoconstriction , Adenosine Triphosphatases/metabolism , Animals , Arterioles/drug effects , Arterioles/physiopathology , Blood Pressure , Calcium Signaling , Cells, Cultured , Connexin 43/deficiency , Connexin 43/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Genotype , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Hydrolysis , Mechanotransduction, Cellular , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Myocardial Infarction/complications , Myocytes, Smooth Muscle/metabolism , Myosin Light Chains/metabolism , Phenotype , Phosphorylation , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7/deficiency , Receptors, Purinergic P2X7/genetics , Uridine Diphosphate/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
10.
PLoS One ; 11(6): e0156803, 2016.
Article in English | MEDLINE | ID: mdl-27254077

ABSTRACT

ATP-mediated signaling is an important regulator of electrolyte transport in the kidney. The purinergic cation channel P2X6 has been previously localized to the distal convoluted tubule (DCT), a nephron segment important for Mg2+ and Na+ reabsorption, but its role in ion transport remains unknown. In this study, P2x6 knockout (P2x6-/-) mice were generated to investigate the role of P2X6 in renal electrolyte transport. The P2x6-/- animals displayed a normal phenotype and did not differ physiologically from wild type mice. Differences in serum concentration and 24-hrs urine excretion of Na+, K+, Mg2+ and Ca2+ were not detected between P2x6+/+, P2x6+/- and P2x6-/- mice. Quantitative PCR was applied to examine potential compensatory changes in renal expression levels of other P2x subunits and electrolyte transporters, including P2x1-5, P2x7, Trpm6, Ncc, Egf, Cldn16, Scnn1, Slc12a3, Slc41a1, Slc41a3, Cnnm2, Kcnj10 and Fxyd2. Additionally, protein levels of P2X2 and P2X4 were assessed in P2x6+/+ and P2x6-/- mouse kidneys. However, significant changes in expression were not detected. Furthermore, no compensatory changes in gene expression could be demonstrated in heart material isolated from P2x6-/- mice. Except for a significant (P<0.05) upregulation of P2x2 in the heart of P2x6-/- mice compared to the P2x6+/+ mice. Thus, our data suggests that purinergic signaling via P2X6 is not significantly involved in the regulation of renal electrolyte handling under normal physiological conditions.


Subject(s)
Electrolytes/metabolism , Homeostasis , Receptors, Purinergic P2/deficiency , Animals , Base Sequence , Behavior, Animal , Breeding , Gene Expression Regulation , HEK293 Cells , Humans , Kidney/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Knockout , Protein Subunits/metabolism , Receptors, Purinergic P2/metabolism
11.
Cardiovasc Res ; 106(2): 314-23, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25770145

ABSTRACT

AIMS: High-density lipoproteins (HDLs) protect against atherosclerosis mainly due to their function in hepatobiliary reverse cholesterol transport (RCT). This is a process whereby excess cholesterol from peripheral tissues is transported by HDL particles to the liver for further metabolism and biliary excretion. Hepatic uptake of HDL holoparticles involves the P2Y13 receptor, independently of the selective cholesteryl ester uptake mediated by scavenger receptor class B, type I (SR-BI). Accordingly, P2Y13-deficient mice (P2Y13 (-/-)) have impaired RCT. This study assessed whether P2Y13 deficiency would affect atherosclerotic development. METHODS AND RESULTS: P2Y13 (-/-) mice were crossbred with atherosclerosis-prone apoE(-/-) mice. When 15 weeks old, P2Y13 (-/-)/apoE(-/-) mice had more aortic sinus lesions than apoE(-/-) mice. Bone marrow transplantation showed that the absence of the P2Y13 receptor in blood cells did not lead to significantly greater atherosclerotic plaque size formation compared with control apoE(-/-) reconstituted animals. Conversely, the absence of the P2Y13 receptor, except in blood cells, resulted in lesion sizes similar to that in P2Y13 (-/-)/apoE(-/-) reconstituted mice, pointing to a role for non-haematopoietic-derived P2Y13. Unexpectedly, P2Y13 (-/-)/apoE(-/-) mice displayed a lower HDL-cholesterol level than apoE(-/-) mice, which might be due to greater SR-BI expression in the liver. However, P2Y13 deficiency in apoE(-/-) mice was translated into reduced biliary and faecal sterol excretion and impaired RCT from macrophage to faeces, suggesting that an alteration in hepatobiliary RCT could be solely responsible for the greater atherosclerosis observed. CONCLUSION: The P2Y13 receptor protects against atherosclerosis, primarily through its role in hepatobiliary RCT.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Receptors, Purinergic P2/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Biological Transport/genetics , Disease Models, Animal , Lipoproteins, HDL/metabolism , Liver/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2/deficiency
12.
J Immunol ; 194(4): 1874-81, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595790

ABSTRACT

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.


Subject(s)
Endothelin-1/biosynthesis , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Receptors, Purinergic P2/deficiency , Animals , Disease Models, Animal , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Mice , Mice, Knockout , Myocardial Infarction/physiopathology , Real-Time Polymerase Chain Reaction , Transcriptome
13.
PLoS One ; 9(10): e111385, 2014.
Article in English | MEDLINE | ID: mdl-25360548

ABSTRACT

BACKGROUND: P2Y(6), a purinergic receptor for UDP, is enriched in atherosclerotic lesions and is implicated in pro-inflammatory responses of key vascular cell types and macrophages. Evidence for its involvement in atherogenesis, however, has been lacking. Here we use cell-based studies and three murine models of atherogenesis to evaluate the impact of P2Y(6) deficiency on atherosclerosis. METHODOLOGY/PRINCIPAL FINDINGS: Cell-based studies in 1321N1 astrocytoma cells, which lack functional P2Y(6) receptors, showed that exogenous expression of P2Y(6) induces a robust, receptor- and agonist-dependent secretion of inflammatory mediators IL-8, IL-6, MCP-1 and GRO1. P2Y(6)-mediated inflammatory responses were also observed, albeit to a lesser extent, in macrophages endogenously expressing P2Y(6) and in acute peritonitis models of inflammation. To evaluate the role of P2Y(6) in atherosclerotic lesion development, we used P2Y(6)-deficient mice in three mouse models of atherosclerosis. A 43% reduction in aortic arch plaque was observed in high fat-fed LDLR knockout mice lacking P2Y(6) receptors in bone marrow-derived cells. In contrast, no effect on lesion development was observed in fat-fed whole body P2Y(6)xLDLR double knockout mice. Interestingly, in a model of enhanced vascular inflammation using angiotensin II, P2Y(6) deficiency enhanced formation of aneurysms and exhibited a trend towards increased atherosclerosis in the aorta of LDLR knockout mice. CONCLUSIONS: P2Y(6) receptor augments pro-inflammatory responses in macrophages and exhibits a pro-atherogenic role in hematopoietic cells. However, the overall impact of whole body P2Y(6) deficiency on atherosclerosis appears to be modest and could reflect additional roles of P2Y(6) in vascular disease pathophysiologies, such as aneurysm formation.


Subject(s)
Atherosclerosis/metabolism , Macrophages/metabolism , Receptors, Purinergic P2/metabolism , Animals , Atherosclerosis/immunology , Cell Line, Tumor , Cytokines/metabolism , Female , Gene Knockout Techniques , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics
14.
Arterioscler Thromb Vasc Biol ; 34(10): 2237-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25104800

ABSTRACT

OBJECTIVE: Nucleotides such as ATP, ADP, UTP, and UDP serve as proinflammatory danger signals via purinergic receptors on their release to the extracellular space by activated or dying cells. UDP binds to the purinergic receptor Y6 (P2Y6) and propagates vascular inflammation by inducing the expression of chemokines such as monocyte chemoattractant protein 1, interleukin-8, or its mouse homologsCCL1 (chemokine [C-C motif] ligand 1)/keratinocyte chemokine, CXCL2 (chemokine [C-X-C motif] ligand 2)/macrophage inflammatory protein 2, and CXCL5 (chemokine [C-X-C motif] ligand 5)/LIX, and adhesion molecules such as vascular cell adhesion molecule 1 and intercellular cell adhesion molecule 1. Thus, P2Y6 contributes to leukocyte recruitment and inflammation in conditions such as allergic asthma or sepsis. Because atherosclerosis is a chronic inflammatory disease driven by leukocyte recruitment to the vessel wall, we hypothesized a role of P2Y6 in atherogenesis. APPROACH AND RESULTS: Intraperitoneal stimulation of wild-type mice with UDP induced rolling and adhesion of leukocytes to the vessel wall as assessed by intravital microscopy. This effect was not present in P2Y6-deficient mice. Atherosclerotic aortas of low-density lipoprotein receptor-deficient mice consuming high-cholesterol diet for 16 weeks expressed significantly more transcripts and protein of P2Y6 than respective controls. Finally, P2Y6 (-/-)/low-density lipoprotein receptor-deficient mice consuming high-cholesterol diet for 16 weeks developed significantly smaller atherosclerotic lesions compared with P2Y6 (+/+)/low-density lipoprotein receptor-deficient mice. Bone marrow transplantation identified a crucial role of P2Y6 on vascular resident cells, most likely endothelial cells, on leukocyte recruitment and atherogenesis. Atherosclerotic lesions of P2Y6-deficient mice contained fewer macrophages and fewer lipids as determined by immunohistochemistry. Mechanistically, RNA expression of vascular cell adhesion molecule 1 and interleukin-6 was decreased in these lesions and P2Y6-deficient macrophages took up less modified low-density lipoprotein cholesterol. CONCLUSIONS: We show for the first time that P2Y6 deficiency limits atherosclerosis and plaque inflammation in mice.


Subject(s)
Aorta/metabolism , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Inflammation/prevention & control , Receptors, Purinergic P2/deficiency , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Bone Marrow Transplantation , Cholesterol, Dietary , Disease Models, Animal , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Leukocyte Rolling , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Purinergic P2/genetics , Signal Transduction , Time Factors , Transendothelial and Transepithelial Migration , Uridine Diphosphate/metabolism
15.
Purinergic Signal ; 10(3): 455-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24510452

ABSTRACT

Gastrointestinal symptoms have a major impact on the quality of life and are becoming more prevalent in the western population. The enteric nervous system (ENS) is pivotal in regulating gastrointestinal functions. Purinergic neurotransmission conveys a range of short and long-term cellular effects. This study investigated the role of the ADP-sensitive P2Y13 receptor in lipid-induced enteric neuropathy. Littermate P2Y13 (+/+) and P2Y13 (-/-) mice were fed with either a normal diet (ND) or high-fat diet (HFD) for 6 months. The intestines were analysed for morphological changes as well as neuronal numbers and relative numbers of vasoactive intestinal peptide (VIP)- and neuronal nitric oxide synthase (nNOS)-containing neurons. Primary cultures of myenteric neurons from the small intestine of P2Y13 (+/+) or P2Y13 (-/-) mice were exposed to palmitic acid (PA), the P2Y13 receptor agonist 2meSADP and the antagonist MRS2211. Neuronal survival and relative number of VIP-containing neurons were analysed. In P2Y13 (+/+), but not in P2Y13 (-/-) mice, HFD caused a significant loss of myenteric neurons in both ileum and colon. In colon, the relative numbers of VIP-containing submucous neurons were significantly lower in the P2Y13 (-/-) mice compared with P2Y13 (+/+) mice. The relative numbers of nNOS-containing submucous colonic neurons increased in P2Y13 (+/+) HFD mice. HFD also caused ileal mucosal thinning in P2Y13 (+/+) and P2Y13 (-/-) mice, compared to ND fed mice. In vitro PA exposure caused loss of myenteric neurons from P2Y13 (+/+) mice while neurons from P2Y13 (-/-) mice were unaffected. Presence of MRS2211 prevented PA-induced neuronal loss in cultures from P2Y13 (+/+) mice. 2meSADP caused no change in survival of cultured neurons. P2Y13 receptor activation is of crucial importance in mediating the HFD- and PA-induced myenteric neuronal loss in mice. In addition, the results indicate a constitutive activation of enteric neuronal apoptosis by way of P2Y13 receptor stimulation.


Subject(s)
Diet, High-Fat/adverse effects , Enteric Nervous System/metabolism , Neurons/metabolism , Palmitic Acid/toxicity , Receptors, Purinergic P2/deficiency , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Myenteric Plexus/drug effects , Myenteric Plexus/metabolism , Myenteric Plexus/pathology , Neurons/drug effects , Organ Culture Techniques
16.
Cell Mol Life Sci ; 71(9): 1775-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24030815

ABSTRACT

The protective effect of high density lipoproteins (HDL) against atherosclerosis is mainly attributed to their capacity to transport excess cholesterol from peripheral tissues back to the liver for further elimination into the bile, a process called reverse cholesterol transport (RCT). Recently, the importance of the P2Y13 receptor (P2Y13-R) was highlighted in HDL metabolism since HDL uptake by the liver was decreased in P2Y13-R deficient mice, which translated into impaired RCT. Here, we investigated for the first time the molecular mechanisms regulating cell surface expression of P2Y13-R. When transiently expressed, P2Y13-R was mainly detected in the endoplasmic reticulum (ER) and strongly subjected to proteasome degradation while its homologous P2Y12 receptor (P2Y12-R) was efficiently targeted to the plasma membrane. We observed an inverse correlation between cell surface expression and ubiquitination level of P2Y13-R in the ER, suggesting a close link between ubiquitination of P2Y13-R and its efficient targeting to the plasma membrane. The C-terminus tail exchange between P2Y13-R and P2Y12-R strongly restored plasma membrane expression of P2Y13-R, suggesting the involvement of the intra-cytoplasmic tail of P2Y13-R in expression defect. Accordingly, proteasomal inhibition increased plasma membrane expression of functionally active P2Y13-R in hepatocytes, and consequently stimulated P2Y13-R-mediated HDL endocytosis. Importantly, proteasomal inhibition strongly potentiated HDL hepatic uptake (>200 %) in wild-type but not in P2Y13-R-deficient mice, thus reinforcing the role of P2Y13-R expression in regulating HDL metabolism. Therefore, specific inhibition of the ubiquitin-proteasome system might be a novel powerful HDL therapy to enhance P2Y13-R expression and consequently promote the overall RCT.


Subject(s)
Lipoproteins, HDL/metabolism , Liver/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Purinergic P2/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Endocytosis , Endoplasmic Reticulum/metabolism , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteasome Endopeptidase Complex/chemistry , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ubiquitination
17.
J Immunol ; 191(12): 5807-10, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24244016

ABSTRACT

Leukotriene C4 (LTC4) and its extracellular metabolites, LTD4 and LTE4, mediate airway inflammation. They signal through three specific receptors (type 1 cys-LT receptor [CysLT1R], CysLT2R, and GPR99) with overlapping ligand preferences. In this article, we demonstrate that LTC4, but not LTD4 or LTE4, activates mouse platelets exclusively through CysLT2R. Platelets expressed CysLT1R and CysLT2R proteins. LTC4 induced surface expression of CD62P by wild-type mouse platelets in platelet-rich plasma (PRP) and caused their secretion of thromboxane A2 and CXCL4. LTC4 was fully active on PRP from mice lacking either CysLT1R or GPR99, but completely inactive on PRP from CysLT2R-null (Cysltr2(-/-)) mice. LTC4/CysLT2R signaling required an autocrine ADP-mediated response through P2Y12 receptors. LTC4 potentiated airway inflammation in a platelet- and CysLT2R-dependent manner. Thus, CysLT2R on platelets recognizes LTC4 with unexpected selectivity. Nascent LTC4 may activate platelets at a synapse with granulocytes before it is converted to LTD4, promoting mediator generation and the formation of leukocyte-platelet complexes that facilitate inflammation.


Subject(s)
Blood Platelets/drug effects , Leukotriene C4/physiology , Receptors, Leukotriene/physiology , Adenosine Diphosphate/pharmacology , Animals , Autocrine Communication , Blood Platelets/metabolism , Leukotriene C4/toxicity , Leukotriene D4/pharmacology , Leukotriene E4/pharmacology , Mice , Mice, Knockout , Ovalbumin/immunology , Ovalbumin/toxicity , P-Selectin/biosynthesis , P-Selectin/genetics , Platelet Activation/drug effects , Platelet Factor 4/metabolism , Platelet-Rich Plasma , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/immunology , Receptors, Leukotriene/deficiency , Receptors, Leukotriene/genetics , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/physiology , Receptors, Thromboxane A2, Prostaglandin H2/deficiency , Thromboxane A2/metabolism
18.
Cell Death Dis ; 4: e703, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23828566

ABSTRACT

In utero exposure of the embryo and fetus to radiation has been implicated in malformations or fetal death, and often produces lifelong health consequences such as cancers and mental retardation. Here we demonstrate that deletion of a G-protein-coupled purinergic receptor, P2Y14, confers potent resistance to in utero radiation. Intriguingly, a putative P2Y14 receptor ligand, UDP-glucose, phenocopies the effect of P2Y14 deficiency. These data indicate that P2Y14 is a receptor governing in utero tolerance to genotoxic stress that may be pharmacologically targeted to mitigate radiation toxicity in pregnancy.


Subject(s)
Hydrocephalus/prevention & control , Prenatal Exposure Delayed Effects/prevention & control , Radiation Injuries, Experimental/prevention & control , Receptors, Purinergic P2/genetics , Uridine Diphosphate Glucose/pharmacology , Animals , Atrophy/prevention & control , Body Weight/drug effects , DNA Damage , Female , Hydrocephalus/metabolism , Male , Maternal Exposure , Mice , Mice, Knockout , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Radiation Injuries, Experimental/metabolism , Radiation Tolerance , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2Y , Spleen/drug effects , Spleen/pathology , Uridine Diphosphate Glucose/therapeutic use , Whole-Body Irradiation
19.
Am J Physiol Heart Circ Physiol ; 303(7): H835-43, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22865387

ABSTRACT

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.


Subject(s)
Cardiomegaly, Exercise-Induced/genetics , Cardiomegaly/prevention & control , Exercise Tolerance/genetics , Gene Deletion , Heart/physiopathology , Myocardium/metabolism , Receptors, Purinergic P2/deficiency , Swimming , Adrenergic Fibers/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Behavior, Animal , Blood Pressure/genetics , Blood Pressure Monitoring, Ambulatory , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Catecholamines/metabolism , Disease Models, Animal , Dobutamine/pharmacology , Exercise Test , Exercise Tolerance/drug effects , Genotype , Heart/innervation , Heart Rate/genetics , Hypothermia/genetics , Hypothermia/metabolism , Hypothermia/physiopathology , Locomotion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Phenotype , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Receptors, Purinergic P2/genetics , Recovery of Function , Telemetry , Time Factors
20.
Mol Endocrinol ; 26(1): 142-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22108801

ABSTRACT

Osteoporosis is a condition of excessive and uncoupled bone turnover, in which osteoclastic resorption exceeds osteoblastic bone formation, resulting in an overall net bone loss, bone fragility, and morbidity. Although numerous treatments have been developed to inhibit bone loss by blocking osteoclastic bone resorption, understanding of the mechanisms behind bone loss is incomplete. The purinergic signaling system is emerging to be a pivotal regulator of bone homeostasis, and extracellular ADP has previously been shown to be a powerful osteolytic agent in vitro. We report here that deletion of the P2Y(13) receptor, a G protein-coupled receptor for extracellular ADP, leads to a 40% reduction in trabecular bone mass, 50% reduction in osteoblast and osteoclast numbers in vivo, as well as activity in vitro, and an overall 50% reduction in the rate of bone remodeling in mice in vivo. Down-regulation of RhoA/ROCK I signaling and a reduced ratio of receptor activator of nuclear factor κB ligand/osteoprotegerin observed in osteoblasts from P2Y(13)R(-/-) mice might explain this bone phenotype. Furthermore, because one of the main causes of osteoporosis in older women is lack of estrogen, we examined the effect of ovariectomy of the P2Y(13)R(-/-) mice and found them to be protected from ovariectomy-induced bone loss by up to 65%. These data confirm a role of purinergic ADP signaling in the skeleton, whereby deletion of the P2Y(13) receptor leads to reduced bone turnover rates, which provide a protective advantage in conditions of accelerated bone turnover such as oestrogen deficiency-induced osteoporosis.


Subject(s)
Bone Remodeling , Bone Resorption , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/physiology , Animals , Bone and Bones/metabolism , Cells, Cultured , Estrogens/blood , Female , Mice , Mice, Knockout , NF-kappa B/biosynthesis , Osteoblasts/metabolism , Osteoblasts/physiology , Osteoclasts/metabolism , Osteoclasts/physiology , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/prevention & control , Osteoprotegerin/biosynthesis , Ovariectomy/adverse effects , Receptors, Purinergic P2/deficiency , Signal Transduction , rho GTP-Binding Proteins/biosynthesis , rho-Associated Kinases/biosynthesis , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...