Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
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.
Free Radic Biol Med ; 61: 416-27, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23624332

ABSTRACT

Transforming growth factor type-ß1 (TGF-ß1) has been recognized as a central mediator in many pathological events related to extracellular matrix (ECM) proteins accumulation, where their locally increased expression has been implicated in the fibrosis process of numerous organs, including glomerular fibrosis in the kidney. We and others have reported the TGF-ß1 synthesis regulation by reactive oxygen species (ROS), and moreover we also described the implication of integrin-linked kinase (ILK) in the AP-1-dependent TGF-ß1 up-regulation. Thus, we propose here that hydrogen peroxide (H2O2)-dependent TGF-ß1 regulation may be mediated by ILK activation. First we confirmed the increase in TGF-ß1 expression in human mesangial cells (HMC) after treatment with H2O2 or with an alternative H2O2-generating system such as the glucose-oxidase enzyme (GOX). By using immunoblotting, immunofluorescence, and ELISA techniques, we demonstrate that extracellular H2O2 up-regulates TGF-ß1 transcription, as well as increases TGF-ß1 promoter activity. Furthermore, catalase-decreased intracellular H2O2 abolished TGF-ß1 up-regulation. The use of pharmacological inhibitors as well as knockdown of ILK with small interfering RNA (siRNA) demonstrated the implication of a PI3K/ILK/AKT/ERK MAPK signaling pathway axis in the H2O2-induced TGF-ß1 overexpression. Finally, we explored the physiological relevance of these findings by treating HMC with angiotensin II, a known stimuli of H2O2 synthesis. Our results confirm the relevance of previous findings after a more physiological stimulus. In summary, our results provide evidence that ILK activity changes may act as a mechanism in response to different stimuli such as H2O2 in the induced TGF-ß1 up-regulation in pathological or even physiological conditions.


Subject(s)
Hydrogen Peroxide/metabolism , Protein Serine-Threonine Kinases/physiology , Transforming Growth Factor beta1/biosynthesis , Angiotensin II/pharmacology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/physiology , Glucose Oxidase/physiology , Humans , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Up-Regulation
3.
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
4.
Free Radic Biol Med ; 47(10): 1362-70, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19596064

ABSTRACT

Hydrogen peroxide (H(2)O(2)) is implicated in the regulation of signaling pathways leading to changes in vascular smooth muscle function. Contractile effects produced by H(2)O(2) are due to the phosphorylation of myosin light chain kinase triggered by increases in intracellular calcium (Ca(2+)) from intracellular stores or influx of extracellular Ca(2+). One mechanism for mobilizing such stores involves the phosphoinositide pathway. Inositol 1,4,5-trisphosphate (IP(3)) mobilizes intracellular Ca(2+) by binding to a family of receptors (IP(3)Rs) on the endoplasmic-sarcoplasmic reticulum that act as ligand-gated Ca(2+) channels. IP(3)Rs can be rapidly ubiquitinated and degraded by the proteasome, causing a decrease in cellular IP(3)R content. In this study we show that IP(3)R(1) and IP(3)R(3) are down-regulated when vascular smooth muscle cells (VSMC) are stimulated by H(2)O(2), through an increase in proteasome activity. Moreover, we demonstrate that the decrease in IP(3)R by H(2)O(2) is accompanied by a reduction in calcium efflux induced by IP(3) in VSMC. Also, we observed that angiotensin II (ANGII) induces a decrease in IP(3)R by activation of NADPH oxidase and that preincubation with H(2)O(2) decreases ANGII-mediated calcium efflux and planar cell surface area in VSMC. The decreased IP(3) receptor content observed in cells was also found in aortic rings, which exhibited a decreased ANGII-dependent contraction after treatment with H(2)O(2). Altogether, these results suggest that H(2)O(2) mediates IP(3)R down-regulation via proteasome activity.


Subject(s)
Down-Regulation/drug effects , Hydrogen Peroxide/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/biosynthesis , Proteasome Endopeptidase Complex/metabolism , Angiotensin II/pharmacology , Animals , Cells, Cultured , Enzyme Activation/drug effects , Inositol 1,4,5-Trisphosphate Receptors/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
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
6.
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
7.
J Biol Chem ; 281(40): 30057-62, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-16887796

ABSTRACT

Chronic activation of the acute phase response (APR) is associated with atherosclerosis. Elevated levels of interleukin-6, the major inducer of the APR, are associated with an increased risk of cardiovascular events. One of the clinical hallmarks of atherogenesis is endothelial dysfunction, characterized by a decrease in endothelial production of nitric oxide (NO). We hypothesized that interleukin-6 (IL-6) decreases endothelial NO synthase (eNOS) expression. We now show that IL-6 treatment of human aortic endothelial cells (HAEC) decreases steady-state levels of human eNOS mRNA and protein. This decrease in eNOS expression is caused in part by IL-6 inhibition of transactivation of the human eNOS promoter. To explore the mechanism by which IL-6 affects eNOS expression, we examined activation of signal transducer and transactivator-3 (Stat3). The IL-6 receptor (IL-6R) is expressed in HAEC, and Stat3 is phosphorylated in response to IL-6 stimulation of the IL-6R. We identified four consensus sequences for Stat3 binding (SIE) in the eNOS promoter at positions -1520, -1024, -840, and -540. Transfection of eNOS promoter mutants revealed that the SIE at -1024 mediates Stat3 inhibition of eNOS promoter activity. Gel-shift analysis of nuclear extracts from HAEC treated with IL-6 confirms that Stat3 binds to a complex containing the SIE at -1024. RNA silencing of STAT3 blocks the inhibitory effect of IL-6 on eNOS expression. Our data show that IL-6 has direct effects upon endothelial cells, inhibiting eNOS expression in part through Stat3. Decreased levels of eNOS may be an important component of the pro-atherogenic effect of the APR.


Subject(s)
Interleukin-6/physiology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , STAT3 Transcription Factor/physiology , Down-Regulation/physiology , Humans , Nitric Oxide Synthase Type III/biosynthesis , Phosphorylation , STAT3 Transcription Factor/metabolism
8.
Am J Physiol Cell Physiol ; 286(6): C1335-43, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14761892

ABSTRACT

Progressive renal diseases are characterized by an increased synthesis of extracellular matrix (ECM) components. The mechanisms involved in the development of these alterations are not completely known, but a crucial role for TGF-beta 1 has been suggested. Moreover, the ability of the ECM to modulate the phenotypic expression of different cell types has been widely described. In experiments presented here, human mesangial cells (HMC) were grown on collagen type I (COL I) or IV (COL IV). ECM protein and TGF-beta 1 mRNA expression were evaluated by Northern blot analysis, and TGF-beta 1 secretion was evaluated by ELISA. The involvement of tyrosine kinase and serine-threonine kinase pathways was studied by Western blot analysis, immunofluorescence, and in vitro kinase assays. HMC cultured on COL I showed an increased mRNA expression of COL I and COL IV, fibronectin, and TGF-beta 1. Both tyrosine phosphorylation and integrin-linked kinase (ILK) activity increased when HMC were cultured on COL I, and blockade of these pathways inhibited the increased secretion of TGF-beta 1. In conclusion, the present results support a role for extracellular COL I in the regulation of TGF-beta 1 synthesis during progressive renal sclerosis and fibrosis and the subsequent increase in newly synthesized ECM proteins. In addition, ILK, along with the tyrosine kinases, participates in the genesis of this effect.


Subject(s)
Collagen Type I/metabolism , Extracellular Matrix/genetics , Fibroblasts/metabolism , Glomerular Mesangium/metabolism , Transforming Growth Factor beta/metabolism , Cells, Cultured , Collagen Type I/pharmacology , Collagen Type IV/metabolism , Collagen Type IV/pharmacology , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Fibroblasts/drug effects , Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Humans , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Tyrosine/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
9.
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
10.
Am J Physiol Cell Physiol ; 285(4): C891-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12814915

ABSTRACT

cGMP is generated in endothelial cells after stimulation of soluble guanylyl cyclase (sGC) by nitric oxide (NO) or of particulate guanylyl cyclase (pGC) by natriuretic peptides (NP). We examined whether localized increases in cytosolic cGMP have distinct regulatory roles on the contraction induced by H2O2 treatment in human umbilical vein endothelial cells. cGMP concentrations and temporal dynamics were different upon NO stimulation of sGC or C-type NP (CNP) activation of pGC and did not correlate with their relaxing effects measured as planar cell surface area after H2O2 challenge. cGMP production due to sGC stimulation was always smaller and more brief than that induced by pGC stimulation with CNP, which was greater and remained elevated longer. The NO effects on cell relaxation were cGMP dependent because they were blocked by sGC inhibition with 1H-(1,2,4)Oxadiazolo(4,3-a)quinoxaline-1-one and mimicked by 8-Br-cGMP. An antagonist of the cGMP-dependent protein kinase type-I (PKG-I) also inhibited the NO-induced effects. The cell contraction induced by H2O2 produces myosin light chain (MLC) phosphorylation and NO prevented it completely, whereas CNP only produced a partial inhibition. Transfection with a dominant negative form of PKG type-I alpha completely reversed the NO-induced effects on MLC phosphorylation, whereas it only partially inhibited the effects due to CNP. Taken together, these results demonstrate that the NO/sGC/cGMP pathway induces endothelial cell relaxation in a more efficient manner than does CNP/pGC/cGMP pathway, an effect that might be related to a selective stimulation of PKG-1 alpha by NO-derived cGMP. Consequently, stimulated PKG-I alpha may phosphorylate important protein targets that are necessary to inhibit the endothelial contractile machinery activated by oxidative stress.


Subject(s)
Endothelium, Vascular/physiology , Guanylate Cyclase/metabolism , Vasodilation/physiology , Cells, Cultured , Cyclic GMP/pharmacology , Cyclic GMP/physiology , Cyclic GMP-Dependent Protein Kinases/metabolism , Drug Combinations , Endothelium, Vascular/cytology , Enzyme Activation/physiology , Humans , Hydrogen Peroxide/pharmacology , Isoenzymes/metabolism , Natriuretic Peptide, C-Type/metabolism , Natriuretic Peptide, C-Type/pharmacology , Nitric Oxide/pharmacology , Nitric Oxide/physiology , Oxidants/pharmacology , Solubility , Vasoconstriction/drug effects , Vasodilation/drug effects
11.
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
12.
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
13.
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
14.
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
15.
J Pineal Res ; 31(1): 46-56, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11485004

ABSTRACT

Since melatonin (N-acetyl-5-methoxytryptamine) decreases locomotor activity and rearing and increases grooming behavior in a similar manner as somatostatin (SRIF), we examined if melatonin could induce these changes through somatostatinergic neurotransmission in the rat frontoparietal cortex. Male Wistar rats (200-250 g) received a single injection of melatonin (25 microg/kg per day) subcutaneously (s.c.) and were sacrificed 5 hr later. Melatonin treatment increased the number of 125I-Tyr11-SRIF receptors in frontoparietal cortical membranes without any changes in the dissociation constant (Kd). The capacity of SRIF to inhibit basal and forskolin (FK)-stimulated adenylyl cyclase (AC) activity was increased in melatonin-treated rats as compared to the control animals. Melatonin administration also induced a lower AC activity, both under basal conditions and after stimulation of the enzyme via stimulatory guanine nucleotide-binding proteins (Gs), or directly with FK. Functional inhibitory guanine nucleotide-binding protein (Gi) activity was increased in frontoparietal cortical membranes from melatonin-treated rats when compared to controls. Western blot analyzes showed that melatonin administration did not alter the amount of the Gialpha1, or Gialpha3 subunits, but reduced Gialpha2 levels in frontoparietal cortical membranes. No significant changes in SRIF-like immunoreactivity content and SRIF mRNA levels were detected in this brain area after melatonin treatment. Administration of the melatonin receptor antagonist luzindole (10 mg/kg, s.c.) 30 min before melatonin injection did not change the melatonin-induced effects on the SRIF receptor effector system. In conclusion, the present results show that acute melatonin administration increases the activity of the SRIF receptor effector system and decreases Gialpha2 levels in the rat frontoparietal cortex. In addition, the coupling of Gs to AC is disturbed by melatonin.


Subject(s)
Frontal Lobe/drug effects , Melatonin/pharmacology , Parietal Lobe/drug effects , Receptors, Somatostatin/drug effects , Adenylyl Cyclases/metabolism , Animals , Frontal Lobe/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Kinetics , Male , Parietal Lobe/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Somatostatin/metabolism , Somatostatin/genetics , Somatostatin/metabolism
18.
Eur J Pharm Sci ; 12(4): 471-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11231114

ABSTRACT

AIM: The nephrotoxicity and immunosuppressive ability of cyclosporine (CyA) incorporated into polycaprolactone nanoparticles (CyA-NP) was assessed in vitro and in vivo and compared to the effects caused by free drug (Sandimmun. METHODS: The in vivo study included four groups (12 Wistar rats each) receiving oral CyA (10 mg/kg/day for 3 days) as an emulsion of Sandimmun in whole milk or CyA-NP and equivalent doses of empty NP or cremophor in milk as controls. CyA concentrations in blood, urine, liver, spleen and kidney at 24 h post-dosing were measured by fluorescence polarization immunoassay (FPIA). The nephrotoxicity induced by each drug treatment was determined by measuring creatinine plasma levels, malonyl dialdehyde production, and H(2)O(2) and reduced glutathione contents in glomeruli. On the other hand, the immunosuppressive effect was estimated in vivo by incubating lymphocyte suspensions obtained from CyA-, CyA-NP- and control-treated rats, as well as in vitro on lymphocyte suspensions from non-treated healthy animals. RESULTS: Significantly higher blood, urine and tissue levels were achieved with CyA-NP compared to free CyA. However, no changes in creatinine plasma levels were detected due to either CyA or CyA-NP treatment. Only the production of H(2)O(2) in the glomeruli exhibited a significant increase as compared to control groups, but no differences could be ascribed to the different drug treatments. In vivo, the immunosuppressive activity was also comparable for both drug treatments. In contrast, CyA-NP showed a better drug uptake in vitro at concentrations above 25 microM. No immunosuppression was detected in control groups. CONCLUSION: NP improve the oral bioavailability of CyA and its uptake by lymphocytes in vitro above 25 microM. On the contrary, specific immunosuppression and adverse effects were not simultaneously increased. Further studies are needed to clarify the results.


Subject(s)
Creatinine/blood , Cyclosporine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Kidney/drug effects , Lymphocytes/metabolism , Oxidants/metabolism , Polyesters/pharmacokinetics , Animals , Capsules , Cyclosporine/pharmacology , Drug Carriers , Female , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Male , Rats , Rats, Wistar , Tissue Distribution
19.
Pharmazie ; 56(1): 78-82, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11210676

ABSTRACT

The present paper reports about the effect of gonadectomy on cyclosporine (CyA) pharmacokinetics in rats. The oral administration of CyA (10 mg/kg b.w.) to male rats caused two-fold higher drug blood levels than those reached by females at 24 h after the last dose (334.10 +/- 126.70 vs. 161.49 +/- 53.39 ng/ml, p < 0.05). These levels increased by about 25% in orchiectomized male rats (419.47 +/- 132.63 ng/ml) but they returned to control values after testosterone treatment (330.99 +/- 130.80 ng/ml). On the other hand, CyA blood levels (90.66 +/- 22.25 ng/ml) decreased after ovariectomy, even more in the case of gonadectomized female rats receiving estradiol replacement (67.83 +/- 24.15 ng/ml). With regards to drug distribution, the concentrations of CyA in the liver, the kidneys and the spleen at 24 h after the last dose were about 8, 5 and 6-fold higher than blood levels, respectively, regardless of animal gender. These partition coefficients were increased to 11, 7 and 9-fold by male castration suggesting a more extensive drug distribution. Contrariwise, drug tissue levels in ovariectomized rats decreased. The changes of drug blood and tissue levels among groups were not associated to the variations of metabolite concentrations in the liver or blood. Therefore, gonadectomy exerts a complex effect on CyA pharmacokinetics in rats and makes complementary studies necessary to clarify how differences in sexual hormone secretion alter CyA disposition.


Subject(s)
Cyclosporine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Algorithms , Animals , Cyclosporine/blood , Female , Fluorescence Polarization Immunoassay , Humans , Immunosuppressive Agents/blood , Male , Orchiectomy , Ovariectomy , Rats , Rats, Wistar , Tissue Distribution
20.
Kidney Int ; 59(1): 87-95, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135061

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) are excessively produced in pathologic states, including many renal diseases. Transforming growth factor-beta (TGF-beta) may mediate renal fibrotic injury, and ROS may act through the TGF-beta pathway to exert a profibrotic effect. METHODS: The expression of TGF-beta1 and extracellular matrix (ECM) components were assessed in cultured human mesangial cells (HMCs) incubated with glucose oxidase (GO), an enzyme that continuously generates hydrogen peroxide from glucose. A neutralizing anti-TGF-beta antibody was added to test the hypothesis that hydrogen peroxide acts through activation of the TGF-beta pathway to stimulate ECM expression. RESULTS: Northern blot analysis revealed significantly increased steady-state levels of TGF-beta1 and ECM proteins (collagen types I, III, and IV, and fibronectin) by approximately twofold. While no significant effect on mRNA stability after treatment with GO was observed, other studies employing promoter-reporter assays, competitive-quantitative reverse transcription-polymerase chain reaction, mink lung epithelial cell proliferation assay, and TGF-beta1 enzyme-linked immunosorbent assay all demonstrated significant stimulation by GO (>1.5-fold) of TGF-beta1 promoter activity, mRNA level, bioactivity, and protein production, respectively. Catalase pretreatment prevented the GO-induced stimulation of TGF-beta1 mRNA. When incubations were performed with a panselective neutralizing anti-TGF-beta antibody, the GO-stimulated expression of ECM molecules was prevented. CONCLUSIONS: GO-induced hydrogen peroxide production induces TGF-beta1 synthesis and thereby increases ECM gene expression in cultured HMCs. These cellular responses may underlie the development and progression of renal diseases characterized by oxidative stress.


Subject(s)
Extracellular Matrix Proteins/genetics , Glomerular Mesangium/metabolism , Hydrogen Peroxide/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor beta/physiology , Catalase/pharmacology , Cells, Cultured , Extracellular Matrix Proteins/metabolism , Glomerular Mesangium/cytology , Glucose Oxidase/antagonists & inhibitors , Glucose Oxidase/pharmacology , Humans , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...