Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters











Publication year range
1.
Mol Neurobiol ; 61(2): 1002-1021, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37676390

ABSTRACT

There are no effective treatments for post-stroke glial scar formation, which inhibits axonal outgrowth and functional recovery after stroke. We investigated whether astrocytic extracellular vesicles (AEVs) regulated by microglia modulate glial scars and improve stroke recovery. We found that peri-infarct glial scars comprised reactive astrocytes with proliferating C3d and decreased S100A10 expression in chronic stroke. In cultured astrocytes, microglia-conditioned media and treatment with P2Y1 receptor antagonists increased and reduced the area of S100A10- and C3d-expressing reactive astrocytes, respectively, by suppressing mitogen-activated protein kinase/nuclear factor-κß (NF-κB)/tumor necrosis factor-α (TNF-α)/interleukin-1ß signaling after oxygen-glucose deprivation. Intracerebral administrations of AEVs enriched miR-146a-5p, downregulated NF-κB, and suppressed TNF-α expressions, by transforming reactive astrocytes to those with S100A10 preponderance, causing functional recovery in rats subjected to middle cerebral artery occlusion. Modulating neuroinflammation in post-stroke glial scars could permit axonal outgrowth, thus providing a basis for stroke recovery with neuroprotective AEVs.


Subject(s)
Extracellular Vesicles , Stroke , Rats , Animals , Microglia/metabolism , NF-kappa B/metabolism , Astrocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Gliosis/pathology , Stroke/pathology , Extracellular Vesicles/metabolism
2.
Cells ; 12(14)2023 07 13.
Article in English | MEDLINE | ID: mdl-37508508

ABSTRACT

Astrocytes are critical players in brain health and disease. Brain pathologies and lesions are usually accompanied by astroglial alterations known as reactive astrogliosis. Sphingosine 1-phosphate lyase (SGPL1) catalysis, the final step in sphingolipid catabolism, irreversibly cleaves its substrate sphingosine 1-phosphate (S1P). We have shown that neural ablation of SGPL1 causes accumulation of S1P and hence neuronal damage, cognitive deficits, as well as microglial activation. Moreover, the S1P/S1P-receptor signaling axis enhances ATP production in SGPL1-deficient astrocytes. Using immunohistochemical methods as well as RNA Seq and CUT&Tag we show how S1P signaling causes activation of the astrocytic purinoreceptor P2Y1 (P2Y1R). With specific pharmacological agonists and antagonists, we uncover the P2Y1R as the key player in S1P-induced astrogliosis, and DDX3X mediated the activation of the NLRP3 inflammasome, including caspase-1 and henceforward generation of interleukin-1ß (IL-1ß) and of other proinflammatory cytokines. Our results provide a novel route connecting S1P metabolism and signaling with astrogliosis and the activation of the NLRP3 inflammasome, a central player in neuroinflammation, known to be crucial for the pathogenesis of numerous brain illnesses. Thus, our study opens the door for new therapeutic strategies surrounding S1P metabolism and signaling in the brain.


Subject(s)
Inflammasomes , Lyases , Brain , Gliosis , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Mice
3.
Cell Signal ; 109: 110805, 2023 09.
Article in English | MEDLINE | ID: mdl-37437828

ABSTRACT

Genetically encoded Ca2+ indicators have become widely used in cell signalling studies as they offer advantages over cell-loaded dye indicators in enabling specific cellular or subcellular targeting. Comparing responses from dye and protein-based indicators may provide information about indicator properties and cell physiology, but side-by-side recordings in cells are scarce. In this study, we compared cytoplasmic Ca2+ concentration ([Ca2+]i) changes in insulin-secreting ß-cells recorded with commonly used dyes and indicators based on circularly permuted fluorescent proteins. Total internal reflection fluorescence (TIRF) imaging of K+ depolarization-triggered submembrane [Ca2+]i increases showed that the dyes Fluo-4 and Fluo-5F mainly reported stable [Ca2+]i elevations, whereas the proteins R-GECO1 and GCaMP5G more often reported distinct [Ca2+]i spikes from an elevated level. [Ca2+]i spiking occurred also in glucose-stimulated cells. The spikes reflected Ca2+ release from the endoplasmic reticulum, triggered by autocrine activation of purinergic receptors after exocytotic release of ATP and/or ADP, and the spikes were consequently prevented by SERCA inhibition or P2Y1-receptor antagonism. Widefield imaging, which monitors the entire cytoplasm, increased the spike detection by the Ca2+ dyes. The indicator-dependent response patterns were unrelated to Ca2+ binding affinity, buffering and mobility, and probably reflects the much slower dissociation kinetics of protein compared to dye indicators. Ca2+ dyes thus report signalling within the submembrane space excited by TIRF illumination, whereas the protein indicators also catch Ca2+ events originating outside this volume. The study highlights that voltage-dependent Ca2+ entry in ß-cells is tightly linked to local intracellular Ca2+ release mediated via an autocrine route that may be more important than previously reported direct Ca2+ effects on phospholipase C or on intracellular channels mediating calcium-induced calcium release.


Subject(s)
Calcium , Insulin-Secreting Cells , Calcium/metabolism , Insulin-Secreting Cells/metabolism , Signal Transduction , Endoplasmic Reticulum/metabolism , Coloring Agents/metabolism , Coloring Agents/pharmacology , Calcium Signaling , Adenosine Triphosphate/metabolism
4.
J Thromb Haemost ; 21(7): 1891-1902, 2023 07.
Article in English | MEDLINE | ID: mdl-36958516

ABSTRACT

BACKGROUND: The hemostatic plug formation at sites of vascular injury is strongly dependent on rapid platelet activation and integrin-mediated adhesion and aggregation. However, to prevent thrombotic complications, platelet aggregate formation must be a self-limiting process. The second-wave mediator adenosine diphosphate (ADP) activates platelets via Gq-coupled P2Y1 and Gi-coupled P2Y12 receptors. After ADP exposure, the P2Y1 receptor undergoes rapid phosphorylation-induced desensitization, a negative feedback mechanism believed to be critical for limiting thrombus growth. OBJECTIVE: The objective of this study was to examine the role of rapid P2Y1 receptor desensitization on platelet function and thrombus formation in vivo. METHODS: We analyzed a novel knock-in mouse strain expressing a P2Y1 receptor variant that cannot be phosphorylated beyond residue 340 (P2Y1340-0P), thereby preventing the desensitization of the receptor. RESULTS: P2Y1340-0P mice followed a Mendelian inheritance pattern, and peripheral platelet counts were comparable between P2Y1340-0P/340-0P and control mice. In vitro, P2Y1340-0P/340-0P platelets were hyperreactive to ADP, showed a robust activation response to the P2Y1 receptor-selective agonist, MRS2365, and did not desensitize in response to repeated ADP challenge. We observed increased calcium mobilization, protein kinase C substrate phosphorylation, alpha granule release, activation of the small GTPase Rap1, and integrin inside-out activation/aggregation. This hyperreactivity, however, did not lead to increased platelet adhesion or excessive plug formation under physiological shear conditions. CONCLUSION: Our studies demonstrate that receptor phosphorylation at the C-terminus is critical for P2Y1 receptor desensitization in platelets and that impaired desensitization leads to increased P2Y1 receptor signaling in vitro. Surprisingly, desensitization of the P2Y1 receptor is not required for limiting platelet adhesion/aggregation at sites of vascular injury, likely because ADP is degraded quickly or washed away in the bloodstream.


Subject(s)
Thrombosis , Vascular System Injuries , Mice , Animals , Platelet Aggregation , Blood Platelets/metabolism , Hemostasis , Thrombosis/genetics , Thrombosis/prevention & control , Thrombosis/metabolism , Adenosine Diphosphate/pharmacology , Integrins/metabolism , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/metabolism
5.
Exp Ther Med ; 25(1): 67, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36605532

ABSTRACT

The aim of the present study was to explore the expression changes of P2Y purinergic receptor 1 (P2Y1) in the distal colonic submucosa of opioid-induced constipation (OIC) rats and its association with the occurrence of OIC, an OIC rat model was generated by intraperitoneal injection of loperamide hydrochloride, a selective agonist of µ-opioid receptors (MORs). At 7 days post-treatment, the model was assessed by analyzing stool scores and calculating the gastrointestinal (GI) transit ratio of rats. The distribution of P2Y1-expressing neurons in the colonic submucosal plexus was demonstrated by immunofluorescence (IF). Western blotting was performed to evaluate the expression changes of MOR, P2Y1 and ATP synthase subunit ß (ATPB) proteins in the colonic submucosa, while reverse transcription-quantitative PCR (RT-qPCR) analysis was performed to determine the relative mRNA expression of MOR and P2Y1. After 7 days, the feces of OIC rats exhibited an appearance of sausage-shaped pieces and both the stool weight and GI transit ratio of OIC rats were significantly decreased. IF revealed co-expression of P2Y1 and calbindin and MOR and ATPB in the nerve cells of the distal colonic submucosal plexus. Moreover, RT-qPCR analysis showed that the MOR mRNA levels were significantly increased in the distal colonic submucosa of OIC rats, while mRNA levels of P2Y1 were decreased. WB showed that in the distal colonic submucosa of OIC rats, MOR protein expression was increased, whereas that of P2Y1 was significantly decreased. GI transit ratio analysis suggested that the P2Y agonist ATP significantly relieved constipation symptoms in rats, while the P2Y inhibitor MRS2179 aggravated these symptoms. Finally, P2Y1 expression change was shown to be associated with the occurrence of OIC, while expression of MOR and P2Y1 was associated with OIC development in rats.

6.
Purinergic Signal ; 19(1): 55-68, 2023 03.
Article in English | MEDLINE | ID: mdl-35094240

ABSTRACT

Depression is a common neuropsychiatric disorder with high incidence and disability. Electroacupuncture (EA) is effective in the treatment of depression. However, the underlying mechanisms are not fully understood. Social isolation stress during post-weaning period can impair purinergic signaling in the brain of rodents and has emerged as a major risk factor for depression. The purpose of this study was to investigate the involvement of P2Y1 receptor (P2Y1R) in the antidepressant-like effects of EA. In this study, C57BL/6 mice were randomly assigned to group-housed (GH) or social isolated (SI) groups at post-natal day 21. After 6 weeks of social isolation, EA was performed on acupoints "Bai-hui" (GV20) and "Yin-tang" (GV29), or non-acupoints for 4 weeks. The SI mice received either intracerebroventricular injection of a selective P2Y1R agonist, MRS2365 (1 nmol); or a selective P2Y1R antagonist, MRS2179 (2 µmol), before and after EA. We found that SI mice exhibited depression-like behaviors accompanied with anxiety-like behaviors. The expressions of P2Y1R were well co-localized with GFAP-positive astrocytes and increased in the prefrontal cortex and hippocampus of SI mice. After treated with MRS2179, the depression-like behaviors of SI mice were attenuated, but not with MRS2365. Meanwhile, we found that EA could attenuate social isolation caused depression- and anxiety-like behaviors, and inhibited the up-regulation of P2Y1R in the prefrontal cortex and hippocampus of SI mice. Notably, the positive effects of EA on depression-like behaviors of SI mice could be reversed by MRS2365, while MRS2365 had no effect on the anxiolytic-like effects of EA. Therefore, we provide new evidence that EA could ameliorate depression- and anxiety-like behaviors in social isolation stress mice, and P2Y1R was involved in the antidepressant-like effects of EA.


Subject(s)
Electroacupuncture , Mice , Animals , Receptors, Purinergic P2Y1/metabolism , Mice, Inbred C57BL , Antidepressive Agents , Hippocampus/metabolism , Receptors, Purinergic/metabolism , Social Isolation
7.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1032094

ABSTRACT

@#Objective To investigate the role of P2Y1 receptors and astrocytes in delayed encephalopathy after acute CO poisoning (DEACMP) and the possible pathogenesis of DEACMP.Methods Male SD rats with acceptable cognitive function were screened by water maze test and randomly divided into two groups:the control group and the CO poisoning group.The poisoning group was subjected to DEACMP model.The behavioral changes,neuronal changes and the expressions of P2Y1 receptor and astrocytes in hippocampus of the two groups were compared at 7,14,21 and 28 d after modeling,respectively.Results Compared with the control group,the escape latencies of rats in the poisoning group were significantly prolonged on the 21st and 28th days after modeling (P<0.05).HE staining showed that the hippocampal pyramidal cells and neurons in the model group exhibited obvious necrosis on days 14,21,and 28 after modeling.The water maze indicated that DEACMP occurred on day 21.Compared with the control group,Western blot analysis showed that the expression levels of P2Y1 and GFAP proteins in the hippocampus of the poisoning group were increased at each time point (P<0.05),which increased first and then decreased.Immunofluorescence showed co-expression of P2Y1 and GFAP in hippocampus.Compared with the control group,the expressions of P2Y1 and GFAP in hippocampal CA1 region were up-regulated at each time point after poisoning (P<0.05). Conclusion The activation of astrocytes by P2Y1 receptor may be one of the pathogenesis of DEACMP,and astrocytes may impair learning and memory ability of co-poisoned rats by mediating immune inflammation,leading to DEACMP.

8.
Dev Cell ; 57(18): 2221-2236.e5, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36108628

ABSTRACT

Pulmonary neuroendocrine cells (PNECs) are rare airway cells with potential sensory capacity linked to vagal neurons and immune cells. How PNECs sense and respond to external stimuli remains poorly understood. We discovered PNECs located within pig and human submucosal glands, a tissue that produces much of the mucus that defends the lung. These PNECs sense succinate, an inflammatory molecule in liquid lining the airway surface. The results indicate that succinate migrates down the submucosal gland duct to the acinus, where it triggers apical succinate receptors, causing PNECs to release ATP. The short-range ATP signal stimulates the contraction of myoepithelial cells wrapped tightly around the submucosal glands. Succinate-triggered gland contraction may complement the action of neurotransmitters that induce mucus release but not gland contraction to promote mucus ejection onto the airway surface. These findings identify a local circuit in which rare PNECs within submucosal glands sense an environmental cue to orchestrate the function of airway glands.


Subject(s)
Neuroendocrine Cells , Adenosine Triphosphate/metabolism , Animals , Humans , Lung/metabolism , Mucus/metabolism , Succinic Acid/metabolism , Swine
9.
J Biomol Struct Dyn ; 40(23): 12908-12916, 2022.
Article in English | MEDLINE | ID: mdl-34542380

ABSTRACT

The human Guanine Protein coupled membrane Receptor 17 (hGPR17), an orphan receptor that activates uracil nucleotides and cysteinyl leukotrienes is considered as a crucial target for the neurodegenerative diseases. Yet, the detailed molecular interaction of potential synthetic ligands of GPR17 needs to be characterized. Here, we have studied a comparative analysis on the interaction specificity of GPR17-ligands with hGPR17 and human purinergic G protein-coupled receptor (hP2Y1) receptors. Previously, we have simulated the interaction stability of synthetic ligands such as T0510.3657, AC1MLNKK, and MDL29951 with hGPR17 and hP2Y1 receptor in the lipid environment. In the present work, we have comparatively studied the protein-ligand interaction of hGPR17-T0510.3657 and P2Y1-MRS2500. Sequence analysis and structural superimposition of hGPR17 and hP2Y1 receptor revealed the similarities in the structural arrangement with the local backbone root mean square deviation (RMSD) value of 1.16 Å and global backbone RMSD value of 5.30 Å. The comparative receptor-ligand interaction analysis between hGPR17 and hP2Y1 receptor exposed the distinct binding sites in terms of geometrical properties. Further, the molecular docking of T0510.3657 with the hP2Y1 receptor have shown non-specific interaction. The experimental validation also revealed that Gi-coupled activation of GPR17 by specific ligands leads to the adenylyl cyclase inhibition, while there is no inhibition upon hP2Y1 activation. Overall, the above findings suggest that T0510.3657-GPR17 binding specificity could be further explored for the treatment of numerous neuronal diseases. Communicated by Ramaswamy H. Sarma.


Subject(s)
Receptors, G-Protein-Coupled , Humans , Receptors, Purinergic P2Y1 , Molecular Docking Simulation , Ligands , Receptors, G-Protein-Coupled/metabolism , Binding Sites
10.
Future Med Chem ; 13(21): 1845-1864, 2021 11.
Article in English | MEDLINE | ID: mdl-34505540

ABSTRACT

Background: (1-(2-hydroxy-5-nitrophenyl)(4-hydroxyphenyl)methyl)indoline-4-carbonitrile (HIC), an agonist of the P2Y1 receptor (P2Y1R), induces cell death in prostate cancer cells. However, the molecular mechanism behind the inhibition of HIC in prostate cancer remains elusive. Methods & results: Here, to outline the inhibitory role of HIC on prostate cancer cells, PC-3 and DU145 cell lines were treated with the respective IC50 concentrations, which reduced cell proliferation, adherence properties and spheroid formation. HIC was able to arrest the cell cycle at G1/S phase and also induced apoptosis and DNA damage, validated by gene expression profiling. HIC inhibited the prostate cancer cells' migration and invasion, revealing its antimetastatic ability. P2Y1R-targeted HIC affects p53, MAPK and NF-κB protein expression, thereby improving the p53 stabilization essential for G1/S arrest and cell death. Conclusion: These findings provide an insight on the potential use of HIC, which remains the mainstay treatment for prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Prostatic Neoplasms/drug therapy , Receptors, Purinergic P2Y1/metabolism , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
11.
J Inflamm Res ; 14: 3367-3392, 2021.
Article in English | MEDLINE | ID: mdl-34305404

ABSTRACT

Treatment of epilepsy remains a clinical challenge, with >30% of patients not responding to current antiseizure drugs (ASDs). Moreover, currently available ASDs are merely symptomatic without altering significantly the progression of the disease. Inflammation is increasingly recognized as playing an important role during the generation of hyperexcitable networks in the brain. Accordingly, the suppression of chronic inflammation has been suggested as a promising therapeutic strategy to prevent epileptogenesis and to treat drug-refractory epilepsy. As a consequence, a strong focus of ongoing research is identification of the mechanisms that contribute to sustained inflammation in the brain during epilepsy and whether these can be targeted. ATP is released in response to several pathological stimuli, including increased neuronal activity within the central nervous system, where it functions as a neuro- and gliotransmitter. Once released, ATP activates purinergic P2 receptors, which are divided into metabotropic P2Y and ionotropic P2X receptors, driving inflammatory processes. Evidence from experimental models and patients demonstrates widespread expression changes of both P2Y and P2X receptors during epilepsy, and critically, drugs targeting both receptor subtypes, in particular the P2Y1 and P2X7 subtypes, have been shown to possess both anticonvulsive and antiepileptic potential. This review provides a detailed summary of the current evidence suggesting ATP-gated receptors as novel drug targets for epilepsy and discusses how P2 receptor-driven inflammation may contribute to the generation of seizures and the development of epilepsy.

12.
Auton Neurosci ; 234: 102830, 2021 09.
Article in English | MEDLINE | ID: mdl-34116466

ABSTRACT

The ATP analogue α,ß-meATP is a potent relaxant of gastrointestinal smooth muscle, but its molecular target is uncertain inside the gut. α,ß-meATP relaxed the carbachol-precontracted guinea-pig taenia coli in a concentration-dependent manner (EC50, 2.0 ± 0.1 µM). A luciferase-based assay confirmed that α,ß-meATP solutions were minimally contaminated with ATP. α,ß-meATP-evoked relaxations were inhibited by the competitive P2Y1 antagonist MRS2179 (pA2 = 5.36), but also by the competitive P2X3 antagonist, A-317491 (pA2 = 5.51). When MRS2179 and A-317491 were applied together, residual α,ß-meATP responses converted from brief to prolonged relaxations. Sodium nitroprusside (a nitric oxide donor) also caused prolonged relaxations. Immunohistochemistry revealed that P2X3 receptors were present in myenteric ganglion cells and their varicose nerve terminals. The amplitude of α,ß-meATP responses was not inhibited by TTX (NaV channel blocker) and ωCgTx (N-type CaV channel blocker). However, responses to α,ß-meATP were inhibited by TEA (non-selective K+-channel blocker), indicating that relaxations involved opening K+-channels. The findings of this study are consistent with the conclusion that α,ß-meATP stimulates Ca2+-permeable P2X3 receptors on varicose nerve terminals to release inhibitory nucleotides: 1) ATP and ß-NAD release results in P2Y1-mediated brief relaxations; 2) another released transmitter (possibly NO) results in prolonged relaxations. Prejunctional P2X3 receptors represent a purinergic feed-forward mechanism to augment the action of inhibitory nerves on gut motility. This positive feed-forward mechanism may counter-balance the known negative feedback mechanism caused by adenosine and prejunctional A1 receptors on inhibitory motor nerves.


Subject(s)
Muscle, Smooth , Receptors, Purinergic P2X3 , Adenosine Triphosphate , Animals , Carbachol , Colon , Guinea Pigs , Receptors, Purinergic
13.
Purinergic Signal ; 17(3): 449-465, 2021 09.
Article in English | MEDLINE | ID: mdl-34050505

ABSTRACT

Extracellular purines are important signaling molecules involved in numerous physiological and pathological processes via the activation of P2 receptors. Information about the spatial and temporal P2 receptor (P2R) expression and its regulation remains crucial for the understanding of the role of P2Rs in health and disease. To identify cells carrying P2X2Rs in situ, we have generated BAC transgenic mice that express the P2X2R subunits as fluorescent fusion protein (P2X2-TagRFP). In addition, we generated a BAC P2Y1R TagRFP reporter mouse expressing a TagRFP reporter for the P2RY1 gene expression. We demonstrate expression of the P2X2R in a subset of DRG neurons, the brain stem, the hippocampus, as well as on Purkinje neurons of the cerebellum. However, the weak fluorescence intensity in our P2X2R-TagRFP mouse precluded tracking of living cells. Our P2Y1R reporter mice confirmed the widespread expression of the P2RY1 gene in the CNS and indicate for the first time P2RY1 gene expression in mouse Purkinje cells, which so far has only been described in rats and humans. Our P2R transgenic models have advanced the understanding of purinergic transmission, but BAC transgenic models appeared not always to be straightforward and permanent reliable. We noticed a loss of fluorescence intensity, which depended on the number of progeny generations. These problems are discussed and may help to provide more successful animal models, even if in future more versatile and adaptable nuclease-mediated genome-editing techniques will be the methods of choice.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Receptors, Purinergic P2X2/biosynthesis , Receptors, Purinergic P2X2/genetics , Receptors, Purinergic P2Y1/biosynthesis , Receptors, Purinergic P2Y1/genetics , Animals , Cells, Cultured , Chromosomes, Artificial, Bacterial/metabolism , Female , Ganglia, Spinal/metabolism , Gene Expression , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Xenopus laevis
14.
Front Pharmacol ; 12: 627773, 2021.
Article in English | MEDLINE | ID: mdl-33679406

ABSTRACT

Cardiac fibroblasts (CFs) activation is a hallmark feature of cardiac fibrosis caused by cardiac remodeling. The purinergic signaling molecules have been proven to participate in the activation of CFs. In this study, we explored the expression pattern of P2Y receptor family in the cardiac fibrosis mice model induced by the transverse aortic constriction (TAC) operation and in the activation of CFs triggered by transforming growth factor ß1 (TGF-ß1) stimulation. We then investigated the role of P2Y1receptor (P2Y1R) in activated CFs. The results showed that among P2Y family members, only P2Y1R was downregulated in the heart tissues of TAC mice. Consistent with our in vivo results, the level of P2Y1R was decreased in the activated CFs, when CFs were treated with TGF-ß1. Silencing P2Y1R expression with siP2Y1R accelerated the effects of TGF-ß1 on CFs activation. Moreover, the P2Y1R selective antagonist BPTU increased the levels of mRNA and protein of profibrogenic markers, such as connective tissue growth factor (CTGF), periostin (POSTN). periostin (POSTN), and α-smooth muscle actin(α-SMA). Further, MRS2365, the agonist of P2Y1R, ameliorated the activation of CFs and activated the p38 MAPK and ERK signaling pathways. In conclusion , our findings revealed that upregulating of P2Y1R may attenuate the abnormal activation of CFs via the p38 MAPK and ERK signaling pathway.

15.
Purinergic Signal ; 16(4): 543-559, 2020 12.
Article in English | MEDLINE | ID: mdl-33129204

ABSTRACT

Rapid phosphoester hydrolysis of endogenous purine and pyrimidine nucleotides has challenged the characterization of the role of P2 receptors in physiology and pathology. Nucleotide phosphoester stabilization has been pursued on a number of medicinal chemistry fronts. We investigated the in vitro and in vivo stability and pharmacokinetics of prototypical nucleotide P2Y1 receptor (P2Y1R) agonists and antagonists. These included the riboside nucleotide agonist 2-methylthio-ADP and antagonist MRS2179, as well as agonist MRS2365 and antagonist MRS2500 containing constrained (N)-methanocarba rings, which were previously reported to form nucleotides that are more slowly hydrolyzed at the α-phosphoester compared with the ribosides. In vitro incubations in mouse and human plasma and blood demonstrated the rapid hydrolysis of these compounds to nucleoside metabolites. This metabolism was inhibited by EDTA to chelate divalent cations required by ectonucleotidases for nucleotide hydrolysis. This rapid hydrolysis was confirmed in vivo in mouse pharmacokinetic studies that demonstrate that MRS2365 is a prodrug of the nucleoside metabolite AST-004 (MRS4322). Furthermore, we demonstrate that the nucleoside metabolites of MRS2365 and 2-methylthio-ADP are adenosine receptor (AR) agonists, notably at A3 and A1ARs. In vivo efficacy of MRS2365 in murine models of traumatic brain injury and stroke can be attributed to AR activation by its nucleoside metabolite AST-004, rather than P2Y1R activation. This research suggests the importance of reevaluation of previous in vitro and in vivo research of P2YRs and P2XRs as there is a potential that the pharmacology attributed to nucleotide agonists is due to AR activation by active nucleoside metabolites.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacokinetics , Adenosine A3 Receptor Agonists/pharmacokinetics , Prodrugs/pharmacokinetics , Purinergic P2Y Receptor Agonists/pharmacokinetics , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacokinetics , Animals , Deoxyadenine Nucleotides/pharmacokinetics , Female , Humans , Mice , Mice, Inbred C57BL , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A3/metabolism , Receptors, Purinergic P2Y1/metabolism
16.
J Neurosci ; 39(27): 5377-5392, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31048325

ABSTRACT

Extracellular ATP activates inflammatory responses to tissue injury. It is also implicated in establishing lasting network hyperexcitability in the brain by acting upon independent receptor systems. Whereas the fast-acting P2X channels have well-established roles driving neuroinflammation and increasing hyperexcitability, the slower-acting metabotropic P2Y receptors have received much less attention. Recent studies of P2Y1 receptor function in seizures and epilepsy have produced contradictory results, suggesting that the role of this receptor during seizure pathology may be highly sensitive to context. Here, by using male mice, we demonstrate that the metabotropic P2Y1 receptor mediates either proconvulsive or anticonvulsive responses, dependent on the time point of activation in relation to the induction of status epilepticus. P2Y1 deficiency or a P2Y1 antagonist (MRS2500) administered before a chemoconvulsant, exacerbates epileptiform activity, whereas a P2Y1 agonist (MRS2365) administered at this time point is anticonvulsant. When these drugs are administered after the onset of status epilepticus, however, their effect on seizure severity is reversed, with the antagonist now anticonvulsant and the agonist proconvulsant. This result was consistent across two different mouse models of status epilepticus (intra-amygdala kainic acid and intraperitoneal pilocarpine). Pharmacologic P2Y1 blockade during status epilepticus reduces also associated brain damage, delays the development of epilepsy and, when applied during epilepsy, suppresses spontaneous seizures, in mice. Our data show a context-specific role for P2Y1 during seizure pathology and demonstrate that blocking P2Y1 after status epilepticus and during epilepsy has potent anticonvulsive effects, suggesting that P2Y1 may be a novel candidate for the treatment of drug-refractory status epilepticus and epilepsy.SIGNIFICANCE STATEMENT This is the first study to fully characterize the contribution of a metabotropic purinergic P2Y receptor during acute seizures and epilepsy. The findings suggest that targeting P2Y1 may offer a potential novel treatment strategy for drug-refractory status epilepticus and epilepsy. Our data demonstrate a context-specific role of P2Y1 activation during seizures, switching from a proconvulsive to an anticonvulsive role depending on physiopathological context. Thus, our study provides a possible explanation for seemingly conflicting results obtained between studies of different brain diseases where P2Y1 targeting has been proposed as a potential treatment strategy and highlights that the timing of pharmacological interventions is of critical importance to the understanding of how receptors contribute to the generation of seizures and the development of epilepsy.


Subject(s)
Brain/physiopathology , Epilepsy/physiopathology , Receptors, Purinergic P2Y1/physiology , Status Epilepticus/physiopathology , Adenosine Diphosphate/administration & dosage , Adenosine Diphosphate/analogs & derivatives , Animals , Brain/drug effects , Deoxyadenine Nucleotides/administration & dosage , Disease Models, Animal , Electroencephalography , Male , Mice, Inbred C57BL , Mice, Knockout , Purinergic P2Y Receptor Agonists/administration & dosage , Purinergic P2Y Receptor Antagonists/administration & dosage , Receptors, Purinergic P2Y1/genetics
17.
Neurosci Lett ; 706: 182-188, 2019 07 27.
Article in English | MEDLINE | ID: mdl-31085293

ABSTRACT

The nodose ganglion (NG) is the main parasympathetic ganglion conveying sensory signals to the CNS from numerous visceral organs including digestive signals such as gastric distension or the release the gastrointestinal peptides. The response characteristics of NG neurons to ATP and ADP and pharmacological interrogation of purinergic receptor subtypes have been previously investigated but often in NG cells of undetermined visceral origin. In this study, we confirmed the presence of P2X3 and P2Y1 receptors and characterized P2X and P2Y responses in gastric-innervating NG neurons. Application of ATP-evoked large inward currents and cytosolic Ca2+ increases in gastric-innervating NG neurons. Despite the expression of P2Y1 receptors, ADP elicited only minor modulation of voltage-gated Ca2+ channels. Considering the sensitivity of NG neurons to comorbidities associated with disease or neural injury, purinergic modulation of gastric NG neurons in disease- or injury-states is worthy of further investigation.


Subject(s)
Nodose Ganglion/metabolism , Receptors, Purinergic/metabolism , Sensory Receptor Cells/metabolism , Stomach/innervation , Vagus Nerve/metabolism , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Male , Nodose Ganglion/drug effects , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects , Stomach/drug effects , Vagus Nerve/drug effects
18.
Mol Neurobiol ; 56(8): 5917-5933, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30689196

ABSTRACT

As mitochondrial dysfunction is evident in neurodegenerative disorders that are accompanied by pain, we generated inducible mutant mice with disruption of mitochondrial respiratory chain complex IV, by COX10 deletion limited to sensory afferent neurons through the use of an Advillin Cre-reporter. COX10 deletion results in a selective energy-deficiency phenotype with minimal production of reactive oxygen species. Mutant mice showed reduced activity of mitochondrial respiratory chain complex IV in many sensory neurons, increased ADP/ATP ratios in dorsal root ganglia and dorsal spinal cord synaptoneurosomes, as well as impaired mitochondrial membrane potential, in these synaptoneurosome preparations. These changes were accompanied by marked pain hypersensitivity in mechanical and thermal (hot and cold) tests without altered motor function. To address the underlying basis, we measured Ca2+ fluorescence responses of dorsal spinal cord synaptoneurosomes to activation of the GluK1 (kainate) receptor, which we showed to be widely expressed in small but not large nociceptive afferents, and is minimally expressed elsewhere in the spinal cord. Synaptoneurosomes from mutant mice showed greatly increased responses to GluK1 agonist. To explore whether altered nucleotide levels may play a part in this hypersensitivity, we pharmacologically interrogated potential roles of AMP-kinase and ADP-sensitive purinergic receptors. The ADP-sensitive P2Y1 receptor was clearly implicated. Its expression in small nociceptive afferents was increased in mutants, whose in vivo pain hypersensitivity, in mechanical, thermal and cold tests, was reversed by a selective P2Y1 antagonist. Energy depletion and ADP elevation in sensory afferents, due to mitochondrial respiratory chain complex IV deficiency, appear sufficient to induce pain hypersensitivity, by ADP activation of P2Y1 receptors.


Subject(s)
Electron Transport Complex IV/genetics , Hypersensitivity/pathology , Mitochondria/metabolism , Mutation/genetics , Neurons, Afferent/pathology , Pain/pathology , Receptors, Purinergic P2Y1/metabolism , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Alkyl and Aryl Transferases/metabolism , Animals , Behavior, Animal , Calcium/metabolism , Cells, Cultured , Electron Transport Complex IV/metabolism , Fluorescence , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hypersensitivity/complications , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Nociception/drug effects , Pain/complications , Phenotype , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Kainic Acid/metabolism , Spinal Cord/pathology , Synapses/drug effects , Synapses/metabolism
19.
Brain Res Bull ; 151: 74-83, 2019 09.
Article in English | MEDLINE | ID: mdl-30593879

ABSTRACT

Nucleotides can contribute to the survival of different glial and neuronal models at the nervous system via activation of purinergic P2X and P2Y receptors. Their activation counteracts different proapoptotic events, such as excitotoxicity, mitochondrial impairment, oxidative stress and DNA damage, which concur to elicit cell loss in different processes of neurodegeneration and brain injury. Thus, it is frequent to find that different neuroprotective mediators converge in the activation of the same intracellular survival pathways to protect cells from death. The present review focuses on the role of P2Y1 and P2Y13 metabotropic receptors, and P2X7 ionotropic receptors to regulate the balance between survival and apoptosis. In particular, we analyze the intracellular pathways involved in the signaling of these nucleotide receptors to elicit survival, including calcium/PLC, PI3K/Akt/GSK3, MAPK cascades, and the expression of antioxidant and antiapoptotic genes. This review emphasizes the novel contribution of nucleotide receptors to maintain cell homeostasis through the regulation of MAP kinases and phosphatases. Unraveling the different roles found for nucleotide receptors in different models and cellular contexts may be crucial to delineate future therapeutic applications based on targeting nucleotide receptors for neuroprotection.


Subject(s)
Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/metabolism , Brain Injuries/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , MAP Kinase Signaling System , Neuroglia/metabolism , Neurons/metabolism , Neuroprotection/physiology , Neuroprotective Agents/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2X/physiology , Receptors, Purinergic P2X7/physiology , Receptors, Purinergic P2Y/metabolism , Receptors, Purinergic P2Y/physiology , Receptors, Purinergic P2Y1/physiology , Signal Transduction
20.
Eur Heart J ; 40(17): 1362-1373, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30395215

ABSTRACT

AIMS: Calcific aortic valve stenosis (CAVS) is characterized by a fibrocalcific process. Studies have shown an association between CAVS and the activation of platelets. It is believed that shear stress associated with CAVS promotes the activation of platelets. However, whether platelets actively participate to the mineralization of the aortic valve (AV) and the progression of CAVS is presently unknown. To identify the role of platelets into the pathobiology of CAVS. METHODS AND RESULTS: Explanted control non-mineralized and mineralized AVs were examined by scanning electron microscope (SEM) for the presence of activated platelets. In-depth functional assays were carried out with isolated human valve interstitial cells (VICs) and platelets as well as in LDLR-/- apoB100/100 IGFII (IGFII) mice. Scanning electron microscope and immunogold markings for glycoprotein IIb/IIIa (GPIIb/IIIa) revealed the presence of platelet aggregates with fibrin in endothelium-denuded areas of CAVS. In isolated VICs, collagen-activated platelets induced an osteogenic programme. Platelet-derived adenosine diphosphate induced the release of autotaxin (ATX) by VICs. The binding of ATX to GPIIb/IIIa of platelets generated lysophosphatidic acid (LysoPA) with pro-osteogenic properties. In IGFII mice with CAVS, platelet aggregates were found at the surface of AVs. Administration of activated platelets to IGFII mice accelerated the development of CAVS by 2.1-fold, whereas a treatment with Ki16425, an antagonist of LysoPA receptors, prevented platelet-induced mineralization of the AV and the progression of CAVS. CONCLUSIONS: These findings suggest a novel role for platelets in the progression of CAVS.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Blood Platelets/metabolism , Calcinosis/metabolism , Osteogenesis , Animals , Aortic Valve/metabolism , Aortic Valve/ultrastructure , Apolipoprotein B-100/metabolism , Disease Progression , Humans , Integrin beta3/metabolism , Lysophospholipids/metabolism , Mice , Microscopy, Electron, Scanning/methods , Phosphoric Diester Hydrolases/metabolism , Platelet Membrane Glycoprotein IIb/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL