Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Elife ; 72018 01 31.
Article in English | MEDLINE | ID: mdl-29384476

ABSTRACT

Perisynaptic glial cells respond to neural activity by increasing cytosolic calcium, but the significance of this pathway is unclear. Terminal/perisynaptic Schwann cells (TPSCs) are a perisynaptic glial cell at the neuromuscular junction that respond to nerve-derived substances such as acetylcholine and purines. Here, we provide genetic evidence that activity-induced calcium accumulation in neonatal TPSCs is mediated exclusively by one subtype of metabotropic purinergic receptor. In P2ry1 mutant mice lacking these responses, postsynaptic, rather than presynaptic, function was altered in response to nerve stimulation. This impairment was correlated with a greater susceptibility to activity-induced muscle fatigue. Interestingly, fatigue in P2ry1 mutants was more greatly exacerbated by exposure to high potassium than in control mice. High potassium itself increased cytosolic levels of calcium in TPSCs, a response which was also reduced P2ry1 mutants. These results suggest that activity-induced calcium responses in TPSCs regulate postsynaptic function and muscle fatigue by regulating perisynaptic potassium.


Subject(s)
Calcium Signaling , Muscle Fatigue , Receptors, Purinergic P2Y1/metabolism , Schwann Cells/physiology , Animals , Mice , Mice, Transgenic , Receptors, Purinergic P2Y1/deficiency
2.
Glia ; 65(7): 1059-1071, 2017 07.
Article in English | MEDLINE | ID: mdl-28370368

ABSTRACT

Nervous tissue is characterized by a tight structural association between glial cells and neurons. It is well known that glial cells support neuronal functions, but their role under pathologic conditions is less well understood. Here, we addressed this question in vivo using an experimental model of retinal ischemia and transgenic mice for glia-specific inhibition of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-dependent exocytosis. Transgene expression reduced glutamate, but not ATP release from single Müller cells, impaired glial volume regulation under normal conditions and reduced neuronal dysfunction and death in the inner retina during the early stages of ischemia. Our study reveals that the SNARE-dependent exocytosis in glial cells contributes to neurotoxicity during ischemia in vivo and suggests glial exocytosis as a target for therapeutic approaches.


Subject(s)
Exocytosis/genetics , Ischemia/complications , Nerve Degeneration/etiology , Retina/pathology , Retinal Ganglion Cells/metabolism , SNARE Proteins/metabolism , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Doxycycline/therapeutic use , Ependymoglial Cells/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Intermediate Filaments/metabolism , Ischemia/pathology , Light , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Protein Kinase C-alpha/metabolism , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , SNARE Proteins/genetics , Vascular Endothelial Growth Factor A/metabolism
3.
Chem Biol Interact ; 259(Pt B): 291-294, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27378627

ABSTRACT

Adenosine 5'-triphosphate (ATP), a neurotransmitter and a neuromodulator, has been shown to be co-stored and co-released with acetylcholine (ACh) at the pre-synaptic vesicles in vertebrate neuromuscular junction (nmj). Several lines of studies demonstrated that binding of ATP to its corresponding P2Y1 receptors (P2Y1R) in muscle and neuron regulated the post-synaptic gene expressions. Indeed, the expression of acetylcholinesterase (AChE) in muscle was markedly decreased in P2Y1R-/- (P2Y1R knock-out) mice. In order to search for possible role of P2Y1R in cholinergic function of the brain, the expression of globular form AChE was determined in the brain of P2Y1R-/- mice. In contrast to that in muscle, the amounts of AChE activity, AChE catalytic subunit, structure subunit PRiMA and the amount of ACh, in the brain were not, significantly, altered, suggesting the role of P2Y1R in neuron could have different function as that in muscle. However, the expressions of a series of neuronal development markers, i.e. neurofilaments, were reduced in P2Y1R-/- mouse brain, indicating P2Y1R may be involved in neuronal development process.


Subject(s)
Acetylcholinesterase/metabolism , Brain/metabolism , Receptors, Purinergic P2Y1/deficiency , Acetylcholine/metabolism , Animals , Intermediate Filaments/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2Y1/metabolism
4.
Cell Biol Toxicol ; 32(3): 229-47, 2016 06.
Article in English | MEDLINE | ID: mdl-27197531

ABSTRACT

Extracellular adenosine-5'-triphosphate (ATP) regulates cell death and survival of neighboring cells. The detailed effects are diverse depending on cell types and extracellular ATP concentration. We addressed the effect of ATP on ethanol-induced cytotoxicity in epithelial cells, the cell type that experiences the highest concentrations of alcohol. Using pancreatic duct epithelial cells (PDEC), we found that a micromolar range of ATP reverses all intracellular toxicity mechanisms triggered by exceptionally high doses of ethanol and, thus, improves cell viability dramatically. Out of the many purinergic receptors expressed in PDEC, the P2Y1 receptor was identified to mediate the protective effect, based on pharmacological and siRNA assays. Activation of P2Y1 receptors increased intracellular cyclic adenosine monophosphate (cAMP). The protective effect of ATP was mimicked by forskolin and 8-Br-cAMP but inhibited by a protein kinase A (PKA) inhibitor, H-89. Finally, ATP reverted leakiness of PDEC monolayers induced by ethanol and helped to maintain epithelial integrity. We suggest that purinergic receptors reduce extreme alcohol-induced cell damage via the cAMP signal pathway in PDEC and some other types of cells.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Cyclic AMP/metabolism , Ethanol/toxicity , Pancreatic Ducts/drug effects , Pancreatic Ducts/metabolism , Receptors, Purinergic P2Y1/metabolism , Animals , Calcium/metabolism , Cell Line , Dogs , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Knockdown Techniques , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Pancreatic Ducts/cytology , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , Signal Transduction/drug effects
5.
J Mol Neurosci ; 57(3): 446-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26036470

ABSTRACT

ATP is co-stored and co-released with acetylcholine (ACh) at the pre-synaptic vesicles in vertebrate neuromuscular junction (nmj). Several lines of studies demonstrated that binding of ATP to its corresponding P2Y1 and P2Y2 receptors in the muscle regulated post-synaptic gene expressions. To further support the notion that P2Y receptors are playing indispensable role in formation of post-synaptic specifications at the nmj, the knock-out mice of P2Y1 receptor (P2Y1R (-/-)) were employed here for analyses. In P2Y1R (-/-) mice, the expression of P2Y2 receptor in muscle was reduced by over 50 %, as compared to P2Y1R (+/+) mice. In parallel, the expression of acetylcholinesterase (AChE) in muscle was markedly decreased. In the analysis of the expression of anchoring subunits of AChE in P2Y1R (-/-) mice, the proline-rich membrane anchor (PRiMA) subunit was reduced by 60 %; while the collagen tail (ColQ) subunit was reduced by 50 %. AChE molecular forms in the muscle were not changed, except the amount of enzyme was reduced. Immuno-staining of P2Y1R (-/-) mice nmj, both AChE and AChR were still co-localized at the nmj, and the staining was diminished. Taken together our data demonstrated that P2Y1 receptor regulated the nmj gene expression.


Subject(s)
Acetylcholinesterase/biosynthesis , Gene Expression Regulation/physiology , Neuromuscular Junction/metabolism , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y2/biosynthesis , Acetylcholinesterase/chemistry , Acetylcholinesterase/genetics , Adenosine Triphosphate/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Protein Subunits , Receptors, Cholinergic/metabolism , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/physiology , Receptors, Purinergic P2Y2/genetics
6.
Arterioscler Thromb Vasc Biol ; 35(3): 637-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25593131

ABSTRACT

OBJECTIVE: Platelet hyperactivity is associated with vascular disease and contributes to the genesis of thrombotic disorders. ADP plays an important role in platelet activation and activates platelets through 2 G-protein-coupled receptors, the Gq-coupled P2Y1 receptor (P2Y1R), and the Gi-coupled P2Y12 receptor. Although the involvement of the P2Y1R in thrombogenesis is well established, there are no antagonists that are currently available for clinical use. APPROACH AND RESULTS: Our goal is to determine whether a novel antibody targeting the ligand-binding domain, ie, second extracellular loop (EL2) of the P2Y1R (EL2Ab) could inhibit platelet function and protect against thrombogenesis. Our results revealed that the EL2Ab does indeed inhibit ADP-induced platelet aggregation, in a dose-dependent manner. Furthermore, EL2Ab was found to inhibit integrin GPIIb-IIIa activation, dense and α granule secretion, and phosphatidylserine exposure. These inhibitory effects translated into protection against thrombus formation, as evident by a prolonged time for occlusion in a FeCl3-induced thrombosis model, but this was accompanied by a prolonged tail bleeding time. We also observed a dose-dependent displacement of the radiolabeled P2Y1R antagonist [(3)H]MRS2500 from its ligand-binding site by EL2Ab. CONCLUSIONS: Collectively, our findings demonstrate that EL2Ab binds to and exhibits P2Y1R-dependent function-blocking activity in the context of platelets. These results add further evidence for a role of the P2Y1R in thrombosis and validate the concept that targeting it is a relevant alternative or complement to current antiplatelet strategies.


Subject(s)
Antibodies/pharmacology , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/drug effects , Animals , Antibodies/metabolism , Antibodies/toxicity , Binding Sites , Binding, Competitive , Blood Platelets/metabolism , Carotid Artery Injuries/blood , Carotid Artery Injuries/drug therapy , Deoxyadenine Nucleotides/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Epitopes , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/toxicity , Hemorrhage/chemically induced , Hemostasis/drug effects , Humans , Ligands , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylserines/blood , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/toxicity , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Purinergic P2Y Receptor Antagonists/metabolism , Purinergic P2Y Receptor Antagonists/toxicity , Receptors, Purinergic P2Y1/blood , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/immunology , Receptors, Purinergic P2Y1/metabolism , Secretory Vesicles/drug effects , Secretory Vesicles/metabolism , Thrombosis/blood , Thrombosis/prevention & control , Time Factors
7.
PLoS One ; 9(10): e110101, 2014.
Article in English | MEDLINE | ID: mdl-25290311

ABSTRACT

High concentration of extracellular ADP has been reported to induce cell apoptosis, but the molecular mechanisms remain not fully elucidated. In this study, we found by serendipity that ADP treatment of human umbilical vein endothelial cells (HUVEC) and human aortic endothelial cells (HAEC) down-regulated the protein level of nucleolin in a dose- and time-dependent manner. ADP treatment did not decrease the transcript level of nucloelin, suggesting that ADP might induce nucleolin protein degradation. HUVEC and HAEC expressed ADP receptor P2Y13 receptor, but did not express P2Y1 or P2Y12 receptors. However, P2Y1, 12, 13 receptor antagonists MRS2179, PSB0739, MRS2211 did not inhibit ADP-induced down-regulation of nucleolin. Moreover, MRS2211 itself down-regulated nucleolin protein level. In addition, 2-MeSADP, an agonist for P2Y1, 12 and 13 receptors, did not down-regulate nucleolin protein. These results suggested that ADP-induced nucleolin down-regulation was not due to the activation of P2Y1, 12, or 13 receptors. We also found that ADP treatment induced cell cycle arrest in S phase, cell apoptosis and cell proliferation inhibition via nucleolin down-regulation. The over-expression of nucleolin by gene transfer partly reversed ADP-induced cell cycle arrest, cell apoptosis and cell proliferation inhibition. Furthermore, ADP sensitized HUVEC to cisplatin-induced cell death by the down-regulation of Bcl-2 expression. Taken together, we found, for the first time to our knowledge, a novel mechanism by which ADP regulates cell proliferation by induction of cell cycle arrest and cell apoptosis via targeting nucelolin.


Subject(s)
Adenosine Diphosphate/pharmacology , Apoptosis/drug effects , Endothelial Cells/drug effects , Phosphoproteins/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , S Phase Cell Cycle Checkpoints/drug effects , Adenosine Diphosphate/analogs & derivatives , Antineoplastic Agents/pharmacology , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Azo Compounds/pharmacology , Cell Line , Cell Proliferation/drug effects , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Purinergic Agonists/pharmacology , Purinergic Antagonists/pharmacology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y12/deficiency , Receptors, Purinergic P2Y12/genetics , S Phase Cell Cycle Checkpoints/genetics , Signal Transduction , Thionucleotides/pharmacology , Nucleolin
8.
J Physiol ; 592(6): 1283-93, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24396055

ABSTRACT

Purines induce transient contraction and prolonged relaxation of detrusor muscles. Transient contraction could be due to activation of inward currents in smooth muscle cells, but the mechanism of purinergic relaxation has not been determined. We recently reported a new class of interstitial cells in detrusor muscles and showed that these cells could be identified with antibodies against platelet-derived growth factor receptor-α (PDGFRα(+) cells). The current density of small conductance Ca(2+)-activated K(+) (SK) channels in these cells is far higher (∼100 times) than in smooth muscle cells. Thus, we examined purinergic receptor (P2Y) mediated SK channel activation as a mechanism for purinergic relaxation. P2Y receptors (mainly P2ry1 gene) were highly expressed in PDGFRα(+) cells. Under voltage clamp conditions, ATP activated large outward currents in PDGFRα(+) cells that were inhibited by blockers of SK channels. ATP also induced significant hyperpolarization under current clamp conditions. A P2Y1 agonist, MRS2365, mimicked the effects of ATP, and a P2Y1 antagonist, MRS2500, inhibited ATP-activated SK currents. Responses to ATP were largely abolished in PDGFRα(+) cells of P2ry1(-/-) mice, and no response was elicited by MRS2365 in these cells. A P2X receptor agonist had no effect on PDGFRα(+) cells but, like ATP, activated transient inward currents in smooth muscle cells (SMCs). A P2Y1 antagonist decreased nerve-evoked relaxation. These data suggest that purines activate SK currents via mainly P2Y1 receptors in PDGFRα(+) cells. Our findings provide an explanation for purinergic relaxation in detrusor muscles and show that there are no discrete inhibitory nerve fibres. A dual receptive field for purines provides the basis for inhibitory neural regulation of excitability.


Subject(s)
Muscle, Smooth/physiology , Receptor, Platelet-Derived Growth Factor alpha/physiology , Receptors, Purinergic P2Y1/physiology , Urinary Bladder/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Deoxyadenine Nucleotides/pharmacology , Electric Stimulation , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Urinary Bladder/cytology , Urinary Bladder/drug effects
9.
Ophthalmic Res ; 50(4): 209-14, 2013.
Article in English | MEDLINE | ID: mdl-24052001

ABSTRACT

BACKGROUND/AIMS: Osmotic swelling of Müller cells is a common phenomenon in animal models of ischemic and diabetic retinopathies. Müller cells possess a swelling-inhibitory purinergic signaling cascade which can be activated by various receptor ligands including vascular endothelial growth factor (VEGF) and glutamate. Here, we investigated whether deletion of P2Y1 (P2Y1R) and adenosine A1 receptors (A1AR), and of inositol-1,4,5-trisphosphate-receptor type 2 (IP3R2), in mice affects the inhibitory action of VEGF and glutamate on Müller cell swelling. METHODS: The cross-sectional area of Müller cell somata was recorded after a 4-min superfusion of retinal slices with a hypoosmotic solution. RESULTS: Hypoosmolarity induced a swelling of Müller cells from P2Y1R(-/-), A1AR(-/-) and IP3R2(-/-) mice, but not from wild-type mice. Swelling of wild-type Müller cells was induced by hypoosmotic solution containing barium chloride. Whereas VEGF inhibited the swelling of wild-type Müller cells, it had no swelling-inhibitory effect in cells from A1AR(-/-) and IP3R2(-/-) mice. Glutamate inhibited the swelling of wild-type Müller cells but not of cells from P2Y1R(-/-), A1AR(-/-) and IP3R2(-/-) animals. CONCLUSION: The swelling-inhibitory effects of VEGF and glutamate in murine Müller cells is mediated by transactivation of P2Y1R and A1AR, as well as by intracellular calcium signaling via activation of IP3R2.


Subject(s)
Ependymoglial Cells/physiology , Neuroglia/physiology , Osmotic Pressure/physiology , Signal Transduction/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Calcium Signaling/physiology , Enzyme Inhibitors/pharmacology , Ependymoglial Cells/drug effects , Glutamic Acid/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/deficiency , Mice , Mice, Knockout , Models, Animal , Neuroglia/drug effects , Osmotic Pressure/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/deficiency , Signal Transduction/drug effects , Vascular Endothelial Growth Factors/pharmacology , Xanthines/pharmacology
10.
ASN Neuro ; 4(6): 357-69, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22894715

ABSTRACT

Previous studies indicated that a ganglioside 9acGD3 (9-O-acetyl GD3) antibody [the J-Ab (Jones antibody)] reduces GCP (granule cell progenitor) migration in vitro and in vivo. We here investigated, using cerebellar explants of post-natal day (P) 6 mice, the mechanism by which 9acGD3 reduces GCP migration. We found that immunoblockade of the ganglioside with the J-Ab or the lack of GD3 synthase reduced GCP in vitro migration and the frequency of Ca(2+) oscillations. Immunocytochemistry and pharmacological assays indicated that GCPs expressed P2Y(1)Rs (P2Y(1) receptors) and that deletion or blockade of these receptors decreased the migration rate of GCPs and the frequency of Ca(2+) oscillations. The reduction in P2Y(1)-mediated calcium signals seen in Jones-treated and GD3 synthase-null GCPs were paralleled by P2Y(1)R internalization. We conclude that 9acGD3 controls GCP migration by influencing P2Y(1)R cellular distribution and function.


Subject(s)
Calcium Signaling/genetics , Cell Movement/physiology , Cerebellum/cytology , Gangliosides/metabolism , Neural Stem Cells/physiology , Receptors, Purinergic P2Y1/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Antibodies/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cell Movement/drug effects , Cell Movement/genetics , Gangliosides/deficiency , Gangliosides/immunology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/drug effects , Organ Culture Techniques , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , Transfection , Tubulin/metabolism
11.
J Neurosci ; 32(27): 9238-47, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22764232

ABSTRACT

In adult mammalian brains, neural stem cells (NSCs) exist in the subventricular zone (SVZ), where persistent neurogenesis continues throughout life. Those NSCs produce neuroblasts that migrate into the olfactory bulb via formation of transit-amplifying cells, which are committed precursor cells of the neuronal lineage. In this SVZ niche, cell-cell communications conducted by diffusible factors as well as physical cell-cell contacts are important for the regulation of the proliferation and fate determination of NSCs. Previous studies have suggested that extracellular purinergic signaling, which is mediated by purine compounds such as ATP, plays important roles in cell-cell communication in the CNS. Purinergic signaling also promotes the proliferation of adult NSCs in vitro. However, the in vivo roles of purinergic signaling in the neurogenic niche still remain unknown. In this study, ATP infusion into the lateral ventricle of the mouse brain resulted in an increase in the numbers of rapidly dividing cells and Mash1-positive transit-amplifying cells (Type C cells) in the SVZ. Mash1-positive cells express the P2Y1 purinergic signaling receptor and infusion of the P2Y1 receptor-specific antagonist MRS2179 decreased the number of rapidly dividing bromodeoxyuridine (BrdU)-positive cells and Type C cells. Moreover, a 17% reduction of rapidly dividing BrdU-positive cells and a 19% reduction of Mash1-positive cells were observed in P2Y1 knock-out mice. Together, these results suggest that purinergic signaling promotes the proliferation of rapidly dividing cells and transit-amplifying cells, in the SVZ niche through the P2Y1 receptor.


Subject(s)
Aging/physiology , Cell Proliferation , Cerebral Ventricles/cytology , Cerebral Ventricles/physiology , Receptors, Purinergic P2Y1/physiology , Signal Transduction/physiology , Aging/genetics , Animals , Cell Division/genetics , Cell Division/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis/physiology , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , Signal Transduction/genetics , Stem Cell Niche/physiology
13.
J Physiol ; 590(8): 1943-56, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22371472

ABSTRACT

Purinergic and nitrergic co-transmission is the dominant mechanism responsible for neural-mediated smooth muscle relaxation in the gastrointestinal tract. The aim of the present paper was to test whether or not P2Y(1) receptors are involved in purinergic neurotransmission using P2Y(1)(−/−) knock-out mice. Tension and microelectrode recordings were performed on colonic strips. In wild type (WT) animals, electrical field stimulation (EFS) caused an inhibitory junction potential (IJP) that consisted of a fast IJP (MRS2500 sensitive, 1 µm) followed by a sustained IJP (N(ω)-nitro-L-arginine (L-NNA) sensitive, 1 mm). The fast component of the IJP was absent in P2Y(1)(−/−) mice whereas the sustained IJP (L-NNA sensitive) was recorded. In WT animals, EFS-induced inhibition of spontaneous motility was blocked by the consecutive addition of L-NNA and MRS2500. In P2Y(1)(−/−) mice, EFS responses were completely blocked by L-NNA. In WT and P2Y(1)(−/−) animals, L-NNA induced a smooth muscle depolarization but 'spontaneous' IJP (MRS2500 sensitive) could be recorded in WT but not in P2Y(1)(−/−) animals. Finally, in WT animals, 1 µm MRS2365 caused a smooth muscle hyperpolarization that was blocked by 1 µm MRS2500. In contrast, 1 µm MRS2365 did not modify smooth muscle resting membrane potential in P2Y(1)(−/−) mice. ß-Nicotinamide adenine dinucleotide (ß-NAD, 1 mm) partially mimicked the effect of MRS2365. We conclude that P2Y(1) receptors mediate purinergic neurotransmission in the gastrointestinal tract and ß-NAD partially fulfils the criteria to participate in rodent purinergic neurotransmission. The P2Y(1)(−/−) mouse is a useful animal model to study the selective loss of purinergic neurotransmission.


Subject(s)
Colon/physiology , Neuromuscular Junction/physiology , Receptors, Purinergic P2Y1/deficiency , Synaptic Transmission/physiology , Animals , Colon/drug effects , Electric Stimulation/methods , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Neuromuscular Junction/drug effects , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Synaptic Transmission/drug effects
14.
Pain ; 153(2): 410-419, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22137295

ABSTRACT

We have recently found that, following complete Freund's adjuvant (CFA)-induced inflammation, cutaneous polymodal nociceptors (CPM) lacking the transient receptor potential vanilloid 1 (TRPV1) are sensitized to heat stimuli. In order to determine possible mechanisms playing a role in this change, we examined gene expression in the L2/L3 sensory ganglia following CFA injection into the hairy hind paw skin and found that G-protein-coupled purinoreceptor P2Y1 expression was increased. This receptor is of particular interest, as most CPMs innervating mouse hairy skin bind isolectin B4, which co-localizes with P2Y1. Additionally, our recent findings have shown that cutaneous CPMs in P2Y1-/- mice displayed significantly reduced thermal sensitivity. Together, these findings suggested a possible role for P2Y1 in inflammation-induced heat sensitization in these fibers. To test this hypothesis, we utilized our in vivo small interfering RNA technique to knock down the inflammation-induced increase in P2Y1 expression and then examined the functional effects using ex vivo recording. We found that the normal reduction of heat thresholds in CPM fibers induced by CFA was completely blocked by inhibition of P2Y1. Surprisingly, inhibition of P2Y1 during inflammation also significantly increased the number of CPM neurons expressing TRPV1 without a change in the total number of TRPV1-positive cells in the L2 and L3 dorsal root ganglia. These results show that the inflammation-induced enhanced expression of P2Y1 is required for normal heat sensitization of cutaneous CPM fibers. They also suggest that P2Y1 plays a role in the maintenance of phenotype in cutaneous afferent fibers containing TRPV1.


Subject(s)
Inflammation Mediators/physiology , Peripheral Nervous System Diseases/physiopathology , Receptors, Purinergic P2Y1/physiology , Sensory Receptor Cells/pathology , Thermosensing/physiology , Animals , Hyperalgesia/metabolism , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Mice , Mice, Knockout , Neuritis/metabolism , Neuritis/pathology , Neuritis/physiopathology , Pain Threshold/physiology , Peripheral Nervous System Diseases/metabolism , Phenotype , Primary Cell Culture , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , Sensory Receptor Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...