Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Leukemia ; 36(1): 248-256, 2022 01.
Article in English | MEDLINE | ID: mdl-34285343

ABSTRACT

Recent evidence indicates that extracellular adenosine triphosphate (eATP), as a major mediator of purinergic signaling, plays an important role in regulating the mobilization and homing of hematopoietic stem progenitor cells (HSPCs). In our previous work we demonstrated that eATP activates the P2X7 ion channel receptor in HSPCs and that its deficiency impairs stem cell trafficking. To learn more about the role of the P2X purinergic receptor family in hematopoiesis, we phenotyped murine and human HSPCs with respect to the seven P2X receptors and observed that, these cells also highly express P2X4 receptors, which shows ~50% sequence similarity to P2X7 subtypes, but that P2X4 cells are more sensitive to eATP and signal much more rapidly. Using the selective P2X4 receptor antagonist PSB12054 as well as P2X4-KO mice, we found that the P2X4 receptor, similar to P2X7 receptor, promotes trafficking of HSPCs in that its deficiency leads to impaired chemotaxis of HSPCs in response to a stromal-derived factor 1 (SDF-1) gradient, less effective pharmacological mobilization, and defective homing and engraftment of HSPCs after transplantation into myeloablated hosts. This correlated with a decrease in SDF-1 expression in the BM microenvironment. Overall, our results confirm the proposed cooperative dependence of both receptors in response to eATP signaling. In G-CSF-induced mobilization, a lack of one receptor is not compensated by the presence of the other one, which supports their mutual dependence in regulating HSPC trafficking.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Receptors, Purinergic P2X4/physiology , Receptors, Purinergic P2X7/metabolism , Stem Cell Niche , Animals , Chemotaxis , Female , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2X7/genetics , Signal Transduction
2.
Front Immunol ; 12: 645834, 2021.
Article in English | MEDLINE | ID: mdl-33897694

ABSTRACT

Extracellular nucleotides are important mediators of activation, triggering various responses through plasma membrane P2 and P1 receptors. P2 receptors are further subdivided into ionotropic P2X receptors and G protein-coupled P2Y receptors. P2X4 is an ATP-gated cation channel broadly expressed in most tissues of the body. Within the P2X family, P2X4 has a unique subcellular distribution, being preferentially localized in lysosomes. In these organelles, high ATP concentrations do not trigger P2X4 because of the low pH. However, when the pH increases to 7.4, P2X4 can be stimulated by intra-lysosomal ATP, which is in its active, tetra-anionic form. Elucidation of P2X4, P2X3 and P2X7 structures has shed some light on the functional differences between these purinergic receptors. The potential interaction between P2X4 and P2X7 has been extensively studied. Despite intensive effort, it has not been possible yet to determine whether P2X4 and P2X7 interact as heterotrimers or homotrimers at the plasma membrane. However, several publications have shown that functional interactions between P2X4 and P2X7 do occur. Importantly, these studies indicate that P2X4 potentiates P2X7-dependent activation of inflammasomes, leading to increased release of IL-1ß and IL-18. The role of P2X4 in various diseases could be beneficial or deleterious even though the pathophysiological mechanisms involved are still poorly defined. However, in diseases whose physiopathology involves activation of the NLRP3 inflammasome, P2X4 was found to exacerbate severity of disease. The recent production of monoclonal antibodies specific for the human and mouse P2X4, some of which are endowed with agonist or antagonist properties, raises the possibility that they could be used therapeutically. Analysis of single nucleotide polymorphisms of the human P2RX4 gene has uncovered the association of P2RX4 gene variants with susceptibility to several human diseases.


Subject(s)
Receptors, Purinergic P2X4/chemistry , Receptors, Purinergic P2X4/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cell Degranulation , Encephalomyelitis, Autoimmune, Experimental/etiology , Ethanol/pharmacology , Humans , Inflammasomes/physiology , Inflammation/etiology , Mast Cells/physiology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Antigen, T-Cell/physiology , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/physiology
3.
J Steroid Biochem Mol Biol ; 202: 105725, 2020 09.
Article in English | MEDLINE | ID: mdl-32652201

ABSTRACT

The family of ATP-gated purinergic P2X receptors comprises seven bunits (P2X1-7) that are unevenly distributed in the central and peripheral nervous systems as well as other organs. Endogenous modulators of P2X receptors are phospholipids, steroids and neurosteroids. Here, we analyzed whether bile acids, which are natural products derived from cholesterol, affect P2X receptor activity. We examined the effects of primary and secondary bile acids and newly synthesized derivatives of lithocholic acid on agonist-induced responses in HEK293T cells expressing rat P2X2, P2X4 and P2X7 receptors. Electrophysiology revealed that low micromolar concentrations of lithocholic acid and its structural analog 4-dafachronic acid strongly inhibit ATP-stimulated P2X2 but potentiate P2X4 responses, whereas primary bile acids and other secondary bile acids exhibit no or reduced effects only at higher concentrations. Agonist-stimulated P2X7 responses are significantly potentiated by lithocholic acid at moderate concentrations. Structural modifications of lithocholic acid at positions C-3, C-5 or C-17 abolish both inhibitory and potentiation effects to varying degrees, and the 3α-hydroxy group contributes to the ability of the molecule to switch between potentiation and inhibition. Lithocholic acid allosterically modulates P2X2 and P2X4 receptor sensitivity to ATP, reduces the rate of P2X4 receptor desensitization and antagonizes the effect of ivermectin on P2X4 receptor deactivation. Alanine-scanning mutagenesis of the upper halve of P2X4 transmembrane domain-1 revealed that residues Phe48, Val43 and Tyr42 are important for potentiating effect of lithocholic acid, indicating that modulatory sites for lithocholic acid and ivermectin partly overlap. Lithocholic acid also inhibits ATP-evoked currents in pituitary gonadotrophs expressing native P2X2, and potentiates ATP currents in nonidentified pituitary cells expressing P2X4 receptors. These results indicate that lithocholic acid is a bioactive steroid that may help to further unveil the importance of the P2X2, and P2X4 receptors in many physiological processes.


Subject(s)
Ion Channel Gating/drug effects , Lithocholic Acid/pharmacology , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X2/physiology , Receptors, Purinergic P2X4/physiology , Animals , Female , HEK293 Cells , Humans , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/physiology , Lithocholic Acid/analogs & derivatives , Male , Neurons/drug effects , Neurons/physiology , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/physiology , Rats, Wistar , Receptors, Purinergic P2X7/physiology
4.
Med Sci (Paris) ; 36(5): 525-528, 2020 May.
Article in French | MEDLINE | ID: mdl-32452377

ABSTRACT

TITLE: Récepteurs purinergiques et fibrose hépatique. ABSTRACT: Pour la cinquième année, dans le cadre du module d'enseignement « Physiopathologie de la signalisation ¼ proposé par l'université Paris-sud, les étudiants du Master « Biologie Santé ¼ de l'université Paris-Saclay se sont confrontés à l'écriture scientifique. Ils ont sélectionné une quinzaine d'articles scientifiques récents dans le domaine de la signalisation cellulaire présentant des résultats originaux, via des approches expérimentales variées, sur des thèmes allant des relations hôte-pathogène aux innovations thérapeutiques, en passant par la signalisation hépatique et le métabolisme. Après un travail préparatoire réalisé avec l'équipe pédagogique, les étudiants, organisés en binômes, ont ensuite rédigé, guidés par des chercheurs, une Nouvelle soulignant les résultats majeurs et l'originalité de l'article étudié. Ils ont beaucoup apprécié cette initiation à l'écriture d'articles scientifiques et, comme vous pourrez le lire, se sont investis dans ce travail avec enthousiasme ! Trois de ces Nouvelles sont publiées dans ce numéro, les autres le seront dans des prochains numéros.


Subject(s)
Liver Cirrhosis , Receptors, Purinergic/physiology , Animals , Calcium Signaling/genetics , End Stage Liver Disease/genetics , End Stage Liver Disease/therapy , Exocytosis/genetics , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Myofibroblasts/metabolism , Myofibroblasts/pathology , Protein Transport/genetics , Receptors, Purinergic P2X4/physiology
5.
Biomed Pharmacother ; 127: 110112, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32294598

ABSTRACT

Akt kinase regulates several cellular processes, among them growth, proliferation and survival, and has been correlated to neoplastic disease. We report here crosstalk between several Akt regulatory phosphatases that controls the level of the activated form (phosphorylated) of Akt and affects tumor cell aggressiveness. In prostate cancer cell lines, we observed that transient transfection of PTEN decreased the endogenous level of PHLPPs and in contrast, the transient transfection of PHLPPs decreased the endogenous level of PTEN. Furthermore, silencing of PTEN by siRNA resulted in increased PHLPP levels. This phenomenon was not seen in non-transformed cells or in prostate stem cells. This crosstalk promoted cancer cell invasion and was controlled by epigenetically regulated processes where activation of miRs (miR-190 and miR214), the polycomb group of proteins and DNA methylation were involved. The purinergic P2X4 receptor, which has been shown to have a role in wound healing, was identified to be the mediator of this crosstalk. We also studied prostate stem cells and found this crosstalk in the TGFß1-activated epithelial-mesenchymal transition (EMT). The crosstalk seemed to be a natural part of EMT. In summary, we identify a crosstalk between Akt phosphatases which is not present in non-transformed prostate cells but occurs in cancer cells and stem cells transformed by TGFß-1. This crosstalk is important for cellular invasion. BACKGROUND: Phosphatases regulate the Akt oncogene. RESULTS: Crosstalk between Akt phosphatases in prostate cancer cells and in TGF-ß1 activated stem cells but not in non-transformed cells. CONCLUSION: This back-up mechanism facilitates invasive migration of prostate stem and cancer cells. SIGNIFICANCE: Characterization of Akt regulation may lead to a better understanding of tumor development and to novel strategies for treatment.


Subject(s)
Nuclear Proteins/physiology , PTEN Phosphohydrolase/physiology , Phosphoprotein Phosphatases/physiology , Stem Cells/metabolism , Cell Line, Tumor , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/physiology , Humans , Neoplasm Invasiveness/physiopathology , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphoprotein Phosphatases/metabolism , RNA, Small Interfering/pharmacology , Receptors, Purinergic P2X4/physiology , Transfection , Transforming Growth Factor beta1
6.
FASEB J ; 34(4): 5465-5482, 2020 04.
Article in English | MEDLINE | ID: mdl-32086866

ABSTRACT

We tested the hypothesis that the P2X4 purinergic receptor (P2X4) exacerbates ischemic acute kidney injury (AKI) by promoting renal tubular inflammation after ischemia and reperfusion (IR). Supporting this, P2X4-deficient (KO) mice were protected against ischemic AKI with significantly attenuated renal tubular necrosis, inflammation, and apoptosis when compared to P2X4 wild-type (WT) mice subjected to renal IR. Furthermore, WT mice treated with P2X4 allosteric agonist ivermectin had exacerbated renal IR injury whereas P2X4 WT mice treated with a selective P2X4 antagonist (5-BDBD) were protected against ischemic AKI. Mechanistically, induction of kidney NLRP3 inflammasome signaling after renal IR was significantly attenuated in P2X4 KO mice. A P2 agonist ATPγS increased NLRP3 inflammasome signaling (NLRP3 and caspase 1 induction and IL-1ß processing) in isolated renal proximal tubule cells from WT mice whereas these increases were absent in renal proximal tubules isolated from P2X4 KO mice. Moreover, 5-BDBD attenuated ATPγS induced NLRP3 inflammasome induction in renal proximal tubules from WT mice. Finally, P2X4 agonist ivermectin induced NLRP3 inflammasome and pro-inflammatory cytokines in cultured human proximal tubule cells. Taken together, our studies suggest that renal proximal tubular P2X4 activation exacerbates ischemic AKI and promotes NLRP3 inflammasome signaling.


Subject(s)
Acute Kidney Injury/pathology , Inflammasomes/metabolism , Inflammation/pathology , Kidney Tubules, Proximal/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Purinergic P2X4/physiology , Reperfusion Injury/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Animals , Apoptosis , Cytokines/metabolism , Inflammation/etiology , Inflammation/metabolism , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
7.
J Headache Pain ; 21(1): 4, 2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31937253

ABSTRACT

BACKGROUND: According to our previous study, microglia P2X4 receptors (P2X4Rs) play a pivotal role in the central sensitization of chronic migraine (CM). However, the molecular mechanism that underlies the crosstalk between microglia P2X4Rs and neurons of the trigeminal nucleus caudalis (TNC) is not fully understood. Therefore, the aim of this study is to examine the exact P2X4Rs signalling pathway in the development of central sensitization in a CM animal model. METHODS: We used an animal model with recurrent intermittent administration of nitroglycerin (NTG), which closely mimics CM. NTG-induced basal mechanical and thermal hypersensitivity were evaluated using a von Frey filament test and an increasing-temperature hot plate apparatus (IITC). We detected P2X4Rs, brain-derived neurotrophic factor (BDNF) and phosphorylated p38 mitogen-activated protein kinase (p-p38-MAPK) expression profiles in the TNC. We investigated the effects of a P2X4R inhibitor (5-BDBD) and an agonist (IVM) on NTG-induced hyperalgesia and neurochemical changes as well as on the expression of p-p38-MAPK and BDNF. We also detected the effects of a tropomyosin-related kinase B (TrkB) inhibitor (ANA-12) on the CM animal model in vivo. Then, we evaluated the effect of 5-BDBD and SB203580 (a p38-MAPK inhibitors) on the release and synthesis of BDNF in BV2 microglia cells treated with 50 µM adenosine triphosphate (ATP). RESULTS: Chronic intermittent administration of NTG resulted in chronic mechanical and thermal hyperalgesia, accompanied by the upregulation of P2X4Rs and BDNF expression. 5-BDBD or ANA-12 prevented hyperalgesia induced by NTG, which was associated with a significant inhibition of the NTG-induced increase in phosphorylated extracellular regulated protein kinases (p-ERK) and calcitonin gene related peptide (CGRP) release in the TNC. Repeated administration of IVM produced sustained hyperalgesia and significantly increased the levels of p-ERK and CGRP release in the TNC. Activating P2X4Rs with ATP triggered BDNF release and increased BDNF synthesis in BV2 microglia, and these results were then reduced by 5-BDBD or SB203580. CONCLUSIONS: Our results indicated that the P2X4R contributes to the central sensitization of CM by releasing BDNF and promoting TNC neuronal hyper-excitability. Blocking microglia P2X4R-BDNF signalling may have an effect on the prevention of migraine chronification.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Central Nervous System Sensitization/physiology , Microglia/physiology , Migraine Disorders/physiopathology , Receptors, Purinergic P2X4/physiology , Signal Transduction/physiology , Adenosine Triphosphate/pharmacology , Animals , Calcitonin Gene-Related Peptide/metabolism , Central Nervous System Sensitization/drug effects , Disease Models, Animal , Hyperalgesia/metabolism , Male , Microglia/metabolism , Migraine Disorders/metabolism , Nitroglycerin/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Trigeminal Caudal Nucleus/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Int J Mol Sci ; 20(20)2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31627451

ABSTRACT

Mast cells (MCs) recognize antigens (Ag) via IgE-bound high affinity IgE receptors (FcεRI) and trigger type I allergic reactions. FcεRI-mediated MC activation is regulated by various G protein-coupled receptor (GPCR) agonists. We recently reported that ionotropic P2X4 receptor (P2X4R) stimulation enhanced FcεRI-mediated degranulation. Since MCs are involved in Ag-independent hypersensitivity, we investigated whether co-stimulation with ATP and GPCR agonists in the absence of Ag affects MC degranulation. Prostaglandin E2 (PGE2) induced synergistic degranulation when bone marrow-derived MCs (BMMCs) were co-stimulated with ATP, while pharmacological analyses revealed that the effects of PGE2 and ATP were mediated by EP3 and P2X4R, respectively. Consistently, this response was absent in BMMCs prepared from P2X4R-deficient mice. The effects of ATP and PGE2 were reduced by PI3 kinase inhibitors but were insensitive to tyrosine kinase inhibitors which suppressed the enhanced degranulation induced by Ag and ATP. MC-dependent PGE2-triggered vascular hyperpermeability was abrogated in a P2X4R-deficient mouse ear edema model. Collectively, our results suggest that P2X4R signaling enhances EP3R-mediated MC activation via a different mechanism to that involved in enhancing Ag-induced responses. Moreover, the cooperative effects of the common inflammatory mediators ATP and PGE2 on MCs may be involved in Ag-independent hypersensitivity in vivo.


Subject(s)
Cell Degranulation , Mast Cells/physiology , Receptors, Prostaglandin E, EP3 Subtype/physiology , Receptors, Purinergic P2X4/physiology , Adenosine Triphosphate/agonists , Animals , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Purinergic P2X4/metabolism , Signal Transduction , Syk Kinase/metabolism
10.
Environ Toxicol ; 33(6): 631-639, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29457680

ABSTRACT

Chronic lead exposure causes peripheral sympathetic nerve stimulation, including increased blood pressure and heart rate. Purinergic receptors are involved in the sympathoexcitatory response induced by myocardial ischemia injury. However, whether P2X4 receptor participates in sympathoexcitatory response induced by chronic lead exposure and the possible mechanisms are still unknown. The aim of this study was to explore the change of the sympathoexcitatory response induced by chronic lead exposure via the P2X4 receptor in the stellate ganglion (SG). Rats were given lead acetate through drinking water freely at doses of 0 g/L (control group), 0.5 g/L (low lead group), and 2 g/L (high lead group) for 1 year. Our results demonstrated that lead exposure caused autonomic nervous dysfunction, including blood pressure and heart rate increased and heart rate variability (HRV) decreased. Western blotting results indicated that after lead exposure, the protein expression levels in the SG of P2X4 receptor, IL-1ß and Cx43 were up-regulated, the phosphorylation of p38 mitogen-activated protein kinase (MAPK) was activated. Real-time PCR results showed that the mRNA expression of P2X4 receptor in the SG was higher in lead exposure group than that in the control group. Double-labeled immunofluorescence results showed that P2X4 receptor was co-expressed with glutamine synthetase (GS), the marker of satellite glial cells (SGCs). These changes were positively correlated with the dose of lead exposure. The up-regulated expression of P2X4 receptor in SGCs of the SG maybe enhance the sympathoexcitatory response induced by chronic lead exposure.


Subject(s)
Lead/toxicity , Receptors, Purinergic P2X4/physiology , Stellate Ganglion/drug effects , Adrenergic Fibers/drug effects , Adrenergic Fibers/physiology , Animals , Blood Pressure/drug effects , Female , Heart Rate/drug effects , Male , Neuroglia/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X4/drug effects , Stellate Ganglion/pathology , Synaptic Transmission/drug effects , Toxicity Tests, Chronic , Up-Regulation/drug effects
11.
J Physiol ; 596(10): 1833-1845, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29063617

ABSTRACT

Ivermectin (IVM) is an antiparasitic drug that is used worldwide and rescues hundreds of millions of people from onchocerciasis and lymphatic filariasis. It was discovered by Satoshi Omura and William C. Campbell, to whom the 2015 Nobel Prize in Physiology or Medicine was awarded. It kills parasites by activating glutamate-gated Cl- channels, and it also targets several ligand-gated ion channels and receptors, including Cys-loop receptors, P2X4 receptors and fernesoid X receptors. Recently, we found that IVM also activates a novel target, the G-protein-gated inwardly rectifying K+ channel, and also identified the structural determinant for the activation. In this review, we aim to provide an update and summary of recent progress in the identification of IVM targets, as well as their modulation mechanisms, through molecular structures, chimeras and site-directed mutagenesis, and molecular docking and modelling studies.


Subject(s)
Antiparasitic Agents/pharmacology , Chloride Channels/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Ion Channel Gating , Ivermectin/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Purinergic P2X4/physiology , Animals , Chloride Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Humans , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Purinergic P2X4/drug effects
12.
Brain Behav Immun ; 66: 302-312, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28751018

ABSTRACT

INTRODUCTION: Acute ischemic injury leads to severe neuronal loss. One of the key mechanisms responsible for this effect is inflammation, which is characterized by the activation of myeloid cells, including resident microglia and infiltrating monocytes/macrophages. P2X4 receptors (P2X4Rs) present on these immune cells modulate the inflammatory response. For example, excessive release of adenosine triphosphate during acute ischemic stroke triggers stimulation of P2X4Rs, leading to myeloid cell activation and proliferation and further exacerbating post-ischemic inflammation. In contrast, during recovery P2X4Rs activation on microglia leads to the release of brain-derived neurotrophic factor (BDNF), which alleviate depression, maintain synaptic plasticity and hasten post-stroke behavioral recovery. Therefore, we hypothesized that deletion of the P2X4R specifically from myeloid cells would have differential effects on acute versus chronic recovery following stroke. METHODS: We subjected global or myeloid-specific (MS) P2X4R knock-out (KO) mice and wild-type littermates of both sexes to right middle cerebral artery occlusion (60min). We performed histological, behavioral (sensorimotor and depressive), and biochemical (quantitative PCR and flow cytometry) analyses to determine the acute (three days after occlusion) and chronic (30days after occlusion) effects of receptor deletion. RESULTS: Global P2X4R deletion led to reduced infarct size in both sexes. In MS P2X4R KO mice, only females showed reduced infarct size, an effect that did not change with ovariectomy. MS P2X4R KO mice of both sexes showed swift recovery from sensorimotor deficits during acute recovery but exhibited a more pronounced post-stroke depressive behavior phenotype that was independent of infarct size. Quantitative PCR analysis of whole cell lysate as well as flow-sorted myeloid cells from the perilesional cortex showed increased cellular interleukin 1 beta (IL-1ß), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α) mRNA levels but reduced plasma levels of these cytokines in MS P2X4R KO mice after stroke. The expression levels of BDNF and other depression-associated genes were reduced in MS P2X4R KO mice after stroke. CONCLUSIONS: P2X4R deletion protects against stroke acutely but predisposes to depression-like behavior chronically after stroke. Thus, a time-sensitive approach should be considered when targeting P2X4Rs after stroke.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/metabolism , Depression/complications , Receptors, Purinergic P2X4/physiology , Stroke/metabolism , Stroke/pathology , Animals , Behavior, Animal , Brain/pathology , Brain Ischemia/complications , Cytokines/metabolism , Depression/genetics , Female , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Microglia/pathology , Phenotype , RNA, Messenger/metabolism , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Recovery of Function , Stroke/complications
13.
PLoS Comput Biol ; 13(7): e1005643, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28708827

ABSTRACT

The P2X4 receptor (P2X4R) is a member of a family of purinergic channels activated by extracellular ATP through three orthosteric binding sites and allosterically regulated by ivermectin (IVM), a broad-spectrum antiparasitic agent. Treatment with IVM increases the efficacy of ATP to activate P2X4R, slows both receptor desensitization during sustained ATP application and receptor deactivation after ATP washout, and makes the receptor pore permeable to NMDG+, a large organic cation. Previously, we developed a Markov model based on the presence of one IVM binding site, which described some effects of IVM on rat P2X4R. Here we present two novel models, both with three IVM binding sites. The simpler one-layer model can reproduce many of the observed time series of evoked currents, but does not capture well the short time scales of activation, desensitization, and deactivation. A more complex two-layer model can reproduce the transient changes in desensitization observed upon IVM application, the significant increase in ATP-induced current amplitudes at low IVM concentrations, and the modest increase in the unitary conductance. In addition, the two-layer model suggests that this receptor can exist in a deeply inactivated state, not responsive to ATP, and that its desensitization rate can be altered by each of the three IVM binding sites. In summary, this study provides a detailed analysis of P2X4R kinetics and elucidates the orthosteric and allosteric mechanisms regulating its channel gating.


Subject(s)
Ion Channel Gating/physiology , Ivermectin/metabolism , Receptors, Purinergic P2X4/metabolism , Receptors, Purinergic P2X4/physiology , Adenosine Triphosphate/metabolism , Algorithms , Binding Sites , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Markov Chains , Patch-Clamp Techniques , Receptors, Purinergic P2X4/drug effects
14.
Yakugaku Zasshi ; 137(5): 563-569, 2017.
Article in Japanese | MEDLINE | ID: mdl-28458288

ABSTRACT

A growing body of evidence indicates that extracellular ATP released or leaked from nonexcitable cells as well as neurons plays important roles in the regulation of neuronal and glial functions in the entire body through ATP receptors. ATP receptors (ionotropic P2X and metabotropic P2Y receptors) are the most abundant receptor families in living organisms. In the central nervous system, these receptors participate in the synaptic transmission and intercellular communications between neurons and glia. The glia cells are classified into three types: astrocytes; oligodendrocytes; and microglia. There are many reports that spinal microglia express ATP receptors (P2X4, P2X7, P2Y6, and P2Y12 receptors) that have very important roles. We reported that several molecules of microglia are activated after peripheral nerve injury in a neuropathic pain model. In particular, P2X4 receptors (P2X4Rs) expressed in microglia play a critical role in evoking neuropathic pain. P2X4Rs are upregulated in spinal microglia after nerve injury by several factors such as the CC chemokine receptor CCR2, fibronectin in the spinal cord, interferon regulatory factor (IRF) 8, and IRF5 expressed in microglia. The inhibition of P2X4R action suppresses the functions of microglia and neuropathic pain. These results indicate that overexpressing P2X4Rs on microglia are a central player in evoking neuropathic pain.


Subject(s)
Microglia/physiology , Neuralgia/genetics , Receptors, Purinergic P2X4/physiology , Animals , Fibronectins/physiology , Gene Expression , Humans , Interferon Regulatory Factors/physiology , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Rats , Receptors, CCR2/physiology , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Up-Regulation
15.
Neurochem Int ; 108: 27-33, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28192150

ABSTRACT

Neuropathic pain is a type of chronic pain caused by nervous system damage and dysfunction. The pathogenesis of chronic pain is complicated, and there are no effective therapies for neuropathic pain. Studies show that the P2X4 receptor expressed in the satellite glial cells (SGCs) of dorsal root ganglia (DRG) is related to neuropathic pain. Artemisinin is a monomeric component extracted from traditional Chinese medicine and has a variety of important pharmacological effects and potential applications. This study observed the effect of artemisinin on neuropathic pain and delineated its possible mechanism. The chronic constriction injury (CCI) rat model was used in this study. The results demonstrated that artemisinin relieved pain behaviors in the CCI rats, inhibited the expression of P2X4 receptor in the DRG, and decreased the ATP-activated currents in HEK293 cells transfected with P2X4 plasmid. Dual-labeling immunofluorescence showed that the coexpression of P2X4 receptor and glial fibrillary acidic protein (GFAP) in the DRG of CCI rats was increased compared to control rats. After CCI rats were treated with artemisinin, the coexpression of P2X4 receptor and GFAP in the DRG was significantly decreased compared to the CCI group. This finding suggested that artemisinin could inhibit the nociceptive transmission mediated by P2X4 receptor in the DRG SGCs and thus relieve pain behaviors in the CCI rats.


Subject(s)
Artemisinins/therapeutic use , Ganglia, Spinal/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Pain Measurement/methods , Receptors, Purinergic P2X4/physiology , Animals , Artemisinins/pharmacology , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Male , Pain Measurement/drug effects , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2X Receptor Antagonists/therapeutic use , Rats , Rats, Sprague-Dawley , Treatment Outcome
16.
Neuropharmacology ; 113(Pt A): 60-70, 2017 02.
Article in English | MEDLINE | ID: mdl-27641072

ABSTRACT

Current pharmacotherapies for alcohol used disorder (AUD) are few and relatively ineffective illustrating the need for the development of new, effective medications. Using a translational approach, our laboratory reported that ivermectin, an FDA-approved, human and animal anti-parasitic agent, can significantly reduce ethanol intake in male and female mice across different drinking paradigms. Extending this line of investigation, the current paper investigated the utility of moxidectin (MOX), an analogue of ivermectin, to reduce ethanol intake. Notably, MOX is widely held to have lower neurotoxicity potential and improved margin of safety compared to ivermectin. Using a 24-h-two-bottle choice paradigm, MOX significantly reduced ethanol intake in a dose dependent manner in both male and female C57BL/6J mice, respectively (1.25-7.5 mg/kg) and (1.25-10 mg/kg). Further, multi-day administration of MOX (2.5 mg/kg; intraperitoneal injection) for 5 consecutive days significantly reduced ethanol intake in both the 24-h-two-bottle choice and Drinking-in-the-Dark paradigms in female mice. No overt signs of behavioral toxicity were observed. Notably in both male and female mice, MOX significantly reduced ethanol intake starting approximately 4 h post-injection. Using a Xenopus oocyte expression system, we found that MOX significantly potentiated P2X4 receptor (P2X4R) function and antagonized the inhibitory effects of ethanol on ATP-gated currents in P2X4Rs. This latter finding represents the first report of MOX having activity on P2X4Rs. In addition, MOX potentiated GABAA receptors, but to a lesser degree as compared to ivermectin supporting the hypothesis that MOX would be advantageous (compared to ivermectin) with respect to reducing contraindications. Overall, the results illustrate the potential for development of MOX as a novel pharmacotherapy for the treatment of AUD.


Subject(s)
Alcohol-Related Disorders/drug therapy , Anthelmintics/administration & dosage , Choice Behavior/drug effects , Ethanol/administration & dosage , Macrolides/administration & dosage , Alcohol Drinking/drug therapy , Alcohol-Related Disorders/physiopathology , Animals , Anthelmintics/therapeutic use , Female , Macrolides/therapeutic use , Male , Mice , Mice, Inbred C57BL , Receptors, GABA-A/physiology , Receptors, Purinergic P2X4/physiology , Xenopus laevis
17.
J Physiol ; 594(15): 4253-66, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27477609

ABSTRACT

KEY POINTS: SLC17A9 proteins function as a lysosomal ATP transporter responsible for lysosomal ATP accumulation. P2X4 receptors act as lysosomal ion channels activated by luminal ATP. SLC17A9-mediated ATP transport across the lysosomal membrane is suppressed by Bafilomycin A1, the V-ATPase inhibitor. SLC17A9 mainly uses voltage gradient but not pH gradient generated by the V-ATPase as the driving force to transport ATP into the lysosome to activate P2X4. ABSTRACT: The lysosome contains abundant ATP which plays important roles in lysosome functions and in cell signalling. Recently, solute carrier family 17 member 9 (SLC17A9, also known as VNUT for vesicular nucleotide transporter) proteins were suggested to function as a lysosomal ATP transporter responsible for lysosomal ATP accumulation, and P2X4 receptors were suggested to be lysosomal ion channels that are activated by luminal ATP. However, the molecular mechanism of SLC17A9 transporting ATP and the regulatory mechanism of lysosomal P2X4 are largely unknown. In this study, we report that SLC17A9-mediated ATP transport across lysosomal membranes is suppressed by Bafilomycin A1, the V-ATPase inhibitor. By measuring P2X4 activity, which is indicative of ATP transport across lysosomal membranes, we further demonstrated that SLC17A9 mainly uses voltage gradient but not pH gradient as the driving force to transport ATP into lysosomes. This study provides a molecular mechanism for lysosomal ATP transport mediated by SLC17A9. It also suggests a regulatory mechanism of lysosomal P2X4 by SLC17A9.


Subject(s)
Adenosine Triphosphatases/physiology , Adenosine Triphosphate/physiology , Lysosomes/physiology , Nucleotide Transport Proteins/physiology , Receptors, Purinergic P2X4/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Gene Knockdown Techniques , Mice , Nucleotide Transport Proteins/genetics
19.
J Neurochem ; 139(1): 134-48, 2016 10.
Article in English | MEDLINE | ID: mdl-27402173

ABSTRACT

Purinergic P2X4 receptors (P2X4Rs) belong to the P2X superfamily of ion channels regulated by ATP. We recently demonstrated that P2X4R knockout (KO) mice exhibited deficits in sensorimotor gating, social interaction, and ethanol drinking behavior. Dopamine (DA) dysfunction may underlie these behavioral changes, but there is no direct evidence for P2X4Rs' role in DA neurotransmission. To test this hypothesis, we measured markers of DA function and dependent behaviors in P2X4R KO mice. P2X4R KO mice exhibited altered density of pre-synaptic markers including tyrosine hydroxylase, dopamine transporter; post-synaptic markers including dopamine receptors and phosphorylation of downstream targets including dopamine and cyclic-AMP regulated phosphoprotein of 32 kDa and cyclic-AMP-response element binding protein in different parts of the striatum. Ivermectin, an allosteric modulator of P2X4Rs, significantly affected dopamine and cyclic AMP regulated phosphoprotein of 32 kDa and extracellular regulated kinase1/2 phosphorylation in the striatum. Sensorimotor gating deficits in P2X4R KO mice were rescued by DA antagonists. Using the 6-hydroxydopamine model of DA depletion, P2X4R KO mice exhibited an attenuated levodopa (L-DOPA)-induced motor behavior, whereas ivermectin enhanced this behavior. Collectively, these findings identified an important role for P2X4Rs in maintaining DA homeostasis and illustrate how this association is important for CNS functions including motor control and sensorimotor gating. We propose that P2X4 receptors (P2X4Rs) regulate dopamine (DA) homeostasis and associated behaviors. Pre-synaptic and post-synaptic DA markers were significantly altered in the dorsal and ventral striatum of P2X4R KO mice, implicating altered DA neurotransmission. Sensorimotor gating deficits in P2X4R KO mice were rescued by DA antagonists. Ivermectin (IVM), a positive modulator of P2X4Rs, enhanced levodopa (L-DOPA)-induced motor behavior. These studies highlight potential interactions between P2X4Rs and DA system.


Subject(s)
Behavior, Animal , Corpus Striatum/metabolism , Dopamine/metabolism , Receptors, Purinergic P2X4/drug effects , Receptors, Purinergic P2X4/physiology , Alcohol Drinking/genetics , Alcohol Drinking/psychology , Animals , Corpus Striatum/drug effects , Dopamine Agents/pharmacology , Homeostasis/genetics , Interpersonal Relations , Ivermectin/pharmacology , Levodopa/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Oxidopamine , Reflex, Startle/drug effects , Synaptic Transmission/genetics
20.
Eur J Neurosci ; 43(10): 1366-78, 2016 05.
Article in English | MEDLINE | ID: mdl-27199162

ABSTRACT

Recent data have provided evidence that microglia, the brain-resident macrophage-like cells, modulate neuronal activity in both physiological and pathophysiological conditions, and microglia are therefore now recognized as synaptic partners. Among different neuromodulators, purines, which are produced and released by microglia, have emerged as promising candidates to mediate interactions between microglia and synapses. The cellular effects of purines are mediated through a large family of receptors for adenosine and for ATP (P2 receptors). These receptors are present at brain synapses, but it is unknown whether they can respond to microglia-derived purines to modulate synaptic transmission and plasticity. Here, we used a simple model of adding immune-challenged microglia to mouse hippocampal slices to investigate their impact on synaptic transmission and plasticity at hippocampal mossy fibre (MF) synapses onto CA3 pyramidal neurons. MF-CA3 synapses show prominent forms of presynaptic plasticity that are involved in the encoding and retrieval of memory. We demonstrate that microglia-derived ATP differentially modulates synaptic transmission and short-term plasticity at MF-CA3 synapses by acting, respectively, on presynaptic P2X4 receptors and on adenosine A1 receptors after conversion of extracellular ATP to adenosine. We also report that P2X4 receptors are densely located in the mossy fibre tract in the dentate gyrus-CA3 circuitry. In conclusion, this study reveals an interplay between microglia-derived purines and MF-CA3 synapses, and highlights microglia as potent modulators of presynaptic plasticity.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine/metabolism , Microglia/metabolism , Mossy Fibers, Hippocampal/physiology , Neuronal Plasticity , Receptors, Purinergic P2X4/physiology , Synaptic Transmission , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Apyrase/pharmacology , Cells, Cultured , Excitatory Postsynaptic Potentials , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Mossy Fibers, Hippocampal/drug effects , Neuronal Plasticity/drug effects , Purinergic P2X Receptor Agonists/pharmacology , Purines/pharmacology , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...