Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Type of study
Publication year range
1.
Comp Biochem Physiol B Biochem Mol Biol ; 275: 111023, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39154851

ABSTRACT

A critical role of omega-3 polyunsaturated fatty acids (PUFA), mainly docosahexaenoic acid 22:6ω3 (DHA), in the development and function of the brain and visual system is well established. DHA, the most abundant omega-3 PUFA in the vertebrate brain, contributes to neuro- and synaptogenesis, neuronal differentiation, synaptic transmission and plasticity, neuronal network formation, memory and behaviour formation. Based on these data, the unique importance of DHA and its irreplaceability in neural and retinal tissues has been postulated. In this review, we consider omega-3 PUFA composition in the brain and retina of various invertebrates, and show that DHA has only been found in marine mollusks and crustaceans. A gradual decrease in the DHA content until its disappearance can be observed in the brain lipids of the series marine-freshwater-terrestrial crustaceans and marine-terrestrial mollusks, suggesting that the transition to the land lifestyle in the evolution of invertebrates, but not vertebrates, was accompanied by a loss of DHA. As with terrestrial crustaceans and mollusks, DHA was not found in insects, either terrestrial or aquatic, or in nematodes. We show that the nervous and visual systems of various DHA-free invertebrates can be highly enriched in alpha-linolenic acid 18:3ω3 or eicosapentaenoic acid 20:5ω3, which affect neurological and visual function, stimulating synaptogenesis, synaptic transmission, visual processing, learning and even cognition. The review data show that, in animals at different levels of organization, omega-3 PUFA are required for the functioning of the nervous and visual systems and that their specific needs can be met by various omega-3 PUFA.

2.
Cells ; 12(4)2023 02 18.
Article in English | MEDLINE | ID: mdl-36831324

ABSTRACT

Impairment of the blood-brain barrier (BBB) integrity is implicated in the numerous neurological disorders associated with neuroinflammation, neurodegeneration and aging. It is now evident that short-chain fatty acids (SCFAs), mainly acetate, butyrate and propionate, produced by anaerobic bacterial fermentation of the dietary fiber in the intestine, have a key role in the communication between the gastrointestinal tract and nervous system and are critically important for the preservation of the BBB integrity under different pathological conditions. The effect of SCFAs on the improvement of the compromised BBB is mainly based on the decrease in paracellular permeability via restoration of junctional complex proteins affecting their transcription, intercellular localization or proteolytic degradation. This review is focused on the revealed and putative underlying mechanisms of the direct and indirect effects of SCFAs on the improvement of the barrier function of brain endothelial cells. We consider G-protein-coupled receptor-mediated effects of SCFAs, SCFAs-stimulated acetylation of histone and non-histone proteins via inhibition of histone deacetylases, and crosstalk of these signaling pathways with transcriptional factors NF-κB and Nrf2 as mainstream mechanisms of SCFA's effect on the preservation of the BBB integrity.


Subject(s)
Blood-Brain Barrier , Microbiota , Endothelial Cells/metabolism , Fatty Acids, Volatile/metabolism , Butyrates/metabolism
3.
Pharmaceutics ; 13(2)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499252

ABSTRACT

Lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, is the most abundant proinflammatory agent. Considerable evidence indicates that LPS challenge inescapably causes oxidative stress and mitochondrial dysfunction, leading to cell and tissue damage. Increased mitochondrial reactive oxygen species (mtROS) generation triggered by LPS is known to play a key role in the progression of the inflammatory response. mtROS at excessive levels impair electron transport chain functioning, reduce the mitochondrial membrane potential, and initiate lipid peroxidation and oxidative damage of mitochondrial proteins and mtDNA. Over the past 20 years, a large number of mitochondria-targeted antioxidants (mito-AOX) of different structures that can accumulate inside mitochondria and scavenge free radicals have been synthesized. Their protective role based on the prevention of oxidative stress and the restoration of mitochondrial function has been demonstrated in a variety of common diseases and pathological states. This paper reviews the current data on the beneficial application of different mito-AOX in animal endotoxemia models, in either in vivo or in vitro experiments. The results presented in our review demonstrate the promising potential of approaches based on mito-AOX in the development of new treatment strategies against Gram-negative infections and LPS per se.

4.
Article in English | MEDLINE | ID: mdl-30660802

ABSTRACT

Previously we showed that arginine-vasotocin (AVT)-stimulated osmotic water permeability (OWP) of the frog urinary bladder was decreased if the mucosal side of the bladder has been naturally colonized by Gram-negative bacteria, or if bacterial lipopolysaccharide (LPS) was introduced into the lumen of the isolated bladder (J. Exp. Zool., 2013, 319, 487-494). Taking into account that in different tissues and cell types, challenge with LPS causes significant metabolic shift and energy deficiency, we hypothesized that an LPS-induced decrease of AVT-stimulated OWP could depend on the reduction of fatty acid oxidation (FAO), which is important for generation of ATP in epithelia. Using an isolated frog Rana temporaria urinary bladder we showed that the AVT-induced increase of OWP did not depend on the external glucose, but was inhibited by oligomycin, an ATP-synthase inhibitor, and by etomoxir, an inhibitor of carnitine palmitoyltransferase-1. In primary cultured epithelial cells isolated from the bladder mucosa, LPS E. coli (25 µg/ml, 21 h), as well as etomoxir (100 µM), decreased FAO accompanied by triacylglycerol accumulation. Both drugs impaired mitochondrial functions demonstrated by decreased ATP production and a reduced maximal oxygen consumption rate (OCR) and OCR directed at ATP synthesis. Additionally, we found that LPS decreased the expression of peroxisome proliferator-activated receptor alpha, a key player in the regulation of FAO. These data indicate that the impairment of AVT-induced water transport in osmoregulatory epithelium caused by LPS depends at least partly on defects in FAO and FAO-dependent energy production.


Subject(s)
Lipopolysaccharides/toxicity , Osmosis/drug effects , Rana temporaria , Urinary Bladder/drug effects , Water/metabolism , Animals , Cells, Cultured , Energy Metabolism , Epithelial Cells/drug effects , Escherichia coli/metabolism , Fatty Acids/metabolism , Lipid Peroxidation , Male , Osmosis/physiology , Urinary Bladder/cytology , Urinary Bladder/physiology
5.
J Lipids ; 2018: 5745790, 2018.
Article in English | MEDLINE | ID: mdl-30245885

ABSTRACT

The effect of bacterial lipopolysaccharide (LPS) on eukaryotic cell could be accompanied by a significant metabolic shift that includes accumulation of triacylglycerol (TAG) in lipid droplets (LD), ubiquitous organelles associated with fatty acid storage, energy regulation and demonstrated tight spatial and functional connections with mitochondria. The impairment of mitochondrial activity under pathological stimuli has been shown to provoke TAG storage and LD biogenesis. However the potential mechanisms that link mitochondrial disturbances and TAG accumulation are not completely understood. We hypothesize that mitochondrial ROS (mROS) may play a role of a trigger leading to subsequent accumulation of intracellular TAG and LD in response to a bacterial stimulus. Using isolated epithelial cells from the frog urinary bladder, we showed that LPS decreased fatty acids oxidation, enhanced TAG deposition, and promoted LD formation. LPS treatment did not affect the mitochondrial membrane potential but increased cellular ROS production and led to impairment of mitochondrial function as revealed by decreased ATP production and a reduced maximal oxygen consumption rate (OCR) and OCR directed at ATP turnover. The mitochondrial-targeted antioxidant MitoQ at a dose of 25 nM did not prevent LPS-induced alterations in cellular respiration, but, in contrast to nonmitochondrial antioxidant α-tocopherol, reduced the effect of LPS on the generation of ROS, restored the LPS-induced decline of fatty acids oxidation, and prevented accumulation of TAG and LD biogenesis. The data obtained indicate the key signaling role of mROS in the lipid metabolic shift that occurs under the impact of a bacterial pathogen in epithelial cells.

6.
Biochim Biophys Acta ; 1851(3): 239-47, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25499607

ABSTRACT

Exogenous gangliosides are known to inhibit the effects of Escherichia coli lipopolysaccharide (LPS) in different cells exhibiting anti-inflammatory and immunosuppressive activities. The mechanisms underlying ganglioside action are not fully understood. Because LPS recognition and receptor complex formation occur in lipid rafts, and gangliosides play a key role in their maintenance, we hypothesize that protective effects of exogenous gangliosides would depend on inhibition of LPS signaling via prevention of TLR4 translocation into lipid rafts. The effect of GM1 and GD1a gangliosides on LPS-induced toxic and inflammatory reactions in PC12 cells, and in epithelial cells isolated from the frog urinary bladder, was studied. In PC12 cells, GD1a and GM1 significantly reduced the effect of LPS on the decrease of cell survival and on stimulation of reactive oxygen species production. In epithelial cells, gangliosides decreased LPS-stimulated iNOS expression, NO, and PGE2 production. Subcellular fractionation, in combination with immunoblotting, showed that pretreatment of cells with GM1, GD1a, or methyl-ß-cyclodextrin, completely eliminated the effect of LPS on translocation of TLR4 into lipid rafts. The results are consistent with the hypothesis that ganglioside-induced prevention of TLR4 translocation into lipid rafts could be a mechanism of protection against LPS in various cells.


Subject(s)
G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Membrane Microdomains/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Cattle , Cell Survival/drug effects , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Escherichia coli/chemistry , Gene Expression Regulation , Lipopolysaccharides/toxicity , Male , Membrane Microdomains/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , PC12 Cells , Primary Cell Culture , Protein Transport , Rana temporaria , Rats , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Urinary Bladder/cytology , Urinary Bladder/drug effects , Urinary Bladder/metabolism , beta-Cyclodextrins/pharmacology
7.
J Exp Zool A Ecol Genet Physiol ; 319(9): 487-94, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23836531

ABSTRACT

In frogs and toads the urinary bladder is very important for the maintenance of water balance due to its ability to store water which can be reabsorbed under the action of arginine-vasotocin (AVT). The usage of isolated bladders as a model for studying the osmotic water permeability (OWP) regulation has a disadvantage which relates to high variability of AVT effect among individuals, some showing insensitivity to the hormone. We hypothesized that the response of the bladder to AVT could depend on the colonization of the mucosal epithelium by Gram-negative bacteria. To test this, paired hemibladders of the frog Rana temporaria were used for measurement of OWP and for analysis of Gram-negative bacteria in the bladder tissue or isolated epithelial cells. Among the 206 frogs studied, 41% were infected by different Enterobacteriaceae, with prevalence of Hafnia alvei and Escherichia coli. In infected bladders the basal level of OWP was unchanged, whereas OWP stimulated by AVT was reduced (non-infected: 2.53 ± 0.13, n = 59, infected: 1.21 ± 0.17 µL min(-1) cm(-2), n = 38, for the 15 min of AVT action, P < 0.001). In the sample, 100% of hemibladders that responded to AVT very weakly (OWP <0.5 µL min(-1) cm(-2)) had a bacterial infection. Overnight treatment of hemibladders with mucosal lipopolysaccharide E. coli decreased OWP induced by AVT, forskolin, or IBMX lowering basal and stimulated level of cAMP. The data obtained indicate that the frog bladder epithelium could be colonized by Gram-negative bacteria, probably of cloacal origin, leading to reduction of sensitivity to AVT and to impairment of the urinary bladder to provide osmoregulation.


Subject(s)
Enterobacteriaceae/pathogenicity , Osmotic Pressure/drug effects , Rana temporaria/microbiology , Urinary Bladder/microbiology , Absorption , Animals , Arginine/administration & dosage , Arginine/metabolism , Enterobacteriaceae/drug effects , Enterobacteriaceae/isolation & purification , Organ Culture Techniques , Osmoregulation/drug effects , Permeability/drug effects , Vasotocin/administration & dosage , Vasotocin/metabolism , Water/chemistry
8.
Am J Physiol Regul Integr Comp Physiol ; 303(10): R1042-52, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23019216

ABSTRACT

As in mammals, epithelium of the amphibian urinary bladder forms a barrier to pathogen entry and is a first line of defense against penetrating microorganisms. We investigated the effect of Escherichia coli LPS on generation of nitric oxide (NO), a critically important mediator during infectious processes, by primary cultured frog (Rana temporaria) urinary bladder epithelial cells (FUBEC). It was found that FUBEC constitutively express Toll-like receptor 4 (TLR4), a receptor of LPS, and respond to LPS (10 µg/ml) by stimulation of inducible nitric oxide synthase (iNOS) mRNA/protein expression and NOS activity measured by nitrite produced in the culture medium and by citrulline assay. We characterized uptake of l-arginine, a precursor in NO synthesis, by FUBEC and showed that it is mediated mainly by the y+ cationic amino acid transport system. LPS stimulated l-arginine uptake, and this effect was blocked by the iNOS inhibitor 1400W. Arginase II was found to be expressed in FUBEC. Inhibition of arginase activity by (S)-(boronoethyl)-l-cysteine increased generation of NO, suggesting contribution of arginase to NO production via competing with NOS for the substrate. LPS altered neither total arginase activity nor arginase II expression. Among epithelial cells, phagocytic macrophage-like cells were observed, but they did not contribute to LPS-induced NO production. These data demonstrate that amphibian urinary bladder epithelial cells recognize LPS and respond to it by increased generation of NO via stimulation of iNOS expression and l-arginine uptake, which appears to be essential for the regulation of the innate immune response and the inflammation in bladder epithelium.


Subject(s)
Epithelial Cells/metabolism , Lipopolysaccharides/toxicity , Ranidae/physiology , Toll-Like Receptor 4/metabolism , Urinary Bladder/physiology , Urothelium/cytology , Amino Acid Sequence , Animals , Arginine/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Male , Molecular Sequence Data , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Toll-Like Receptor 4/genetics
9.
Br J Pharmacol ; 167(4): 826-38, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22612416

ABSTRACT

BACKGROUND AND PURPOSE: cGMP is involved in the regulation of many cellular processes including cardiac and smooth muscle contractility, aldosterone synthesis and inhibition of platelet activation. Intracellular effects cGMP are mediated by cGMP-dependent PKs, cGMP-regulated PDEs and cGMP-gated ion channels. PKG inhibitors are widely used to discriminate PKG-specific effects. They can be divided into cyclic nucleotide-binding site inhibitors such as Rp-phosphorothioate analogues (Rp-cGMPS), ATP-binding site inhibitors such as KT5823, and substrate binding site inhibitors represented by the recently described DT-oligopeptides. As it has been shown that Rp-cGMPS and KT5823 have numerous non-specific effects, we analysed the pharmacological properties of the oligopeptide (D)-DT-2 described as a highly specific, membrane-permeable, PKG inhibitor. EXPERIMENTAL APPROACH: Specificity and potency of (D)-DT-2 to inhibit PKG activity was evaluated using biochemical assays in vitro and by substrate phosphorylation analysis in various cell types including human platelets, rat mesangial cells and rat neonatal cardiomyocytes. KEY RESULTS: Despite potent inhibition of PKGI in vitro, (D)-DT-2 lost specificity for PKG in cell homogenates and particularly in living cells, as demonstrated by phosphorylation of different substrates. Instead, (D)-DT-2 modulated activity of other kinases including ERK, p38, PKB and PKC, thereby inducing unpredicted and often opposing functional effects. CONCLUSIONS AND IMPLICATIONS: We conclude that DT-oligopeptides, as other inhibitors, cannot be used to specifically inhibit PKG in intact cells. Therefore, no specific pharmacological PKG inhibitors are available, and reliable studies of PKG signalling can only be made by using RNA knockdown or genetic deletion methods.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Oligopeptides/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , HEK293 Cells , Humans , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Platelet Aggregation/drug effects , Rats , Rats, Sprague-Dawley
10.
Am J Physiol Regul Integr Comp Physiol ; 293(1): R528-37, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17363677

ABSTRACT

PGE(2) is a well-known inhibitor of the antidiuretic hormone-induced increase of osmotic water permeability (OWP) in different osmoregulatory epithelia; however, the mechanisms underlying this effect of PGE(2) are not completely understood. Here, we report that, in the frog Rana temporaria urinary bladder, EP(1)-receptor-mediated inhibition of arginine-vasotocin (AVT)-induced OWP by PGE(2) is attributed to increased generation of nitric oxide (NO) in epithelial cells. It was shown that the inhibitory effect of 17-phenyl-trinor-PGE(2) (17-ph-PGE(2)), an EP(1) agonist, on AVT-induced OWP was significantly reduced in the presence of 7-nitroindazole (7-NI), a neuronal NO synthase (nNOS) inhibitor. NO synthase (NOS) activity in both lysed and intact epithelial cells measured as a rate of conversion of l-[(3)H]arginine to l-[(3)H]citrulline was Ca(2+) dependent and inhibited by 7-NI. PGE(2) and 17-ph-PGE(2), but not M&B-28767 (EP(3) agonist) or butaprost (EP(2) agonist), stimulated NOS activity in epithelial cells. The above effect of PGE(2) was abolished in the presence of SC-19220, an EP(1) antagonist. 7-NI reduced the stimulatory effect of 17-ph-PGE(2) on NOS activity. 17-ph-PGE(2) increased intracellular Ca(2+) concentration and cGMP in epithelial cells. Western blot analysis revealed an nNOS expression in epithelial cells. These results show that the inhibitory effect of PGE(2) on AVT-induced OWP in the frog urinary bladder is based at least partly on EP(1)-receptor-mediated activation of the NO/cGMP pathway, suggesting a novel cross talk between AVT, PGE(2), and nNOS that may be important in the regulation of water transport.


Subject(s)
Cyclic AMP/physiology , Dinoprostone/pharmacology , Nitric Oxide/physiology , Oxytocics/pharmacology , Receptors, Prostaglandin E/physiology , Signal Transduction/drug effects , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Vasotocin/antagonists & inhibitors , Vasotocin/pharmacology , Water-Electrolyte Balance/drug effects , Animals , Blotting, Western , Brain Chemistry/drug effects , Calcium/metabolism , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/analogs & derivatives , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , In Vitro Techniques , Indazoles/pharmacology , Male , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Permeability/drug effects , Rana temporaria , Receptors, Prostaglandin E, EP1 Subtype
11.
Pflugers Arch ; 448(2): 197-203, 2004 May.
Article in English | MEDLINE | ID: mdl-14722776

ABSTRACT

The present study addressed the question of whether nitric oxide (NO) participates in regulation of osmotic water permeability in the urinary bladder of the frog Rana temporaria L. Experiments were carried out on isolated, paired hemi-bladders filled with amphibian Ringer solution diluted 1:10 with distilled water. Sodium nitroprusside (SNP, 125-250 micro M), an NO donor, markedly attenuated the increase of osmotic water flow elicited by arginine-vasotocin (AVT) (AVT 10(-10) M: 2.20+/-0.26; AVT plus 200 micro M SNP: 1.21+/-0.15 micro l/min cm(2), n=20, P<0.001). This effect of SNP was apparent only in the presence of 50 micro M zaprinast, an inhibitor of the cGMP-specific phosphodiesterase-5 (PDE5). In the presence of zaprinast, SNP elevated cGMP production significantly both in control and AVT-stimulated urinary bladders, but had no effect on the level of cAMP (AVT 5 x 10(-10) M: 7.6+/-0.6; AVT plus SNP 200 micro M: 7.5+/-0.4 pmol/mg protein, n=8, N.S.). 1 H-[1,2,4]-oxadiazole-[4,3-a]-quinoxalin-1-one (ODQ, 25-100 micro M), an inhibitor of soluble guanylate cyclase, enhanced the AVT-induced water flow, decreased the SNP-stimulated increase of cGMP in the bladder tissue and almost abolished the inhibitory effect of SNP on the AVT-induced hydroosmotic response. 8-( p-Chlorophenylthio)-cGMP (8-pCPT-cGMP, 25 or 50 micro M), a membrane-permeable cGMP analogue specific for cGMP-dependent protein kinase (PKG), inhibited, whereas 2 micro M KT-5823, an inhibitor of PKG, significantly stimulated the increase of water flow induced by AVT. The inhibitory effect of SNP on AVT-induced water flow was almost completely reversed by KT-5823, but not by 50-100 micro M erythro-9-[2-hydroxy-3-nonyl]adenine (EHNA), an inhibitor of cGMP-activated PDE2. Immunohistochemistry of urinary bladder slices with antibodies against different types of NO synthase (NOS) revealed a positive immunostaining for neuronal NOS (nNOS) in the mucosal epithelium. These results suggest that in the frog urinary bladder endogenous NO is involved in regulation of water osmotic permeability. NO inhibits the AVT-induced increase of water flow at least partly by activation of PKG, which interferes with the hydroosmotic effect of AVT probably at (a) post-cAMP step(s).


Subject(s)
Adenine/analogs & derivatives , Nitric Oxide/pharmacology , Urinary Bladder/metabolism , Vasotocin/pharmacology , Adenine/pharmacology , Animals , Carbazoles/pharmacology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Exonucleases/antagonists & inhibitors , Immunohistochemistry , In Vitro Techniques , Indicators and Reagents , Indoles/pharmacology , Male , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitroprusside/pharmacology , Osmotic Pressure , Permeability , Phosphodiesterase Inhibitors/pharmacology , Rana temporaria , Urinary Bladder/drug effects
12.
J Am Soc Nephrol ; 14(5): 1107-15, 2003 May.
Article in English | MEDLINE | ID: mdl-12707381

ABSTRACT

Aldosterone exerts its effects through interactions with two types of binding sites, the mineralocorticoid (MR) and the glucocorticoid (GR) receptors. Although both receptors are known to be involved in the anti-natriuretic response to aldosterone, the mechanisms of signal transduction leading to modulation of electrolyte transport are not yet fully understood. This study measured the Na(+) and K(+) urinary excretion and the mRNA levels of three known aldosterone-induced transcripts, the serum and glucocorticoid-induced kinase (Sgk-1), the alpha subunit of the epithelial Na(+) channel (alphaENaC), and the glucocorticoid-induced-leucine-zipper protein (GILZ) in the whole kidney and in isolated cortical collecting tubules of adrenalectomized rats treated with low doses of aldosterone and/or dexamethasone. The resulting plasma concentrations of both steroids were close to 1 nmol/L. Aldosterone, given with or without dexamethasone, induced anti-natriuresis and kaliuresis, whereas dexamethasone alone did not. GILZ and alphaENaC transcripts were higher after treatment with either or both hormones, whereas the mRNA abundance of Sgk-1 was increased in the cortical collecting tubule by aldosterone but not by dexamethasone. We conclude the increased expression of Sgk-1 in the cortical collecting tubules is a primary event in the early antinatriuretic and kaliuretic responses to physiologic concentrations of aldosterone. Induction of alphaENaC and/or GILZ mRNAs may play a permissive role in the enhancement of the early and/or late responses; these effects may be necessary for a full response but do not by themselves promote early changes in urinary Na(+) and K(+) excretion.


Subject(s)
Aldosterone/pharmacology , Kidney Tubules, Collecting/metabolism , Nuclear Proteins , Protein Serine-Threonine Kinases/genetics , Sodium Channels/genetics , Transcription Factors/genetics , Aldosterone/blood , Animals , Dexamethasone/blood , Dexamethasone/pharmacology , Epithelial Sodium Channels , Gene Expression/drug effects , Gene Expression/physiology , Glucocorticoids/blood , Glucocorticoids/pharmacology , Immediate-Early Proteins , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Kidney Tubules, Collecting/drug effects , Male , Potassium/urine , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/analysis , Rats , Rats, Wistar , Sodium/urine , Sodium Channels/metabolism , Transcription Factors/metabolism
13.
Pflugers Arch ; 444(1-2): 159-66, 2002 May.
Article in English | MEDLINE | ID: mdl-11976928

ABSTRACT

The present study was performed to investigate the role of prostaglandin E(2) (PGE(2)) in the regulation of urea transport in the frog urinary bladder, which is known to occur via a specialized arginine-vasotocin- (AVT-) regulated urea transporter. The bladders isolated from Rana temporaria L. were filled with amphibian Ringer solution containing 370 Bq/ml (0.01 microCi/ml) of [14C]urea, and urea permeability ( P(urea)) was determined by sampling the serosal and mucosal bathing medium at 30-min intervals for measurement of radioactivity. It was found that, from the serosal side, PGE(2) (10 nM to 1 microM) caused a dose-dependent increase in P(urea) [(7.2+/-1.8)x10(-6) cm/s in the presence of 0.5 microM PGE(2)versus (1.0+/-0.2)x10(-6) cm/s in control, n=9, P<0.001]. As in response to AVT, the PGE(2)-induced P(urea)reached a maximum in 1-1.5 h after the agonist was added. The stimulatory effects of PGE(2) and AVT applied together were not additive. PGE(2)-induced urea transport was strongly inhibited by nearly 75% in the presence of mucosal or serosal phloretin (10(-4) M). P(urea) was enhanced up to (4.7+/-0.8)x10(-6) cm/s (n=12, P<0.001) by butaprost (5 x 10(-6) M), a selective EP(2) receptor agonist, while sulprostone (EP(1)/EP(3) agonist, 10(-6) M) caused no changes in P(urea). PGE(2)dose-dependently increased the content of cAMP in mucosal epithelial cells (control: 18.0+/-1.8; 10(-6) M PGE(2): 74.2+/-9.3 pmol cAMP/mg protein per 30 min, n=7, P<0.001). Phorbol esters did not alter PGE(2)-induced P(urea), whereas H-89 (20 microM), a protein kinase A inhibitor, reduced it by 45.1+/-9.9% ( n=5, P<0.05). PGE(2)did not change the AVT-stimulated P(urea) measured in isoosmotic conditions, but inhibited the last one in the presence of a serosa-to-mucosa osmotic gradient. The data obtained show that, in the frog urinary bladder, PGE(2)is a stimulator of phloretin-inhibitable urea transport. Its effect seems to be mediated by EP(2) receptor-coupled generation of intracellular cAMP.


Subject(s)
Dinoprostone/pharmacology , Sulfonamides , Urea/metabolism , Urinary Bladder/metabolism , Algorithms , Animals , Biological Transport, Active/drug effects , Cyclic AMP/metabolism , Dinoprostone/metabolism , In Vitro Techniques , Indicators and Reagents , Isoquinolines/pharmacology , Male , Osmolar Concentration , Phloretin/pharmacology , Rana temporaria , Receptors, Prostaglandin/agonists , Tetradecanoylphorbol Acetate/pharmacology , Urinary Bladder/drug effects , Vasotocin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL