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










Publication year range
1.
J Immunol Res ; 2019: 1094520, 2019.
Article in English | MEDLINE | ID: mdl-31236418

ABSTRACT

Cardiotonic steroids, such as ouabain and digoxin, are known to bind to Na+/K+-ATPase and to promote several biological activities, including anti-inflammatory activity. However, there are still no reports in the literature about inflammation and marinobufagenin, a cardiotonic steroid from the bufadienolide family endogenously found in mammals. Therefore, the aim of this work was to analyze, in vivo and in vitro, the role of marinobufagenin in acute inflammation. Swiss mice were treated with 0.56 mg/kg of marinobufagenin intraperitoneally (i.p.) and zymosan (2 mg/mL, i.p.) was used to induce peritoneal inflammation. Peritoneal fluid was collected and used for counting cells by optical microscopy and proinflammatory cytokine quantification (IL-1ß, IL-6, and TNF-α) by immunoenzymatic assay (ELISA). Zymosan stimulation, as expected, induced increased cell migration and proinflammatory cytokine levels in the peritoneum. Marinobufagenin treatment reduced polymorphonuclear cell migration and IL-1ß and IL-6 levels in the peritoneal cavity, without interfering in TNF-α levels. In addition, the effect of marinobufagenin was evaluated using peritoneal macrophages stimulated by zymosan (0.2 mg/mL) in vitro. Marinobufagenin treatment at different concentrations (10, 100, 1000, and 10000 nM) showed no cytotoxic effect on peritoneal macrophages. Interestingly, the lowest concentration, which did not inhibit Na+/K+-ATPase activity, attenuated proinflammatory cytokines IL-1ß, IL-6, and TNF-α levels. To investigate the putative mechanism of action of marinobufagenin, the expression of surface molecules (TLR2 and CD69) and P-p38 MAPK were also evaluated, but no significant effect was observed. Thus, our results suggest that marinobufagenin has an anti-inflammatory role in vivo and in vitro and reveals a novel possible endogenous function of this steroid in mammals.


Subject(s)
Bufanolides/pharmacology , Chemotaxis, Leukocyte/drug effects , Cytokines/biosynthesis , Enzyme Inhibitors/pharmacology , Inflammation Mediators/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Animals , Biomarkers , Cell Survival/drug effects , Cytotoxicity, Immunologic/drug effects , Female , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Phosphorylation , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors
2.
Front Physiol ; 7: 195, 2016.
Article in English | MEDLINE | ID: mdl-27313535

ABSTRACT

Decreased Na(+), K(+)-ATPase (NKA) activity causes energy deficiency, which is commonly observed in neurodegenerative diseases. The NKA is constituted of three subunits: α, ß, and γ, with four distinct isoforms of the catalytic α subunit (α1-4). Genetic mutations in the ATP1A2 gene and ATP1A3 gene, encoding the α2 and α3 subunit isoforms, respectively can cause distinct neurological disorders, concurrent to impaired NKA activity. Within the central nervous system (CNS), the α2 isoform is expressed mostly in glial cells and the α3 isoform is neuron-specific. Mutations in ATP1A2 gene can result in familial hemiplegic migraine (FHM2), while mutations in the ATP1A3 gene can cause Rapid-onset dystonia-Parkinsonism (RDP) and alternating hemiplegia of childhood (AHC), as well as the cerebellar ataxia, areflexia, pescavus, optic atrophy and sensorineural hearing loss (CAPOS) syndrome. Data indicates that the central glutamatergic system is affected by mutations in the α2 isoform, however further investigations are required to establish a connection to mutations in the α3 isoform, especially given the diagnostic confusion and overlap with glutamate transporter disease. The age-related decline in brain α2∕3 activity may arise from changes in the cyclic guanosine monophosphate (cGMP) and cGMP-dependent protein kinase (PKG) pathway. Glutamate, through nitric oxide synthase (NOS), cGMP and PKG, stimulates brain α2∕3 activity, with the glutamatergic N-methyl-D-aspartate (NMDA) receptor cascade able to drive an adaptive, neuroprotective response to inflammatory and challenging stimuli, including amyloid-ß. Here we review the NKA, both as an ion pump as well as a receptor that interacts with NMDA, including the role of NKA subunits mutations. Failure of the NKA-associated adaptive response mechanisms may render neurons more susceptible to degeneration over the course of aging.

3.
Planta Med ; 81(3): 200-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25615276

ABSTRACT

Diene valepotriates obtained from Valeriana glechomifolia present antidepressant-like activity, mediated by dopaminergic and noradrenergic neurotransmissions. Also, previous studies have shown inhibitory activity of diene valepotriates towards Na(+)/K(+)-ATPase from the rat brain in vitro. Nevertheless, in vivo studies regarding the action of diene valepotriates on this enzyme are still lacking. Considering that Na(+)/K(+)-ATPase cerebral activity is involved in depressive disorders, the aim of this study was to investigate the effects of acute (5 mg/kg, p. o.) and repeated (5 mg/kg, p. o., once a day for three days) diene valepotriate administration on Na(+)/K(+)-ATPase activity in the cortex and hippocampus of mice submitted or not submitted to the forced swimming test. In addition, the protein expression of Na(+)/K(+)-ATPase α1, α2, and α3 isoforms in the cortex of mice repeatedly treated with diene valepotriates (and submitted or not submitted to the forced swimming test) was investigated. Diene valepotriates significantly decreased mice immobility time in the forced swimming test when compared to the control group. Only the animals repeatedly treated with diene valepotriates presented increased Na(+)/K(+)-ATPase activity in the cerebral cortex, and the exposure to the forced swimming test counteracted the effects of the diene valepotriates. No alterations in the hippocampal Na(+)/K(+)-ATPase activity were observed. Repeated diene valepotriate administration increased the cortical content of the α2 isoform, but the α3 isoform protein expression was augmented only in mice repeatedly treated with diene valepotriates and forced to swim. Mice treated with the vehicle and submitted to the forced swimming test also presented an increase in the content of the α2 isoform, but no alterations in Na(+)/K(+)-ATPase activity. These results suggest that cortical Na(+)/K(+)-ATPase may represent a molecular target of the diene valepotriates in vivo and long-term regulatory mechanisms are involved in this effect. Also, the forced swimming test per se influences the protein expression of Na(+)/K(+)-ATPase isoforms and counteracts the effects of the diene valepotriates on cortical Na(+)/K(+)-ATPase.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Iridoids/pharmacology , Motor Activity/drug effects , Plant Extracts/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Valerian/chemistry , Animals , Antidepressive Agents/therapeutic use , Brain/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Iridoids/therapeutic use , Male , Mice , Phytotherapy , Plant Extracts/therapeutic use , Protein Isoforms , Stress, Psychological/drug therapy , Swimming
4.
PLoS One ; 9(10): e108776, 2014.
Article in English | MEDLINE | ID: mdl-25290152

ABSTRACT

Cardiotonic steroids are used to treat heart failure and arrhythmia and have promising anticancer effects. The prototypic cardiotonic steroid ouabain may also be a hormone that modulates epithelial cell adhesion. Cardiotonic steroids consist of a steroid nucleus and a lactone ring, and their biological effects depend on the binding to their receptor, Na,K-ATPase, through which, they inhibit Na+ and K+ ion transport and activate of several intracellular signaling pathways. In this study, we added a styrene group to the lactone ring of the cardiotonic steroid digoxin, to obtain 21-benzylidene digoxin (21-BD), and investigated the effects of this synthetic cardiotonic steroid in different cell models. Molecular modeling indicates that 21-BD binds to its target Na,K-ATPase with low affinity, adopting a different pharmacophoric conformation when bound to its receptor than digoxin. Accordingly, 21-DB, at relatively high µM amounts inhibits the activity of Na,K-ATPase α1, but not α2 and α3 isoforms. In addition, 21-BD targets other proteins outside the Na,K-ATPase, inhibiting the multidrug exporter Pdr5p. When used on whole cells at low µM concentrations, 21-BD produces several effects, including: 1) up-regulation of Na,K-ATPase expression and activity in HeLa and RKO cancer cells, which is not found for digoxin, 2) cell specific changes in cell viability, reducing it in HeLa and RKO cancer cells, but increasing it in normal epithelial MDCK cells, which is different from the response to digoxin, and 3) changes in cell-cell interaction, altering the molecular composition of tight junctions and elevating transepithelial electrical resistance of MDCK monolayers, an effect previously found for ouabain. These results indicate that modification of the lactone ring of digoxin provides new properties to the compound, and shows that the structural change introduced could be used for the design of cardiotonic steroid with novel functions.


Subject(s)
Apoptosis/drug effects , Digoxin/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Tight Junctions/drug effects , Animals , Cardenolides/metabolism , Cardenolides/pharmacology , Cell Line, Tumor , Digoxin/analogs & derivatives , Digoxin/chemistry , Enzyme Activation/drug effects , Humans , Mice , Models, Molecular , Molecular Conformation , Neoplasms/genetics , Neoplasms/metabolism , Rats , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics
5.
PLoS One ; 9(6): e99510, 2014.
Article in English | MEDLINE | ID: mdl-24941071

ABSTRACT

LASSBio-1135 is an imidazo[1,2-a]pyridine derivative with high efficacy in screening models of nociception and inflammation, presumed as a weak COX-2 inhibitor. In order to tease out its mechanism of action, we investigated others possible target for LASSBio-1135, such as TNF-α and TRPV1, to better characterize it as a multitarget compound useful in the treatment of chronic pain. TRPV1 modulation was assessed in TRPV1-expressing Xenopus oocytes against capsaicin and low pH-induced current. Modulation of TNF-α production was evaluated in culture of macrophages stimulated with LPS. In vivo efficacy of LASSBio-1135 was investigated in carrageenan and partial sciatic ligation-induced thermal hyperalgesia and mechanical allodynia. Corroborating its previous demonstration of efficacy in a model of capsaicin-induced hyperalgesia, LASSBio-1135 blocks capsaicin-elicited currents in a non-competitive way with an IC50 of 580 nM as well as low pH-induced current at 50 µM. As an additional action, LASSBio-1135 inhibited TNF-α release in these cells stimulated by LPS with an IC50 of 546 nM by reducing p38 MAPK phosphorilation. Oral administration of 100 µmol x Kg(-1) LASSBio-1135 markedly reduced thermal hyperalgesia induced by carrageenan, however at 10 µmol x Kg(-1) only a partial reduction was observed at the 4th h. Neutrophil recruitment and TNF-α production after carrageenan stimulus was also inhibited by the treatment with LASSBio-1135. Modulating TRPV1 and TNF-α production, two key therapeutic targets of neuropathic pain, 100 µmol x Kg(-1) LASSBio-1135 was orally efficacious in reversing thermal hyperalgesia and mechanical allodynia produced by partial sciatic ligation 7-11 days after surgery without provoking hyperthermia, a common side effect of TRPV1 antagonists. In conclusion LASSBio-1135, besides being a weak COX-2 inhibitor, is a non-competitive TRPV1 antagonist and a TNF-α inhibitor. As a multitarget compound, LASSBio-1135 is orally efficacious in a model of neuropathic pain without presenting hyperthermia.


Subject(s)
Imidazoles/administration & dosage , Imidazoles/therapeutic use , Inflammation/drug therapy , Neuralgia/drug therapy , Pyridines/administration & dosage , Pyridines/therapeutic use , TRPV Cation Channels/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Administration, Oral , Animals , Capsaicin/pharmacology , Carrageenan , Disease Models, Animal , Enzyme Activation/drug effects , Female , Hydrogen-Ion Concentration , Hyperalgesia/complications , Hyperalgesia/drug therapy , Imidazoles/pharmacology , Inflammation/complications , MAP Kinase Signaling System/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice, Inbred BALB C , Neuralgia/complications , Neutrophil Infiltration/drug effects , Oocytes/metabolism , Pyridines/pharmacology , Rats , TRPV Cation Channels/metabolism , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis , Xenopus laevis , p38 Mitogen-Activated Protein Kinases/metabolism
6.
J Physiol Biochem ; 69(2): 207-14, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23011780

ABSTRACT

The effect of long-lasting in vivo restriction of nitric oxide (NO) bioavailability on cardiac and renal P-type ATPases critical for intracellular ion homeostasis is controversial. Previous work has shown in eNOS knockout (eNOS(-/-)) mice hearts that Na(+)/K(+)- and Ca(2+)-ATPase activities were depressed but the underlying mechanisms are still unclear. The goal of this study was to characterize potential alterations responsible for impaired enzyme activity in eNOS(-/-) mice. Na(+)/K(+)-ATPase activity from crude preparations of adult male eNOS(-/-) mice hearts and kidneys was reduced compared with wild-type animals (32 %, p < 0.05 and 16 %, p < 0.0001, respectively). Immunoblot analysis showed that although the expression of the predominant (or exclusive, for the kidney) Na(+)/K(+)-ATPase α1 isoform was not significantly changed, there was an important downregulation of the less abundant α2 isoform in the heart (57 %, p < 0.0001). In addition, although cardiac Ca(2+)-ATPase activity was unaltered, the expression of sarco/endoplasmic reticulum Ca(2+)-ATPase 2 protein in eNOS(-/-) mice was very high (290 % compared with wild-type animals, p < 0.0001) without any significant change in phospholamban expression. Consistent with these findings, the content of cardiac and renal free sulfhydryl groups, essential for the catalytic function of such ATPases, was decreased (23 %, p < 0.01 and 35 %, p < 0.05, respectively). Altogether, the present results suggest that the absence of eNOS promotes a compartmentalized altered redox balance that affects the activity and expression of ion transport ATPases.


Subject(s)
Calcium-Transporting ATPases/metabolism , Nitric Oxide Synthase Type III/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Male , Mice , Mice, Knockout , Myocardium/enzymology , Myocardium/metabolism , Nitric Oxide Synthase Type III/metabolism , Phenotype
7.
Reproduction ; 144(6): 737-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23028124

ABSTRACT

Novel roles for the interaction of cardiotonic steroids to Na(+)/K(+)-ATPase have been established in recent years. The aim of this study was to investigate the intracellular signaling events downstream the action of ouabain on Na(+)/K(+)-ATPase in Sertoli cell obtained from immature rats. Treatment of Sertoli cells with ouabain (1 µM) induced a rapid and transient increase in the extracellular signal-regulated kinase (ERK1/2 or MAPK3/1) and phosphatidylinositol 3-kinase (PI3K)/serine-threonine protein kinase (AKT) phosphorylation. Also, ouabain upregulated the expression of cyclin D1 and incorporation of [methyl-(3)H]thymidine, both of which were dependent on MAPK3/1 but not AKT intracellular cascade, as shown by pretreatment with MEK (MAP2K1/2) inhibitor U0126 and PI3K inhibitor wortmannin respectively. Moreover, the effect of ouabain on these proliferation parameters was completely prevented by phospho-cAMP response element-binding protein (CREB)/CREB-binding protein complex inhibitor KG501 and only partially by nuclear factor κB nuclear translocation inhibitor SN50. Pretreatment with estrogen receptor antagonist ICI 182780 showed that MAPK3/1 activation by ouabain does not involve this receptor. The Na(+)/K(+)-ATPase α1 isoform, but not α4, was detected in Sertoli cells, suggesting that ouabain effects in Sertoli cells are mediated via α1. Taken together, these results show a rapid ouabain action in the Sertoli cells, which in turn can modulate nuclear transcriptional events essential for Sertoli cell proliferation in a critical period of testicular development. Our findings are important to understand the role of ouabain in the testis and its possible implications in male infertility.


Subject(s)
Cardiac Glycosides/pharmacology , MAP Kinase Signaling System/drug effects , Ouabain/pharmacology , Sertoli Cells/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin D1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , NF-kappa B/metabolism , Phosphorylation , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Sertoli Cells/enzymology
8.
Am J Physiol Renal Physiol ; 302(8): F959-66, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22237798

ABSTRACT

Bufadienolides are structurally related to the clinically relevant cardenolides (e.g., digoxin) and are now considered as endogenous steroid hormones. Binding of ouabain to Na(+)-K(+)-ATPase has been associated, in kidney cells, to the activation of the Src kinase pathway and Na(+)-K(+)-ATPase internalization. Nevertheless, whether the activation of this cascade also occurs with other cardiotonic steroids and leads to diuresis and natriuresis in the isolated intact kidney is still unknown. In the present work, we perfused rat kidneys for 120 min with bufalin (1, 3, or 10 µM) and measured its vascular and tubular effects. Thereafter, we probed the effect of 10 µM 3-(4-chlorophenyl)1-(1,1-dimethylethyl)-1H-pyrazolo[3,4-d]pyrimidin-4amine (PP2), a Src family kinase inhibitor, and 1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio] butadiene (UO126), a highly selective inhibitor of both MEK1 and MEK2, on bufalin-induced renal alterations. Bufalin at 3 and 10 µM profoundly increased several parameters of renal function in a time- and/or concentration-dependent fashion. At a concentration that produced similar inhibition of the rat kidney Na(+)-K(+)-ATPase, ouabain had a much smaller diuretic and natriuretic effect. Although bufalin fully inhibited the rat kidney Na(+)-K(+)-ATPase in vitro, its IC(50) (33 ± 1 µM) was threefold higher than the concentration used ex vivo and all its renal effects were blunted by PP2 and UO126. Furthermore, the phosphorylated (activated) ERK1/2 expression was increased after bufalin perfusion and this effect was totally prevented after PP2 pretreatment. The present study shows for the first time the direct diuretic, natriuretic, and kaliuretic effects of bufalin in isolated rat kidney and the relevance of Na(+)-K(+)-ATPase-mediated signal transduction.


Subject(s)
Bufanolides/pharmacology , Enzyme Inhibitors/pharmacology , Kidney/drug effects , Natriuretic Agents/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , src-Family Kinases/metabolism , Animals , Butadienes/pharmacology , Diuresis/drug effects , Kidney/enzymology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Male , Natriuresis/drug effects , Nitriles/pharmacology , Ouabain , Potassium/urine , Pyrimidines/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors
9.
Pharmacol Rep ; 63(4): 1029-39, 2011.
Article in English | MEDLINE | ID: mdl-22001991

ABSTRACT

We investigated the effects of LASSBio-998 (L-998), a compound designed to be a p38 MAPK (mitogen-activated protein kinase) inhibitor, on lipopolysaccharide (LPS)-induced acute lung inflammation in vivo. BALB/c mice were challenged with aerosolized LPS inhalation (0.5 mg/ml) 4 h after oral administration of L-998. Three hours after LPS inhalation, bronchoalveolar lavage fluid was obtained to measure the levels of the proinflammatory cytokines TNF-α (tumor necrosis factor-α) and IL-1 (interleukin-1) and the chemokines MCP-1 (monocyte chemoattractant protein-1) and KC (keratinocyte chemoattractant). In addition, neutrophil infiltration and p38 MAPK phosphorylation was measured. L-998 inhibited LPS-induced production of TNF-α and IL-1ß and did not alter KC and MCP-1 levels. Furthermore, L-998 also significantly decreased neutrophil accumulation in lung tissues. As expected, L-998 diminished p38 MAPK phosphorylation and reduced acute lung inflammation. Inhibition of p38 MAPK phosphorylation by L-998 was also demonstrated in LPS-challenged murine C57BL/6 peritoneal macrophages in vitro, with concentration-dependent effects. L-998 suppressed LPS-induced lung inflammation, most likely by inhibition of the cytokine-p38 MAPK pathway, and we postulate that L-998 could be a clinically relevant anti-inflammatory drug candidate.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Quinolines/pharmacology , Urea/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/administration & dosage , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Dose-Response Relationship, Drug , In Vitro Techniques , Inflammation/pathology , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Lung/drug effects , Lung/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Phosphorylation/drug effects , Quinolines/administration & dosage , Urea/administration & dosage
10.
PLoS One ; 6(8): e23547, 2011.
Article in English | MEDLINE | ID: mdl-21853150

ABSTRACT

BACKGROUND AND AIMS: Schistosomiasis is an intravascular parasitic disease associated with inflammation. Endothelial cells control leukocyte transmigration and vascular permeability being modulated by pro-inflammatory mediators. Recent data have shown that endothelial cells primed in vivo in the course of a disease keep the information in culture. Herein, we evaluated the impact of schistosomiasis on endothelial cell-regulated events in vivo and in vitro. METHODOLOGY AND PRINCIPAL FINDINGS: The experimental groups consisted of Schistosoma mansoni-infected and age-matched control mice. In vivo infection caused a marked influx of leukocytes and an increased protein leakage in the peritoneal cavity, characterizing an inflamed vascular and cellular profile. In vitro leukocyte-mesenteric endothelial cell adhesion was higher in cultured cells from infected mice as compared to controls, either in the basal condition or after treatment with the pro-inflammatory cytokine tumor necrosis factor (TNF). Nitric oxide (NO) donation reduced leukocyte adhesion to endothelial cells from control and infected groups; however, in the later group the effect was more pronounced, probably due to a reduced NO production. Inhibition of control endothelial NO synthase (eNOS) increased leukocyte adhesion to a level similar to the one observed in the infected group. Besides, the adhesion of control leukocytes to endothelial cells from infected animals is similar to the result of infected animals, confirming that schistosomiasis alters endothelial cells function. Furthermore, NO production as well as the expression of eNOS were reduced in cultured endothelial cells from infected animals. On the other hand, the expression of its repressor protein, namely caveolin-1, was similar in both control and infected groups. CONCLUSION/SIGNIFICANCE: Schistosomiasis increases vascular permeability and endothelial cell-leukocyte interaction in vivo and in vitro. These effects are partially explained by a reduced eNOS expression. In addition, our data show that the disease primes endothelial cells in vivo, which keep the acquired phenotype in culture.


Subject(s)
Cell Communication , Endothelial Cells/pathology , Leukocytes/pathology , Schistosomiasis/pathology , Animals , Caveolin 1/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Endothelial Cells/enzymology , Male , Mesentery/pathology , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Peritoneal Cavity/pathology , Schistosoma mansoni/physiology , Schistosomiasis/enzymology
11.
Planta Med ; 77(15): 1702-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21567360

ABSTRACT

Valepotriates are iridoids found in variable amounts in Valerianaceae and might be among the bioactive compounds which confer anxiolytic properties to the Valeriana species. On the other hand, unspecific cytotoxicity has also been described. Presently, however, no particular molecular target has been defined for these compounds. Here we studied the effect of valtrate, acevaltrate, and 1- ß-acevaltrate isolated from Valeriana glechomifolia on the enzymatic activity of rat P-type ATPases. Valepotriates did not affect rat skeletal muscle sarco/endoplasmic reticulum Ca²âº-ATPase (SERCA) activity at the highest concentration used (100 µM). In contrast, the same concentration inhibited roughly half of the total H⁺/K⁺-ATPase activity from rat gastric epithelium (valtrate 54.6 ± 3.2 %, acevaltrate 60.7 ± 7.3 %, 1- ß-acevaltrate 50.2 ± 3.1 %; mean ± SEM, n = 3-5). Finally, these substances showed the highest inhibitory potency toward Na⁺/K⁺-ATPase, and the inhibition curves obtained provided a similar IC50 (in µM) for rat kidney α1 isoform (valtrate 21.2, acevaltrate 22.8, 1- ß-acevaltrate 24.4) and brain hemispheres α2/ α3 isoforms (valtrate 19.4, acevaltrate 42.3, 1- ß-acevaltrate 38.3). Our results suggest that P-type ATPases are differentially inhibited by valepotriates and that Na⁺/K⁺-ATPase might be one of their molecular targets in vivo.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Iridoids/pharmacology , Valerian/chemistry , Adenosine Triphosphatases/drug effects , Adenosine Triphosphatases/metabolism , Animals , Brain/enzymology , Epithelium/enzymology , H(+)-K(+)-Exchanging ATPase/drug effects , H(+)-K(+)-Exchanging ATPase/metabolism , Inhibitory Concentration 50 , Iridoids/chemistry , Iridoids/isolation & purification , Kidney/enzymology , Male , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Stomach/enzymology
12.
J Mol Cell Cardiol ; 49(3): 525-31, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20451529

ABSTRACT

Recent studies have demonstrated that the Na(+)/K(+)-ATPase is not only an ion pump, but also a membrane receptor that confers the ligand-like effects of cardiotonic steroids (CTS) such as ouabain on protein kinases and cell growth. Because CTS have been implicated in cardiac fibrosis, this study examined the role of caveolae in the regulation of Na(+)/K(+)-ATPase function and CTS signaling in cardiac fibroblasts. In cardiac fibroblasts prepared from wild-type and caveolin-1 knockout [Cav-1(-/-)] mice, we found that the absence of caveolin-1 did not affect total cellular amount or surface expression of Na(+)/K(+)-ATPase alpha1 subunit. However, it did increase ouabain-sensitive (86)Rb(+) uptake. While knockout of caveolin-1 increased basal activities of Src and ERK1/2, it abolished the activation of these kinases induced by ouabain but not angiotensin II. Finally, ouabain stimulated collagen synthesis and cell proliferation in wild type but not Cav-1(-/-) cardiac fibroblasts. Thus, we conclude that caveolae are important for regulating both pumping and signal transducing functions of Na(+)/K(+)-ATPase. While depletion of caveolae increases the pumping function of Na(+)/K(+)-ATPase, it suppresses CTS-induced signal transduction, growth, and collagen production in cardiac fibroblasts.


Subject(s)
Caveolin 1/physiology , Fibroblasts/metabolism , Heart/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Western , Cardiotonic Agents/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation , Collagen/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Heart/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Ouabain/pharmacology , Signal Transduction
13.
Biochem Pharmacol ; 77(6): 1029-39, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19161990

ABSTRACT

Neutrophil accumulation response to cigarette smoke (CS) in humans and animal models is believed to play an important role in pathogenesis of many tobacco-related lung diseases. Here we evaluated the lung anti-inflammatory effect of aspirin and indomethacin in mice exposed to CS. C57BL/6 mice were exposed to four cigarettes per day during 4 days and were treated i.p. with aspirin or indomethacin, administered each day 1h before CS exposure. Twenty four hours after the last exposure, cells and inflammatory mediators were assessed in bronchoalveolar lavage (BAL) fluid and the lungs used for evaluation of lipid peroxidation, p38 mitogen-activated protein kinase (MAPK) phosphorylation and nuclear transcription factor kappaB (NF-kappaB) activation. Exposure to CS resulted in a marked lung neutrophilia. Moreover, the levels of oxidative stress-related lipid peroxidation, prostaglandin E(2) (PGE(2)), interleukin 1beta (IL-1beta), monocyte chemotactic protein 1 (MCP-1), and activated NF-kappaB and p38 MAPK were greatly increased in CS group. Aspirin or indomethacin treatment led to a significant reduction of neutrophil influx, but only aspirin resulted in dramatic decrease of inflammatory mediators. Moreover, both drugs reduced lung p38 MAPK and NF-kappaB activation induced by CS. These results demonstrate that short-term CS exposure has profound airway inflammatory effects counteracted by the anti-inflammatory agents aspirin and indomethacin, probably through COX-dependent and -independent mechanisms.


Subject(s)
Aspirin/therapeutic use , Indomethacin/therapeutic use , Nicotiana/adverse effects , Pneumonia/pathology , Pneumonia/prevention & control , Smoke/adverse effects , Animals , Bronchoalveolar Lavage Fluid/cytology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pneumonia/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...