Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Publication year range
1.
Cell Biosci ; 12(1): 10, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35090553

ABSTRACT

BACKGROUND: Obesity is caused by the enlargement of the white adipose tissue (WAT) depots, characterized by the hypertrophic enlargement of malfunctioning adipocytes within WAT which increases the storage of triglycerides (TG) in the lipid droplets (LD). Adipogenesis pathways as well as the expression and activity of some extracellular matrix receptors integrins are upregulated. Integrinß1 (INTB1) is the main isoform involved in WAT remodeling during obesity and insulin resistance-related diseases. We recently described Integrin Linked Kinase (ILK), a scaffold protein recruited by INTB1, as an important mediator of WAT remodeling and insulin resistance. As the few approved drugs to fight obesity have brought long-term cardiovascular side effects and given that the consideration of INTB1 and/or ILK modulation as anti-obesogenic strategies remains unexplored, we aimed to evaluate the anti-obesogenic capacity of the clinically approved anticoagulant Tirofiban (TF), stated in preclinical studies as a cardiovascular protector. METHODS: Fully differentiated adipocytes originating from C3H10T1/2 were exposed to TF and were co-treated with specific INTB1 blockers or with siRNA-based knockdown ILK expression. Lipid-specific dyes were used to determine the TG content in LD. The genetic expression pattern of ILK, pro-inflammatory cytokines (MCP1, IL6), adipogenesis (PPARγ, Leptin), thermogenesis (UCP1), proliferation (PCNA), lipid metabolism (FASN, HSL, ATGL), and metabolite transporters (FABP4, FAT, AQP7) were detected using quantitative PCR. Cytoskeletal actin polymerization was detected by confocal microscopy. Immunoblotting was performed to detect INTB1 phosphorylation at Thr788/9 and ILK activity as phosphorylation levels of protein kinase B (AKT) in Ser473 and glycogen synthase kinase 3ß (GSK3ß) at Ser9. TF was intraperitoneally administered once per day to wildtype and ILK knockdown mice (cKDILK) challenged with a high-fat diet (HFD) or control diet (STD) for 2 weeks. Body and WAT weight gains were compared. The expression of ILK and other markers was determined in the visceral epididymal (epi) and inguinal subcutaneous (sc) WAT. RESULTS: TF reduced TG content and the expression of adipogenesis markers and transporters in adipocytes, while UCP-1 expression was increased and the expression of lipases, cytokines or PCNA was not affected. Mechanistically, TF rapidly increased and faded the intracellular phosphorylation of INTB1 but not AKT or GSK3ß. F-actin levels were rapidly decreased, and INTB1 blockade avoided the TF effect. After 24 h, ILK expression and phosphorylation rates of AKT and GSK3ß were upregulated, while ILK silencing increased TG content. INTB1 blockade and ILK silencing avoided TF effects on the TG content and the transcriptional expression of PPARγ and UCP1. In HFD-challenged mice, the systemic administration of TF for several days reduced the weight gain on WAT depots. TF reduced adipogenesis and pro-inflammatory biomarkers and increased lipolysis markers HSL and FAT in epiWAT from HFD, while increased UCP1 in scWAT. In both WATs, TF upregulated ILK expression and activity, while no changes were observed in other tissues. In HFD-fed cKDILK, the blunted ILK in epiWAT worsened weight gain and avoided the anti-obesogenic effect of in vivo TF administration. CONCLUSIONS: ILK downregulation in WAT can be considered a biomarker of obesity establishment. Via an INTB1-ILK axis, TF restores malfunctioning hypertrophied WAT by changing the expression of adipocyte-related genes, increasing ILK expression and activity, and reducing TG storage. TF prevents obesity, a property to be added to its anticoagulant and cardiovascular protective advantages.

2.
Am J Physiol Cell Physiol ; 300(3): C683-91, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21160032

ABSTRACT

The nitric oxide (NO)-soluble guanylate cyclase (sGC) pathway exerts most of its cellular actions through the activation of the cGMP-dependent protein kinase (PKG). Accumulation of extracellular matrix is one of the main structural changes in pathological conditions characterized by a decreased activity of this pathway, such as hypertension, diabetes, or aging, and it is a well-known fact that extracellular matrix proteins modulate cell phenotype through the interaction with membrane receptors such as integrins. The objectives of this study were 1) to evaluate whether extracellular matrix proteins, particularly fibronectin (FN), modulate PKG expression in contractile cells, 2) to analyze the mechanisms involved, and 3) to evaluate the functional consequences. FN increased type I PKG (PKG-I) protein content in human mesangial cells, an effect dependent on the interaction with ß(1)-integrin. The FN upregulation of PKG-I protein content was due to increased mRNA expression, determined by augmented transcriptional activity of the PKG-I promoter region. Akt and the transcription factor CCAAT enhancer-binding protein (C/EBP) mediated the genesis of these changes. FN also increased PKG-I in another type of contractile cell, rat vascular smooth muscle cells (RVSMC). Tirofiban, a pharmacological analog of FN, increased PKG-I protein content in RVSMC and rat aortic walls and magnified the hypotensive effect of dibutyryl cGMP in conscious Wistar rats. The present results provide evidence of a mechanism able to increase PKG-I protein content in contractile cells. Elucidation of this novel mechanism provides a rationale for future pharmacotherapy in certain vascular diseases.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Fibronectins/physiology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Transcriptional Activation/physiology , Up-Regulation/physiology , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/genetics , Fibronectins/metabolism , Humans , Male , Mesangial Cells/cytology , Mesangial Cells/enzymology , Mesangial Cells/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
3.
Int J Biochem Cell Biol ; 39(1): 133-45, 2007.
Article in English | MEDLINE | ID: mdl-16978906

ABSTRACT

While arginine-glycine-aspartic acid-based peptidomimetics have been employed for the treatment of cardiovascular disorders and cancer, their use in other contexts remains to be explored. Arginine-glycine-aspartic acid-serine induces Transforming growth factor-beta1 transcription in human mesangial cells, but the molecular mechanisms involved have not been studied extensively. We explored whether this effect could be due to Activator protein-1 activation and studied the potential pathways involved. Addition of arginine-glycine-aspartic acid-serine promoted Activator protein-1 binding to its cognate sequence within the Transforming growth factor-beta1 promoter as well as c-jun and c-fos protein abundance. Moreover, this effect was suppressed by curcumin, a c-Jun N terminal kinase inhibitor, and was absent when the Activator protein-1 cis-regulatory element was deleted. Activator protein-1 binding was dependent on the activity of integrin linked kinase, as transfection with a dominant negative mutant suppressed both Activator protein-1 binding and c-jun and c-fos protein increment. Integrin linked kinase was, in turn, dependent on Phosphoinositol-3 kinase activity. Arginine-glycine-aspartic acid-serine stimulated Phosphoinositol-3 kinase activity, and Transforming growth factor-beta1 promoter activation was abrogated by the use of Phosphoinositol-3 kinase specific inhibitors. In summary, we propose that arginine-glycine-aspartic acid-serine activates Integrin linked kinase via the Phosphoinositol-3 kinase pathway and this leads to activation of c-jun and c-fos and increased Activator protein-1 binding and Transforming growth factor-beta1 promoter activity. These data may contribute to understand the molecular mechanisms involved in the cellular actions of arginine-glycine-aspartic acid-related peptides and enhance their relevance as these products evolve into clinical therapeutic use.


Subject(s)
Mesangial Cells/metabolism , Peptides, Cyclic/pharmacology , Promoter Regions, Genetic , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/biosynthesis , Up-Regulation/drug effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta1/genetics
4.
Br J Pharmacol ; 152(3): 313-22, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17643133

ABSTRACT

BACKGROUND AND PURPOSE: CGS-26303 inhibits endothelin converting enzyme (ECE)-1 more specifically than phosphoramidon. We have studied the effect of CGS-26303 on ECE-1 expression in bovine aortic endothelial cells. METHODS: ECE-1 activity and big endothelin (ET)-1 levels were measured by ELISA, ECE-1 expression using western and northern blot and promoter activity using transfection assays. KEY RESULTS: ECE-1 activity was completely inhibited by CGS-26303 25 microM and phosphoramidon 100 microM. CGS-26303 and phosphoramidon, though not thiorphan, a neutral endopeptidase (NEP) inhibitor, stimulated ECE-1 expression in cells (maximal effect at 16 h, 25 microM). Cycloheximide abolished that effect. CGS-26303 induced ECE-1 mRNA expression and ECE-1 promoter activity. CGS-35066, a selective ECE-1 inhibitor, mimicked the effects of CGS-26303, suggesting that the effect was specific to ECE-1 inhibition. Big ET-1 accumulated in the cells and in the supernatants after CGS-26303 treatment. Neither exogenously added ET-1 nor the blockade of their receptors with bosentan modified ECE-1 protein. When big ET-1 was added to cells, significant increases in ECE-1 protein content and ECE-1 promoter activity were found. Bosentan did not block those effects. CGS-26303 did not modify prepro-ET-1 expression. CGS-26303 and big ET-1 induced the same effects in human endothelial cells, at lower doses. CONCLUSIONS: These results suggest that the accumulation of big ET-1 is responsible for the effects of CGS-26303 on ECE-1 and they did not depend on NEP blockade. Changes in ECE-1 protein after the administration of CGS-26303 could lead to a decreased response in long-term treatments.


Subject(s)
Aspartic Acid Endopeptidases/drug effects , Aspartic Acid Endopeptidases/metabolism , Endothelin-1/drug effects , Metalloendopeptidases/drug effects , Metalloendopeptidases/metabolism , Organophosphonates/pharmacology , Protease Inhibitors/pharmacology , Tetrazoles/pharmacology , Animals , Aorta, Thoracic , Blotting, Northern , Blotting, Western , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelin-1/metabolism , Endothelin-Converting Enzymes , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Humans , Methyltransferases/drug effects , Methyltransferases/metabolism , Neprilysin/antagonists & inhibitors , Organophosphonates/administration & dosage , Promoter Regions, Genetic/drug effects , Protease Inhibitors/administration & dosage , Tetrazoles/administration & dosage , Transfection
5.
Circ Res ; 90(5): 539-45, 2002 Mar 22.
Article in English | MEDLINE | ID: mdl-11909817

ABSTRACT

Endothelial dysfunction, considered as a defective vascular dilatation after certain stimuli, is characteristic of different pathological conditions, such as hypertension, atherosclerosis, or diabetes. A decreased synthesis or an increased degradation of nitric oxide (NO) has been postulated as the mechanism responsible for this alteration. The present experiments were designed to test the hypothesis that the presence of an abnormal extracellular matrix in vessel walls could be responsible for the decreased NO synthesis observed in these pathological conditions. Experiments were performed in cultured human umbilical vein endothelial cells (HUVECs) grown on type IV (Col. IV) or type I (Col. I) collagen. Cells seeded on Col. I showed decreased nitrite synthesis, nitric oxide synthase activity, eNOS protein content, and eNOS mRNA expression when compared with cells grown on Col. IV. Moreover, cells grown on Col. I failed to respond to glucose oxidase activation of the eNOS system. In both cases, the changes in the eNOS mRNA expression seemed to depend on the modulation of eNOS promoter activity. The downregulation of eNOS induced by Col. I was blocked by D6Y, a peptide that interferes with the Col. I-dependent signals through integrins, as well as by specific anti-integrin antibodies. Moreover, a decreased activation of integrin-linked kinase (ILK) may explain the effects observed in Col. I-cultured cells because the activity of this kinase was decreased in these cells and ILK modulation prevented the Col. I-induced changes in HUVECs. Taken together, these findings may contribute to explaining the basis of endothelial dysfunction in some vascular diseases.


Subject(s)
Collagen Type I/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Cells, Cultured , Citrulline/metabolism , Collagen Type I/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Integrins/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Nitrites/metabolism , Peptides/pharmacology , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Biochim Biophys Acta ; 1643(1-3): 105-12, 2003 Dec 07.
Article in English | MEDLINE | ID: mdl-14654233

ABSTRACT

Natriuretic peptides (NP) activate particulate guanylate cyclase (pGC) and nitric oxide (NO) activates soluble guanylate cyclase (sGC). Both guanylate cyclases catalyse the formation of the same second messenger, cyclic guanosine 3',5'-monophosphate (cGMP), which activates the cGMP-dependent protein kinases (PKG). PKG then starts a signalling cascade that mediates many cardiovascular and renal effects, such as smooth muscle relaxation and diuresis. Many cell types possess both sGC and pGC. Because both GC-cGMP systems play complementary roles, an interaction between the two pathways might represent an important physiological control mechanism. In this report we demonstrate an interaction between the two pathways. C-type natriuretic peptide (CNP) decreased the beta-subunit of sGC (sGC-beta) steady-state protein levels and enzymatic activity in cultured human mesangial cells (HMC) in a time- and dose-dependent manner. This down-regulation was not dependent on changes in sGC-beta mRNA levels. Treatment of the cells with the stable cGMP analogue 8-Br-cGMP or the phosphodiesterase type-5 inhibitor Zaprinast produced the same down-regulatory effect. Inhibition of PKG or proteasome activity prevented the CNP-induced reduction of sGC-beta protein levels and activity. Taken together, these results demonstrate that pGC activation induces a post-transductional down-regulation of sGC by a mechanism involving PKG and the proteasome pathway.


Subject(s)
Cysteine Endopeptidases/metabolism , Guanylate Cyclase/metabolism , Multienzyme Complexes/metabolism , Natriuretic Peptide, C-Type/pharmacology , Cells, Cultured , Cyclic GMP , Down-Regulation/drug effects , Feedback, Physiological , Glomerular Mesangium/cytology , Guanylate Cyclase/analysis , Humans , Nitric Oxide/pharmacology , Proteasome Endopeptidase Complex , RNA, Messenger/analysis , Solubility
7.
Biochim Biophys Acta ; 1316(2): 102-8, 1996 Jun 07.
Article in English | MEDLINE | ID: mdl-8672546

ABSTRACT

Hyperplasia of the pancreatic tissue during late lactation (third week) and lasting for at least the first two weeks after weaning has been observed by several authors. Since the tetradecapeptide somatostatin (SS) inhibits pancreatic growth and its plasma levels are elevated during these periods, the aim of the present study was to determine the possible implication of the somatostatinergic system in the pancreatic changes cited above. Thus, the present study investigated 125I-Tyr(11)-somatostatin (125I-Tyr(11)-SS) binding and the effects of SS on guanylate cyclase activity as well as pancreatic somatostatin-like immunoreactivity (SSLI) levels in pancreatic acinar membranes from control, lactating and weaning rats. SS receptors were identified using 125I-Tyr(11)-SS and isolated pancreatic acinar membranes in vitro. There was an increase in the number of SS receptors after the third week of lactation (244 +/- 6 vs. 155 +/- 12 fmol/mg protein, P < 0.01) and the first two weeks after weaning (327 +/- 8 vs. 164 +/-10 fmol/mg protein, P < 0.001). No change in the affinity of the receptor site was detected at either study time. In addition, SS-stimulated guanylate cyclase activity was markedly increased at the third week of lactation (119%) and at the second week after weaning (158%) when compared with the control group. In contrast, basal guanylate cyclase activity was not modified at either study period. Thus, SS-stimulated guanylate cyclase activity is increased in pancreatic acinar membranes at late lactation and at the second week after beginning weaning probably due to an increase in the number of SS receptors. Significant decreases in SSLI content were observed at the third week of lactation (69%) and the second week after weaning (37%) when compared with the respective controls. The present results suggest that pancreatic acinar cell growth observed at the third week of lactation and the second week after weaning is associated with up-regulation of SS receptors which would represent a mechanism promoted by the cell that would negatively regulate the mitogenic activity of the increased number of pancreatic growth factors observed during both periods.


Subject(s)
Guanylate Cyclase/metabolism , Lactation , Pancreas/metabolism , Receptors, Somatostatin/metabolism , Weaning , Animals , Cell Membrane/metabolism , Cyclic AMP/metabolism , Female , Radioligand Assay , Rats , Rats, Wistar , Somatostatin/metabolism
8.
FASEB J ; 17(11): 1529-31, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12824296

ABSTRACT

Extracellular matrix (ECM) components, through specific peptide motifs such as Arg-Gly-Asp (RGD), interact with integrins and can modify the behavior of cells. Transforming growth factor-beta1 (TGF-beta1) is the main cytokine involved in the synthesis of ECM proteins. We analyzed the effect of a RGD-containing peptide, as Arg-Gly-Asp-Ser (RGDS), on the regulation of TGF-beta1 secretion in cultured human mesangial cells. We found that RGDS increased mRNA expression and secretion of TGF-beta1 by stimulating the TGF-beta1 gene promoter. This effect was dependent on the interaction of RGDS with integrins. We evaluated the signaling pathways implicated in TGF-beta1 production by analyzing the effect of RGDS on kinase-related integrins. RGDS stimulated tyrosine phosphorylation as well as integrin-linked kinase (ILK) activity. However, tyrosine kinase inhibitors did not prevent the RGDS effect. In contrast, the inhibition of ILK by cell transfection with a kinase dead-ILK completely abolished the increased TGF-beta1 secretion and promoter activity in the presence of RGDS. Thus RGDS modulates the secretion of TGF-beta1, probably through increased synthesis by interacting with integrins and activating ILK. This supports a role for ECM components in the regulation of their own secretion.


Subject(s)
Integrins/metabolism , Oligopeptides/pharmacology , Transforming Growth Factor beta/biosynthesis , Cells, Cultured , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Humans , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/biosynthesis , Transcriptional Activation , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Tyrosine/metabolism
9.
Endocrinology ; 136(8): 3444-51, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7628380

ABSTRACT

The present experiments were devoted to analyzing the hypothesis that somatostatin (SS) could modulate glomerular filtration rate by interacting with mesangial cells. Studies were performed in cultured human mesangial cells, passages 3-5. Radioligand experiments demonstrated the presence in the cells of two kinds of receptors, with high (dissociation constant 14 pM. Number of sites: 426 fmol/mg) and low (dissociation constant 56 pM. Number of sites: 20, 111 fmol/mg) affinity. SS prevented in a dose-dependent manner the reduction in planar cell surface area induced by 100 nM Angiotensin II (AII). This effect was not inhibited by the blockade of the vasorelaxing prostaglandins (indomethacin, 10 microM), nitric oxide (L-N-methyl-arginine, 0.2 mM), adenylate cyclase (2,5'-dideoxyadenosine, 0.1 mM), or guanylate cyclase (Methylene blue, 30 microM; LY-83583, 10 microM), but it was potentiated by zaprinast, an inhibitor of the cyclic GMP (cGMP)-specific phosphodiesterase. SS also blocked the increase in myosin light chain phosphorylation induced by AII. SS increased cGMP synthesis by cultured human mesangial cells, an effect that seemed to be dependent on the stimulation of a particulate guanylate cyclase. Preincubation of the cells with pertussis toxin (0.5 microgram/ml) inhibited the effect of SS on the AII-dependent changes in planar cell surface area, as well as the SS-dependent cGMP stimulation. In summary, these results demonstrate the ability of SS to relax cultured human mesangial cells, thus supporting a role for this peptide in the regulation of the glomerular filtration rate. The SS-dependent mesangial cell relaxation may be due to changes in the intracellular concentrations of cGMP, as a consequence of the activation of a particulate guanylate cyclase.


Subject(s)
Glomerular Mesangium/drug effects , Somatostatin/pharmacology , Angiotensin II/antagonists & inhibitors , Angiotensin II/pharmacology , Cells, Cultured , Glomerular Filtration Rate/drug effects , Glomerular Mesangium/cytology , Humans , Octreotide/analogs & derivatives , Octreotide/metabolism , Precipitin Tests
10.
Free Radic Biol Med ; 22(1-2): 49-56, 1997.
Article in English | MEDLINE | ID: mdl-8958129

ABSTRACT

The mechanisms responsible for age-related glomerular sclerosis (GS) have not been clearly identified. The present experiments were aimed at assessing the importance of the oxidant/antioxidant balance in the early stages of this process. For this purpose, the renal function (biochemical and clearance studies), some characteristics of isolated glomeruli, and reactive oxygen production (superoxide anion, hydrogen peroxide) as well as the antioxidant ability (superoxide dismutase, catalase, glutathione peroxidase) of glomeruli and cultured mesangial cells were studied in 3- and 18-month-old Fischer 344 rats (YOUNG and OLD rats, respectively). OLD animals show a normal renal function, increased urine protein excretion, and augmented protein glomerular content, an indirect index of GS. Isolated glomeruli from these rats produced increased amounts of superoxide anion and hydrogen peroxide, and catalase activity was increased. The glomerular thiobarbituric acid-reactive substances (TBARS) content was higher in OLD than in YOUNG animals. Similar results were obtained in cultured mesangial cells. In summary, the present results demonstrate, at an early stage of rat GS development, an association between the functional and structural changes of this process and an increased TBARS content (likely indicative of lipid oxidative damage) at the glomerular structures as well as in cultured mesangial cells. More extensive studies are needed to confirm the nature of this association.


Subject(s)
Antioxidants/metabolism , Glomerular Mesangium/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Kidney Glomerulus/metabolism , Oxidants/metabolism , Reactive Oxygen Species/metabolism , Aging/metabolism , Aging/pathology , Animals , Cells, Cultured , Glomerular Mesangium/cytology , Glomerulosclerosis, Focal Segmental/pathology , In Vitro Techniques , Male , Rats , Rats, Inbred F344
11.
Free Radic Biol Med ; 32(5): 406-13, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11864780

ABSTRACT

Vascular injury leads to the production of reactive oxygen species (ROS), but the mechanisms by which ROS contribute to vascular pathology are not completely understood. We hypothesized that ROS increase endothelin converting enzyme (ECE-1) expression. We found that glucose oxidase (GO) increases ECE-1 mRNA, protein, and activity in bovine aortic endothelial cells. Catalase abolishes this effect. Glucose oxidase treatment of endothelial cells transactivates the ECE-1 promoter. The ECE-1 promoter element that mediates this response to GO is located between -444 and -216 bp. This region contains a STAT response element, and GO activates STAT-3 binding to this STAT response element. Our data suggest that STAT3 mediates hydrogen peroxide induction of ECE-1 expression.


Subject(s)
Antioxidants/pharmacology , Aspartic Acid Endopeptidases/metabolism , Endothelium, Vascular/enzymology , Gene Expression Regulation, Enzymologic/physiology , Glucose Oxidase/pharmacology , Hydrogen Peroxide/pharmacology , Promoter Regions, Genetic/genetics , Reactive Oxygen Species/metabolism , Animals , Aorta/metabolism , Aspartic Acid Endopeptidases/genetics , Blotting, Western , Catalase/metabolism , Cattle , Cell Nucleus , Cells, Cultured , Cytosol , DNA Primers/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Endothelin-1/metabolism , Endothelin-Converting Enzymes , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , HeLa Cells , Humans , Luciferases/metabolism , Metalloendopeptidases , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , STAT3 Transcription Factor , Sequence Deletion , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection
12.
Free Radic Biol Med ; 26(5-6): 501-10, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10218638

ABSTRACT

The importance of endothelial contraction in the genesis of inflammatory edema has been reported. ROS are metabolites synthesized in pathological conditions in that a significant intravascular fluid leak occurs, such as ischemia-reperfusion. Present experiments were designed to test the hypothesis that ROS, particularly H2O2, may elicit the contraction of endothelial cells, and to explore the mechanisms involved. Bovine aortic endothelial cells incubated with H2O2 showed a significant reduction in planar cell surface area (PCSA), and a significant increase in myosin light chain phosphorylation (MLCP), with a time- and dose-dependent pattern, without any significant toxicity. This effect of H2O2 was not blocked by sulotroban (TxA2 antagonist) or BN 52021 (PAF antagonist). Lanthanum chloride (calcium channel blocker) and EGTA partially inhibited the increase in MLCP induced by H2O2. H7 and staurosporine, PKC inhibitors, and PKC down-regulation (phorbol myristate acetate treatment, 24 h) also blocked H2O2-dependent endothelial contraction, measured as PCSA or MLCP. H2O2 increased the intracellular calcium concentration, an effect blunted by EGTA and lanthanum chloride. H2O2 also increased the phosphorylation of an 80 kD polypeptide, probably MARCKS, a PKC substrate. In summary, the present results demonstrate the ROS-dependent contraction of endothelial cells, an effect that could explain the intravascular fluid leak observed in some pathophysiological situations. Calcium and PKC may be involved in the development of this contraction.


Subject(s)
Diterpenes , Endothelium, Vascular/drug effects , Hydrogen Peroxide/pharmacology , Animals , Aorta , Cattle , Cell Size/drug effects , Cells, Cultured , Cyclic GMP/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Free Radical Scavengers/pharmacology , Ginkgolides , Kinetics , Lactones/pharmacology , Myosin Light Chains/metabolism , Phosphorylation , Reactive Oxygen Species , Sulfonamides/pharmacology
13.
Antioxid Redox Signal ; 4(6): 869-75, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12573135

ABSTRACT

Vascular smooth muscle cells (VSMC) exhibit a hypertrophic and contractile response after angiotensin II (Ang II) treatment, and the NADH/NADPH oxidase-dependent synthesis of hydrogen peroxide (H(2)O(2)) seems to play a central role in these responses. Present experiments were designed to analyze the mechanisms responsible for the rapid changes induced by Ang II in the intracellular H(2)O(2) concentration in VSMC. Ang II induced a quick and transient increase of dichlorodihydrofluorescein (DCHF) fluorescence in VSMC, an effect that was completely abolished by catalase and by diethyldithiocarbamate, a cell-permeable superoxide dismutase inhibitor. Losartan and pertussis toxin prevented the stimulatory effect of Ang II. Both diphenylene iodonium (NADH/NADPH oxidase blocker) and 3-(4-octadecylbenzoyl)acrylic acid (phospholipase A2 blocker) inhibited the changes in DCHF fluorescence induced by Ang II, in a dose-dependent fashion, and the effects of both inhibitors were additive. These data demonstrate that Ang II induces a very quick and transient increase of H(2)O(2) in VSMC. This effect depends on the receptor type 1, is linked to a G protein, and involves both NADH/NADPH oxidase and phospholipase A2 activation. The mechanism may be related to the previously proposed role of H(2)O(2) in the genesis of the Ang II-induced cell contraction.


Subject(s)
Angiotensin II/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Reactive Oxygen Species/metabolism , Acrylates/pharmacology , Angiotensin II/metabolism , Animals , Benzoates , Catalase/metabolism , Catalase/pharmacology , Cells, Cultured , Ditiocarb/pharmacology , Enzyme Inhibitors/pharmacology , Fluoresceins/chemistry , Fluoresceins/metabolism , Fluorescence , Hydrogen Peroxide/metabolism , Indomethacin/pharmacology , Losartan/pharmacology , Muscle, Smooth, Vascular/cytology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/metabolism , Onium Compounds/pharmacology , Pertussis Toxin/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A/metabolism , Phospholipases A2 , Rats , Rats, Wistar
14.
Transplantation ; 66(10): 1325-9, 1998 Nov 27.
Article in English | MEDLINE | ID: mdl-9846517

ABSTRACT

BACKGROUND: We report an investigation of the effects of cyclosporine (CsA) on kidney function, the glomerular synthesis of reactive oxygen species, the peroxidation of lipids, and the levels of thromboxane B2 (TXB2). The effect of the simultaneous administration of the antioxidant vitamin E (Vit E) and CsA in rats was also evaluated. METHODS: Adult male Wistar rats were treated for 30 days with CsA (30 mg/kg/day), with Vit E (0.05 mg/ml), with CsA plus Vit E, or with the vehicle used for administration of CsA, namely 12.6% ethanol. RESULTS: CsA induced kidney failure and increased the glomerular synthesis of superoxide anion, H2O2, malonyldialdehyde, and TXB2. Vit E minimized the adverse effects of CsA on kidney function and the glomerular synthesis of these compounds. CONCLUSIONS: Our results suggest that the acute decrease in glomerular filtration rate induced by CsA might be mediated by the synthesis of reactive oxygen species and subsequent peroxidation of lipids, which increases the levels of TXB2. Treatment with Vit E prevented these effects, suggesting a possible role for antioxidants in the prevention of CsA nephrotoxicity.


Subject(s)
Cyclosporine/pharmacology , Kidney Glomerulus/metabolism , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Cyclosporine/adverse effects , Cyclosporine/blood , Kidney Diseases/chemically induced , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Rats , Rats, Wistar , Vitamin E/pharmacology
15.
Br J Pharmacol ; 124(3): 447-54, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9647467

ABSTRACT

1. Cyclosporine A (CsA) increases eNOS mRNA expression in bovine cultured aortic endothelial cells (BAEC). As some effects of CsA may be mediated by reactive oxygen species (ROS), present experiments were devoted to test the hypothesis that the CsA-induced eNOS up-regulation could be dependent on an increased synthesis of ROS. 2. CsA induced a dose-dependent increase of ROS synthesis, with the two fluorescent probes used, DHR123 (CsA 1 microM: 305+/-7% over control) and H2DCFDA (CsA 1 microM: 178+/-6% over control). 3. Two ROS generating systems, xanthine plus xanthine oxidase (XXO) and glucose oxidase (GO), increased the expression of eNOS mRNA in BAEC, an effect which was maximal after 8 h of incubation (XXO: 168+/-21% of control values. GO: 208+/-18% of control values). The ROS-dependent increased eNOS mRNA expression was followed by an increase in eNOS activity. 4. The effect of CsA on eNOS mRNA expression was abrogated by catalase, and superoxide dismutase (SOD). In contrast, the antioxidant PDTC augmented eNOS mRNA expression, both in basal conditions and in the presence of CsA. 5. The potential participation of the transcription factor AP-1 was explored. Electrophoretic mobility shift assays were consistent with an increase in AP-1 DNA-binding activity in BAEC treated with CsA or glucose oxidase. 6. The present results support a role for ROS, particularly superoxide anion and hydrogen peroxide, as mediators of the CsA-induced eNOS mRNA up-regulation. Furthermore, they situate ROS as potential regulators of gene expression in endothelial cells, both in physiological and pathophysiological situations.


Subject(s)
Cyclosporine/pharmacology , Endothelium, Vascular/enzymology , Immunosuppressive Agents/pharmacology , Nitric Oxide Synthase/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Cattle , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/drug effects , Nitric Oxide Synthase Type III , Transcription Factor AP-1/metabolism , Up-Regulation/drug effects
16.
Kidney Int Suppl ; 68: S20-4, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9839278

ABSTRACT

Cyclosporine A (CsA) and FK506 increase endothelial nitric oxide synthase (eNOS) mRNA expression in cultured bovine aortic endothelial cells (BAEC). CsA appears to increase eNOS mRNA levels mainly by increasing the rate of transcription, although a small contribution of mRNA stabilization could not be ruled out. CsA and FK506 induced an increase of ROS synthesis with the fluorescent probe used, DHR123. The ROS generating system glucose oxidase (GO) increased the expression of eNOS mRNA in BAEC. This upregulation of eNOS mRNA by CsA or GO was abrogated by catalase. As AP-1 is a redox-sensitive transcription factor and the bovine eNOS promoter has an AP-1 consensus sequence, a role of this factor in the up-regulation of eNOS mRNA was studied. Electrophoretic mobility shift assays were consistent with an increase in AP-1 DNA-binding activity in BAEC treated with CsA or glucose oxidase. The potential participation of ROS and the transcription factor AP-1 in the regulation of eNOS gene expression is suggested.


Subject(s)
Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Nitric Oxide Synthase/genetics , Reactive Oxygen Species/metabolism , Tacrolimus/pharmacology , Transcription Factor AP-1/metabolism , Animals , Aorta/cytology , Blotting, Northern , Calcineurin Inhibitors , Catalase/pharmacology , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Glucose Oxidase/pharmacology , Nitric Oxide Synthase Type III , Oxidation-Reduction , RNA, Messenger/analysis
17.
Peptides ; 20(8): 929-35, 1999.
Article in English | MEDLINE | ID: mdl-10503770

ABSTRACT

In experimental models and in humans, somatostatin (SRIF) is able to contract certain vascular structures. The present experiments were designed to assess the capacity of SRIF to contract cultured rat aortic vascular smooth muscle cells (VSMC), and to analyze the possible mechanisms involved. Cells incubated with SRIF showed a significant reduction in planar cell surface area, in a time- and dose-dependent manner. This effect was partially blocked by preincubating the cells with a combination of calcium antagonists (10 microM verapamil, plus 10 microM 3,4,5-Trimethoxybenzoic acid 3-(diethylanino) octyl ester TMB)-8). SRIF was also able to stimulate myosin light-chain phosphorylation in VSMC. A small but significant increase of intracellular calcium concentration, and decreased levels of cAMP, without changes in cGMP, were detected in VSMC incubated with SRIF. A search for the known SRIF receptors present in these cells, by reverse transcription-polymerase chain reaction, only SRIF receptor-4 was found to be present. These results demonstrate the ability of SRIF to contract cultured rat VSMC. The contraction observed in these cells appears to be due to a mixed mechanism, that involves [Ca2+]i and cAMP as second messengers, and is likely mediated via SRIF receptor-4.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Somatostatin/pharmacology , Animals , Base Sequence , Cells, Cultured , DNA Primers , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/physiology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
18.
Peptides ; 16(8): 1461-7, 1995.
Article in English | MEDLINE | ID: mdl-8745059

ABSTRACT

Gastrectomy increased pancreatic growth and this effect was associated with an increase in the number of somatostatin-14 (SS) receptors (146% of control) without altering their affinity. SS increased guanylate cyclase activity twofold in pancreatic acinar membranes from gastrectomized rats. The gastrectomy decreased pancreatic SS-like immunoreactivity (SS-LI) content (55% of control levels) and tyrosine phosphatase activity (74% of control levels). Administration of proglumide (20 mg/kg, IP), a gastrin/cholecystokinin (CCK) receptor antagonist, suppressed the inhibitory effect of gastrectomy on basal tyrosine phosphatase activity and SS-LI content, which returned to control levels. Furthermore, proglumide suppressed the increase of the number of SS receptors and of SS-stimulated guanylate cyclase activity induced by gastrectomy. All this suggests that pancreatic acinar cell growth is associated with upregulation of SS receptors, which could represent a mechanism promoted by the cell to negatively regulate the mitogenic activity of pancreatic growth factors such as CCK. In addition, the results also suggest that the negative regulation of tyrosine phosphatase activity may be important in the events involved in the pancreatic hyperplasia observed after gastrectomy.


Subject(s)
Guanylate Cyclase/metabolism , Pancreas/metabolism , Pancreas/pathology , Protein Tyrosine Phosphatases/metabolism , Somatostatin/metabolism , Animals , Gastrectomy , Hyperplasia , Male , Proglumide/pharmacology , Rats , Rats, Wistar , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Somatostatin/metabolism
19.
Prostaglandins Leukot Essent Fatty Acids ; 49(2): 597-602, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8415809

ABSTRACT

Platelets seem to be involved in the pathogenesis of some kidney diseases, but the exact relationships between platelets and the changes in renal function are incompletely known. Mesangial cells (MC) were incubated with platelet-supernatants (PS) and cellular surface area (CSA) and myosin light-chain phosphorylation (MLCP) were measured. CSA of PS-incubated MC (PS-MC) significantly diminished, as compared to control MC (70 +/- 6% vs. 100 +/- 5%). PS induced a significant increase in MLCP with respect to control cells (150 +/- 23% vs. 100 +/- 18%). When platelets were pretreated with indomethacin, the PS-dependent contraction was abolished. Pretreatment with sulotroban (SU) or BN-52021 (BN), a thromboxane A2 (TXA2) and a platelet-activating factor (PAF) receptor blocker respectively, also completely blocked the PS effects. In other experiments, platelets were activated with thrombin (T), adding the so obtained PS to MC. Moreover, cells were also preincubated with T and then added PS. No changes in CSA were observed in either case. It may be concluded that PS contracted cultured MC, and these changes could be related to the decreased glomerular filtration rate (GFR) observed in some diseases in which platelets seem to be involved. TXA2 and PAF may be responsible for this effect. In contrast, T incubation inhibited the effect of PS, perhaps through a direct relaxing effect of T in MC.


Subject(s)
Blood Platelets/physiology , Glomerular Mesangium/cytology , Glomerular Mesangium/physiology , Animals , Cells, Cultured , Glomerular Mesangium/drug effects , Glomerular Mesangium/ultrastructure , Myosins/metabolism , Phosphorylation , Platelet Activating Factor/pharmacology , Rats , Rats, Wistar , Thrombin/pharmacology
20.
Eur J Pharmacol ; 336(2-3): 251-6, 1997 Oct 08.
Article in English | MEDLINE | ID: mdl-9384240

ABSTRACT

The present experiments were devoted to analyzing the mechanisms involved in the captopril-dependent inhibition of human mesangial cell proliferation. Studies were performed in cultured human mesangial cells incubated with captopril, an angiotensin II-converting enzyme inhibitor with antioxidant properties, lisinopril, a non-antioxidant angiotensin II-converting enzyme inhibitor, and tocopherol, a pure antioxidant. Both angiotensin II-converting enzyme inhibitors significantly inhibited fetal calf serum-induced [3H]thymidine uptake by human mesangial cells, in a dose- and time-dependent manner, an effect which was not observed with tocopherol. The antiproliferative effect of captopril and its ability to block tyrosine phosphorylation of human mesangial cells proteins were significantly greater than those of lisinopril. Moreover, captopril significantly prevented the fetal calf serum-induced tyrosine phosphorylation of pp60(c-src). The present results suggest that the antiproliferative ability of captopril does not completely depend on its angiotensin II-converting enzyme inhibitor properties, pointing to a possible interaction of the drug with the intracellular mechanisms responsible for the transmission of the proliferative signals.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antioxidants/pharmacology , Captopril/pharmacology , Glomerular Mesangium/drug effects , Proto-Oncogene Proteins pp60(c-src)/metabolism , Cell Division/drug effects , Cells, Cultured , Glomerular Mesangium/metabolism , Humans , Lisinopril/pharmacology , Phosphorylation , Vitamin E/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL