Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 59
1.
Microvasc Res ; 139: 104256, 2022 01.
Article En | MEDLINE | ID: mdl-34530027

BACKGROUND: The purine adenosine triphosphate (ATP) plays a significant role in retinal blood flow regulation and recent evidence suggests that the vasoactive effect of the compound differs in vessels at different branching level. However, the cellular basis for the regulation of retinal blood flow mediated by ATP has only been scarcely studied. METHODS: Perfused porcine hemiretinas (n = 60) were loaded with the calcium-sensitive fluorophore Oregon Green ex vivo. Spontaneous oscillations in fluorescence were studied in perivascular cells at five different vascular branching levels ranging from the main arteriole to the capillaries, before and after the addition of intra- and extravascular ATP alone or in the presence of a P2-purinergic receptor antagonist. RESULTS: Intravascular ATP induced an overall significant (p < 0.01) constriction of (mean ± SD) -9.79 ± 13.40% and extravascular ATP an overall significant (p < 0.01) dilatation of (mean ± SD) 19.62 ± 13.47%. Spontaneous oscillations of fluorescence in perivascular cells were significantly more intense around third order arterioles than around vessels at both lower and higher branching levels (p < 0.05 for all comparisons). ATP increased intracellular fluorescence in perivascular cells of first and second order arterioles after extravascular application, and the increase correlated with the accompanying vasodilatation (p < 0.03). Blocking of P2-receptors reduced oscillating fluorescence in pre-capillary arterioles secondary to intravascular ATP (p = 0.03). CONCLUSIONS: Spontaneous oscillations of calcium-sensitive fluorescence in perivascular retinal cells differ at different vascular branching levels. Extravascular ATP increases fluorescence in cells around the larger retinal arterioles exposed to the retinal surface. Future studies should investigate calcium signaling activity in perivascular retinal cells during interventions that simulate retinal pathology such as hypoxia.


Adenosine Triphosphate/pharmacology , Arterioles/drug effects , Calcium Signaling/drug effects , Capillaries/drug effects , Purinergic P2 Receptor Agonists/pharmacology , Retinal Vessels/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects , Animals , Arterioles/metabolism , Capillaries/metabolism , Cellular Microenvironment , Purinergic P2 Receptor Antagonists/pharmacology , Retinal Vessels/metabolism , Sus scrofa
2.
Physiol Rep ; 9(11): e14888, 2021 06.
Article En | MEDLINE | ID: mdl-34110719

Hypercholesterolemia and oxidative stress may lead to disturbances in the renal microvasculature in response to vasoactive agents, including P2 receptors (P2R) agonists. We investigated the renal microvascular response to diadenosine tetraphosphate (Ap4 A), an agonist of P2R, in diet-induced hypercholesteremic rats over 28 days, supplemented in the last 10 days with tempol (2 mM) or DL-buthionine-(S,R)-sulfoximine (BSO, 20 mM) in the drinking water. Using laser Doppler flowmetry, renal blood perfusion in the cortex and medulla (CBP, MBP) was measured during the infusion of Ap4 A. This induced a biphasic response in the CBP: a phase of rapid decrease was followed by one of rapid increase extended for 30 min in both the normocholesterolemic and hypercholesterolemic rats. The phase of decreased CBP was not affected by tempol or BSO in either group. Early and extended increases in CBP were prevented by tempol in the hypercholesterolemia rats, while, in the normocholesterolemic rats, only the extended increase in CBP was affected by tempol; BSO prevented extended increase in CBP in normocholesterolemic rats. MBP response is not affected by hypercholesterolemia. The hypercholesterolemic rats were characterized by increased urinary albumin and 8-isoPGF2α excretion. Moreover, BSO increased the urinary excretion of nephrin in the hypercholesterolemic rats but, similar to tempol, did not affect the excretion of albumin in their urine. The results suggest the important role of redox balance in the extracellular nucleotide regulation of the renal vasculature and glomerular injury in hypercholesterolemia.


Dinucleoside Phosphates/pharmacology , Hemodynamics/drug effects , Hypercholesterolemia/complications , Kidney/drug effects , Oxidation-Reduction/drug effects , Purinergic P2 Receptor Agonists/pharmacology , Animals , Diet, High-Fat/adverse effects , Hypercholesterolemia/metabolism , Hypercholesterolemia/physiopathology , Kidney/blood supply , Kidney/physiopathology , Lipids/blood , Male , Rats , Rats, Wistar , Receptors, Purinergic P2/drug effects , Renal Circulation/drug effects
3.
Int J Mol Sci ; 22(2)2021 Jan 16.
Article En | MEDLINE | ID: mdl-33467058

Vascular dysfunction in cardiovascular diseases includes vasomotor response impairments, endothelial cells (ECs) activation, and smooth muscle cells (SMCs) proliferation and migration to the intima. This results in intimal hyperplasia and vessel failure. We previously reported that activation of the P2Y11 receptor (P2Y11R) in human dendritic cells, cardiofibroblasts and cardiomyocytes was protective against hypoxia/reoxygenation (HR) lesions. In this study, we investigated the role of P2Y11R signaling in vascular dysfunction. P2Y11R activity was modulated using its pharmacological agonist NF546 and antagonist NF340. Rat aortic rings were exposed to angiotensin II (AngII) and evaluated for their vasomotor response. The P2Y11R agonist NF546 reduced AngII-induced vascular dysfunction by promoting EC-dependent vasorelaxation, through an increased nitric oxide (NO) bioavailability and reduced AngII-induced H2O2 release; these effects were prevented by the use of the P2Y11R antagonist NF340. Human vascular SMCs and ECs were subjected to AngII or H/R simulation in vitro. P2Y11R agonist modulated vasoactive factors in human ECs, that is, endothelial nitric oxide synthase (eNOS) and endothelin-1, reduced SMC proliferation and prevented the switch towards a synthetic phenotype. H/R and AngII increased ECs secretome-induced SMC proliferation, an effect prevented by P2Y11R activation. Thus, our data suggest that P2Y11R activation may protect blood vessels from HR-/AngII-induced injury and reduce vascular dysfunctions. These results open the way for new vasculoprotective interventions.


Diphosphonates/pharmacology , Naphthalenesulfonates/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P2/metabolism , Reperfusion Injury/metabolism , Tunica Intima/pathology , Angiotensin II/toxicity , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Diphosphonates/therapeutic use , Endothelin-1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hyperplasia/prevention & control , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Naphthalenesulfonates/therapeutic use , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Purinergic P2 Receptor Agonists/therapeutic use , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Tunica Intima/drug effects , Tunica Intima/metabolism , Vasodilation , Water/metabolism
4.
J Vasc Res ; 58(1): 38-48, 2021.
Article En | MEDLINE | ID: mdl-33207336

Diabetes through adenosine A1 receptor (A1R) and P2 receptors (P2Rs) may lead to disturbances in renal microvasculature. We investigated the renal microvascular response to Ap4A, an agonist of P2Rs, in streptozotocin-induced diabetic rats. Using laser Doppler flowmetry, renal blood perfusion (RBP) was measured during infusion of Ap4A alone or in the presence of A1R antagonist, either DPCPX (8-cyclopentyl-1,3-dipropylxanthine) or 8-cyclopentyltheophylline (CPT). Ap4A induced a biphasic response in RBP: a phase of rapid decrease was followed by a rapid increase, which was transient in diabetic rats but extended for 30 min in nondiabetic rats. Phase of decreased RBP was not affected by DPCPX or CPT in either group. Early and extended increases in RBP were prevented by DPCPX and CPT in nondiabetic rats, while in diabetic rats, the early increase in RBP was not affected by these antagonists. A1R mRNA and protein levels were increased in isolated glomeruli of diabetic rats, but no changes were detected in P2Y1R and P2Y2R mRNA. Presence of unblocked A1R is a prerequisite for the P2R-mediated relaxing effect of Ap4A in nondiabetic conditions, but influence of A1R on P2R-mediated renal vasorelaxation is abolished under diabetic conditions.


Acid Anhydride Hydrolases/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Kidney Cortex/blood supply , Kidney Medulla/blood supply , Purinergic P2 Receptor Agonists/pharmacology , Receptor, Adenosine A1/metabolism , Renal Circulation/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Blood Flow Velocity , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Male , Rats, Wistar , Receptor Cross-Talk , Receptors, Purinergic P2/metabolism , Signal Transduction
5.
Int J Mol Sci ; 21(17)2020 Aug 27.
Article En | MEDLINE | ID: mdl-32867347

BACKGROUND: Human mesenchymal stem cells (hMSCs) have shown their multipotential including differentiating towards endothelial and smooth muscle cell lineages, which triggers a new interest for using hMSCs as a putative source for cardiovascular regenerative medicine. Our recent publication has shown for the first time that purinergic 2 receptors are key players during hMSC differentiation towards adipocytes and osteoblasts. Purinergic 2 receptors play an important role in cardiovascular function when they bind to extracellular nucleotides. In this study, the possible functional role of purinergic 2 receptors during MSC endothelial and smooth muscle differentiation was investigated. METHODS AND RESULTS: Human MSCs were isolated from liposuction materials. Then, endothelial and smooth muscle-like cells were differentiated and characterized by specific markers via Reverse Transcriptase-PCR (RT-PCR), Western blot and immunochemical stainings. Interestingly, some purinergic 2 receptor subtypes were found to be differently regulated during these specific lineage commitments: P2Y4 and P2Y14 were involved in the early stage commitment while P2Y1 was the key player in controlling MSC differentiation towards either endothelial or smooth muscle cells. The administration of natural and artificial purinergic 2 receptor agonists and antagonists had a direct influence on these differentiations. Moreover, a feedback loop via exogenous extracellular nucleotides on these particular differentiations was shown by apyrase digest. CONCLUSIONS: Purinergic 2 receptors play a crucial role during the differentiation towards endothelial and smooth muscle cell lineages. Some highly selective and potent artificial purinergic 2 ligands can control hMSC differentiation, which might improve the use of adult stem cells in cardiovascular tissue engineering in the future.


Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Receptors, Purinergic P2/metabolism , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Endothelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Lipectomy , Mesenchymal Stem Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Young Adult
6.
Fitoterapia ; 146: 104709, 2020 Oct.
Article En | MEDLINE | ID: mdl-32829014

In recent years, interest in the research of P2 receptor (P2R)-mediated responses has grown significantly due to the recognition of the involvement of these receptors in various physiological and pathological processes. Despite all the progress made in the functional characterization of P2Rs, purinergic signaling research is still limited by the lack of selective or efficient ligands for different receptor subtypes. In this sense, several molecules have been tested towards these receptors as agonists or antagonists. Historically, natural products have always been sources of new bioactive substances for diverse purposes. However, compared to synthetic molecules, the number of natural products assessed for P2R ligands is still low. In this review, we present examples of studies that demonstrated plant natural products acting directly on P2R and modulating their functionality. In some cases, we highlight that the pharmacological activity previously described for the original organism could be correlated to an agonist or antagonist activity of a specific natural product on these receptors. These examples reinforce the need for more studies to investigate the pharmacological potential of new or known natural compounds targeting P2 receptors.


Phytochemicals/pharmacology , Receptors, Purinergic P2/metabolism , Signal Transduction , Biological Products/pharmacology , Clinical Trials as Topic , Humans , Ligands , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology
7.
J Med Chem ; 63(17): 9990-10029, 2020 09 10.
Article En | MEDLINE | ID: mdl-32787112

Lysophosphatidylserine (LysoPS), an endogenous ligand of G protein-coupled receptors, consists of l-serine, glycerol, and fatty acid moieties connected by phosphodiester and ester linkages, respectively. An ester linkage of phosphatidylserine can be hydrolyzed at the 1-position or at the 2-position to give 2-acyl lysophospholipid or 1-acyl lysophospholipid, respectively. 2-Acyl lysophospholipid is in nonenzymatic equilibrium with 1-acyl lysophospholipid in vivo. On the other hand, 3-acyl lysophospholipid is not found, at least in mammals, raising the question of whether the reason for this might be that the 3-acyl isomer lacks the biological activities of the other isomers. Here, to test this idea, we designed and synthesized a series of new 3-acyl lysophospholipids. Structure-activity relationship studies of more than 100 "glycol surrogate" derivatives led to the identification of potent and selective agonists for LysoPS receptors GPR34 and P2Y10. Thus, the non-natural 3-acyl compounds are indeed active and appear to be biologically orthogonal with respect to the physiologically relevant 1- and 2-acyl lysophospholipids.


Lysophospholipids/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Lysophospholipid/agonists , Receptors, Purinergic P2/metabolism , HEK293 Cells , Humans , Isomerism , Lysophospholipids/chemical synthesis , Molecular Conformation , Molecular Docking Simulation , Purinergic P2 Receptor Agonists/chemical synthesis , Structure-Activity Relationship
8.
Sci Rep ; 10(1): 8292, 2020 05 19.
Article En | MEDLINE | ID: mdl-32427920

The P2X receptor family of ATP-gated cation channels are attractive drug targets for pain and inflammatory disease, but no subtype-selective agonists, and few partially selective agonists have been described to date. As proof-of-concept for the discovery of novel P2X receptor agonists, here we demonstrate the use of Drosophila taste neurons heterologously expressing rat P2X2 receptors as a screening platform. We demonstrate that wild-type rat P2X2 expressed in Drosophila is fully functional (ATP EC50 8.7 µM), and that screening of small (2 µl) volumes of a library of 80 adenosine nucleotide analogues is rapid and straightforward. We have determined agonist potency and specificity profiles for rat P2X2 receptors; triphosphate-bearing analogues display broad activity, tolerating a number of substitutions, and diphosphate and monophosphate analogues display very little activity. While several ATP analogues gave responses of similar magnitude to ATP, including the previously identified agonists ATPγS and ATPαS, we were also able to identify a novel agonist, the synthetic analogue 2-fluoro-ATP, and to confirm its agonist activity on rat P2X2 receptors expressed in human cells. These data validate our Drosophila platform as a useful tool for the analysis of agonist structure-activity relationships, and for the screening and discovery of novel P2X receptor agonists.


Adenosine Triphosphate/analogs & derivatives , Neurons/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P2X2/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/pharmacology , Animals , Animals, Genetically Modified , Drosophila , HEK293 Cells , Humans , Neurons/drug effects , Proof of Concept Study , Purinergic P2 Receptor Agonists/chemistry , Rats , Receptors, Purinergic P2X2/genetics , Structure-Activity Relationship , Taste
9.
Int J Mol Sci ; 22(1)2020 Dec 29.
Article En | MEDLINE | ID: mdl-33383710

ATP is a major energy source in the mammalian cells, but it is an extracellular chemical messenger acting on P2 purinergic receptors. A line of evidence has shown that ATP is released from many different types of cells including neurons, endothelial cells, and muscle cells. In this review, we described the distribution of P2 receptor subtypes in the cardiac cells and their physiological and pathological roles in the heart. So far, the effects of external application of ATP or its analogues, and those of UTP on cardiac contractility and rhythm have been reported. In addition, specific genetic alterations and pharmacological agonists and antagonists have been adopted to discover specific roles of P2 receptor subtypes including P2X4-, P2X7-, P2Y2- and P2Y6-receptors in cardiac cells under physiological and pathological conditions. Accumulated data suggest that P2X4 receptors may play a beneficial role in cardiac muscle function, and that P2Y2- and P2Y6-receptors can induce cardiac fibrosis. Recent evidence further demonstrates P2Y1 receptor and P2X4 receptor as important mechanical signaling molecules to alter membrane potential and Ca2+ signaling in atrial myocytes and their uneven expression profile between right and left atrium.


Mechanotransduction, Cellular , Myocytes, Cardiac/metabolism , Receptors, Purinergic P2/metabolism , Signal Transduction , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Disease Susceptibility , Extracellular Space/metabolism , Gene Expression Regulation , Heart Rate/drug effects , Heart Rate/genetics , Humans , Mechanotransduction, Cellular/drug effects , Myocardial Contraction , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2/genetics , Signal Transduction/drug effects , Stress, Physiological/drug effects , Stress, Physiological/genetics
10.
Methods Mol Biol ; 2041: 45-64, 2020.
Article En | MEDLINE | ID: mdl-31646479

Membrane receptors that are activated by the purine nucleoside adenosine (adenosine receptors) or by purine or pyrimidine nucleotides (P2Y and P2X receptors) transduce extracellular signals to the cytosol. They play important roles in physiology and disease. The G protein-coupled adenosine receptors comprise four subtypes: A1, A2A, A2B, and A3. The G-protein-coupled P2Y receptors are subdivided into eight subtypes: P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14, while the P2X receptors represent ATP-gated homomeric or heteromeric ion channels consisting of three subunits; the most important subunits are P2X1, P2X2, P2X3, P2X4, and P2X7. This chapter provides guidance for selecting suitable tool compounds for studying these large and important purine receptor families.


Adenosine Triphosphate/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic/chemistry , Receptors, Purinergic/physiology , Animals , Humans , Signal Transduction
11.
Sci Rep ; 9(1): 17224, 2019 11 20.
Article En | MEDLINE | ID: mdl-31748569

Myocardial ischemia evokes powerful reflex responses through activation of vagal and sympathetic afferents in the heart through the release of ischemic metabolites. We have demonstrated that extracellular ATP stimulates cardiac sympathetic afferents through P2 receptor-mediated mechanism, and that opioid peptides suppress these afferents' activity. However, the roles of both P2 receptor and endogenous opioids in cardiac sympathoexcitatory reflex (CSR) responses remain unclear. We therefore hypothesized that activation of cardiac P2 receptor evokes CSR responses by stimulating cardiac sympathetic afferents and these CSR responses are modulated by endogenous opioids. We observed that intrapericardial injection of α,ß-methylene ATP (α,ß-meATP, P2X receptor agonist), but not ADP (P2Y receptor agonist), caused a graded increase in mean arterial pressure in rats with sinoaortic denervation and vagotomy. This effect of α,ß-meATP was abolished by blockade of cardiac neural transmission with intrapericardial procaine treatment and eliminated by intrapericardial A-317491, a selective P2X2/3 and P2X3 receptor antagonist. Intrapericardial α,ß-meATP also evoked CSR response in vagus-intact rats. Furthermore, the P2X receptor-mediated CSR responses were enhanced by intrapericardial naloxone, a specific opioid receptor antagonist. These data suggest that stimulation of cardiac P2X2/3 and P2X3, but not P2Y receptors, powerfully evokes CSR responses through activation of cardiac spinal afferents, and that endogenous opioids suppress the P2X receptor-mediated CSR responses.


Heart/physiology , Receptors, Opioid/metabolism , Receptors, Purinergic P2X/metabolism , Reflex/physiology , Sympathetic Nervous System/physiology , Anesthetics, Local/pharmacology , Animals , Gene Expression Regulation , Heart/drug effects , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Procaine/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid/genetics , Receptors, Purinergic P2X/chemistry , Receptors, Purinergic P2X/genetics , Reflex/drug effects , Sympathetic Nervous System/drug effects
12.
Sci Rep ; 9(1): 11613, 2019 08 12.
Article En | MEDLINE | ID: mdl-31406184

Sterile inflammation is a key determinant of myocardial reperfusion injuries. It participates in infarct size determination in acute myocardial infarction and graft rejection following heart transplantation. We previously showed that P2Y11 exerted an immunosuppressive role in human dendritic cells, modulated cardiofibroblasts' response to ischemia/reperfusion in vitro and delayed graft rejection in an allogeneic heterotopic heart transplantation model. We sought to investigate a possible role of P2Y11 in the cellular response of cardiomyocytes to ischemia/reperfusion. We subjected human AC16 cardiomyocytes to 5 h hypoxia/1 h reoxygenation (H/R). P2Y11R (P2Y11 receptor) selective agonist NF546 and/or antagonist NF340 were added at the onset of reoxygenation. Cellular damages were assessed by LDH release, MTT assay and intracellular ATP level; intracellular signaling pathways were explored. The role of P2Y11R in mitochondria-derived ROS production and mitochondrial respiration was investigated. In vitro H/R injuries were significantly reduced by P2Y11R stimulation at reoxygenation. This protection was suppressed with P2Y11R antagonism. P2Y11R stimulation following H2O2-induced oxidative stress reduced mitochondria-derived ROS production and damages through PKCε signaling pathway activation. Our results suggest a novel protective role of P2Y11 in cardiomyocytes against reperfusion injuries. Pharmacological post-conditioning targeting P2Y11R could therefore contribute to improve myocardial ischemia/reperfusion outcomes in acute myocardial infarction and cardiac transplantation.


Myocytes, Cardiac/drug effects , Protein Kinase C-epsilon/metabolism , Receptors, Purinergic P2/drug effects , Reperfusion Injury/prevention & control , Signal Transduction , Adenosine Triphosphate/administration & dosage , Cardiotonic Agents/pharmacology , Heart Transplantation , Humans , Myocardial Infarction/prevention & control , Myocytes, Cardiac/enzymology , Oxygen/metabolism , Purinergic P2 Receptor Agonists/pharmacology
13.
Cephalalgia ; 39(11): 1421-1434, 2019 Oct.
Article En | MEDLINE | ID: mdl-31104506

BACKGROUND: The current understanding of mechanisms behind migraine pain has been greatly enhanced with the recent therapies targeting calcitonin gene-related peptide and its receptor. The clinical efficacy of calcitonin gene-related peptide-blocking drugs indicates that, at least in a considerable proportion of patients, calcitonin gene-related peptide is a key molecule in migraine pain. There are several receptors and molecular pathways that can affect the release of and response to calcitonin gene-related peptide. One of these could be purinergic receptors that are involved in nociception, but these are greatly understudied with respect to migraine. OBJECTIVE: We aimed to explore purinergic receptors as potential anti-migraine targets. METHODS: We used the human middle meningeal artery as a proxy for the trigeminal system to screen for possible anti-migraine candidates. The human findings were followed by intravital microscopy and calcitonin gene-related peptide release measurements in rodents. RESULTS: We show that the purinergic P2Y13 receptor fulfills all the features of a potential anti-migraine target. The P2Y13 receptor is expressed in both the human trigeminal ganglion and middle meningeal artery and activation of this receptor causes: a) middle meningeal artery contraction in vitro; b) reduced dural artery dilation following periarterial electrical stimulation in vivo and c) a reduction of CGRP release from both the dura and the trigeminal ganglion in situ. Furthermore, we show that P2X3 receptor activation of the trigeminal ganglion causes calcitonin gene-related peptide release and middle meningeal artery dilation. CONCLUSION: Both an agonist directed at the P2Y13 receptor and an antagonist of the P2X3 receptor seem to be viable potential anti-migraine therapies.


Meningeal Arteries/drug effects , Migraine Disorders/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Aged , Aged, 80 and over , Animals , Calcitonin Gene-Related Peptide/drug effects , Calcitonin Gene-Related Peptide/metabolism , Disease Models, Animal , Female , Humans , Male , Meningeal Arteries/metabolism , Middle Aged , Rats, Sprague-Dawley , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/metabolism , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism
14.
J Thorac Cardiovasc Surg ; 158(3): 780-790.e1, 2019 09.
Article En | MEDLINE | ID: mdl-30711276

OBJECTIVE: Myocardial ischemia reperfusion is a major cause of cell injury during cardiac transplantation and is responsible for increased graft rejection. Several in vitro studies demonstrated the protective effect of P2Y11-like purinoreceptor stimulation in the context of myocardial ischemia/reperfusion. In this study, we hypothesized a possible cardioprotective role of P2Y11R stimulation against ischemia/reperfusion lesions and validated its clinical effect in vivo in a heart transplantation model. METHODS: We subjected H9c2 rat cardiomyocyte-derived cell line to 5 hours of hypoxia and 1 hour of reoxygenation. P2Y11R selective agonist NF546 and antagonist NF340 were added at the onset of reoxygenation. Cell injuries were assessed by microculture tetrazolium reduction and intracellular adenosine triphosphate level. Clinical effect of P2Y11R stimulation was further investigated in vivo. Hearts from BALB/c mice were transplanted intra-abdominally into allogenic C57BL/6 mice (n = 104). Recipient mice were injected with P2Y11R agonist. Mice in the sham group were injected with saline solution. In the control group, hearts from C57BL/6 were transplanted into syngeneic C57BL/6 mice. Rejection lesions were investigated using histology and immunohistochemistry at days 3, 5, and 7 after transplantation. We measured caspase activities to quantify apoptosis. Production of proinflammatory and anti-inflammatory cytokines was investigated. RESULTS: P2Y11R stimulation at the onset of reoxygenation significantly reduced in vitro hypoxia/reoxygenation injuries. This protection was suppressed with P2Y11R antagonist. In vivo, cardiac allograft survival was significantly prolonged after P2Y11R stimulation. Rejection lesions, classified according to the International Society of Heart Lung Transplantation guidelines and quantified using the mean number of inflammatory cells per field, were significantly reduced in the treated group. At day 5 after transplantation, P2Y11R agonist pretreated allografts also demonstrated less apoptotic lesions. CONCLUSIONS: Our data suggest a novel cardioprotective role of P2Y11R at the onset of reoxygenation/reperfusion against reperfusion injuries. Pharmacologic conditioning using P2Y11 agonist may be beneficial after cardiac transplantation in improving myocardial ischemia/reperfusion outcomes and decreasing graft rejection lesions.


Diphosphonates/pharmacology , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Naphthalenesulfonates/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P2/drug effects , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Line , Cytokines/metabolism , Disease Models, Animal , Female , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Receptors, Purinergic P2/metabolism , Signal Transduction/drug effects , Time Factors
15.
J Neurosci Res ; 96(2): 253-264, 2018 02.
Article En | MEDLINE | ID: mdl-28752899

Oxidative stress and neural degeneration have been shown to be involved in the pathogenesis of Parkinson's disease (PD). The P2Y6 purinergic receptor (P2Y6R) has been shown to participate in the activation of microglia and the production of pro-inflammatory factors induced by lipopolysaccharide to cause neuronal loss. However, the function of P2Y6R during oxidative stress in neurons is unclear. In the present study, 1-methyl-4-phenylpyridinium (MPP+ ) treatment increased the level of UDP/P2Y6R on neuronal SH-SY5Y cells. Importantly, pharmacological inhibition of P2Y6R or knockdown of P2Y6R using a siRNA exerted an increased protective effect by preventing MPP+ -induced increases in the levels of reactive oxygen species (ROS), superoxide anion, inducible nitric oxide synthase (iNOS), and malondialdehyde (MDA) and down-regulation of superoxide dismutase 1 (SOD1) expression. UDP, an agonist of P2Y6R, enhanced the effects of MPP+ , which was also inhibited by apyrase or MRS2578. Additionally, P2Y6R knockdown also significantly reversed both the loss of cell viability and the increase in the levels of phosphorylated extracellular signal-regulated protein kinase (p-ERK1/2) and p38 (p-p38) caused by MPP+ stimulation. However, the inhibition of the ERK1/2 and p38 kinase signaling pathways had no effect on P2Y6R expression. Taken together, these results support the hypothesis that P2Y6R expressed on neuronal SH-SY5Y cell is associated with the progression of oxidative stress and cell death induced by MPP+ , suggesting that P2Y6R may play an important role in the pathogenesis of PD.


Cell Death/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Herbicides/pharmacology , Oxidative Stress/drug effects , Receptors, Purinergic P2/metabolism , 1-Methyl-4-phenylpyridinium/pharmacology , Apyrase/pharmacology , Cell Line, Tumor , Humans , Isothiocyanates/pharmacology , Malondialdehyde/metabolism , Neuroblastoma/pathology , Nitric Oxide Synthase Type II/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2/genetics , Superoxide Dismutase-1/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology , Transfection , Uridine Diphosphate/pharmacology
16.
Sci Rep ; 7(1): 6050, 2017 07 20.
Article En | MEDLINE | ID: mdl-28729619

Purinergic P2Y 2 receptors, G-protein coupled receptors that primarily couple with Gαq/11-proteins, are activated equipotently by adenosine-5'-triphosphate (ATP) and uridine-5'-triphosphate. Evidence suggests that P2Y 2 agonists make potential drug candidates for the treatment of cardiovascular diseases. However, selective non-nucleotide, small-molecule P2Y 2 agonists have yet to be developed. In this report, we discuss Compound 89, a novel non-nucleotide allosteric P2Y 2 agonist that was active in signal transduction and gene induction, and in our in vitro cardiac hypertrophy model. Compound 89 exhibited selective P2Y 2 agonistic activity and potentiated responses to the endogenous agonist ATP, while exhibiting no agonistic activities for four other Gαq/11-coupled human P2Y (hP2Y) receptors and one representative Gαi/o-coupled hP2Y12 receptor. Its P2Y 2 agonistic effect on mouse P2Y 2 receptors suggested non-species-specific activity. Compound 89 acted as a pure positive allosteric modulator in a Ca2+ mobilization assay of neonatal rat cardiomyocytes; it potentiated ATP-induced expression of genes in the nuclear receptor 4A family (negative regulators of hypertrophic stimuli in cardiomyocytes). Additionally, Compound 89 attenuated isoproterenol-induced cardiac hypertrophy, presumably through dose-dependent interaction with pericellular ATP. These results indicate that Compound 89 is potentially efficacious against cardiomyocytes and therefore a good proof-of-concept tool for elucidating the therapeutic potential of P2Y2 activation in various cardiovascular diseases.


Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Quinolones/pharmacology , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Animals , Animals, Newborn , Calcium/metabolism , Female , Gene Expression Regulation/drug effects , Purinergic P2 Receptor Agonists/chemistry , Quinolones/chemistry , Rats
17.
Am J Physiol Renal Physiol ; 313(2): F361-F369, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28468962

We recently reported that natriuresis produced by renal medullary salt loading is dependent on endothelin (ET)-1 and purinergic (P2) receptors in male rats. Because sex differences in ET-1 and P2 signaling have been reported, we decided to test whether ovarian sex hormones regulate renal medullary ET-1 and P2-dependent natriuresis. The effect of medullary NaCl loading on Na+ excretion was determined in intact and ovariectomized (OVX) female Sprague-Dawley rats with and without ET-1 or P2 receptor antagonism. Isosmotic saline (284 mosmol/kgH2O) was infused in the renal medullary interstitium of anesthetized rats during a baseline urine collection period, followed by isosmotic or hyperosmotic saline (1,800 mosmol/kgH2O) infusion. Medullary NaCl loading significantly enhanced Na+ excretion in intact and OVX female rats. ETA+B or P2 receptor blockade did not attenuate the natriuretic effect of medullary NaCl loading in intact females, whereas ETA+B or P2 receptor blockade attenuated the natriuretic response to NaCl loading in OVX rats. Activation of medullary P2Y2 and P2Y4 receptors by UTP infusion had no significant effect in intact females but enhanced Na+ excretion in OVX rats. Combined ETA+B receptor blockade significantly inhibited the natriuretic response to UTP observed in OVX rats. These data demonstrate that medullary NaCl loading induces ET-1 and P2-independent natriuresis in intact females. In OVX, activation of medullary P2 receptors promotes ET-dependent natriuresis, suggesting that ovarian hormones may regulate the interplay between the renal ET-1 and P2 signaling systems to facilitate Na+ excretion.


Endothelin-1/metabolism , Kidney Medulla/metabolism , Natriuresis , Ovariectomy , Receptors, Purinergic P2Y2/metabolism , Receptors, Purinergic P2/metabolism , Renal Elimination , Sodium/urine , Animals , Endothelin Receptor Antagonists/pharmacology , Endothelin-1/genetics , Female , Kidney Medulla/drug effects , Natriuresis/drug effects , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Rats, Sprague-Dawley , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2Y2/drug effects , Renal Elimination/drug effects , Signal Transduction , Sodium Chloride/administration & dosage , Sodium Chloride/metabolism , Time Factors
18.
Am J Physiol Renal Physiol ; 311(2): F260-7, 2016 08 01.
Article En | MEDLINE | ID: mdl-27226106

Renal endothelin-1 (ET-1) and purinergic signaling systems regulate Na(+) reabsorption in the renal medulla. A link between the renal ET-1 and purinergic systems was demonstrated in vitro, however, the in vivo interaction between these systems has not been defined. To test whether renal medullary activation of purinergic (P2) receptors promotes ET-dependent natriuresis, we determined the effect of increased medullary NaCl loading on Na(+) excretion and inner medullary ET-1 mRNA expression in anesthetized adult male Sprague-Dawley rats in the presence and absence of purinergic receptor antagonism. Isosmotic saline (NaCl; 284 mosmol/kgH2O) was infused into the medullary interstitium (500 µl/h) during a 30-min baseline urine collection period, followed by isosmotic or hyperosmotic saline (1,800 mosmol/kgH2O) for two further 30-min urine collection periods. Na(+) excretion was significantly increased during intramedullary infusion of hyperosmotic saline. Compared with isosmotic saline, hyperosmotic saline infused into the renal medulla caused significant increases in inner medullary ET-1 mRNA expression. Renal intramedullary infusion of the P2 receptor antagonist suramin inhibited the increase in Na(+) excretion and inner medullary ET-1 mRNA expression induced by NaCl loading in the renal medulla. Activation of medullary P2Y2/4 receptors by infusion of UTP increased urinary Na(+) excretion. Combined ETA and ETB receptor blockade abolished the natriuretic response to intramedullary infusion of UTP. These data demonstrate that activation of medullary P2 receptors promotes ET-dependent natriuresis in male rats, suggesting that the renal ET-1 and purinergic signaling systems interact to efficiently facilitate excretion of a NaCl load.


Endothelin-1/physiology , Kidney Medulla/drug effects , Natriuresis/drug effects , Purinergic P2 Receptor Agonists/pharmacology , Animals , Endothelin-1/biosynthesis , Male , Osmolar Concentration , Potassium/urine , Purinergic P2 Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A/drug effects , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/drug effects , Receptor, Endothelin B/metabolism , Receptors, Purinergic P2X4/drug effects , Sodium/metabolism , Sodium/urine , Sodium Chloride/pharmacology , Uridine Triphosphate/pharmacology
19.
Am J Physiol Renal Physiol ; 311(1): F103-11, 2016 07 01.
Article En | MEDLINE | ID: mdl-27076649

Purinoceptors (adrengeric receptors and P2 receptors) are expressed on the cellular components of the glomerular filtration barrier, and their activation may affect glomerular permeability to albumin, which may ultimately lead to albuminuria, a well-established risk factor for the progression of chronic kidney disease and development of cardiovascular diseases. We investigated the mechanisms underlying the in vitro and in vivo purinergic actions on glomerular filter permeability to albumin by measuring convectional albumin permeability (Palb) in a single isolated rat glomerulus based on the video microscopy method. Primary cultured rat podocytes were used for the analysis of Palb, cGMP accumulation, PKG-Iα dimerization, and immunofluorescence. In vitro, natural nucleotides (ATP, ADP, UTP, and UDP) and nonmetabolized ATP analogs (2-meSATP and ATP-γ-S) increased Palb in a time- and concentration-dependent manner. The effects were dependent on P2 receptor activation, nitric oxide synthase, and cytoplasmic guanylate cyclase. ATP analogs significantly increased Palb, cGMP accumulation, and subcortical actin reorganization in a PKG-dependent but nondimer-mediated route in cultured podocytes. In vivo, 2-meSATP and ATP-γ-S increased Palb but did not significantly affect urinary albumin excretion. Both agonists enhanced the clathrin-mediated endocytosis of albumin in podocytes. A product of adenine nucleotides hydrolysis, adenosine, increased the permeability of the glomerular barrier via adrenergic receptors in a dependent and independent manner. Our results suggest that the extracellular nucleotides that stimulate an increase of glomerular Palb involve nitric oxide synthase and cytoplasmic guanylate cyclase with actin reorganization in podocytes.


Albumins/metabolism , Albuminuria/metabolism , Kidney Glomerulus/metabolism , Purines/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Albuminuria/pathology , Animals , Cyclic GMP/metabolism , Endocytosis/drug effects , Female , Guanylate Cyclase/biosynthesis , In Vitro Techniques , Kidney Glomerulus/drug effects , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Permeability/drug effects , Podocytes/drug effects , Podocytes/metabolism , Primary Cell Culture , Purinergic P2 Receptor Agonists/pharmacology , Rats , Rats, Wistar
20.
J Leukoc Biol ; 99(2): 289-99, 2016 Feb.
Article En | MEDLINE | ID: mdl-26382298

Macrophages represent a highly heterogenic cell population of the innate immune system, with important roles in the initiation and resolution of the inflammatory response. Purinergic signaling regulates both M1 and M2 macrophage function at different levels by controlling the secretion of cytokines, phagocytosis, and the production of reactive oxygen species. We found that extracellular nucleotides arrest macrophage differentiation from bone marrow precursors via adenosine and P2 receptors. This results in a mature macrophage with increased expression of M2, but not M1, genes. Similar to adenosine and ATP, macrophage growth arrested with LPS treatment resulted in an increase of the M2-related marker Ym1. Recombinant Ym1 was able to affect macrophage proliferation and could, potentially, be involved in the arrest of macrophage growth during hematopoiesis.


Inflammation/immunology , Macrophage Activation/drug effects , Macrophages/drug effects , Myelopoiesis/drug effects , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P1/physiology , Receptors, Purinergic P2/physiology , Adenine/pharmacology , Adenine Nucleotides/pharmacology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Inflammation/pathology , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Lectins/biosynthesis , Lectins/genetics , Lectins/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/classification , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/biosynthesis , Receptors, Purinergic P2X7/deficiency , Receptors, Purinergic P2X7/drug effects , Receptors, Purinergic P2X7/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Specific Pathogen-Free Organisms , beta-N-Acetylhexosaminidases/biosynthesis , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/pharmacology
...