Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
Add more filters










Publication year range
1.
Brain Res ; 1636: 193-199, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26874070

ABSTRACT

Recently it has been reported that prolactin (PRL) exerts a neuroprotective effect against excitotoxicity in hippocampus in the rat in vivo models. However, the exact mechanism by which PRL mediates this effect is not completely understood. The aim of our study was to assess whether prolactin exerts neuroprotection against excitotoxicity in an in vitro model using primary cell cultures of hippocampal neurons, and to determine whether this effect is mediated via the prolactin receptor (PRLR). Primary cell cultures of rat hippocampal neurons were used in all experiments, gene expression was evaluated by RT-qPCR, and protein expression was assessed by Western blot analysis and immunocytochemistry. Cell viability was assessed by using the MTT method. The results demonstrated that PRL treatment of neurons from primary cultures did not modify cell viability, but that it exerted a neuroprotective effect, with cells treated with PRL showing a significant increase of viability after glutamate (Glu)--induced excitotoxicity as compared with neurons treated with Glu alone. Cultured neurons expressed mRNA for both PRL and its receptor (PRLR), and both PRL and PRLR expression levels changed after the excitotoxic insult. Interestingly, the PRLR protein was detected as two main isoforms of 100 and 40 kDa as compared with that expressed in hypothalamic cells, which was present only as a 30 kDa variant. On the other hand, PRL was not detected in neuron cultures, either by western blot or by immunohistochemistry. Neuroprotection induced by PRL was significantly blocked by specific oligonucleotides against PRLR, thus suggesting that the PRL role is mediated by its receptor expressed in these neurons. The overall results indicated that PRL induces neuroprotection in neurons from primary cell cultures.


Subject(s)
Hippocampus/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Prolactin/pharmacology , Receptors, Prolactin/metabolism , Analysis of Variance , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Excitatory Amino Acid Agonists/toxicity , Gene Expression Regulation/drug effects , Glutamic Acid/toxicity , Oligonucleotides/pharmacology , Prolactin/genetics , Prolactin/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Prolactin/genetics
2.
Neurochem Res ; 39(7): 1219-31, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24706094

ABSTRACT

Thrombin effect increasing swelling-induced glutamate efflux was examined in cultured cortical astrocytes, cerebellar granule neurons (CGN), hippocampal and cortical neurons. Hypotonic glutamate efflux (monitored by D-[(3)H]aspartate) from cortical astrocytes was increased by thrombin (5 U/mL) to reach 16% of the cell pool in 5 min. Thrombin had lower effects in CGN, and marginal effects in hippocampal and cortical neurons. These differences were related to the magnitude of thrombin-evoked cytosolic calcium rise. The protease-activated receptor 1 is expressed in astrocytes and neurons. In astrocytes exposed to chemical ischemia (sodium iodoacetate plus sodium azide) D-[(3)H]aspartate release showed a first phase (20-40 min) of initial low efflux which progressively increases; and a second phase (40-60 min) of larger efflux coincident with cell swelling. Efflux at the first phase was 52% inhibited by the glutamate transporter blocker DL-threo-ß-benzyloxyaspartic-acid (TBOA) and 11% by the volume-sensitive pathway blocker phloretin. At the second phase, efflux was reduced 52 and 38% respectively, by these blockers. In CGN D-[(3)H]aspartate efflux increased sharply and then decreased. This efflux was 32% reduced by calcium omission, 46% by TBOA and 32% by 4-[(2-butyl-6,7dichloro-2-cyclopentyl-2,3-dihydro-1oxo-1H-inden-5-yl)oxy] butanoic-acid. Thrombin enhanced this release by 32%. Ischemic treatment increased astrocyte mortality from 4% in controls to 39 and 61% in ischemia and ischemia plus thrombin, respectively. Cell death was prevented by phloretin. CGN viability was unaffected by the treatment. These results suggest that coincidence of swelling and thrombin during ischemia elevates extracellular glutamate prominently from astrocyte efflux, which may endanger neurons in vivo.


Subject(s)
Aspartic Acid/metabolism , Astrocytes/metabolism , Hyponatremia/metabolism , Neurons/metabolism , Thrombin/toxicity , Tritium/metabolism , Animals , Astrocytes/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Neurons/drug effects , Rats , Rats, Wistar
3.
Neuroscience ; 168(4): 871-84, 2010 Jul 28.
Article in English | MEDLINE | ID: mdl-20004708

ABSTRACT

Cerebral water control is critical to maintain neuronal excitability, and to prevent injuries derived from brain swelling or shrinkage. The influence of aquaporins (AQPs) in the balance of water distribution between intracranial compartments is getting much experimental support. The importance of AQPs in fluid clearance during vasogenic brain edema seems well established but their role in cytotoxic swelling and in brain cell shrinkage is not known in detail. The main AQPs function as water channels anticipates their influence on cell volume changes as well as on the mechanisms of volume recovery, which include notably the osmolyte translocation across the cell membrane. Osmolyte fluxes permit the reestablishment of an osmotic balance and volume recovery in anisosmotic-elicited cell volume changes, but are also causal factors per se of brain cell swelling or shrinkage in pathological situations. This review aims to inform on the so far described functional interactions between AQPs and osmolyte fluxes and their volume-sensitive pathways. It also points to the coincidence of AQPs and activation of osmolyte fluxes in physiological and pathological conditions and to the importance of finding possible functional links between these two events, thus enlarging the possibilities via AQP manipulations, to prevent the adverse consequences of cell volume changes in brain.


Subject(s)
Aquaporin 4/metabolism , Water/metabolism , Animals , Aquaporin 4/chemistry , Aquaporin 4/genetics , Humans , Metals/metabolism , Permeability , Phosphorylation
4.
Neurochem Res ; 33(8): 1518-24, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18357525

ABSTRACT

Activation of protein-activated receptor (PAR-1) by thrombin potentiates the hyposmotic efflux of (3)H-D-aspartate and (3)H-taurine from cultured cerebellar astrocytes. This effect is mediated by a thrombin-elicited increase in cytosolic Ca(2+) levels [Ca(2+)](i) and the activation of phosphoinositide-3-kinase (PI3K). These signalling pathways operate independently showing additive effects if prevented simultaneously. The contribution of the Ca(2+)-mediated pathway to thrombin-increased D-aspartate or taurine efflux, evaluated by the inhibitory effect of preventing [Ca(2+)](i) rise, was higher for D-aspartate (64% efflux decrease) than for taurine (40% decrease). The PI3K blocker decreased 48% and 36% D-aspartate and taurine efflux, respectively. Hyposmolarity increases phosphorylation of EGFR and c-src, but thrombin did not enhance this effect. Blockade of EGFR/src phosphorylation marginally reduced (11-14%) the hyposmolarity plus thrombin efflux of D-aspartate; taurine efflux was more sensitive to these blockers (18-26%). Since thrombin has no effect increasing EGFR/src phosphorylation in astrocytes, the contribution of this transactivation pathway may represent the inhibition of the hyposmotic efflux solely.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Signal Transduction/physiology , Taurine/metabolism , Thrombin/metabolism , Animals , Astrocytes/cytology , Calcium/metabolism , Cells, Cultured , ErbB Receptors/metabolism , Humans , Osmolar Concentration , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , src-Family Kinases/metabolism
5.
Pflugers Arch ; 455(5): 859-72, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17899168

ABSTRACT

The present study in Swiss3T3 fibroblasts examines the effect of thrombin on hyposmolarity-induced osmolyte fluxes and RVD, and the contribution of the src/EGFR pathway. Thrombin (5 U/ml) added to a 30% hyposmotic medium markedly increased hyposmotic 3H-taurine efflux (285%), accelerated the volume-sensitive Cl- current (ICI-swell) and increased RVD rate. These effects were reduced (50-65%) by preventing the thrombin-induced intracellular Ca2+ [Ca2+]i rise with EGTA-AM, or with the phospholipase C (PLC) blocker U73122. Ca2+calmodulin (CaM) and calmodulin kinase II (CaMKII) also participate in this Ca2+-dependent pathway. Thrombin plus hyposmolarity increased src and EGFR phosphorylation, whose blockade by PP2 and AG1478, decreased by 30-50%, respectively, the thrombin effects on hyposmotic taurine efflux, ICI-swell and RVD. Ca2+- and src/EGFR-mediated pathways operate independently as shown by (1) the persistence of src and EGFR activation when [Ca2+]i rise is prevented and (2) the additive effect on taurine efflux, ICI-swell or RVD by simultaneous inhibition of the two pathways, which essentially suppressed these events. PLC-Ca2+- and src/EGFR-signaling pathways operate in the hyposmotic condition and because thrombin per se failed to increase taurine efflux and ICI-swell under isosmotic condition it seems that it is merely amplifying these previously activated mechanisms. The study shows that thrombin potentiates hyposmolarity-induced osmolyte fluxes and RVD by increasing src/EGFR-dependent signaling, in addition to the Ca2+-dependent pathway.


Subject(s)
Cell Size , Chlorides/metabolism , Hemostatics/metabolism , Taurine/pharmacokinetics , Thrombin/metabolism , Animals , Calcium/metabolism , Chloride Channels/physiology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Estrenes/pharmacology , Hemostatics/pharmacology , Mice , Osmotic Pressure/drug effects , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrrolidinones/pharmacology , Quinazolines , Signal Transduction/drug effects , Signal Transduction/physiology , Swiss 3T3 Cells , Thrombin/pharmacology , Tritium , Tyrphostins/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
6.
Acta Physiol (Oxf) ; 187(1-2): 93-102, 2006.
Article in English | MEDLINE | ID: mdl-16734746

ABSTRACT

Recent evidence documents the involvement of protein tyrosine kinases (TK) in the signalling network activated by hyposmotic swelling and regulatory volume decrease. Both receptor type and cytosolic TK participate as signalling elements in the variety of cell adaptive responses to volume changes, which include adhesion reactions, reorganization of the cytoskeleton, temporal deformation/remodelling of the membrane and stress-detecting mechanisms. The present review refers to the influence of TK on the activation/operation of the osmolyte efflux pathways, ultimately leading to cell volume recovery, i.e. the osmosensitive Cl- channel (Cl-swell), the K+ channels activated by swelling in the different cell types and the taurine efflux pathway as representative of the organic osmolyte pathway.


Subject(s)
Cell Physiological Phenomena , Ion Channels/physiology , Protein-Tyrosine Kinases/metabolism , Taurine/metabolism , Water-Electrolyte Balance/physiology , Animals , Cell Membrane/metabolism , Cell Size , Enzyme Activation , Humans
7.
Neurochem Res ; 30(12): 1589-97, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16362778

ABSTRACT

Changes in external osmolarity, including both hyper- or hyposmotic conditions, elicit the tyrosine phosphorylation of a number of tyrosine kinase receptors (TKR). We show here that the epidermal growth factor receptor (EGFR) is activated by both cell swelling (hyposmolarity, isosmotic urea, hyperosmotic sorbitol) or shrinkage (hyperosmotic NaCl or raffinose) and discuss the mechanisms by which these apparently opposed conditions come to the same effect, i.e., EGFR activation. Evidence suggests that this results from early activation of integrins, p38 and tyrosine kinases of the Src family, which are all activated in the two anisosmotic conditions. TKR transactivation by integrins and p38 is likely occurring via an effect on the metalloproteinases. Information discussed in this review, points to TKR as elements in osmotransduction as a useful mechanism to amplify and diversify the initial response to anisosmolarity and cell volume changes, due to their privileged situation as convergence point for numerous intracellular signaling pathways. The variety of effector pathways connected to TKR is advantageous for the cell to cope with the changes in cell volume including adaptation to stress, cytoskeleton remodeling, adhesion reactions, cell survival and the adaptive mechanisms to ultimately restore the original cell volume.


Subject(s)
Cell Size , ErbB Receptors/physiology , Signal Transduction/physiology , Animals , Humans , Osmolar Concentration , Phosphorylation
8.
Eur J Neurosci ; 22(7): 1636-42, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16197504

ABSTRACT

Osmolarity reduction (20%) elicited 3H-norepinephrine (NE) efflux from rat cortical synaptosomes. The hyposmotic NE release resulted from the following events: (i) a Na+-dependent and La3+-, Gd3+- and ruthenium red-sensitive depolarization; (ii) a cytosolic Ca2+ ([Ca2+]i) rise with contributions from external Ca2+ influx and internal Ca2+ release, probably through the mitochondrial Na+-Ca2+ exchanger; and (iii) activation of a [Ca2+]i-evoked, tetanus toxin (TeTX)-sensitive, PKC-modulated NE efflux mechanism. This sequence was established from results showing a drop in the hyposmotic [Ca2+]i rise by preventing depolarization with La3+, and by the inhibitory effects of Ca2+-free medium (EGTA; 50%), CGP37157 (the mitochondrial Na+-Ca2+ exchanger blocker; 48%), EGTA + CGP37157 or by EGTA-AM (> 95% in both cases). In close correspondence with these effects, NE efflux was 92% decreased by Na+ omission, 75% by La3+, 47% by EGTA, 50% by CGP37157, 90% by EGTA + CGP37157 and 88% by EGTA-AM. PKC influenced the intracellular Ca2+ release and, mainly through this action, modulated NE efflux. TeTX suppressed NE efflux. The K+-stimulated NE release, studied in parallel, was unaffected by Na+ omission, or by La3+, Gd3+ or ruthenium red. It was fully dependent on external Ca2+, insensitive to CGP37157 and abolished by TeTX. These results suggest that the hyposmotic events, although different from the K+-evoked depolarization and [Ca2+]i rise mechanisms, are able to trigger a depolarization-dependent, Ca2+-dependent and TeTX-sensitive mechanism for neurotransmitter release.


Subject(s)
Calcium/metabolism , Cerebral Cortex/cytology , Exocytosis/physiology , Norepinephrine/metabolism , Synaptosomes/physiology , Animals , Barium/pharmacology , Cadmium/pharmacology , Calcium Channel Blockers/pharmacology , Carbazoles/pharmacology , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Drug Interactions , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Exocytosis/drug effects , Indoles/pharmacology , Lanthanum/pharmacology , Osmolar Concentration , Phorbol Esters/pharmacology , Potassium Chloride/pharmacology , Protein Kinase C/pharmacology , Rats , Rats, Wistar , Ruthenium Red/pharmacology , Sodium/pharmacology , Sodium-Calcium Exchanger/pharmacology , Spectrometry, Fluorescence/methods , Synaptosomes/drug effects , Tetanus Toxin/pharmacology , Thiazepines/pharmacology , Time Factors , Tritium/metabolism , omega-Conotoxins/pharmacology
9.
Pflugers Arch ; 449(6): 526-36, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15630602

ABSTRACT

An increase in extracellular KCl ([KCl]o) occurs under various pathological conditions in the retina, leading to retinal swelling and possible neuronal damage. The mechanisms of this KCl o-induced retinal swelling were investigated in the present study, with emphasis on the Cl- transport mechanisms. Increasing [KCl]o (from 5 to 70 mM) led to concentration-dependent swelling in chicken retinas. The curve relating the degree of swelling to [KCl]o was biphasic, with one component from 5 to 35 mM [KCl]o and another from 35 to 100 mM. As Cl- omission prevented swelling in all conditions, the effect of cotransporter or Cl- channel blockers was examined to investigate the mechanisms of Cl- influx. The cotransporter blockers bumetanide and DIOA reduced swelling by 68% and 76%, respectively at [KCl]o 25 mM (K25), and by 14-17% at [KCl]o 54 mM (K54). The Cl- channel blockers NPPB and niflumic acid did not affect swelling at K25 but reduced it by 90-95% at K54 (NPPB IC50 60.7 microM). Furosemide showed an atypical effect, decreasing swelling by 14% at K25 and by 95% at K54 (IC50 173.9 microM). Na+ omission decreased swelling at K25 but not at K54. These results suggest the contribution of cotransporters to Cl- influx at K25 and of Cl- channels at K54. At K25, swelling was found in the ganglion cell layer and in the lower half of the inner nuclear layer. With K54, swelling occurred in all inner retinal layers. The ganglion cell layer swelling was due to both Muller cell end-foot and ganglion cell soma swelling. K54 also induced ganglion cell damage as shown by disorganized, pyknotic and refringent nuclei.


Subject(s)
Chlorides/metabolism , Papilledema/metabolism , Potassium Chloride/pharmacology , Retinal Ganglion Cells/metabolism , Acetates/pharmacology , Animals , Bumetanide/pharmacology , Chickens , Chloride Channels/antagonists & inhibitors , Chloride Channels/metabolism , Diuretics/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Indenes/pharmacology , Nitrobenzoates/pharmacology , Papilledema/chemically induced , Papilledema/pathology , Potassium/metabolism , Receptors, Glutamate/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Rubidium Radioisotopes , Sodium Potassium Chloride Symporter Inhibitors , Sodium-Potassium-Chloride Symporters/metabolism
10.
Am J Physiol Cell Physiol ; 286(6): C1399-409, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14736709

ABSTRACT

Volume changes and whole cell ionic currents activated by gradual osmolarity reductions (GOR) of 1.8 mosM/min were characterized in C6 glioma cells. Cells swell less in GOR than after sudden osmolarity reductions (SOR), the extent of swelling being partly Ca(2+) dependent. In nominally Ca(2+)-free conditions, GOR activated predominantly whole cell outward currents. Cells depolarized from the initial -79 mV to a steady state of -54 mV reached at 18% osmolarity reduction [hyposmolarity of -18% (H-18%)]. Recordings of Cl(-) and K(+) currents showed activation at H-3% of an outwardly rectifying Cl(-) current, with conductance of 1.6 nS, sensitive to niflumic acid and 5-nitro-2-(3-phenylpropylamino)benzoic acid, followed at H-18% by an outwardly rectifying K(+) current with conductance of 4.1 nS, inhibited by clofilium but insensitive to the typical K(+) channel blockers. With 200 nM Ca(2+) in the patch pipette, whole cell currents activated at H-3% and at H-13% cells depolarized from -77 to -63 mV. A K(+) current activated at H-1%, showing a rapid increase in conductance, suppressed by charybdotoxin and insensitive to clofilium. These results show the operation of two different K(+) channels in response to GOR in the same cell type, activated by Ca(2+) and osmolarity and with different osmolarity activation thresholds. Taurine and glutamate efflux, monitored by labeled tracers, showed delayed osmolarity thresholds of H-39 and H-33%, respectively. This observation clearly separates the Cl(-) and amino acid osmosensitive pathways. The delayed amino acid efflux may contribute to counteract swelling at more stringent osmolarity reductions.


Subject(s)
Cell Membrane/metabolism , Glioma/metabolism , Potassium Channels/metabolism , Water-Electrolyte Balance/physiology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Size/drug effects , Cell Size/physiology , Chloride Channels/drug effects , Chloride Channels/metabolism , Glutamic Acid/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Osmolar Concentration , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Rats , Taurine/metabolism , Water-Electrolyte Balance/drug effects
12.
J Neurochem ; 79(1): 143-51, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595766

ABSTRACT

Cultured cerebellar granule neurons exposed to gradual reductions in osmolarity (-1.8 mOsm/min) maintained constant volume up to -50% external osmolarity (pi(o)), showing the occurrence of isovolumetric regulation (IVR). Amino acids, Cl-, and K+ contributed at different phases of IVR, with early efflux threshold for [3H]taurine, D-[3H]aspartate (as marker for glutamate) of pi(o) -2% and -19%, respectively, and more delayed thresholds of -30% for [3H]glycine and -25% and -29%, respectively, for Cl- (125I) and K+ (86Rb). Taurine seems preferentially involved in IVR, showing the lowest threshold, the highest efflux rate (five-fold over other amino acids) and the largest cell content decrease. Taurine and Cl- efflux were abolished by niflumic acid and 86Rb by 15 mM Ba2+. Niflumic acid essentially prevented IVR in all ranges of pi(o). Cl--free medium impaired IVR when pi(o) decreased to -24% and Ba2+ blocked it only at a late phase of -30% pi(o). These results indicate that in cerebellar granule neurons: (i) IVR is an active process of volume regulation accomplished by efflux of intracellular osmolytes; (ii) the volume regulation operating at small changes of pi(o) is fully accounted for by mechanisms sensitive to niflumic acid, with contributions of both Cl- and amino acids, particularly taurine; (iii) Cl- contribution to IVR is delayed with respect to other niflumic acid-sensitive osmolyte fluxes (osmolarity threshold of -25% pi(o)); and (iv), K+ fluxes do not contribute to IVR until a late phase (< -30% pi(o)).


Subject(s)
Cell Size , Cerebellum/cytology , Neurons/physiology , Water-Electrolyte Balance , Amino Acids/metabolism , Animals , Barium/pharmacology , Cells, Cultured , Chlorides/administration & dosage , Chlorides/metabolism , Culture Media , Iodine Radioisotopes/metabolism , Niflumic Acid/pharmacology , Osmolar Concentration , Potassium/metabolism , Rats , Rubidium Radioisotopes/metabolism , Taurine/metabolism
13.
Pflugers Arch ; 442(5): 791-800, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11512036

ABSTRACT

A 30% decrease in osmolarity stimulated 3H-taurine, 3H-GABA and glutamate (followed as 3H-D-aspartate) efflux from rat hippocampal slices. 3H-taurine efflux was activated rapidly but inactivated slowly. It was decreased markedly by 100 microM 5-nitro-(3-phenylpropylamino)benzoic acid (NPPB) and 600 microM niflumic acid and inhibited strongly by tyrphostins AG18, AG879 and AG112 (25-100 microM), suggesting a tyrosine kinase-mediated mechanism. Hyposmolarity activated the mitogen-activated protein kinases (MAPK) extracellular-signal-related kinase-1/2 (ERK1/ERK2) and p38, but blockade of this reaction did not affect 3H-taurine efflux. Hyposmosis also activated phosphatidylinositide 3-kinase (PI3K) and its prevention by wortmannin (100 nM) essentially abolished 3H-taurine efflux. 3H-taurine efflux was insensitive to the protein kinase C (PKC) blocker chelerythrine (2.5 microM) or to cytochalasin E (3 microM). The release of 3H-GABA and 3H-D-aspartate occurred by a different mechanism, characterized by rapid activation and inactivation, insensitivity to NPPB, niflumic acid, tyrphostins or wortmannin. 3H-GABA and 3H-D-aspartate efflux was not due to external [NaCl] decrease, cytosolic Ca2+ increase or depolarization, or to reverse operation of the carrier. This novel mechanism of amino acid release may be mediated by Ca2+-independent exocytosis and modulated by PKC and actin cytoskeleton disruption, as suggested by its inhibition by chelerythrine and potentiation by 100 nM phorbol-12-myristate-13 acetate (PMA) and cytochalasin E. GABA and glutamate osmosensitive efflux may explain the hyposmolarity-elicited increase in amplitude of inhibitory and excitatory postsynaptic potentials in hippocampal slices as well as the hyperexcitability associated with hyponatraemia.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/metabolism , Osmolar Concentration , Taurine/metabolism , gamma-Aminobutyric Acid/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Androstadienes/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cytochalasins/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Glutamic Acid/chemistry , Hippocampus/drug effects , Imidazoles/pharmacology , In Vitro Techniques , Mitogen-Activated Protein Kinases/metabolism , Niflumic Acid/pharmacology , Nitrobenzoates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Pyridines/pharmacology , Rats , Taurine/chemistry , Tritium/chemistry , Tritium/metabolism , Tyrphostins/pharmacology , Wortmannin , gamma-Aminobutyric Acid/chemistry
14.
Biochim Biophys Acta ; 1538(2-3): 313-20, 2001 Apr 23.
Article in English | MEDLINE | ID: mdl-11336802

ABSTRACT

The role of phospholemman (PLM) in taurine and Cl(-) efflux elicited by 30% hyposmotic solution was studied in cultured cerebellar astrocytes with reduced PLM expression by antisense oligonucleotide (AO) treatment. PLM, a substrate for protein kinases (PK) C and A, is a protein that increases an anion current in Xenopus oocytes and forms taurine-selective channels in lipid bilayers. Taurine contributes as an osmolyte to regulatory volume decrease (RVD) and is highly permeable through PLM channels in bilayers. Two antisense oligonucleotides (AO1 and AO2) effectively decreased the expression of the PLM protein by 40% and 30%, respectively, and markedly reduced [(3)H]taurine efflux by 67% and 62%. AO treatment also decreased the osmosensitive release of Cl(-), followed as (125)I. The inhibition of Cl(-) efflux (23% for AO1 and 13% for AO2) was notably lower than for [(3)H]taurine. The contribution of PKC and PKA in the function of PLM was also evaluated in astrocytes. Pharmacological activation or inhibition of PKC and PKA revealed that the osmosensitive taurine efflux is essentially PKC-independent while (125)I efflux is reduced by the PKC blockers H-7 (21%) and Gö6983 (41%). The PKA activator forskolin and dbcAMP increased taurine efflux by 66-70% and (125)I efflux by 21-45%. Norepinephrine increased the osmosensitive taurine efflux at about the same extent as dbcAMP and forskolin, and this was reduced by PKA blockers. These results suggest that PLM plays a role in RVD in astrocytes by predominantly influencing taurine fluxes, which are modulated by PKA but not PKC.


Subject(s)
Astrocytes/metabolism , Membrane Proteins/biosynthesis , Phosphoproteins/biosynthesis , Taurine/metabolism , Animals , Animals, Newborn , Cells, Cultured , Chlorides/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gene Expression , Iodine Radioisotopes , Membrane Proteins/analysis , Membrane Proteins/genetics , Norepinephrine/pharmacology , Oligonucleotides, Antisense/pharmacology , Osmolar Concentration , Phosphoproteins/analysis , Phosphoproteins/genetics , Protein Kinase C/antagonists & inhibitors , Rats , Tritium
15.
Neurochem Int ; 38(2): 153-61, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11137884

ABSTRACT

The role of phosphorylation events on the activation and modulation of the osmosensitive (3)H-taurine release (OTR) was examined in cultured cerebellar granule neurons (CGN) stimulated with 30% hyposmotic solutions. OTR was not decreased when [Ca(2+)](i) rise evoked by hyposmolarity was prevented by EGTA-AM (50 microM) or depleted by treatment with 1 microM ionomycin in Ca(2+)-free medium. Accordingly, OTR was not inhibited by Ca(2+)-dependent signaling events. The calmodulin (CAM) blocker W-7 (50 microM) potentiated OTR while the Ca(2+)/CAM kinase blocker KN-93 (10 microM) was without effect. Blockade of PKC by H-7, H-8 (50 microM) and Gö6976 (1 microM), as well as activation by phorbol myristate acetate (PMA) (100 nM) did not influence OTR, but chronic treatment to down regulate PKC decreased it by 30%. Forskolin (20 microM) and 8-BrcAMP (10 microM) did not change OTR. Protein tyrosine phosphorylation seems to be of crucial importance in the activation and modulation of OTR, as it was markedly inhibited (90%) by tyrphostine A23 (50 microM) and potentiated by the tyrosine phosphatase inhibitor ortho-vanadate (100 microM). The PI3 kinase blocker wortmannin 100 nM essentially abolished OTR but LY294002 (10-100 microM) was without effect. This difference may be accounted for PI3K isoforms in neurons with different sensitivity to the blockers. Alternatively, the effect of wortmannin may be exerted not in PI3 kinase but instead on phospholipases, which are also sensitive to this blocker. The hyposmotic stimulus induced activation of Erk1/Erk2, but blockade of this effect by PD 98059 (50 microM) only marginally decreased OTR suggesting that the Erk1/Erk2 is an epiphenomenon, not directly involved in OTR activation.


Subject(s)
Cerebellum/metabolism , Neurons/metabolism , Protein Kinases/physiology , Taurine/metabolism , Androstadienes/pharmacology , Animals , Calcium/metabolism , Cerebellum/cytology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Intracellular Membranes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Osmolar Concentration , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Rats , Tyrosine/metabolism , Water-Electrolyte Balance , Wortmannin
16.
Nephron ; 86(4): 414-27, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11124589

ABSTRACT

In most cell types, hyposmotic swelling consistently elicits an increase in the concentration of cytosolic Ca2+ - [Ca2+]i - with contributions of extracellular and intracellular sources. The mechanisms of Ca2+ entry and release from endogenous sources are not fully clarified and may be cell specific. The ubiquity of the swelling-evoked [Ca2+]i rise makes Ca2+ a likely candidate for a role as osmotransducing signal. However, the regulatory volume decrease (RVD) which follows swelling and the osmolyte fluxes involved in this process are not always Ca2+ dependent. It was found that, with a few exceptions, in most cell types the osmosensitive Cl- efflux pathway and the swelling-activated organic osmolyte fluxes are Ca2+ independent. In contrast, Ca2+-dependent or Ca2+-independent K+ fluxes activated by swelling are detected, depending on the cell type. The close correlation found in this review between the Ca2+ dependence of RVD and that of the K+ channels activated by swelling led to the conclusion that it is the type of osmosensitive K+ pathway which largely confers the Ca2+ dependence to RVD. Interestingly, this coincidence of Ca2+-dependent K+ efflux and RVD is found predominantly in epithelial cells, whereas in nonepithelial cells both processes are largely Ca2+ independent. In these cells, the [Ca2+]i rise elicited by swelling may be an epiphenomenon.


Subject(s)
Calcium Signaling/physiology , Calcium/physiology , Kidney/cytology , Potassium Channels/physiology , Water-Electrolyte Balance/physiology , Animals , Humans
17.
Cell Physiol Biochem ; 10(5-6): 361-70, 2000.
Article in English | MEDLINE | ID: mdl-11125217

ABSTRACT

Brain adaptation to hyposmolarity is accomplished by loss of both electrolytes and organic osmolytes, including amino acids, polyalcohols and methylamines. In brain in vivo, the organic osmolytes account for about 35% of the total solute loss. This review focus on the role of amino acids in cell volume regulation, in conditions of sudden hyposmosis, when cells respond by active regulatory volume decrease (RVD) or after gradual exposure to hyposmotic solutions, a condition where cell volume remains unchanged, named isovolumetric regulation (IVR). The amino acid efflux pathway during RVD is passive and is similar in many respects to the volume-activated anion pathway. The molecular identity of this pathway is still unknown, but the anion exchanger and the phospholemman are good candidates in certain cells. The activation trigger of the osmosensitive amino acid pathway is unclear, but intracellular ionic strength seems to be critically involved. Tyrosine protein kinases markedly influence amino acid efflux during RVD and may play an important role in the transduction signaling cascades for osmosensitive amino acid fluxes. During IVR, amino acids, particularly taurine are promptly released with an efflux threshold markedly lower than that of K(+), emphasizing their contribution (possibly as well as of other organic osmolytes) vs inorganic ions, in the osmolarity range corresponding to physiopathological conditions. Amino acid efflux also occurs in response to isosmotic swelling as that associated with ischemia or trauma. Characterization of the pathway involved in this type of swelling is hampered by the fact that most osmolyte amino acids are also neuroactive amino acids and may be released in response to stimuli concurrent with swelling, such as depolarization or intracellular Ca(++) elevation.


Subject(s)
Amino Acids/metabolism , Brain/metabolism , Brain/cytology , Cell Size , Osmolar Concentration
18.
Neurochem Res ; 25(9-10): 1301-14, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11059803

ABSTRACT

Brain cell swelling compromises neuronal function and survival by the risk of generation of ischemia episodes as compression of small vessels occurs due to the limits to expansion imposed by the rigid skull. External osmolarity reductions or intracellular accumulation of osmotically active solutes result in cell swelling which can be counteracted by extrusion of osmolytes through specific efflux pathways. Characterization of these pathways has received considerable attention, and there is now interest in the understanding of the intracellular signaling events involved in their activation and regulation. Calcium and calmodulin, phosphoinositides and cAMP may act as second messengers, carrying the information about a cell volume change into signaling enzymes. Small GTPases, protein tyrosine kinases and phospholipases, also appear to be part of the signaling cascades ultimately modulating the osmolyte efflux pathways. This review focus on i) the influence of hyposmotic and isosmotic swelling on these signaling events and molecules and ii) the effects of manipulating their function on the osmolyte fluxes, particularly K+, CI- and amino acids, and on the consequent efficiency of cell volume adjustment.


Subject(s)
Brain/cytology , Brain/physiology , Neurons/cytology , Neurons/physiology , Signal Transduction/physiology , Animals , Cell Size/physiology , Humans
19.
J Neurosci Res ; 61(6): 701-11, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10972967

ABSTRACT

The efflux of potassium (K(+)) and amino acids from hippocampal slices was measured after sudden exposure to 10% (270 mOsm), 25% (225 mOsm) or 50% (150 mOsm) hyposmotic solutions or after gradual decrease (-2.5 mOsm/min) in external osmolarity. In slices suddenly exposed to 50% hyposmotic solutions, swelling was followed by partial (74%) cell volume recovery, suggesting regulatory volume decrease (RVD). With gradual hyposmotic changes, no increase in cell water content was observed even when the solution at the end of the experiment was 50% hyposmotic, showing the occurrence of isovolumic regulation (IVR). The gradual decrease in osmolarity elicited the efflux of (3)H-taurine with a threshold at -5 mOsm and D-[(3)H]aspartate (as marker for glutamate) and at -20 mOsm for [(3)H]GABA. The efflux rate of [(3)H]taurine was always notably higher than those of [(3)H]GABA and D-[(3)H]aspartate, with a maximal increase over the isosmotic efflux of about 7-fold for [(3)H]taurine and 3- and 2-fold for [(3)H]GABA and D-[(3)H]aspartate, respectively. The amino acid content in slices exposed to 50% hyposmotic solutions (abrupt change) during 20 min decreased by 50. 6% and 62.6% (gradual change). Taurine and glutamate showed the largest decrease. An enhancement in (86)Rb efflux and a corresponding decrease in K(+) tissue content was seen in association with RVD but not with IVR. These results demonstrate the contribution of amino acids to IVR and indicate their involvement in this mechanism of cell volume control.


Subject(s)
Amino Acids/metabolism , Hippocampus/physiology , Neurons/metabolism , Water-Electrolyte Balance/physiology , Animals , Aspartic Acid/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Chloride Channels/antagonists & inhibitors , Chloride Channels/metabolism , Colforsin/analogs & derivatives , Colforsin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Hippocampus/cytology , Hypotonic Solutions/pharmacology , Male , Neurons/cytology , Niflumic Acid/pharmacology , Nitrobenzoates/pharmacology , Organ Culture Techniques , Osmolar Concentration , Potassium/metabolism , Rats , Rats, Wistar , Rubidium Radioisotopes , Taurine/metabolism , Tritium , Water/metabolism , Water-Electrolyte Balance/drug effects , gamma-Aminobutyric Acid/metabolism
20.
Biochim Biophys Acta ; 1496(2-3): 252-60, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10771093

ABSTRACT

The role of the phospholemman (PLM) on the efflux of taurine and chloride induced by swelling was studied in HEK293 cells overexpressing stable transfected PLM. PLM, a substrate for protein kinases C and A, is a protein that induces an anion current in Xenopus oocytes and forms taurine-selective channels in lipid bilayers. Taurine contributes as an osmolyte to regulatory volume decrease (RVD) and is highly permeable through PLM channels in bilayers. In PLM-overexpressing cells the process of RVD was more rapid and efficient (75%) than in control cells (44%). Also, [(3)H]taurine and (125)I efflux induced by hyposmolarity were markedly increased (30-100%) in two subclones of cells overexpressing PLM. This increased efflux was sensitive to the Cl channel blockers DDF, NPPB and DIDS. Acute treatment of control cells with isoproterenol and norepinephrine induced a significant potentiation (50-60%) of [(3)H]taurine release induced by hyposmolarity. In PLM-overexpressing cells the potentiation by these drugs was higher (100%). Insulin induced also an increase in [(3)H]taurine release, but only in PLM-overexpressing cells (50%). These results indicate that PLM may play a role in the RVD and that its phosphorylation may have a physiological significance during this process. The mechanisms involved in this process could include the activation of PLM itself as channel or the modulation of other preexisting channels.


Subject(s)
Ion Channels/drug effects , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Taurine/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Cell Line , Cell Size/drug effects , Colforsin/analogs & derivatives , Colforsin/pharmacology , Down-Regulation , Humans , Iodine Radioisotopes , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Nitrobenzoates/pharmacology , Osmolar Concentration , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Taurine/analysis , Transfection , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...