Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(3): 574-589.e14, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259485

ABSTRACT

Sensory neurons initiate defensive reflexes that ensure airway integrity. Dysfunction of laryngeal neurons is life-threatening, causing pulmonary aspiration, dysphagia, and choking, yet relevant sensory pathways remain poorly understood. Here, we discover rare throat-innervating neurons (∼100 neurons/mouse) that guard the airways against assault. We used genetic tools that broadly cover a vagal/glossopharyngeal sensory neuron atlas to map, ablate, and control specific afferent populations. Optogenetic activation of vagal P2RY1 neurons evokes a coordinated airway defense program-apnea, vocal fold adduction, swallowing, and expiratory reflexes. Ablation of vagal P2RY1 neurons eliminates protective responses to laryngeal water and acid challenge. Anatomical mapping revealed numerous laryngeal terminal types, with P2RY1 neurons forming corpuscular endings that appose laryngeal taste buds. Epithelial cells are primary airway sentinels that communicate with second-order P2RY1 neurons through ATP. These findings provide mechanistic insights into airway defense and a general molecular/genetic roadmap for internal organ sensation by the vagus nerve.


Subject(s)
Glossopharyngeal Nerve/physiology , Pharynx/innervation , Vagus Nerve/physiology , Afferent Pathways , Animals , Female , Gene Expression Regulation/genetics , Glossopharyngeal Nerve/metabolism , Larynx/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Sensory Receptor Cells/metabolism , Vagus Nerve/metabolism
2.
Arch Biochem Biophys ; 751: 109844, 2024 01.
Article in English | MEDLINE | ID: mdl-38043889

ABSTRACT

The current study aimed to investigate the hypothesis that purinergic receptors P2Y1 and P2Y2 play a regulatory role in gene expression in unloaded muscle. ATP is released from cells through pannexin channels, and it interacts with P2Y1 and P2Y2 receptors, leading to the activation of markers of protein catabolism and a reduction in protein synthesis. To test this hypothesis thirty-two rats were randomly divided into four groups (8 per group): a non-treated control group (C), a group subjected to three days of hindlimb unloading with a placebo (HS), a group subjected to three days of hindlimb unloading treated with a P2Y1 receptor inhibitor, MRS2179 (HSM), and a group subjected to three days of hindlimb unloading treated with a P2Y2 receptor inhibitor, AR-C 118925XX (HSA). This study revealed several key findings following three days of soleus muscle unloading: 1: Inhibition of P2Y1 or P2Y2 receptors prevented the accumulation of ATP, the increase in IP3 receptor content, and the decrease in the phosphorylation of GSK-3beta. This inhibition also mitigated the reduction in the rate of protein synthesis. However, it had no significant effect on the markers of mTORC1-dependent signaling. 2: Blocking P2Y1 receptors prevented the unloading-induced upregulation of phosphorylated p38MAPK and partially reduced the increase in MuRF1mRNA expression. 3: Blocking P2Y2 receptors prevented muscle atrophy during unloading, partially maintained the levels of phosphorylated ERK1/2, reduced the increase in mRNA expression of MAFbx, ubiquitin, and IL-6 receptor, prevented the decrease in phosphorylated AMPK, and attenuated the increase in phosphorylated p70S6K. Taken together, these results suggest that the prevention of muscle atrophy during unloading, as achieved by the P2Y2 receptor inhibitor, is likely mediated through a reduction in catabolic processes and maintenance of energy homeostasis. In contrast, the P2Y1 receptor appears to play a relatively minor role in muscle atrophy during unloading.


Subject(s)
Muscle, Skeletal , Signal Transduction , Animals , Rats , Adenosine Triphosphate/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/metabolism
3.
Biol Pharm Bull ; 47(7): 1307-1313, 2024.
Article in English | MEDLINE | ID: mdl-39019610

ABSTRACT

Airway ciliary cells are components of the mucociliary transport system and play an important role in sweeping out small particles, such as bacteria and viruses, towards the oropharynx by the action of beating cilia. Several lines of evidence have shown that the ciliary beat is under the regulation of the purinergic system; however, the subtype of receptor and the intracellular signaling pathways involved in the activation of ciliary movement remain to be elucidated. In addition, although the activity of ciliary movement comprises two parameters, the ciliary beat frequency (CBF) and ciliary bend angle (CBA), few reports have analyzed CBA. In this study, we examined the effects of ATP and other purinergic ligands on both CBF and CBA and demonstrated that the purinergic signaling requirements for CBF and CBA are different, with CBF mediated by P2Y1 receptor activation and CBA mediated by the P2X4 receptor.


Subject(s)
Adenosine Triphosphate , Bronchi , Cilia , Animals , Cilia/metabolism , Cilia/physiology , Adenosine Triphosphate/metabolism , Mice , Bronchi/cytology , Mucociliary Clearance/physiology , Male , Receptors, Purinergic P2X4/metabolism , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic/metabolism , Signal Transduction
4.
BMC Biol ; 21(1): 14, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36721118

ABSTRACT

BACKGROUND: Purinergic P2Y1 and P2Y12 receptors (P2Y1-R and P2Y12-R) are G protein-coupled receptors (GPCR) activated by adenosine diphosphate (ADP) to mediate platelet activation, thereby playing a pivotal role in hemostasis and thrombosis. While P2Y12-R is the major target of antiplatelet drugs, no P2Y1-R antagonist has yet been developed for clinical use. However, accumulating data suggest that P2Y1-R inhibition would ensure efficient platelet inhibition with minimal effects on bleeding. In this context, an accurate characterization of P2Y1-R antagonists constitutes an important preliminary step. RESULTS: Here, we investigated the pharmacology of P2Y1-R signaling through Gq and ß-arrestin pathways in HEK293T cells and in mouse and human platelets using highly sensitive resonance energy transfer-based technologies (BRET/HTRF). We demonstrated that at basal state, in the absence of agonist ligand, P2Y1-R activates Gq protein signaling in HEK293T cells and in mouse and human platelets, indicating that P2Y1-R is constitutively active in physiological conditions. We showed that P2Y1-R also promotes constitutive recruitment of ß-arrestin 2 in HEK293T cells. Moreover, the P2Y1-R antagonists MRS2179, MRS2279 and MRS2500 abolished the receptor dependent-constitutive activation, thus behaving as inverse agonists. CONCLUSIONS: This study sheds new light on P2Y1-R pharmacology, highlighting for the first time the existence of a constitutively active P2Y1-R population in human platelets. Given the recent interest of P2Y12-R constitutive activity in patients with diabetes, this study suggests that modification of constitutive P2Y1-R signaling might be involved in pathological conditions, including bleeding syndrome or high susceptibility to thrombotic risk. Thus, targeting platelet P2Y1-R constitutive activation might be a promising and powerful strategy for future antiplatelet therapy.


Subject(s)
Drug Inverse Agonism , GTP-Binding Proteins , Receptors, Purinergic P2Y1 , Signal Transduction , beta-Arrestin 2 , Animals , Humans , Mice , beta-Arrestin 2/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Receptors, Purinergic P2Y1/metabolism , Blood Platelets
5.
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
6.
Purinergic Signal ; 19(1): 305-313, 2023 03.
Article in English | MEDLINE | ID: mdl-35902482

ABSTRACT

Muscle regeneration is indispensable for skeletal muscle health and daily life when injury, muscular disease, and aging occur. Among the muscle regeneration, muscle stem cells' (MuSCs) activation, proliferation, and differentiation play a key role in muscle regeneration. Purines bind to its specific receptors during muscle development, which transmit environmental stimuli and play a crucial role of modulator of muscle regeneration. Evidences proved P2R expression during development and regeneration of skeletal muscle, both in human and mouse. In contrast to P2XR, which have been extensively investigated in skeletal muscles, the knowledge of P2YR in this tissue is less comprehensive. This review summarized muscle regeneration via P2Y1R and P2Y2R and speculated that P2Y1R and P2Y2R might be potential molecular triggers for MuSCs' activation and proliferation via the p-ERK1/2 and PLC pathways, explored their cascade effects on skeletal muscle, and proposed P2Y1/2 receptors as potential pharmacological targets in muscle regeneration, to advance the purinergic signaling within muscle and provide promising strategies for alleviating muscular disease.


Subject(s)
Muscle, Skeletal , Muscular Diseases , Animals , Humans , Mice , Cell Differentiation , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Regeneration/physiology , Signal Transduction , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y2/metabolism
7.
Proc Natl Acad Sci U S A ; 117(20): 10876-10887, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32354994

ABSTRACT

We have described multipotent progenitor-like cells within the major pancreatic ducts (MPDs) of the human pancreas. They express PDX1, its surrogate surface marker P2RY1, and the bone morphogenetic protein (BMP) receptor 1A (BMPR1A)/activin-like kinase 3 (ALK3), but not carbonic anhydrase II (CAII). Here we report the single-cell RNA sequencing (scRNA-seq) of ALK3bright+-sorted ductal cells, a fraction that harbors BMP-responsive progenitor-like cells. Our analysis unveiled the existence of multiple subpopulations along two major axes, one that encompasses a gradient of ductal cell differentiation stages, and another featuring cells with transitional phenotypes toward acinar tissue. A third potential ducto-endocrine axis is revealed upon integration of the ALK3bright+ dataset with a single-cell whole-pancreas transcriptome. When transplanted into immunodeficient mice, P2RY1+/ALK3bright+ populations (enriched in PDX1+/ALK3+/CAII- cells) differentiate into all pancreatic lineages, including functional ß-cells. This process is accelerated when hosts are treated systemically with an ALK3 agonist. We found PDX1+/ALK3+/CAII- progenitor-like cells in the MPDs of types 1 and 2 diabetes donors, regardless of the duration of the disease. Our findings open the door to the pharmacological activation of progenitor cells in situ.


Subject(s)
Pancreas/cytology , Pancreatic Ducts/cytology , Single-Cell Analysis/methods , Stem Cells/cytology , Activins/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Differentiation , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Female , Humans , Insulin-Secreting Cells , Islets of Langerhans Transplantation , Male , Mice , Models, Animal , Receptors, Purinergic P2Y1/metabolism , Transcriptome
8.
Platelets ; 33(8): 1301-1306, 2022 Nov 17.
Article in English | MEDLINE | ID: mdl-35514261

ABSTRACT

Platelet activation by adenosine diphosphate (ADP) is mediated through two G-protein-coupled receptors, P2Y1 and P2Y12, which signal through Gq and Gi, respectively. P2Y1 stimulation leads to phospholipase C activation and an increase in cytosolic calcium necessary for CalDAG-GEF1 activation. Engagement of P2Y12 inhibits adenylate cyclase, which reduces cAMP, and activation of PI3-kinase, which inhibits RASA3 resulting in sustained activated Rap1b. In this study we activated human platelets with 2-MeSADP in the presence of LY294002, a PI3-kinase inhibitor, AR-C69931MX, a P2Y12 antagonist or MRS2179, a P2Y1 antagonist. We measured the phosphorylation of Akt on Ser473 as an indicator of PI3-kinase activity. As previously shown, LY294002 and ARC69931MX abolished 2MeSADP-induced Akt phosphorylation. MRS2179 reduced ADP-induced Akt phosphorylation but did not abolish it. Rap1b activation, however, was only reduced, but not ablated, using LY294002 and was completely inhibited by ARC69931MX or MRS2179. Furthermore, 2MeSADP-induced Rap1b activation was abolished in either P2Y1 or P2Y12 null platelets. These data suggest that ADP-induced Rap1b activation requires both P2Y1 and P2Y12. In addition, although stimulation of P2Y12 results in PI3-kinase activation leading to Akt phosphorylation and Rap1b activation, Rap1b activation can occur independently of PI3-kinase downstream of P2Y12. Thus, we propose that the P2Y12 receptor can regulate Rap1b, possibly through RASA3, in a pathway independent of PI3-kinase.


Subject(s)
Phosphatidylinositol 3-Kinases , Receptors, Purinergic P2 , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Adenylyl Cyclases/metabolism , Blood Platelets/metabolism , Calcium/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Purinergic P2Y Receptor Antagonists , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y12/metabolism , Thionucleotides , Type C Phospholipases/metabolism , rap GTP-Binding Proteins/metabolism
9.
Am J Physiol Heart Circ Physiol ; 320(2): H699-H712, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33306443

ABSTRACT

Brain capillary pericytes have been suggested to play a role in the regulation of cerebral blood flow under physiological and pathophysiological conditions. ATP has been shown to cause constriction of capillaries under ischemic conditions and suggested to be involved in the "no-reflow" phenomenon. To investigate the effects of extracellular ATP on pericyte cell contraction, we studied purinergic receptor activation of cultured bovine brain capillary pericytes. We measured intracellular Ca2+ concentration ([Ca2+]i) responses to purinergic agonists with the fluorescent indicators fura-2 and Cal-520 and estimated contraction of pericytes as relative change in cell area, using real-time confocal imaging. Addition of ATP caused an increase in cytosolic calcium and contraction of the brain capillary pericytes, both reversible and inhibited by the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS). Furthermore, we demonstrated that ATP-induced contraction could be eliminated by intracellular calcium chelation with BAPTA, indicating that the contraction was mediated via purinergic P2-type receptor-mediated [Ca2+]i signaling. ATP stimulation induced inositol triphosphate signaling, consistent with the notion of P2Y receptor activation. Receptor profiling studies demonstrated the presence of P2Y1 and P2Y2 receptors, using ATP, UTP, ADP, and the subtype specific agonists MRS2365 (P2Y1) and 2-thio-UTP (P2Y2). Addition of specific P2X agonists only caused an [Ca2+]i increase at high concentrations, attributed to activation of inositol triphosphate signaling. Our results suggest that contraction of brain capillary pericytes in vitro by activation of P2Y-type purinergic receptors is caused by intracellular calcium release. This adds more mechanistic understanding of the role of pericytes in vessel constriction and points toward P2Y receptors as potential therapeutic targets.NEW & NOTEWORTHY The study concerns brain capillary pericytes, which have been suggested to play a role in the regulation of cerebral blood flow. We show that extracellular ATP causes contraction of primary brain pericytes by stimulation of purinergic receptors and subsequent release of intracellular Ca2+ concentration ([Ca2+]i). The contraction is mainly mediated through activation of P2Y-receptor subtypes, including P2Y1 and P2Y2. These findings add more mechanistic understanding of the role of pericytes in regulation of capillary blood flow. ATP was earlier suggested to be involved in capillary constriction in brain pathologies, and our study gives a detailed account of a part of this important mechanism.


Subject(s)
Adenosine Triphosphate/pharmacology , Brain/blood supply , Calcium Signaling/drug effects , Cell Shape/drug effects , Pericytes/drug effects , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Animals , Capillaries/cytology , Cattle , Cells, Cultured , Inositol 1,4,5-Trisphosphate/metabolism , Pericytes/metabolism , Phenotype , Receptors, Purinergic P2Y/metabolism , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y2/drug effects , Receptors, Purinergic P2Y2/metabolism
10.
Purinergic Signal ; 17(1): 25-31, 2021 03.
Article in English | MEDLINE | ID: mdl-33125617

ABSTRACT

The synaptic event called the inhibitory junction potential (IJP) was arguably one of the more important discoveries made by Burnstock and arguably one of his finer legacies. The discovery of the IJP fundamentally changed how electromechanical coupling was visualised in gastrointestinal smooth muscle. Its discovery also set in motion the search for novel inhibitory neurotransmitters in the enteric nervous system, eventually leading to proposal that ATP or a related nucleotide was a major inhibitory transmitter. The subsequent development of purinergic signalling gave impetus to expanding the classification of surface receptors for extracellular ATP, not only in the GI tract but beyond, and then led to successive phases of medicinal chemistry as the P2 receptor field developed. Ultimately, the discovery of the IJP led to the successful cloning of the first P2Y receptor (chick P2Y1) and expansion of mammalian ATP receptors into two classes: metabotropic P2Y receptors (encompassing P2Y1, P2Y2, P2Y4, P2Y6, P2Y11-14 receptors) and ionotropic P2X receptors (encompassing homomeric P2X1-P2X7 receptors). Here, the causal relationship between the IJP and P2Y1 is explored, setting out the milestones reached and achievements made by Burnstock and his colleagues.


Subject(s)
Adenosine Triphosphate/metabolism , Enteric Nervous System/metabolism , Muscle, Smooth/metabolism , Receptors, Purinergic P2Y1/metabolism , Synaptic Transmission/physiology , Animals , Humans , Signal Transduction/physiology
11.
Nature ; 520(7547): 317-21, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25822790

ABSTRACT

In response to adenosine 5'-diphosphate, the P2Y1 receptor (P2Y1R) facilitates platelet aggregation, and thus serves as an important antithrombotic drug target. Here we report the crystal structures of the human P2Y1R in complex with a nucleotide antagonist MRS2500 at 2.7 Å resolution, and with a non-nucleotide antagonist BPTU at 2.2 Å resolution. The structures reveal two distinct ligand-binding sites, providing atomic details of P2Y1R's unique ligand-binding modes. MRS2500 recognizes a binding site within the seven transmembrane bundle of P2Y1R, which is different in shape and location from the nucleotide binding site in the previously determined structure of P2Y12R, representative of another P2YR subfamily. BPTU binds to an allosteric pocket on the external receptor interface with the lipid bilayer, making it the first structurally characterized selective G-protein-coupled receptor (GPCR) ligand located entirely outside of the helical bundle. These high-resolution insights into P2Y1R should enable discovery of new orthosteric and allosteric antithrombotic drugs with reduced adverse effects.


Subject(s)
Deoxyadenine Nucleotides/chemistry , Deoxyadenine Nucleotides/metabolism , Purinergic P2Y Receptor Antagonists/chemistry , Receptors, Purinergic P2Y1/chemistry , Receptors, Purinergic P2Y1/metabolism , Uracil/analogs & derivatives , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Binding Sites , Crystallography, X-Ray , Deoxyadenine Nucleotides/pharmacology , Humans , Ligands , Models, Molecular , Molecular Conformation , Purinergic P2Y Receptor Antagonists/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Thionucleotides/chemistry , Thionucleotides/metabolism , Uracil/chemistry , Uracil/metabolism , Uracil/pharmacology
12.
Anesth Analg ; 133(3): 794-810, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34166321

ABSTRACT

BACKGROUND: Remifentanil can induce postinfusion cold hyperalgesia. N-methyl-d-aspartate receptor (NMDAR) activation and upregulation of transient receptor potential melastatin 8 (TRPM8) membrane trafficking in dorsal root ganglion (DRG) are critical to cold hyperalgesia derived from neuropathic pain, and TRPM8 activation causes NMDAR-dependent cold response. Contribution of P2Y1 purinergic receptor (P2Y1R) activation in DRG to cold pain hypersensitivity and NMDAR activation induced by P2Y1R upregulation in neurons are also unraveled. This study explores whether P2Y1R contributes to remifentanil-induced cold hyperalgesia via TRPM8-dependent regulation of NMDAR phosphorylation in DRG. METHODS: Rats with remifentanil-induced cold hyperalgesia were injected with TRPM8 antagonist or P2Y1R antagonist at 10 minutes before remifentanil infusion. Cold hyperalgesia (paw lift number and withdrawal duration on cold plate) was measured at -24, 2, 6, 24, and 48 hours following remifentanil infusion. After the last behavioral test, P2Y1R expression, TRPM8 expression and membrane trafficking, and NMDAR subunit (NR1 and NR2B) expression and phosphorylation in DRG were detected by western blot, and colocalization of P2Y1R with TRPM8 was determined by double-labeling immunofluorescence. Two-way repeated measures analysis of variance (ANOVA) or 2 × 2 factorial design ANOVA with repeated measures was used to analyze behavioral data of cold hyperalgesia. One-way ANOVA followed by Bonferroni post hoc comparisons was used to analyze the data in western blot and immunofluorescence. RESULTS: Remifentanil infusion (1 µg·kg-1·min-1 for 60 minutes) induced cold hyperalgesia (hyperalgesia versus control, paw lift number and withdrawal duration on cold plate at 2-48 hours, P < .0001) with upregulated NR1 (hyperalgesia versus naive, 48 hours, mean ± standard deviation [SD], 114.00% ± 12.48% vs 41.75% ± 5.20%, P < .005) and NR2B subunits expression (104.13% ± 8.37% vs 24.63% ± 4.87%, P < .005), NR1 phosphorylation at Ser896 (91.88% ± 7.08% vs 52.00% ± 7.31%, P < .005) and NR2B phosphorylation at Tyr1472 (115.75% ± 8.68% vs 59.75% ± 7.78%, P < .005), TRPM8 expression (115.38% ± 9.27% vs 40.50% ± 4.07%, P < .005) and membrane trafficking (112.88% ± 5.62% vs 48.88% ± 6.49%, P < .005), and P2Y1R expression (128.25% ± 14.86% vs 45.13% ± 7.97%, P < .005) in DRG. Both TRPM8 and P2Y1R antagonists attenuated remifentanil-induced cold hyperalgesia and downregulated increased NR1 and NR2B expression and phosphorylation induced by remifentanil (remifentanil + RQ-00203078 versus remifentanil + saline, NR1 phosphorylation, 69.38% ± 3.66% vs 92.13% ± 4.85%; NR2B phosphorylation, 72.25% ± 6.43% vs 111.75% ± 11.00%, P < .0001). NMDAR activation abolished inhibition of TRPM8 and P2Y1R antagonists on remifentanil-induced cold hyperalgesia. P2Y1R antagonist inhibited remifentanil-evoked elevations in TRPM8 expression and membrane trafficking and P2Y1R-TRPM8 coexpression (remifentanil + 2'-deoxy-N6-methyl adenosine 3',5'-diphosphate [MRS2179] versus remifentanil + saline, coexpression, 8.33% ± 1.33% vs 22.19% ± 2.15%, P < .0001). CONCLUSIONS: Attenuation of remifentanil-induced cold hyperalgesia by P2Y1R inhibition is attributed to downregulations in NMDAR expression and phosphorylation via diminishing TRPM8 expression and membrane trafficking in DRG.


Subject(s)
Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Pain Threshold , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P2Y1/metabolism , TRPM Cation Channels/metabolism , Analgesics/pharmacology , Animals , Behavior, Animal , Cold Temperature , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiopathology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Male , Pain Threshold/drug effects , Phosphorylation , Protein Transport , Purinergic P2Y Receptor Antagonists/pharmacology , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1/drug effects , Remifentanil , Signal Transduction , TRPM Cation Channels/antagonists & inhibitors
13.
Int J Mol Sci ; 22(2)2021 Jan 10.
Article in English | MEDLINE | ID: mdl-33435130

ABSTRACT

Endothelial cells and astrocytes preferentially express metabotropic P2Y nucleotide receptors, which are involved in the maintenance of vascular and neural function. Among these, P2Y1 and P2Y2 receptors appear as main actors, since their stimulation induces intracellular calcium mobilization and activates signaling cascades linked to cytoskeletal reorganization. In the present work, we have analyzed, by means of atomic force microscopy (AFM) in force spectroscopy mode, the mechanical response of human umbilical vein endothelial cells (HUVEC) and astrocytes upon 2MeSADP and UTP stimulation. This approach allows for simultaneous measurement of variations in factors such as Young's modulus, maximum adhesion force and rupture event formation, which reflect the potential changes in both the stiffness and adhesiveness of the plasma membrane. The largest effect was observed in both endothelial cells and astrocytes after P2Y2 receptor stimulation with UTP. Such exposure to UTP doubled the Young's modulus and reduced both the adhesion force and the number of rupture events. In astrocytes, 2MeSADP stimulation also had a remarkable effect on AFM parameters. Additional studies performed with the selective P2Y1 and P2Y13 receptor antagonists revealed that the 2MeSADP-induced mechanical changes were mediated by the P2Y13 receptor, although they were negatively modulated by P2Y1 receptor stimulation. Hence, our results demonstrate that AFM can be a very useful tool to evaluate functional native nucleotide receptors in living cells.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Astrocytes/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2/metabolism , Thionucleotides/metabolism , Uridine Triphosphate/metabolism , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Astrocytes/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Microscopy, Atomic Force , Signal Transduction , Thionucleotides/pharmacology , Uridine Triphosphate/pharmacology
14.
J Neurosci ; 39(31): 6038-6048, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31147524

ABSTRACT

Glutamate is one of the most important neurotransmitters in the process of signal transduction in the CNS. Excessive amounts of this neurotransmitter lead to glutamate excitotoxicity, which is accountable for neuronal death in acute neurological disorders, including stroke and trauma, and in neurodegenerative diseases. Inorganic polyphosphate (PolyP) plays multiple roles in the mammalian brain, including function as a calcium-dependent gliotransmitter mediating communication between astrocytes, while its role in the regulation of neuronal activity is unknown. Here we studied the effect of PolyP on glutamate-induced calcium signal in primary rat neurons in both physiological and pathological conditions. We found that preincubation of primary neurons with PolyP reduced glutamate-induced and AMPA-induced but not the NMDA-induced calcium signal. However, in rat hippocampal acute slices, PolyP reduced ion flux through NMDA and AMPA receptors in native neurons. The effect of PolyP on glutamate and specifically on the AMPA receptors was dependent on the presence of P2Y1 but not of P2X receptor inhibitors and also could be mimicked by P2Y1 agonist 2MeSADP. Preincubation of cortical neurons with PolyP significantly reduced the initial calcium peak as well as the number of neurons with delayed calcium deregulation in response to high concentrations of glutamate and resulted in protection of neurons against glutamate-induced cell death. As a result, activation of P2Y1 receptors by PolyP reduced calcium signal acting through AMPA receptors, thus protecting neurons against glutamate excitotoxicity by reduction of the calcium overload and restoration of mitochondrial function.SIGNIFICANCE STATEMENT One of the oldest polymers in the evolution of living matter is the inorganic polyphosphate (PolyP). It is shown to play a role of gliotransmitter in the brain; however, the role of polyphosphate in neuronal signaling is not clear. Here we demonstrate that inorganic polyphosphate is able to reduce calcium signaling induced by physiological or high concentrations of glutamate. The effect of polyphosphate on glutamate-induced calcium signal in neurons is due to the effect of this polymer on the AMPA receptors. The effect of PolyP on glutamate-induced and AMPA-induced calcium signal is dependent on P2Y receptor antagonist. The ability of PolyP to restrict the glutamate-induced calcium signal lies in the basis of its protection of neurons against glutamate excitotoxicity.


Subject(s)
Glutamic Acid/metabolism , Neurons/metabolism , Polyphosphates/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P2Y1/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Female , Glutamic Acid/toxicity , Male , Neurons/drug effects , Polyphosphates/pharmacology , Rats , Rats, Sprague-Dawley
15.
Neurobiol Dis ; 146: 105132, 2020 12.
Article in English | MEDLINE | ID: mdl-33049315

ABSTRACT

Epilepsy is characterized by a progressive predisposition to suffer seizures due to neuronal hyperexcitability, and one of its most common co-morbidities is cognitive decline. In animal models of chronic epilepsy, such as kindling, electrically induced seizures impair long-term potentiation (LTP), deteriorating learning and memory performance. Astrocytes are known to actively modulate synaptic plasticity and neuronal excitability through Ca2+-dependent gliotransmitter release. It is unclear, however, if astroglial Ca2+ signaling could contribute to the development of synaptic plasticity alterations in the epileptic hippocampus. By employing electrophysiological tools and Ca2+ imaging, we found that glutamatergic CA3-CA1 synapses from kindled rats exhibit an impairment in theta burst (TBS) and high frequency stimulation (HFS)-induced LTP, which is accompanied by an increased probability of neurotransmitter release (Pr) and an abnormal pattern of astroglial Ca2+-dependent transients. Both the impairment in LTP and the Pr were reversed by inhibiting purinergic P2Y1 receptors (P2Y1R) with the specific antagonist MRS2179, which also restored the spontaneous and TBS-induced pattern of astroglial Ca2+-dependent signals. Two consecutive, spaced TBS protocols also failed to induce LTP in the kindled group, however, this impairment was reversed and a strong LTP was induced when the second TBS was applied in the presence of MRS2179, suggesting that the mechanisms underlying the alterations in TBS-induced LTP are likely associated with an aberrant modulation of the induction threshold for LTP. Altogether, these results indicate that P2Y1R inhibition rescues both the pattern of astroglial Ca2+-activity and the plastic properties of CA3-CA1 synapses in the epileptic hippocampus, suggesting that astrocytes might take part in the mechanisms that deteriorate synaptic plasticity and thus cause cognitive decline in epileptic patients.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Epilepsy/physiopathology , Neuronal Plasticity/physiology , Receptors, Purinergic P2Y1/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials/physiology , Long-Term Potentiation/physiology , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology
16.
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
17.
Arterioscler Thromb Vasc Biol ; 39(4): e118-e129, 2019 04.
Article in English | MEDLINE | ID: mdl-30816804

ABSTRACT

Objective- Leukocyte flux contributes to thrombus formation in deep veins under pathological conditions, but mechanisms that inhibit venous thrombosis are incompletely understood. Ectonucleotide di(tri)phosphohydrolase 1 ( ENTPD1 or Cd39), an ectoenzyme that catabolizes extracellular adenine nucleotides, is embedded on the surface of endothelial cells and leukocytes. We hypothesized that under venous stasis conditions, CD39 regulates inflammation at the vein:blood interface in a murine model of deep vein thrombosis. Approach and Results- CD39-null mice developed significantly larger venous thrombi under venous stasis, with more leukocyte recruitment compared with wild-type mice. Gene expression profiling of wild-type and Cd39-null mice revealed 76 differentially expressed inflammatory genes that were significantly upregulated in Cd39-deleted mice after venous thrombosis, and validation experiments confirmed high expression of several key inflammatory mediators. P-selectin, known to have proximal involvement in venous inflammatory and thrombotic events, was upregulated in Cd39-null mice. Inferior vena caval ligation resulted in thrombosis and a corresponding increase in both P-selectin and VWF (von Willebrand Factor) levels which were strikingly higher in mice lacking the Cd39 gene. These mice also manifest an increase in circulating platelet-leukocyte heteroaggregates suggesting heterotypic crosstalk between coagulation and inflammatory systems, which is amplified in the absence of CD39. Conclusions- These data suggest that CD39 mitigates the venous thromboinflammatory response to flow interruption.


Subject(s)
Antigens, CD/physiology , Apyrase/physiology , Chemotaxis, Leukocyte/physiology , Hemorheology , Vasculitis/enzymology , Venous Thrombosis/enzymology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/genetics , Apyrase/deficiency , Apyrase/genetics , Blood Platelets/physiology , Cell Adhesion , Gene Expression Regulation , Gene Regulatory Networks , Ligation , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/biosynthesis , P-Selectin/genetics , Receptors, Purinergic P2Y1/metabolism , Vasculitis/physiopathology , Vena Cava, Inferior , Venous Thrombosis/physiopathology , von Willebrand Factor/biosynthesis , von Willebrand Factor/genetics
18.
Int J Mol Sci ; 21(17)2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32867347

ABSTRACT

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.


Subject(s)
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
19.
Int J Mol Sci ; 21(16)2020 Aug 06.
Article in English | MEDLINE | ID: mdl-32781764

ABSTRACT

Urosepsis is a potentially life-threatening, systemic reaction to uropathogenic bacteria entering the bloodstream of the host. One of the hallmarks of sepsis is early thrombocyte activation with a following fall in circulating thrombocytes as a result of intravascular aggregation and sequestering of thrombocytes in the major organs. Development of a thrombocytopenic state is associated with a poorer outcome of sepsis. Uropathogenic Escherichia coli frequently produce the pore-forming, virulence factor α-haemolysin (HlyA), of which the biological effects are mediated by ATP release and subsequent activation of P2 receptors. Thus, we speculated that inhibition of thrombocyte P2Y1 and P2Y12 receptors might ameliorate the septic response to HlyA-producing E. coli. The study combined in vitro measurements of toxin-induced thrombocyte activation assessed as increased membrane abundance of P-selectin, fibronectin and CD63 and data from in vivo murine model of sepsis-induced by HlyA-producing E. coli under infusion of P2Y1 and P2Y12 antagonists. Our data show that the P2Y1 receptor antagonist almost abolishes thrombocyte activation by pore-forming bacterial toxins. Inhibition of P2Y1, by constant infusion of MRS2500, markedly increased the survival in mice with induced sepsis. Moreover, MRS2500 partially prevented the sepsis-induced depletion of circulating thrombocytes and dampened the sepsis-associated increase in proinflammatory cytokines. In contrast, P2Y12 receptor inhibition had only a marginal effect in vivo and in vitro. Taken together, inhibition of the P2Y1 receptor gives a subtle dampening of the thrombocyte activation and the cytokine response to bacteraemia, which may explain the improved survival observed by P2Y1 receptor antagonists.


Subject(s)
Bacterial Toxins/toxicity , Blood Platelets/pathology , Receptors, Purinergic P2Y12/metabolism , Receptors, Purinergic P2Y1/metabolism , Sepsis/pathology , Urinary Tract Infections/pathology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/therapeutic use , Animals , Deoxyadenine Nucleotides/pharmacology , Disease Models, Animal , Escherichia coli Proteins/metabolism , Hemolysin Proteins/metabolism , Humans , Male , Mice, Inbred BALB C , Sepsis/complications , Sepsis/drug therapy , Treatment Outcome , Urinary Tract Infections/complications , Urinary Tract Infections/drug therapy , Uropathogenic Escherichia coli/drug effects
20.
J Cell Physiol ; 234(4): 4528-4539, 2019 04.
Article in English | MEDLINE | ID: mdl-30206934

ABSTRACT

Periodontal ligament (PDL) cells are mechanosensitive and have the potential to differentiate into osteoblast-like cells under the influence of cyclic tensile force (CTF). CTF modulates the expression of regulatory proteins including bone morphogenetic proteins (BMPs), which are essential for the homeostasis of the periodontium. Among the BMPs, BMP9 is one of the most potent osteogenic BMPs. It is yet unknown whether CTF affects the expression of BMP9 and mineralization. Here, we demonstrated that continuously applied CTF for only the first 6 hr stimulated the synthesis of BMP9 and induced mineral deposition within 14 days by human PDL cells. Stimulation of BMP9 expression depended on ATP and P2Y 1 receptors. Apyrase, an ecto-ATPase, inhibited CTF-mediated ATP-induced BMP9 expression. The addition of ATP increased the expression of BMP9. Loss of function experiments using suramin (a broad-spectrum P2Y antagonist), MRS2179 (a specific P2Y 1 receptor antagonist), MRS 2365 (a specific P2Y 1 agonist), U-73122 (a phospholipase C [PLC] inhibitor), and thapsigargin (enhancer of intracytosolic calcium) revealed the participation of P2Y 1 in regulating the expression of BMP9. This was mediated by an increased level of intracellular Ca 2+ through the PLC pathway. A neutralizing anti-BMP9 antibody decreased mineral deposition, which was stimulated by CTF for almost 45% indicating a role of BMP9 in an in vitro mineralization. Collectively, our findings suggest an essential modulatory role of CTF in the homeostasis and regeneration of the periodontium.


Subject(s)
Calcification, Physiologic , Growth Differentiation Factor 2/biosynthesis , Mechanotransduction, Cellular , Periodontal Ligament/metabolism , Adenosine Triphosphate/metabolism , Calcium Signaling , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Growth Differentiation Factor 2/genetics , Homeostasis , Humans , Periodontal Ligament/cytology , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Stress, Mechanical , Time Factors , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL