ABSTRACT
BACKGROUND: The ATP-gated ionotropic P2X7 receptor (P2X7R) has the unusual ability to function as a small cation channel and a trigger for permeabilization of plasmalemmal membranes. In murine microglia, P2X7R-mediated permeabilization is fundamental to microglial activation, proliferation, and IL-1ß release. However, the role of the P2X7R in primary adult human microglia is poorly understood. METHODS: We used patch-clamp electrophysiology to record ATP-gated current in cultured primary human microglia; confocal microscopy to measure membrane blebbing; fluorescence microscopy to demonstrate membrane permeabilization, caspase-1 activation, phosphatidylserine translocation, and phagocytosis; and kit-based assays to measure cytokine levels. RESULTS: We found that ATP-gated inward currents facilitated with repetitive applications of ATP as expected for current through P2X7Rs and that P2X7R antagonists inhibited these currents. P2X7R antagonists also prevented the ATP-induced uptake of large cationic fluorescent dyes whereas drugs that target pannexin-1 channels had no effect. In contrast, ATP did not induce uptake of anionic dyes. The uptake of cationic dyes was blocked by drugs that target Cl- channels. Finally, we found that ATP activates caspase-1 and inhibits phagocytosis, and these effects are blocked by both P2X7R and Cl- channel antagonists. CONCLUSIONS: Our results demonstrate that primary human microglia in culture express functional P2X7Rs that stimulate both ATP-gated cationic currents and uptake of large molecular weight cationic dyes. Importantly, our data demonstrate that hypotheses drawn from work on murine immune cells accurately predict the essential role of P2X7Rs in a number of human innate immune functions such as phagocytosis and caspase-1 activation. Therefore, the P2X7R represents an attractive target for therapeutic intervention in human neuroinflammatory disorders.
Subject(s)
Microglia/physiology , Receptors, Purinergic P2X7/metabolism , Action Potentials/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacokinetics , Adenosine Triphosphate/pharmacology , Adult , Annexin A5/metabolism , Calcium/metabolism , Caspase 1/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Hydrogen-Ion Concentration , Interleukin-1beta/metabolism , Ionophores/pharmacology , Male , Microglia/drug effects , Nigericin/pharmacology , Phagocytosis/drug effects , Purinergic Agents/pharmacologyABSTRACT
The need for bone grafts is high, due to age-related diseases, such as tumor resections, but also accidents, risky sports, and military conflicts. The gold standard for bone grafting is the use of autografts from the iliac crest, but the limited amount of accessible material demands new sources of bone replacement. The use of mesenchymal stem cells or their descendant cells, namely osteoblast, the bone-building cells and endothelial cells for angiogenesis, combined with artificial scaffolds, is a new approach. Mesenchymal stem cells (MSCs) can be obtained from the patient themselves, or from donors, as they barely cause an immune response in the recipient. However, MSCs never fully differentiate in vitro which might lead to unwanted effects in vivo. Interestingly, purinergic receptors can positively influence the differentiation of both osteoblasts and endothelial cells, using specific artificial ligands. An overview is given on purinergic receptor signaling in the most-needed cell types involved in bone metabolism-namely osteoblasts, osteoclasts, and endothelial cells. Furthermore, different types of scaffolds and their production methods will be elucidated. Finally, recent patents on scaffold materials, as wells as purinergic receptor-influencing molecules which might impact bone grafting, are discussed.
Subject(s)
Bone Regeneration , Guided Tissue Regeneration/methods , Purinergic Agents/pharmacology , Receptors, Purinergic/metabolism , Tissue Scaffolds/chemistry , Animals , Clinical Trials as Topic , Humans , Osteogenesis/drug effectsABSTRACT
Phrenic motor facilitation (pMF), a form of respiratory plasticity, can be elicited by acute intermittent hypoxia (i.e., phrenic long-term facilitation, pLTF) or direct application of drugs to the cervical spinal cord. Moderate acute intermittent hypoxia (mAIH; 3 × 5-min episodes of 35-50 mmHg arterial Po2, 5-min normoxic intervals) induces pLTF by a serotonin-dependent mechanism; mAIH-induced pLTF is abolished by mild systemic inflammation induced by a low dose of lipopolysaccharide (LPS; 100 µg/kg ip). In contrast, severe acute intermittent hypoxia (sAIH; 3 × 5-min episodes of 25-30 mmHg arterial Po2, 5-min normoxic intervals) elicits pLTF by a distinct, adenosine-dependent mechanism. Since it is not known if systemic LPS blocks the mechanism giving rise to sAIH-induced pLTF, we tested the hypothesis that sAIH-induced pLTF and adenosine 2A (A2A) receptor-induced pMF are insensitive to mild systemic inflammation elicited by the same low dose of LPS. In agreement with our hypothesis, neither sAIH-induced pLTF nor cervical intrathecal A2A receptor agonist (CGS-21680; 200 µM, 10 µl × 3)-induced pMF were affected 24 h post-LPS. Pretreatment with intrathecal A2A receptor antagonist injections (MSX-3; 10 µM, 12 µl) blocked sAIH-induced pLTF 24 h post LPS, confirming that pLTF was adenosine dependent. Our results give insights concerning the differential impact of systemic inflammation and the functional significance of multiple cascades capable of giving rise to phrenic motor plasticity. The relative resistance of adenosine-dependent pMF to inflammation suggests that it provides a "backup" system in animals lacking serotonin-dependent pMF due to ongoing inflammation associated with systemic infections and/or neural injury.NEW & NOTEWORTHY This study gives novel insights concerning how a mild systemic inflammation impacts phrenic motor plasticity (pMF), particularly adenosine-dependent pMF. We suggest that since this adenosine-dependent pathway is insensitive to systemic inflammation, it represents an alternative or "backup" mechanism of pMF when other mechanisms are suppressed.
Subject(s)
Adenosine/metabolism , Long-Term Potentiation/physiology , Phrenic Nerve/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Analysis of Variance , Animals , Blood Glucose/drug effects , Blood Glucose/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Hypoxia/complications , Lipopolysaccharides/toxicity , Long-Term Potentiation/drug effects , Male , Phenethylamines/pharmacology , Purinergic Agents/pharmacology , Rats , Rats, Sprague-Dawley , Systemic Inflammatory Response Syndrome/chemically induced , Systemic Inflammatory Response Syndrome/etiology , Time Factors , Xanthines/pharmacologyABSTRACT
Severe pulmonary infection or vigorous cyclic deformation of the alveolar epithelial type I (AT I) cells by mechanical ventilation leads to massive extracellular ATP release. High levels of extracellular ATP saturate the ATP hydrolysis enzymes CD39 and CD73 resulting in persistent high ATP levels despite the conversion to adenosine. Above a certain level, extracellular ATP molecules act as danger-associated molecular patterns (DAMPs) and activate the pro-inflammatory response of the innate immunity through purinergic receptors on the surface of the immune cells. This results in lung tissue inflammation, capillary leakage, interstitial and alveolar oedema and lung injury reducing the production of surfactant by the damaged AT II cells and deactivating the surfactant function by the concomitant extravasated serum proteins through capillary leakage followed by a substantial increase in alveolar surface tension and alveolar collapse. The resulting inhomogeneous ventilation of the lungs is an important mechanism in the development of ventilation-induced lung injury. The high levels of extracellular ATP and the upregulation of ecto-enzymes and soluble enzymes that hydrolyse ATP to adenosine (CD39 and CD73) increase the extracellular adenosine levels that inhibit the innate and adaptive immune responses rendering the host susceptible to infection by invading microorganisms. Moreover, high levels of extracellular adenosine increase the expression, the production and the activation of pro-fibrotic proteins (such as TGF-ß, α-SMA, etc.) followed by the establishment of lung fibrosis.
Subject(s)
Immunity, Innate/immunology , Inflammation/immunology , Lung Injury/etiology , Purinergic Agents/pharmacology , Receptors, Purinergic/metabolism , Adenosine/metabolism , Animals , Humans , Lung Injury/immunology , Lung Injury/pathologyABSTRACT
Based on promising preclinical evidence, microglial P2X7 has increasingly being recognized as a target for therapeutic intervention in neurological and psychiatric diseases. However, despite this knowledge no P2X7-related drug has yet entered clinical trials with respect to CNS diseases. We here discuss the current literature on P2X7 being a drug target and identify unsolved issues and still open questions that have hampered the development of P2X7 dependent therapeutic approaches for CNS diseases. It is concluded here that the lack of brain penetrating P2X7 antagonists is a major obstacle in the field and that central P2X7 is a yet untested clinical drug target. In the CNS, microglial P2X7 activation causes neuroinflammation, which in turn plays a role in various CNS disorders. This has resulted in a surge of brain penetrant P2X7 antagonists. P2X7 is a viable, clinically untested CNS drug target. GLIA 2016;64:1772-1787.
Subject(s)
Central Nervous System Diseases/pathology , Microglia/metabolism , Purinergic Agents/therapeutic use , Receptors, Purinergic P2X7/metabolism , Animals , Central Nervous System Diseases/drug therapy , Humans , Microglia/drug effects , Purinergic Agents/pharmacologyABSTRACT
OBJECTIVE: The need for alternative pharmacologic strategies in treatment of epilepsies is pressing for about 30% of patients with epilepsy who do not experience satisfactory seizure control with present treatments. In temporal lobe epilepsy (TLE) even up to 80% of patients are pharmacoresistant, and surgical resection of the ictogenic tissue is only possible for a minority of TLE patients. In this study we investigate purinergic modulation of drug-resistant seizure-like events (SLEs) in human temporal cortex slices. METHODS: Layer V/VI field potentials from a total of 77 neocortical slices from 17 pharmacoresistant patients were recorded to monitor SLEs induced by application of 8 mM [K(+) ] and 50 µm bicuculline. RESULTS: Activating A1 receptors with a specific agonist completely suppressed SLEs in 73% of human temporal cortex slices. In the remaining slices, incidence of SLEs was markedly reduced. Because a subportion of slices can be pharmacosensitive, we tested effects of an A1 agonist, in slices insensitive to a high dose of carbamazepine (50 µm). Also in these cases the A1 agonist was equally efficient. Moreover, ATP and adenosine blocked or modulated SLEs, an effect mediated not by P2 receptors but rather by adenosine A1 receptors. SIGNIFICANCE: Selective activation of A1 receptors mediates a strong anticonvulsant action in human neocortical slices from pharmacoresistant patients. We propose that our human slice model of seizure-like activity is a feasible option for future studies investigating new antiepileptic drug (AED) candidates.
Subject(s)
Drug Resistant Epilepsy/pathology , Neocortex/drug effects , Neocortex/metabolism , Receptors, Purinergic P1/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Triphosphate/pharmacology , Adult , Bicuculline/analogs & derivatives , Bicuculline/pharmacology , Carbamazepine/adverse effects , Carbamazepine/pharmacology , Drug Resistant Epilepsy/drug therapy , Electric Stimulation , Evoked Potentials/drug effects , Female , Humans , In Vitro Techniques , Male , Middle Aged , Potassium/pharmacology , Purinergic Agents/pharmacology , Time Factors , Young AdultABSTRACT
Inflammation exerts a crucial pathogenic role in the development of hypertension. Hence, the aim of the present study was to investigate the effects of ginger (Zingiber officinale) and turmeric (Curcuma longa) on enzyme activities of purinergic and cholinergic systems as well as inflammatory cytokine levels in Nω-nitro-L-arginine methyl ester hydrochloride-induced hypertensive rats. The rats were divided into seven groups (n = 10); groups 1-3 included normotensive control rats, hypertensive (Nω-nitro-L-arginine methyl ester hydrochloride) rats, and hypertensive control rats treated with atenolol (an antihypertensive drug), while groups 4 and 5 included normotensive and hypertensive (Nω-nitro-L-arginine methyl ester hydrochloride) rats treated with 4â% supplementation of turmeric, respectively, and groups 6 and 7 included normotensive and hypertensive rats treated with 4â% supplementation of ginger, respectively. The animals were induced with hypertension by oral administration of Nω-nitro-L-arginine methyl ester hydrochloride, 40 mg/kg body weight. The results revealed a significant increase in ATP and ADP hydrolysis, adenosine deaminase, and acetylcholinesterase activities in lymphocytes from Nω-nitro-L-arginine methyl ester hydrochloride hypertensive rats when compared with the control rats. In addition, an increase in serum butyrylcholinesterase activity and proinflammatory cytokines (interleukin-1 and - 6, interferon-γ, and tumor necrosis factor-α) with a concomitant decrease in anti-inflammatory cytokines (interleukin-10) was observed in Nω-nitro-L-arginine methyl ester hydrochloride hypertensive rats. However, dietary supplementation of both rhizomes was efficient in preventing these alterations in hypertensive rats by decreasing ATP hydrolysis, acetylcholinesterase, and butyrylcholinesterase activities and proinflammatory cytokines in hypertensive rats. Thus, these activities could suggest a possible insight about the protective mechanisms of the rhizomes against hypertension-related inflammation.
Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Curcuma , Cytokines/metabolism , Hypertension/diet therapy , Plant Preparations/therapeutic use , Zingiber officinale , Animals , Cholinergic Agents/isolation & purification , Cholinergic Agents/pharmacology , Hypertension/enzymology , Male , Purinergic Agents/isolation & purification , Purinergic Agents/pharmacology , Rats , Rats, Wistar , Rhizome , T-Lymphocytes/drug effects , T-Lymphocytes/metabolismABSTRACT
D-serine is a coagonist of N-methyl-d-aspartate (NMDA) subtype of glutamate receptor and plays a role in regulating activity-dependent synaptic plasticity. In this study, we examined the mechanism by which extracellular ATP triggers the release of d-serine from astrocytes and discovered a novel Ca(2+) -independent release mechanism mediated by P2X7 receptors (P2X7 R). Using [(3) H] d-serine, which was loaded into astrocytes via the neutral amino acid transporter 2 (ASCT2), we observed that ATP and a potent P2X7 R agonist, 2'(3')-O-(4-benzoylbenzoyl)adenosine-5'-triphosphate (BzATP), stimulated [(3) H]D-serine release and that were abolished by P2X7 R selective antagonists and by shRNAs, whereas enhanced by removal of intracellular or extracellular Ca(2+) . The P2X7 R-mediated d-serine release was inhibited by pannexin-1 antagonists, such as carbenoxolone (CBX), probenecid (PBN), and (10) Panx-1 peptide, and shRNAs, and stimulation of P2X7 R induced P2X7 R-pannexin-1 complex formation. Simply incubating astrocytes in Ca(2+) /Mg(2+) -free buffer also induced the complex formation, and that enhanced basal d-serine release through pannexin-1. The P2X7 R-mediated d-serine release assayed in Ca(2+) /Mg(2+) -free buffer was enhanced as well, and that was inhibited by CBX. Treating astrocytes with general protein kinase C (PKC) inhibitors, such as chelerythrine, GF109203X, and staurosporine, but not Ca(2+) -dependent PKC inhibitor, Gö6976, inhibited the P2X7 R-mediated d-serine release. Thus, we conclude that in astrocytes, P2X7 R-pannexin-1 complex formation is crucial for P2X7 R-mediated d-serine release through pannexin-1 hemichannel. The release is Ca(2+) -independent and regulates by a Ca(2+) -independent PKC. The activated P2X7 R per se is also functioned as a permeation channel to release d-serine in part. This P2X7 R-mediated d-serine release represents an important mechanism for activity-dependent neuron-glia interaction.
Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Purinergic P2X7/metabolism , Serine/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Amino Acid Transport System ASC/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Male , Minor Histocompatibility Antigens , Purinergic Agents/pharmacology , Rats , Tritium/metabolismABSTRACT
A great body of evidence points toward a functional interaction between metabotropic glutamate 5 receptors (mGluR5) and NMDA receptors (NMDAR) that enhances synaptic plasticity and cognition. However, the molecular mechanism underlying this interaction remains unclear. Here, we show that co-activation of mGluR5 and NMDAR in hippocampal slices synergistically leads to a robust phosphorylation of NR2B (Tyr1472), which is Src kinase dependent and is enabled by endogenous adenosine acting on A2A receptors. As it is well known, NR2B (Tyr1472) phosphorylation anchors NR2B-containing NMDARs to the surface of post-synaptic membranes, preventing their internalization. This is supported by our electrophysiological experiments showing that co-activation of mGluR5 and NMDARs robustly enhances NMDAR-dependent neuronal excitability recorded in CA1 hippocampal region, which temporally coincides with the robust increase in NR2B (Tyr1472) phosphorylation, depends on Src kinases and is also permitted by A2A receptors. Thus, we strongly suggest that NR2B (Tyr1472) phosphorylation constitutes, at least to some extent, the molecular mechanism underlying the mGluR5-mediated enhancement of NMDAR-dependent responses, which is modulated by A2A receptors. A better understanding of the molecular basis of mGluR5/NMDAR interaction would elucidate their role in synaptic plasticity processes as well as in pathological conditions. We propose the following molecular mechanism by which metabotropic Glutamate Receptor 5 (mGluR5) potentiate ionotropic Glutamate N-Methyl-D-Aspartate Receptor (NMDAR) responses in rat hippocampus. Co-activation of mGLUR5/NMDAR activates Src kinases, leading to NR2B(Tyr1472) phosphorylation, which anchors NR2B-containing NMDAR to the plasma membrane, thus inducing a robust increase in the NMDA-dependent excitability. Interestingly, adenosine A2A receptors license the mGluR5-induced NR2B(Tyr1472) phosphorylation.
Subject(s)
Hippocampus/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Adenosine A2/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Tyrosine/metabolism , Animals , Dose-Response Relationship, Drug , Drug Interactions , Excitatory Amino Acid Agents/pharmacology , Hippocampus/drug effects , In Vitro Techniques , Male , Patch-Clamp Techniques , Phosphorylation/drug effects , Phosphorylation/physiology , Purinergic Agents/pharmacology , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Adenosine A2/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Statistics, NonparametricABSTRACT
Adenosine receptors (ARs) are present in the motor terminals at the mouse neuromuscular junction. ARs and the presynaptic muscarinic acetylcholine receptors (mAChRs) share the functional control of the neuromuscular junction. We analysed their mutual interaction in transmitter release modulation. In electrophysiological experiments with unaltered synaptic transmission (muscles paralysed by blocking the voltage-dependent sodium channel of the muscle cells with µ-conotoxin GIIIB), we found that: (i) a collaborative action between different AR subtypes reduced synaptic depression at a moderate activity level (40 Hz); (ii) at high activity levels (100 Hz), endogenous adenosine production in the synaptic cleft was sufficient to reduce depression through A1 -type receptors (A1 Rs) and A2 A-type receptors (A2 A Rs); (iii) when the non-metabolizable 2-chloroadenosine (CADO) agonist was used, both the quantal content and depression were reduced; (iv) the protective effect of CADO on depression was mediated by A1 Rs, whereas A2 A Rs seemed to modulate A1 Rs; (v) ARs and mAChRs absolutely depended upon each other for the modulation of evoked and spontaneous acetylcholine release in basal conditions and in experimental conditions with CADO stimulation; (vi) the purinergic and muscarinic mechanisms cooperated in the control of depression by sharing a common pathway although the purinergic control was more powerful than the muscarinic control; and (vii) the imbalance of the ARs created by using subtype-selective and non-selective inhibitory and stimulatory agents uncoupled protein kinase C from evoked transmitter release. In summary, ARs (A1 Rs, A2 A Rs) and mAChRs (M1 , M2 ) cooperated in the control of activity-dependent synaptic depression and may share a common protein kinase C pathway.
Subject(s)
Acetylcholine/metabolism , Neuromuscular Junction/metabolism , Receptors, Muscarinic/metabolism , Receptors, Purinergic P1/metabolism , 2-Chloroadenosine/pharmacology , Animals , Conotoxins/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Interactions , Electric Stimulation , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Male , Mice , Muscarinic Antagonists/pharmacology , Neuromuscular Junction/drug effects , Protein Kinase C/metabolism , Purinergic Agents/pharmacologyABSTRACT
The hypothalamic suprachiasmatic nuclei (SCN), the circadian master clock in mammals, releases ATP in a rhythm, but the role of extracellular ATP in the SCN is still unknown. In this study, we examined the expression and function of ATP-gated P2X receptors (P2XRs) in the SCN neurons of slices isolated from the brain of 16- to 20-day-old rats. Quantitative RT-PCR showed that the SCN contains mRNA for P2X 1-7 receptors and several G-protein-coupled P2Y receptors. Among the P2XR subunits, the P2X2 > P2X7 > P2X4 mRNAs were the most abundant. Whole-cell patch-clamp recordings from SCN neurons revealed that extracellular ATP application increased the frequency of spontaneous GABAergic IPSCs without changes in their amplitudes. The effect of ATP appears to be mediated by presynaptic P2X2Rs because ATPγS and 2MeS-ATP mimics, while the P2XR antagonist PPADS blocks, the observed enhancement of the frequency of GABA currents. There were significant differences between two SCN regions in that the effect of ATP was higher in the ventrolateral subdivision, which is densely innervated from outside the SCN. Little evidence was found for the presence of P2XR channels in somata of SCN neurons as P2X2R immunoreactivity colocalized with synapsin and ATP-induced current was observed in only 7% of cells. In fura-2 AM-loaded slices, BzATP as well as ADP stimulated intracellular Ca(2+) increase, indicating that the SCN cells express functional P2X7 and P2Y receptors. Our data suggest that ATP activates presynaptic P2X2Rs to regulate inhibitory synaptic transmission within the SCN and that this effect varies between regions.
Subject(s)
Adenosine Triphosphate/pharmacology , Neural Inhibition/drug effects , Neurons/drug effects , Suprachiasmatic Nucleus/cytology , Synaptic Transmission/drug effects , Animals , Animals, Newborn , Biophysical Phenomena/drug effects , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , In Vitro Techniques , Male , Patch-Clamp Techniques , Platelet Aggregation Inhibitors/pharmacology , Purinergic Agents/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/metabolism , Sodium Channel Blockers/pharmacology , Synaptic Potentials/drug effects , Tetrodotoxin/pharmacology , gamma-Aminobutyric Acid/pharmacologyABSTRACT
Parkinsonian symptoms arise due to over-activity of the indirect striatal output pathway, and under-activity of the direct striatal output pathway. l-DOPA-induced dyskinesia (LID) is caused when the opposite circuitry problems are established, with the indirect pathway becoming underactive, and the direct pathway becoming over-active. Here, we define synaptic plasticity abnormalities in these pathways associated with parkinsonism, symptomatic benefits of l-DOPA, and LID. We applied spike-timing dependent plasticity protocols to cortico-striatal synapses in slices from 6-OHDA-lesioned mouse models of parkinsonism and LID, generated in BAC transgenic mice with eGFP targeting the direct or indirect output pathways, with and without l-DOPA present. In naïve mice, bidirectional synaptic plasticity, i.e. LTP and LTD, was induced, resulting in an EPSP amplitude change of approximately 50% in each direction in both striatal output pathways, as shown previously. In parkinsonism and dyskinesia, both pathways exhibited unidirectional plasticity, irrespective of stimulation paradigm. In parkinsonian animals, the indirect pathway only exhibited LTP (LTP protocol: 143.5±14.6%; LTD protocol 177.7±22.3% of baseline), whereas the direct pathway only showed LTD (LTP protocol: 74.3±4.0% and LTD protocol: 63.3±8.7%). A symptomatic dose of l-DOPA restored bidirectional plasticity on both pathways to levels comparable to naïve animals (Indirect pathway: LTP protocol: 124.4±22.0% and LTD protocol: 52.1±18.5% of baseline. Direct pathway: LTP protocol: 140.7±7.3% and LTD protocol: 58.4±6.0% of baseline). In dyskinesia, in the presence of l-DOPA, the indirect pathway exhibited only LTD (LTP protocol: 68.9±21.3% and LTD protocol 52.0±14.2% of baseline), whereas in the direct pathway, only LTP could be induced (LTP protocol: 156.6±13.2% and LTD protocol 166.7±15.8% of baseline). We conclude that normal motor control requires bidirectional plasticity of both striatal outputs, which underlies the symptomatic benefits of l-DOPA. Switching from bidirectional to unidirectional plasticity drives global changes in striatal pathway excitability, and underpins parkinsonism and dyskinesia.
Subject(s)
Antiparkinson Agents/adverse effects , Corpus Striatum/pathology , Dyskinesia, Drug-Induced/pathology , Levodopa/adverse effects , Neural Pathways/pathology , Neuronal Plasticity/physiology , Animals , Animals, Newborn , Disease Models, Animal , Dopamine Agents/pharmacology , Dyskinesia, Drug-Induced/etiology , Excitatory Postsynaptic Potentials/drug effects , Functional Laterality , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Mice , Mice, Transgenic , Neuronal Plasticity/drug effects , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Purinergic Agents/pharmacology , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolismABSTRACT
Endo- as well as exogenous adenosine exhibits anticonvulsant action. Participation of individual types of adenosine receptors was studied in present experiments in immature rats. Cortical epileptic afterdischarges were used as a model in rat pups 12, 18 and 25 days old. CCPA, an agonist of A1 adenosine receptors, decreased markedly duration of afterdischarges whereas DPCPX, an antagonist of A1 receptors, exhibited strong proconvulsant action. Action of either drug was best expressed in 12-day-old rats and it decreased with age. Drugs influencing A2A adenosine receptors (agonist CGS21680 and antagonist ZM241385) did not exhibit systematic effects in our model. Motor phenomena accompanying cortical stimulation or epileptic afterdischarge were never influenced by any of the four drugs studied. A1 adenosine receptors are important in the model of cortical seizures, especially in the youngest group studied.
Subject(s)
Action Potentials/physiology , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Receptor, Adenosine A1/metabolism , Receptors, Adenosine A2/metabolism , Action Potentials/drug effects , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Biophysics , Dose-Response Relationship, Drug , Electric Stimulation , Male , Purinergic Agents/pharmacology , Rats , Rats, WistarABSTRACT
Electrical coupling of photoreceptors through gap junctions suppresses voltage noise, routes rod signals into cone pathways, expands the dynamic range of rod photoreceptors in high scotopic and mesopic illumination, and improves detection of contrast and small stimuli. In essentially all vertebrates, connexin 35/36 (gene homologs Cx36 in mammals, Cx35 in other vertebrates) is the major gap junction protein observed in photoreceptors, mediating rod-cone, cone-cone, and possibly rod-rod communication. Photoreceptor coupling is dynamically controlled by the day/night cycle and light/dark adaptation, and is directly correlated with phosphorylation of Cx35/36 at two sites, serine110 and serine 276/293 (homologous sites in teleost fish and mammals, respectively). Activity of protein kinase A (PKA) plays a key role during this process. Previous studies have shown that activation of dopamine D4 receptors on photoreceptors inhibits adenylyl cyclase, down-regulates cAMP and PKA activity, and leads to photoreceptor uncoupling, imposing the daytime/light condition. In this study, we explored the role of adenosine, a nighttime signal with a high extracellular concentration at night and a low concentration in the day, in regulating photoreceptor coupling by examining photoreceptor Cx35 phosphorylation in zebrafish retina. Adenosine enhanced photoreceptor Cx35 phosphorylation in daytime, but with a complex dose-response curve. Selective pharmacological manipulations revealed that adenosine A2a receptors provide a potent positive drive to phosphorylate photoreceptor Cx35 under the influence of endogenous adenosine at night. A2a receptors can be activated in the daytime as well by micromolar exogenous adenosine. However, the higher affinity adenosine A1 receptors are also present and have an antagonistic though less potent effect. Thus, the nighttime/darkness signal adenosine provides a net positive drive on Cx35 phosphorylation at night, working in opposition to dopamine to regulate photoreceptor coupling via a push-pull mechanism. However, the lower concentration of adenosine present in the daytime actually reinforces the dopamine signal through action on the A1 receptor.
Subject(s)
Adenosine/pharmacology , Gap Junctions/drug effects , Photoreceptor Cells/cytology , Purinergic Agents/pharmacology , Retina/cytology , Adaptation, Ocular/drug effects , Adaptation, Ocular/physiology , Analysis of Variance , Animals , Connexins/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Eye/cytology , Eye Proteins/metabolism , In Vitro Techniques , Photoreceptor Cells/drug effects , Retina/drug effects , Zebrafish , Zebrafish Proteins/metabolismABSTRACT
Ligand-gated ion channels are prototypic oligomeric membrane proteins whose stoichiometry determines their functional properties and subcellular localization. Deciphering the quaternary structure of such protein complexes is an arduous task and usually requires the combination of multiple approaches. ATP-gated P2X receptors are formed by the association of three subunits, but the quaternary arrangement of the seven P2X subunits at the plasma membrane remains poorly characterized. By combining bioluminescence resonance energy transfer, bifunctional fluorescence complementation and protein biochemistry, we developed an experimental approach that allows precise determination of rat P2X receptor quaternary assembly. We found that P2X5 subunits associate with P2X1, P2X2, and P2X4 subunits. We demonstrate that P2X5 and P2X2 subunits interact to form as yet uncharacterized heteromeric receptors with alternate stoichiometries, both present at the plasma membrane. P2X2/5 receptors display functional properties such as pore dilatation, membrane blebbing, and phosphatidylserine exposure that were previously thought to be characteristic hallmarks of the P2X7 receptor. In mouse, P2X2 and P2X5 subunits colocalize and physically interact in specific neuronal populations suggesting that other P2X receptors might contribute to cellular responses typically attributed to P2X7 receptor.
Subject(s)
Protein Subunits/metabolism , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X5/metabolism , Receptors, Purinergic P2X7/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Annexin A5/metabolism , Benzoxazoles/metabolism , Bioluminescence Resonance Energy Transfer Techniques/methods , Brain/metabolism , Enzyme-Linked Immunosorbent Assay , Ganglia, Spinal/cytology , HEK293 Cells , Humans , Immunoprecipitation , Luminescent Proteins/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mutagenesis, Site-Directed/methods , Mutation/genetics , Patch-Clamp Techniques , Protein Subunits/genetics , Purinergic Agents/pharmacology , Quinolinium Compounds/metabolism , Receptors, Purinergic P2X2/genetics , Receptors, Purinergic P2X5/genetics , Transfection , Video Recording , Xenopus laevisABSTRACT
Platelet-derived growth factor receptor α positive (PDGFRα(+)) cells are suggested to mediate purinergic inputs in GI muscles, but the responsiveness of these cells to purines in situ has not been evaluated. We developed techniques to label and visualize PDGFRα(+) cells in murine gastric fundus, load cells with Ca(2+) indicators, and follow their activity via digital imaging. Immunolabelling demonstrated a high density of PDGFRα(+) cells in the fundus. Cells were isolated and purified by fluorescence-activated cell sorting (FACS) using endogenous expression of enhanced green fluorescent protein (eGFP) driven off the Pdgfra promoter. Quantitative PCR showed high levels of expression of purinergic P2Y1 receptors and SK3 K(+) channels in PDGFRα(+) cells. Ca(2+) imaging was used to characterize spontaneous Ca(2+) transients and responses to purines in PDGFRα(+) cells in situ. ATP, ADP, UTP and ß-NAD elicited robust Ca(2+) transients in PDGFRα(+) cells. Ca(2+) transients were also elicited by the P2Y1-specific agonist (N)-methanocarba-2MeSADP (MRS-2365), and inhibited by MRS-2500, a P2Y1-specific antagonist. Responses to ADP, MRS-2365 and ß-NAD were absent in PDGFRα(+) cells from P2ry1((-/-)) mice, but responses to ATP were retained. Purine-evoked Ca(2+) transients were mediated through Ca(2+) release mechanisms. Inhibitors of phospholipase C (U-73122), IP3 (2-APB), ryanodine receptors (Ryanodine) and SERCA pump (cyclopiazonic acid and thapsigargin) abolished Ca(2+) transients elicited by purines. This study provides a link between purine binding to P2Y1 receptors and activation of SK3 channels in PDGFRα(+) cells. Activation of Ca(2+) release is likely to be the signalling mechanism in PDGFRα(+) cells responsible for the transduction of purinergic enteric inhibitory input in gastric fundus muscles.
Subject(s)
Calcium Signaling , Calcium/metabolism , Fibroblasts/metabolism , Gastric Fundus/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Adenosine Diphosphate/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Fibroblasts/drug effects , Flow Cytometry/methods , Gastric Fundus/metabolism , Mice , Mice, Inbred C57BL , NAD/pharmacology , Purinergic Agents/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Type C Phospholipases/antagonists & inhibitorsABSTRACT
The mechanisms behind the therapeutic effects of electrical stimulation of the brain in epilepsy and other disorders are poorly understood. Previous studies in vitro have shown that uniform electric fields can suppress epileptiform activity through a direct polarizing effect on neuronal membranes. Such an effect depends on continuous DC stimulation with unbalanced charge. Here we describe a suppressive effect of a brief (10 ms) DC field on stimulus-evoked epileptiform activity in rat hippocampal brain slices exposed to Cs(+) (3.5 mM). This effect was independent of field polarity, was uncorrelated to changes in synchronized population activity, and persisted during blockade of synaptic transmission with Cd(2+) (500 µM). Antagonists of A(1), P(2X), or P(2Y) receptors were without effect. The suppressive effect depended on the alignment of the external field with the somato-dendritic axis of CA1 pyramidal cells; however, temporal coincidence with the epileptiform activity was not essential, as suppression was detectable for up to 1 s after the field. Pyramidal cells, recorded during epileptiform activity, showed decreased discharge duration and truncation of depolarizing plateau potentials in response to field application. In the absence of hyperactivity, the applied field was followed by slow membrane potential changes, accompanied by decreased input resistance and attenuation of the depolarizing afterpotential following action potentials. These effects recovered over a 1-s period. The study suggests that a brief electric field induces a prolonged suppression of epileptiform activity, which can be related to changes in neuronal membrane properties, including attenuation of signals depending on the persisting Na(+) current.
Subject(s)
Action Potentials , CA1 Region, Hippocampal/physiology , Electric Stimulation , Excitatory Postsynaptic Potentials , Animals , CA1 Region, Hippocampal/drug effects , Cadmium/pharmacology , Cesium/pharmacology , Male , Membrane Potentials , Purinergic Agents/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Wistar , Sodium/metabolismABSTRACT
Astrocytes play a key role in modulating synaptic transmission by controlling the available extracellular GABA via the GAT-1 and GAT-3 GABA transporters (GATs). Using primary cultures of rat astrocytes, we show here that an additional level of regulation of GABA uptake occurs via modulation of the GATs by the adenosine A(1) (A(1)R) and A(2A) (A(2A)R) receptors. This regulation occurs through a complex of heterotetramers (two interacting homodimers) of A(1)R-A(2A)R that signal via two different G-proteins, G(s) and G(i/o), and either enhances (A(2A)R) or inhibits (A(1)R) GABA uptake. These results provide novel mechanistic insight into how G-protein-coupled receptor heteromers signal. Furthermore, we uncover a previously unknown mechanism in which adenosine, in a concentration-dependent manner, acts via a heterocomplex of adenosine receptors in astrocytes to significantly contribute to neurotransmission at the tripartite (neuron-glia-neuron) synapse.
Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Adenosine A2/metabolism , gamma-Aminobutyric Acid/metabolism , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Bacterial Proteins/genetics , Biotinylation , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , GABA Agents/pharmacology , GABA Plasma Membrane Transport Proteins/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Regulation/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Humans , Luminescent Proteins/genetics , Models, Biological , Nipecotic Acids/pharmacology , Phenylisopropyladenosine/metabolism , Protein Binding/drug effects , Purinergic Agents/pharmacology , Rats , Rats, Wistar , Receptors, Adenosine A2/genetics , Recombinant Proteins/metabolism , Time Factors , Transfection/methods , Tritium/metabolismABSTRACT
Parasympathetic control of murine urinary bladder consists of contractile components mediated by both muscarinic and purinergic receptors. Using intracellular recording techniques, the purinergic component of transmission was measured as both evoked excitatory junctional potentials (EJPs) in response to electrical field stimulation and spontaneous events [spontaneous EJPs (sEJPs)]. EJPs, but not sEJPs, were abolished by the application of the Na(+) channel blocker tetrodotoxin and the Ca(2+) channel blocker Cd(2+). Both EJPs and sEJPs were abolished by the application of the P2X(1) antagonist 8,8'-[carbonylbis(imino-4,1-phenylenecarbonylimino-4,1-phenylenecarbonylimino)]bis-1,3,5-naphthalenetrisulfonic acid hexasodium salt (NF279). Application of phorbol dibutyrate (PDBu) increased electrically evoked EJP amplitudes with no effect on mean sEJP amplitudes. Similar increases in EJP amplitudes were produced by PDBu in the presence of either the nonselective protein kinase inhibitor staurosporine or the specific protein kinase C (PKC) inhibitor 2-[1-(3-dimethylaminopropyl)indol-3-yl]-3-(indol-3-yl) maleimide (GF109203X). These results suggest that PDBu increases the purinergic component of detrusor transmission through increasing neurogenic ATP release via a PKC-independent mechanism.
Subject(s)
Neuromuscular Junction/drug effects , Neurotransmitter Agents/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/metabolism , Receptors, Purinergic/metabolism , Urinary Bladder/drug effects , Adenosine Triphosphate/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Electric Stimulation/methods , Membrane Potentials/drug effects , Mice , Neuromuscular Junction/metabolism , Phorbol Esters/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Purinergic Agents/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Suramin/analogs & derivatives , Suramin/pharmacology , Synaptic Transmission/drug effects , Urinary Bladder/enzymology , Urinary Bladder/metabolismABSTRACT
BACKGROUND: ATP is an extracellular signaling molecule with many ascribed functions in sensory systems, including the olfactory epithelium. The mechanism(s) by which ATP is released in the olfactory epithelium has not been investigated. Quantitative luciferin-luciferase assays were used to monitor ATP release, and confocal imaging of the fluorescent ATP marker quinacrine was used to monitor ATP release via exocytosis in Swiss Webster mouse neonatal olfactory epithelial slices. RESULTS: Under control conditions, constitutive release of ATP occurs via exocytosis, hemichannels and ABC transporters and is inhibited by vesicular fusion inhibitor Clostridium difficile toxin A and hemichannel and ABC transporter inhibitor probenecid. Constitutive ATP release is negatively regulated by the ATP breakdown product ADP through activation of P2Y receptors, likely via the cAMP/PKA pathway. In vivo studies indicate that constitutive ATP may play a role in neuronal homeostasis as inhibition of exocytosis inhibited normal proliferation in the OE. ATP-evoked ATP release is also present in mouse neonatal OE, triggered by several ionotropic P2X purinergic receptor agonists (ATP, αßMeATP and Bz-ATP) and a G protein-coupled P2Y receptor agonist (UTP). Calcium imaging of P2X2-transfected HEK293 "biosensor" cells confirmed the presence of evoked ATP release. Following purinergic receptor stimulation, ATP is released via calcium-dependent exocytosis, activated P2X1,7 receptors, activated P2X7 receptors that form a complex with pannexin channels, or ABC transporters. The ATP-evoked ATP release is inhibited by the purinergic receptor inhibitor PPADS, Clostridium difficile toxin A and two inhibitors of pannexin channels: probenecid and carbenoxolone. CONCLUSIONS: The constitutive release of ATP might be involved in normal cell turn-over or modulation of odorant sensitivity in physiological conditions. Given the growth-promoting effects of ATP, ATP-evoked ATP release following injury could lead to progenitor cell proliferation, differentiation and regeneration. Thus, understanding mechanisms of ATP release is of paramount importance to improve our knowledge about tissue homeostasis and post-injury neuroregeneration. It will lead to development of treatments to restore loss of smell and, when transposed to the central nervous system, improve recovery following central nervous system injury.