Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Publication year range
1.
Article in English | MEDLINE | ID: mdl-28428949

ABSTRACT

α-haemolysin (HlyA)-producing Escherichia coli commonly inflict severe urinary tract infections, including pyelonephritis, which comprises substantial risk for sepsis. In vitro, the cytolytic effect of HlyA is mainly mediated by ATP release through the HlyA pore and subsequent P2X1/P2X7 receptor activation. This amplification of the lytic process is not unique to HlyA but is observed by many other pore-forming proteins including complement-induced haemolysis. Since free hemoglobin in the blood is known to be associated with a worse outcome in sepsis one could speculate that inhibition of P2X receptors would ameliorate the course of sepsis. Surprisingly, this study demonstrates that [Formula: see text] and [Formula: see text] mice are exceedingly sensitive to sepsis with uropathogenic E. coli. These mice have markedly lower survival, higher cytokine levels and activated intravascular coagulation. Quite the reverse is seen in [Formula: see text] mice, which had markedly lower cytokine levels and less coagulation activation compared to controls after exposure to uropathogenic E. coli. The high cytokine levels in the [Formula: see text] mouse are unexpected, since P2X7 is implicated in caspase-1-dependent IL-1ß production. Here, we demonstrate that IL-1ß production during sepsis with uropathogenic E. coli is mediated by caspase-8, since caspase-8 and RIPK3 double knock out mice show substantially lower cytokine during sepsis and increased survival after injection of TNFα. These data support that P2X7 and P2X4 receptor activation has a protective effect during severe E. coli infection.


Subject(s)
Disease Susceptibility , Escherichia coli Infections/pathology , Receptors, Purinergic P2X1/deficiency , Receptors, Purinergic P2X4/deficiency , Receptors, Purinergic P2X7/deficiency , Sepsis/pathology , Animals , Disease Models, Animal , Escherichia coli Infections/genetics , Mice , Mice, Knockout , Survival Analysis , Treatment Outcome
2.
Clin Calcium ; 26(12): 1757-1764, 2016.
Article in Japanese | MEDLINE | ID: mdl-27885188

ABSTRACT

Vascular endothelial cells(ECs)play a critical role in controlling a variety of vascular functions including maintenance of the vascular tone, blood coagulation and fibrinolysis, and selective permeability of proteins. It has recently become apparent that ECs respond to hemodynamic forces, namely, shear stress and stretch, by altering their morphology, functions and gene expression profile. These responses also play important roles in maintaining normal circulatory system functions and homeostasis, and their impairment leads to various vascular diseases, including hypertension, aneurysm and atherosclerosis. The mechanisms underlying the mechanotransduction, however, are not yet clearly understood. In this article, we review the literature on the EC responses to mechanical forces and their roles in the regulation of the circulatory system, while also discussing the mechanosensing mechanisms of ECs.


Subject(s)
Endothelial Cells/metabolism , Homeostasis , Mechanotransduction, Cellular , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling , Humans , Receptors, Purinergic P2X4/deficiency , Receptors, Purinergic P2X4/metabolism
3.
Oncotarget ; 7(49): 80288-80297, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27863396

ABSTRACT

Compelling evidences point out a crucial role for extracellular nucleotides such as adenosine triphosphate (ATP) during inflammatory conditions. Once released into the extracellular space, ATP modulates migration, maturation and function of various inflammatory cells via activating of purinergic receptors of the P2Y- and P2X- family. P2RX4 is an ATP-guided ion channel expressed on structural cells such as alveolar epithelial and smooth muscle cells as well as inflammatory cells including macrophages, dendritic cells (DCs) and T cells. P2RX4 has been shown to interact with P2RX7 and promote NLRP3 inflammasome activation. Although P2RX7 has already been implicated in allergic asthma, the role of P2RX4 in airway inflammation has not been elucidated yet. Therefore, we used a selective pharmacological antagonist and genetic ablation to investigate the role of P2RX4 in an ovalbumin (OVA) driven model of allergen-induced airway inflammation (AAI). Both, P2RX4 antagonist 5-BDBD treatment and P2rx4 deficiency resulted in an alleviated broncho alveolar lavage fluid eosinophilia, peribronchial inflammation, Th2 cytokine production and bronchial hyperresponsiveness. Furthermore, P2rx4-deficient bone marrow derived DCs (BMDCs) showed a reduced IL-1ß production in response to ATP accompanied by a decreased P2rx7 expression and attenuated Th2 priming capacity compared to wild type (WT) BMDCs in vitro. Moreover, mice adoptively transferred with P2rx4-deficient BMDCs exhibit a diminished AAI in vivo. In conclusion our data suggests that P2RX4-signaling contributes to AAI pathogenesis by regulating DC mediated Th2 cell priming via modulating IL-1ß secretion and selective P2RX4-antagonists might be a new therapeutic option for allergic asthma.


Subject(s)
Allergens , Bronchial Hyperreactivity/prevention & control , Lung/metabolism , Pneumonia/prevention & control , Receptors, Purinergic P2X4/deficiency , Adenosine Triphosphate/pharmacology , Adoptive Transfer , Animals , Benzodiazepinones/pharmacology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoconstriction , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Lung/drug effects , Lung/immunology , Lung/physiopathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin , Phenotype , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Pyroglyphidae/immunology , Receptors, Purinergic P2X4/drug effects , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
4.
Neurochem Res ; 39(6): 1127-39, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24671605

ABSTRACT

P2X receptors (P2XRs) are a family of cation-permeable ligand-gated ion channels activated by synaptically released extracellular adenosine 5'-triphosphate. The P2X4 subtype is abundantly expressed in the central nervous system and is sensitive to low intoxicating ethanol concentrations. Genetic meta-analyses identified the p2rx4 gene as a candidate gene for innate alcohol intake and/or preference. The current study used mice lacking the p2rx4 gene (knockout, KO) and wildtype (WT) C57BL/6 controls to test the hypothesis that P2X4Rs contribute to ethanol intake. The early acquisition and early maintenance phases of ethanol intake were measured with three different drinking procedures. Further, we tested the effects of ivermectin (IVM), a drug previously shown to reduce ethanol's effects on P2X4Rs and to reduce ethanol intake and preference, for its ability to differentially alter stable ethanol intake in KO and WT mice. Depending on the procedure and the concentration of the ethanol solution, ethanol intake was transiently increased in P2X4R KO versus WT mice during the acquisition of 24-h and limited access ethanol intake. IVM significantly reduced ethanol intake in P2X4R KO and WT mice, but the degree of reduction was 50 % less in the P2X4R KO mice. Western blot analysis identified significant changes in γ-aminobutyric acidA receptor α1 subunit expression in brain regions associated with the regulation of ethanol behaviors in P2X4R KO mice. These findings add to evidence that P2X4Rs contribute to ethanol intake and indicate that there is a complex interaction between P2X4Rs, ethanol, and other neurotransmitter receptor systems.


Subject(s)
Alcohol Drinking/metabolism , Ethanol/administration & dosage , Receptors, Purinergic P2X4/deficiency , Alcohol Drinking/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2X4/genetics
5.
Circ Heart Fail ; 7(3): 510-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24622244

ABSTRACT

BACKGROUND: Heart failure (HF), despite continuing progress, remains a leading cause of mortality and morbidity. P2X4 receptors (P2X4R) have emerged as potentially important molecules in regulating cardiac function and as potential targets for HF therapy. Transgenic P2X4R overexpression can protect against HF, but this does not explain the role of native cardiac P2X4R. Our goal is to define the physiological role of endogenous cardiac myocyte P2X4R under basal conditions and during HF induced by myocardial infarction or pressure overload. METHODS AND RESULTS: Mice established with conditional cardiac-specific P2X4R knockout were subjected to left anterior descending coronary artery ligation-induced postinfarct or transverse aorta constriction-induced pressure overload HF. Knockout cardiac myocytes did not show P2X4R by immunoblotting or by any response to the P2X4R-specific allosteric enhancer ivermectin. Knockout hearts showed normal basal cardiac function but depressed contractile performance in postinfarct and pressure overload models of HF by in vivo echocardiography and ex vivo isolated working heart parameters. P2X4R coimmunoprecipitated and colocalized with nitric oxide synthase 3 (eNOS) in wild-type cardiac myocytes. Mice with cardiac-specific P2X4R overexpression had increased S-nitrosylation, cyclic GMP, NO formation, and were protected from postinfarct and pressure overload HF. Inhibitor of eNOS, L-N(5)-(1-iminoethyl)ornithine hydrochloride, blocked the salutary effect of cardiac P2X4R overexpression in postinfarct and pressure overload HF as did eNOS knockout. CONCLUSIONS: This study establishes a new protective role for endogenous cardiac myocyte P2X4R in HF and is the first to demonstrate a physical interaction between the myocyte receptor and eNOS, a mediator of HF protection.


Subject(s)
Heart Failure/metabolism , Heart Failure/prevention & control , Myocardial Infarction/complications , Myocytes, Cardiac/metabolism , Receptors, Purinergic P2X4/metabolism , Animals , Coronary Vessels/physiopathology , Disease Models, Animal , Female , Ligation/adverse effects , Male , Mice , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/etiology , Nitric Oxide Synthase Type III/metabolism , Receptors, Purinergic P2X4/deficiency , Receptors, Purinergic P2X4/genetics
6.
PLoS Biol ; 12(1): e1001747, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24409095

ABSTRACT

Communication between neuronal and glial cells is important for many brain functions. Astrocytes can modulate synaptic strength via Ca(2+)-stimulated release of various gliotransmitters, including glutamate and ATP. A physiological role of ATP release from astrocytes was suggested by its contribution to glial Ca(2+)-waves and purinergic modulation of neuronal activity and sleep homeostasis. The mechanisms underlying release of gliotransmitters remain uncertain, and exocytosis is the most intriguing and debated pathway. We investigated release of ATP from acutely dissociated cortical astrocytes using "sniff-cell" approach and demonstrated that release is vesicular in nature and can be triggered by elevation of intracellular Ca(2+) via metabotropic and ionotropic receptors or direct UV-uncaging. The exocytosis of ATP from neocortical astrocytes occurred in the millisecond time scale contrasting with much slower nonvesicular release of gliotransmitters via Best1 and TREK-1 channels, reported recently in hippocampus. Furthermore, we discovered that elevation of cytosolic Ca(2+) in cortical astrocytes triggered the release of ATP that directly activated quantal purinergic currents in the pyramidal neurons. The glia-driven burst of purinergic currents in neurons was followed by significant attenuation of both synaptic and tonic inhibition. The Ca(2+)-entry through the neuronal P2X purinoreceptors led to phosphorylation-dependent down-regulation of GABAA receptors. The negative purinergic modulation of postsynaptic GABA receptors was accompanied by small presynaptic enhancement of GABA release. Glia-driven purinergic modulation of inhibitory transmission was not observed in neurons when astrocytes expressed dn-SNARE to impair exocytosis. The astrocyte-driven purinergic currents and glia-driven modulation of GABA receptors were significantly reduced in the P2X4 KO mice. Our data provide a key evidence to support the physiological importance of exocytosis of ATP from astrocytes in the neocortex.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Neocortex/metabolism , Neural Inhibition/physiology , Neurons/metabolism , Animals , Astrocytes/cytology , Bestrophins , Calcium/metabolism , Cell Communication , Exocytosis , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Regulation , Glutamic Acid/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Membrane Potentials/physiology , Mice , Mice, Transgenic , Neocortex/cytology , Neurons/cytology , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, Purinergic P2X4/deficiency , Receptors, Purinergic P2X4/genetics , SNARE Proteins/genetics , SNARE Proteins/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
7.
Glia ; 62(4): 592-607, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24470356

ABSTRACT

ATP is an important regulator of microglia and its effects on microglial cytokine release are currently discussed as important contributors in a variety of brain diseases. We here analyzed the effects of ATP on the production of six inflammatory mediators (IL-6, IL-10, CCL2, IFN-γ, TNF-α, and IL-12p70) in cultured mouse primary microglia. Stimulation of P2X7 receptor by ATP (1 mM) or BzATP (500 µM) evoked the mRNA expression and release of proinflammatory cytokines IL-6, TNF-α, and the chemokine CCL2 in WT cells but not in P2X7(-/-) cells. The effects of ATP and BzATP were inhibited by the nonselective P2 receptor antagonists PPADs and suramin. Various selective P2X7 receptor antagonists blocked the P2X7-dependent release of IL-6 and CCL2, but, surprisingly, had no effect on BzATP-induced release of TNF-α in microglia. Calcium measurements confirmed that P2X7 is the main purine receptor activated by BzATP in microglia and showed that all P2X7 antagonists were functional. It is also presented that pannexin-1 hemichannel function and potential P2X4/P2X7 heterodimers are not involved in P2X7-dependent release of IL-6, CCL2, and TNF-α in microglia. How P2X7-specific antagonists only affect P2X7-dependent IL-6 and CCL2 release, but not TNF-α release is at the moment unclear, but indicates that the P2X7-dependent release of cytokines in microglia is differentially regulated.


Subject(s)
Cell Differentiation/physiology , Chemokine CCL2/metabolism , Interleukin-6/metabolism , Microglia/physiology , Receptors, Purinergic P2X7/physiology , Tumor Necrosis Factor-alpha/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Brain/cytology , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Connexins/genetics , Connexins/metabolism , Dose-Response Relationship, Drug , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/deficiency , Receptors, Purinergic P2X7/deficiency , Receptors, Purinergic P2X7/genetics , Tumor Necrosis Factor-alpha/genetics
8.
Glia ; 61(8): 1306-19, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23828736

ABSTRACT

Within the central nervous system, functions of the ATP-gated receptor-channel P2X4 (P2X4R) are still poorly understood, yet P2X4R activation in neurons and microglia coincides with high or pathological neuronal activities. In this study, we investigated the potential involvement of P2X4R in microglial functions in a model of kainate (KA)-induced status epilepticus (SE). We found that SE was associated with an induction of P2X4R expression in the hippocampus, mostly localized in activated microglial cells. In P2X4R-deficient mice, behavioral responses during KA-induced SE were unaltered. However, 48h post SE specific features of microglial activation, such as cell recruitment and upregulation of voltage-dependent potassium channels were impaired in P2X4R-deficient mice, whereas the expression and function of other microglial purinergic receptors remained unaffected. Consistent with the role of P2X4R in activity-dependent degenerative processes, the CA1 area was partially protected from SE-induced neuronal death in P2X4R-deficient mice compared with wild-type animals. Our findings demonstrate that P2X4Rs are brought into play during neuronal hyperexcitability and that they control specific aspects of microglial activation. Our results also suggest that P2X4Rs contribute to excitotoxic damages by regulating microglial activation.


Subject(s)
Hippocampus/metabolism , Microglia/metabolism , Receptors, Purinergic P2X4/physiology , Status Epilepticus/metabolism , Animals , Cell Proliferation , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Organ Culture Techniques , Receptors, Purinergic P2X4/biosynthesis , Receptors, Purinergic P2X4/deficiency , Status Epilepticus/pathology , Up-Regulation/physiology
9.
Int J Neuropsychopharmacol ; 16(5): 1059-70, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23174033

ABSTRACT

Purinergic ionotropic P2X receptors are a family of cation-permeable channels that bind extracellular adenosine 5'-triphosphate. In particular, convergent lines of evidence have recently highlighted P2X(4) receptors as a potentially critical target in the regulation of multiple nervous and behavioural functions, including pain, neuroendocrine regulation and hippocampal plasticity. Nevertheless, the role of the P2X(4) receptor in behavioural organization remains poorly investigated. To study the effects of P2X(4) activation, we tested the acute effects of its potent positive allosteric modulator ivermectin (IVM, 2.5-10 mg/kg i.p.) on a broad set of paradigms capturing complementary aspects of perceptual, emotional and cognitive regulation in mice. In a novel open field, IVM did not induce significant changes in locomotor activity, but increased the time spent in the peripheral zone. In contrast, IVM produced anxiolytic-like effects in the elevated plus maze and marble burying tasks, as well as depression-like behaviours in the tail-suspension and forced swim tests. The agent induced no significant behavioural changes in the conditioned place preference test and in the novel object recognition task. Finally, the drug induced a dose-dependent decrease in sensorimotor gating, as assessed by pre-pulse inhibition (PPI) of the acoustic startle reflex. In P2X(4) knockout mice, the effects of IVM in the open field and elevated plus maze were similar to those observed in wild type mice; conversely, the drug significantly increased startle amplitude and failed to reduce PPI. Taken together, these results suggest that P2X(4) receptors may play a role in the regulation of sensorimotor gating.


Subject(s)
Behavior, Animal/drug effects , Insecticides/pharmacology , Ivermectin/pharmacology , Receptors, Purinergic P2X4/metabolism , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hindlimb Suspension , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/drug effects , Psychomotor Performance/drug effects , Receptors, Purinergic P2X4/deficiency , Sensory Gating/drug effects , Statistics, Nonparametric , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL