Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Kidney Int ; 74(5): 585-95, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18509316

ABSTRACT

Epithelial to mesenchymal transdifferentiation is a novel mechanism that promotes renal fibrosis and here we investigated whether known causes of renal fibrosis (angiotensin II and transforming growth factor beta, TGFbeta) act through this pathway. We infused angiotensin II into rats for 1 day and found that it activated the Smad pathway which persisted for up to 2 weeks in chronically infused rats. Renal TGF-beta mRNA expression was increased at 3 days and its protein at 2 weeks suggesting Smad pathway activation occurred earlier than TGF-beta upregulation. In cultured human tubuloepithelial cells, angiotensin II caused a rapid activation of Smad signaling independent of TGF-beta however, Smad-dependent transcription after 1 day was TGF-beta mediated. Two weeks of angiotensin II infusion activated genes associated with epithelial mesenchymal transdifferentiation. Stimulation with angiotensin II for 3 days caused transdifferentiation of the cultured epithelial cells by TGF-beta-mediated processes; however, early changes were independent of endogenous TGF-beta. Smad7 overexpression, which blocks Smad2/3 activation, diminished angiotensin II-induced epithelial mesenchymal transdifferentiation. Our results show that angiotensin II activates the Smad signaling system by TGF-beta-independent processes, in vivo and in vitro, causing renal fibrosis.


Subject(s)
Angiotensin II/pharmacology , Kidney/cytology , Kidney/drug effects , Smad Proteins/metabolism , Angiotensin II/administration & dosage , Animals , Cell Differentiation/drug effects , Cell Transdifferentiation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibrosis , Humans , Kidney/metabolism , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , MAP Kinase Signaling System , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/physiology , Signal Transduction , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
2.
Am J Physiol Renal Physiol ; 294(1): F149-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17989112

ABSTRACT

Connective tissue growth factor (CTGF) is overexpressed in kidney diseases associated with extracellular matrix accumulation. Angiotensin II (ANG II) participates in renal fibrosis by the upregulation of growth factors, including CTGF, and extracellular matrix proteins, such as type IV collagen. During renal injury, ANG II and the macrophage-produced cytokine interleukin-1beta (IL-1beta) may be present simultaneously in the glomerular environment. However, there are no studies about the interaction between ANG II and IL-1beta in renal fibrosis. For this reason, in cultured mesangial cells (MC), we investigated whether IL-1beta could regulate ANG II-mediated collagen accumulation and the mechanisms underlying this process. In MC, CTGF is a downstream mediator of type IV collagen production induced by ANG II. IL-1beta did not increase the production of CTGF and type IV collagen but significantly inhibited ANG II-induced CTGF and type IV collagen overexpression. Moreover, IL-1beta also inhibited type IV collagen upregulation caused by exogenous recombinant CTGF. Matrix metalloproteinase-9 (MMP-9) is the main enzyme involved in type IV collagen degradation. In MC, coincubation of IL-1beta and ANG II caused a synergistic increase in MMP-9 gene expression and activity, associated with type IV collagen inhibition. The described IL-1beta effects were dependent on activation of ERK/MAPK but independent p38-MAPK, JNK, phosphatidylinositol 3-kinase/Akt, and Rho-associated kinase pathways. In summary, these data indicate that IL-1beta inhibited ANG II-mediated type IV collagen production, via CTGF downregulation, and increased type IV collagen degradation, through MMP-9 upregulation. Our in vitro data show that the proinflammatory cytokine IL-1beta abrogates ANG II-induced CTGF production, describing antagonistic activities of proinflammatory cytokines on ANG II actions.


Subject(s)
Angiotensin II/pharmacology , Collagen Type IV/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-1beta/pharmacology , Mesangial Cells/metabolism , Animals , Cells, Cultured , Collagen Type IV/genetics , Connective Tissue Growth Factor , Drug Synergism , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Matrix Metalloproteinase 9/metabolism , Mesangial Cells/pathology , Rats , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
3.
Clín. investig. arterioscler. (Ed. impr.) ; 19(5): 232-239, sept. 2007. ilus
Article in Es | IBECS | ID: ibc-056551

ABSTRACT

Introducción. El factor de crecimiento de tejido conectivo (CTGF) está implicado en diversas enfermedades, como la aterosclerosis, la fibrosis de la piel y diversas nefritis experimentales y humanas. Sin embargo, el papel de este factor profibrótico en el daño vascular asociado a hipertensión no se conoce completamente. Objetivo. Estudiar el posible papel del CTGF en el daño vascular asociado a hipertensión en ratas, así como la posible interacción con la aldosterona. Método. Se utilizaron ratas macho espontáneamente hipertensas (SHR) tratadas durante 10 semanas con 2 dosis de eplerenona, un antagonista selectivo de los receptores de mineralocorticoides (30 y 100 mg/kg/día), y ratas normotensas (WKY) como grupo control. Al final del tratamiento se midió la presión arterial sistólica (PAS) y la reactividad vascular en anillos de aorta. Se determinó la expresión vascular y los valores de proteína del CTGF, así como la morfometría de la aorta. Se estudió también el efecto directo de la aldosterona en células de músculo liso vascular (CMLV). Resultados. Las SHR presentaron unos valores de PAS mayores que las ratas controles WKY. Sólo el tratamiento con la dosis alta de eplerenona redujo significativamente estos valores. La expresión vascular génica y los valores de proteínas del CTGF aumentaron significativamente en las SHR respecto a las WKY. El tratamiento con ambas dosis de eplerenona disminuyó significativamente estos parámetros. La relajación dependiente del endotelio fue menor en SHR que en WKY, y el tratamiento con eplerenona normalizó esta respuesta. Las áreas del vaso, la luz y la media aumentaron significativamente en las SHR respecto a las WKY, así como la relación media/luz. El tratamiento con eplerenona redujo todas las áreas estudiadas y normalizó la relación media/luz. La incubación de CMLV con aldosterona aumentó la expresión de CTGF de forma dependiente de la dosis. Conclusiones. La aldosterona participa en las alteraciones tanto funcionales como estructurales asociadas a la hipertensión arterial. El CTGF es uno de los factores implicados en el proceso fibrótico vascular asociado a hipertensión arterial (AU)


Introduction. Connective tissue growth factor (CTGF) is associated with distinct diseases, including atherosclerosis, skin fibrosis, and several human and experimental nephritides. However, the role of this profibrotic factor in the vascular damage associated with hypertension is not well known. Objective. To study the role of CTGF in vascular alterations associated with hypertension in rats, as well as its possible interaction with aldosterone. Method. Male spontaneously hypertensive rats (SHR) were treated with 2 doses (30 and 100 mg/Kg/day) of the mineralocorticoid receptor antagonist eplerenone for 10 weeks. Normotensive rats (WKY) were used as a control group. At the end of the treatment, systolic blood pressure (SBP) and vascular reactivity in aortic rings were measured. In addition, vascular expression and protein levels of CTGF, as well as morphological lesions in the aorta, were evaluated. The direct effect of aldosterone on vascular smooth muscle cells was also studied. Results. SBP was higher in SHR than in WKY and only the high dose of eplerenone significantly reduced SBP. In the aorta of SHR, CTGF mRNA expression and protein levels were upregulated compared with WKY. Both doses of eplerenone similarly and significantly diminished CTGF upregulation. Endothelium-dependent relaxation was lower in SHR than in WKY and treatment with eplerenone normalized this response. Vessel area, lumen area and media area, as well as the media to lumen ratio, were significantly increased in SHR compared with WKY. Treatment with eplerenone reduced all the parameters studied and normalized the media to lumen ratio. Incubation of cultured vascular smooth muscle cells with aldosterone increased CTGF production in a dose-dependent manner. Conclusions. Aldosterone participates in both the functional and structural alterations associated with hypertension. CTGF is one of the factors implicated in the vascular fibrotic process associated with hypertension (AU)


Subject(s)
Animals , Rats , Hypertension/drug therapy , Hypertension/physiopathology , Aldosterone/adverse effects , Aldosterone/biosynthesis , Mineralocorticoid Receptor Antagonists , Arteriosclerosis/diagnosis , Immunohistochemistry/veterinary , Blood Pressure , Connective Tissue/anatomy & histology , Connective Tissue/growth & development , Polymerase Chain Reaction/methods , Connective Tissue Diseases/complications , Immunohistochemistry , Polymerase Chain Reaction/veterinary , Blotting, Western/methods , Blotting, Western/veterinary , Regulatory Sequences, Ribonucleic Acid/physiology
4.
Hypertension ; 50(2): 377-83, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17592071

ABSTRACT

3-Hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) reductase inhibitors (statins) present beneficial effects in cardiovascular diseases. Angiotensin II (Ang II) contributes to cardiovascular damage through the production of profibrotic factors, such as connective tissue growth factor (CTGF). Our aim was to investigate whether HMG-CoA reductase inhibitors could modulate Ang II responses, evaluating CTGF expression and the mechanisms underlying this process. In cultured vascular smooth muscle cells (VSMCs) atorvastatin and simvastatin inhibited Ang II-induced CTGF production. The inhibitory effect of statins on CTGF upregulation was reversed by mevalonate and geranylgeranylpyrophosphate, suggesting that RhoA inhibition could be involved in this process. In VSMCs, statins inhibited Ang II-induced Rho membrane localization and activation. In these cells Ang II regulated CTGF via RhoA/Rho kinase activation, as shown by inhibition of Rho with C3 exoenzyme, RhoA dominant-negative overexpression, and Rho kinase inhibition. Furthermore, activation of p38MAPK and JNK, and redox process were also involved in Ang II-mediated CTGF upregulation, and were downregulated by statins. In rats infused with Ang II (100 ng/kg per minute) for 2 weeks, treatment with atorvastatin (5 mg/kg per day) diminished aortic CTGF and Rho activation without blood pressure modification. Rho kinase inhibition decreased CTGF upregulation in rat aorta, mimicking statin effect. CTGF is a vascular fibrosis mediator. Statins diminished extracellular matrix (ECM) overexpression caused by Ang II in vivo and in vitro. In summary, HMG-CoA reductase inhibitors inhibit several intracellular signaling systems activated by Ang II (RhoA/Rho kinase and MAPK pathways and redox process) involved in the regulation of CTGF. Our results may explain, at least in part, some beneficial effects of statins in cardiovascular diseases.


Subject(s)
Angiotensin II/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/drug effects , rho GTP-Binding Proteins/metabolism , Animals , Cell Movement/drug effects , Cell Proliferation , Cells, Cultured , Connective Tissue Growth Factor , Disease Models, Animal , Immediate-Early Proteins/drug effects , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinases/drug effects , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Signal Transduction , rho GTP-Binding Proteins/administration & dosage
5.
J Hypertens ; 25(3): 629-38, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17278980

ABSTRACT

OBJECTIVE: The aim of the present study was to investigate possible inter-relationships between connective tissue growth factor (CTGF) and aldosterone in vascular and renal damage associated with hypertension. METHOD: Spontaneously hypertensive rats (SHR) were treated with two doses (100 and 30 mg/kg per day) of the mineralocorticoid receptor antagonist eplerenone, or with antihypertensive therapy (HHR) (20 mg/kg per day hydralazine + 7 mg/kg per day hydrochlorothiazide + 0.15 mg/kg per day reserpine). RESULTS: CTGF mRNA expression and protein levels in the aorta of SHR were upregulated (P < 0.05) compared with Wistar-Kyoto rats. Both doses of eplerenone similarly and significantly diminished CTGF upregulation, correlated with amelioration of aortic remodelling and endothelium-dependent relaxations. Only high-dose eplerenone and HHR significantly reduced arterial blood pressure. HHR treatment also diminished CTGF overexpression, suggesting a blood-pressure-mediated effect in CTGF regulation. This reduction, however, was lower (P < 0.05) than that produced by eplerenone (100 mg/kg per day). The direct effect of aldosterone on vascular smooth muscle cells was also studied. Incubation of cultured vascular smooth muscle cells with aldosterone increased CTGF production in a dose-related manner, but was reduced (P < 0.05) by the mineralocorticoid receptor antagonist spironolactone. Renal CTGF mRNA and protein levels were higher in SHR than in Wistar-Kyoto rats (P < 0.05), and were similarly diminished by all treatments (P < 0.05). CONCLUSIONS: These data show that aldosterone and haemodynamic stress from elevated blood pressure levels regulate vascular and renal CTGF in SHR. The results suggest that aldosterone, through CTGF stimulation, could participate in vascular and renal structural alterations associated with hypertension, describing a novel mechanism of aldosterone in hypertensive target organ damage.


Subject(s)
Aldosterone/physiology , Aorta/pathology , Blood Pressure/physiology , Hypertension/complications , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Muscle, Smooth, Vascular/pathology , Animals , Aorta/metabolism , Blood Pressure Determination , Cells, Cultured , Connective Tissue Growth Factor , Eplerenone , Immediate-Early Proteins/genetics , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , Up-Regulation
6.
Curr Opin Nephrol Hypertens ; 15(2): 159-66, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16481883

ABSTRACT

PURPOSE OF REVIEW: We will focus on the recent findings concerning the inflammatory response in vascular and renal tissues caused by hypertension. RECENT FINDINGS: Angiotensin II is one of the main factors involved in hypertension-induced tissue damage. This peptide regulates the inflammatory process. Angiotensin II activates circulating cells, and participates in their adhesion to the activated endothelium and subsequent transmigration through the synthesis of adhesion molecules, chemokines and cytokines. Among the intracellular signals involved in angiotensin II-induced inflammation, the production of reactive oxygen species and the activation of nuclear factor-kappaB are the best known. SUMMARY: The pharmacological blockade of angiotensin II actions, by angiotensin-converting enzyme inhibitors or angiotensin receptor antagonists, results in beneficial organ protective effects, in addition to the effects of these agents on blood pressure control, that can be explained by the blockade of the angiotensin II-induced pro-inflammatory response. These data provide a rationale for the use of blockers of the renin-angiotensin system to prevent vascular and renal inflammation in patients with hypertension.


Subject(s)
Angiotensin II/metabolism , Hypertension, Renal/complications , Hypertension/complications , Inflammation Mediators/metabolism , Inflammation/etiology , Chemokines/metabolism , Cytokines/metabolism , Female , Humans , Hypertension/metabolism , Hypertension, Renal/metabolism , Inflammation/metabolism , Male , Peptidyl-Dipeptidase A/metabolism , Renin-Angiotensin System/physiology , Risk Factors , Sensitivity and Specificity
7.
Nephrol Dial Transplant ; 21(1): 16-20, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16280370

ABSTRACT

Angiotensin II (AngII) participates in the pathogenesis of renal diseases, through the regulation of two key processes inflammation and fibrosis. AT1 and AT2 are the main receptors of AngII. AT1 mediates most of the actions of AngII. This receptor regulates the expression of profibrotic factors, such as connective tissue growth factor (CTGF). The Smad signalling pathway and the Rho/Rho kinase system are two novel mechanisms involved in AngII-induced matrix regulation recently described. The role of AT2 receptors in renal pathophysiological processes is not fully elucidated. Experimental data suggest that AT2 receptors through activation of nuclear factor-kappaB participate in renal inflammatory cell recruitment. Studies in animal models of kidney injury have shown that the combined blockade of both AT1 and AT2 receptors, as well as the inhibition of the NF-kappaB pathway are necessary to stop the inflammatory process fully. On the whole, these data highlight the complex signalling systems activated by AngII and suggest novel potential targets to block fibrosis and inflammation in renal diseases.


Subject(s)
Angiotensin II/metabolism , Inflammation Mediators/analysis , Kidney Diseases/pathology , NF-kappa B/metabolism , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II/analysis , Cytokines/analysis , Cytokines/metabolism , Female , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/physiopathology , Humans , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Kidney Function Tests , Male , NF-kappa B/analysis , Prognosis , Receptor, Angiotensin, Type 2/analysis , Risk Assessment , Sensitivity and Specificity , Severity of Illness Index , Signal Transduction
8.
J Renin Angiotensin Aldosterone Syst ; 7(4): 192-200, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17318787

ABSTRACT

We have evaluated the role of connective tissue growth factor (CTGF) in vascular and renal damage associated with hypertension and possible interactions with angiotensin II (Ang II). Spontaneously hypertensive rats (SHR) were treated with either the Ang II receptor antagonist candesartan (C;2 mg/Kg(-1)/day(-1)) or antihypertensive triple therapy (TT; in mg/Kg(-1)/day(-1);20 hydralazine +7 hydrochlorothiazide +0.15 reserpine) for 10 weeks. Wistar Kyoto rats were used as a normotensive control group. Hypertension was associated with an increase in aortic media area, media-to-lumen ratio and collagen density. Kidneys from SHR showed minimum renal alterations. Aorta and renal gene expression and immunostaining of CTGF were higher in SHR. Candesartan decreased arterial pressure, aortic media area, media-to-lumen ratio and collagen density. However, although arterial pressure decrease was comparable for both treatments, TT partially reduced these parameters. Candesartan-treated rats showed lower levels of vascular CTGF expression, aortic media area, media-to-lumen ratio and collagen density than TT-treated animals. Treatments improve renal damage and reduce renal gene expression and CTGF immunostaining in SHR in a similar manner. The results show that vascular and renal damage is associated with stimulation of CTGF gene and protein content. These results also might suggest that CTGF could be one downstream mediator of Ang II in hypertension-associated organ damage in SHR.


Subject(s)
Angiotensin II/physiology , Aorta/physiopathology , Hypertension/physiopathology , Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Kidney/physiopathology , Animals , Aorta/pathology , Blood Pressure/physiology , Body Weight/physiology , Connective Tissue Growth Factor , Fibrosis/pathology , Hypertension/metabolism , Hypertension/pathology , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/pathology , Kidney Cortex/metabolism , Male , Organ Size/physiology , Rats , Rats, Inbred SHR , Rats, Inbred WKY
9.
Kidney Int Suppl ; (99): S39-45, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16336575

ABSTRACT

BACKGROUND: Angiotensin II (AngII) is a key factor in the pathogenesis of renal damage. AngII via AngII type 1 receptors activates several intracellular signaling systems, including the small guanosine triphosphatase Rho and its downstream effector Rho-dependent serine-threonine kinase (Rho-kinase). The Rho/Rho-kinase pathway contributes to inflammatory and proliferative changes observed in cardiovascular diseases. However, the data on renal diseases are scarce. The aim of this study was to investigate the effect of Rho-kinase inhibition in AngII-induced renal damage. METHODS: We used the model of systemic AngII infusion into normal rats (100 ng/kg per minute; subcutaneous osmotic minipumps), and some animals were treated with the Rho-kinase inhibitor Y-27632 (30 mg/kg per day). In the kidneys of these animals, we evaluated renal lesions, transcription factor activity (by electrophoretic mobility shift assay), and messenger RNA (by polymerase chain reaction) and protein expression levels (by Western blot and/or immunohistochemistry) of proinflammatory and profibrotic factors. RESULTS: Rats infused with AngII for three days present renal inflammatory cell infiltration and slight tubular damage, which were diminished by treatment with the Rho-kinase inhibitor Y-27632. AngII activates nuclear factor-kappaB and causes overexpression of proinflammatory factors, including cytokines (tumor necrosis factor alpha) and chemokines (monocyte chemotactic protein-1), and of profibrotic factors (connective tissue growth factor). Treatment of AngII-infused rats with Y-27632 decreases the upregulation of these proinflammatory and profibrotic mediators. CONCLUSION: These data demonstrate that the Rho-kinase pathway is involved in renal damage caused by AngII through the regulation of proinflammatory and profibrotic mediators. These results suggest that inhibition of the Rho-kinase pathway represents a novel therapy for renal diseases associated with local AngII generation.


Subject(s)
Angiotensin II/metabolism , Kidney Diseases/etiology , Kidney Diseases/physiopathology , Kidney/physiopathology , Protein Serine-Threonine Kinases/metabolism , Amides/pharmacology , Angiotensin II/pharmacology , Animals , Blotting, Western , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Connective Tissue Growth Factor , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Intracellular Signaling Peptides and Proteins , Kidney/drug effects , Kidney/pathology , NF-kappa B/physiology , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Rho Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/drug effects , rho-Associated Kinases
10.
Circ Res ; 97(2): 125-34, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15976312

ABSTRACT

Endothelin (ET)-1 is a potent vasoconstrictor that participates in cardiovascular diseases. Connective tissue growth factor (CTGF) is a novel fibrotic mediator that is overexpressed in human atherosclerotic lesions, myocardial infarction, and experimental models of hypertension. In vascular smooth muscle cells (VSMCs), CTGF regulates cell proliferation/apoptosis, migration, and extracellular matrix (ECM) accumulation. Our aim was to investigate whether ET-1 could regulate CTGF and to investigate the potential role of ET-1 in vascular fibrosis. In growth-arrested rat VSMCs, ET-1 upregulated CTGF mRNA expression, promoter activity, and protein production. The blockade of CTGF by a CTGF antisense oligonucleotide decreased FN and type I collagen expression in ET-1-treated cells, showing that CTGF participates in ET-1-induced ECM accumulation. The ETA, but not ETB, antagonist diminished ET-1-induced CTGF expression gene and production. Several intracellular signals elicited by ET-1, via ETA receptors, are involved in CTGF synthesis, including activation of RhoA/Rho-kinase and mitogen-activated protein kinase and production of reactive oxygen species. CTGF is a mediator of TGF-beta- and angiotensin (Ang) II-induced fibrosis. In VSMCs, ET-1 did not upregulate TGF-beta gene or protein. The presence of neutralizing transforming growth factor (TGF)-beta antibody did not modify ET-1-induced CTGF production, showing a TGF-beta-independent regulation. We have also found an interrelationship between Ang II and ET-1 because the ETA antagonist diminished CTGF upregulation caused by Ang II. Collectively, our results show that, in cultured VSMCs, ET-1, independently of TGF-beta and through the activation of several intracellular signals via ETA receptors, regulates CTGF. This novel finding suggests that CTGF could be a mediator of the profibrotic effects of ET-1 in vascular diseases.


Subject(s)
Endothelin-1/pharmacology , Gene Expression Regulation/drug effects , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Receptor, Endothelin A/physiology , Transforming Growth Factor beta/physiology , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Angiotensin I/pharmacology , Animals , Cells, Cultured , Connective Tissue Growth Factor , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Immediate-Early Proteins/analysis , Intercellular Signaling Peptides and Proteins/analysis , Promoter Regions, Genetic , RNA, Messenger/analysis , Rats , Rats, Inbred WKY , rhoA GTP-Binding Protein/physiology
11.
Circulation ; 111(19): 2509-17, 2005 May 17.
Article in English | MEDLINE | ID: mdl-15883213

ABSTRACT

BACKGROUND: Angiotensin II (Ang II) participates in vascular fibrosis. Transforming growth factor-beta (TGF-beta) is considered the most important fibrotic factor, and Smad proteins are essential components of the TGF-beta signaling system. Our aim was to investigate whether Ang II activates the Smad pathway in vascular cells and its potential role in fibrosis, evaluating connective tissue growth factor (CTGF) and extracellular matrix (ECM) proteins. METHODS AND RESULTS: Systemic infusion of Ang II into Wistar rats increased aortic Smad2, phosphorylated-Smad2, and Smad4 expression, associated with CTGF upregulation. In growth-arrested vascular smooth muscle cells, Ang II treatment for 20 minutes caused Smad2 phosphorylation, nuclear translocation of phosphorylated-Smad2 and Smad4, and increased Smad DNA-binding activity. Ang II also caused Smad overexpression and Smad-dependent gene transcription. The AT1 antagonist losartan diminished Ang II-induced Smad activation. The blockade of endogenous TGF-beta did not modify the activation of Smad caused by Ang II. The p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 diminished Ang II-induced Smad2 phosphorylation. These data show that Ang II activates the Smad pathway via AT1 receptors and MAPK activation independently of TGF-beta. Transient transfection with Smad7, which interferes with receptor-mediated activation of Smad2, diminished Ang II-induced CTGF promoter activation, gene and protein expression, and fibronectin and type-1 procollagen overexpression, showing that Smad activation is involved in Ang II-induced fibrosis. CONCLUSIONS: Our results show that Ang II activates the Smad signaling system in vascular cells in vivo and in vitro. Smad proteins are involved in Ang II-induced CTGF and ECM overexpression independently of TGF-beta. This novel finding suggests that Smad activation could be involved in the profibrogenic effects of Ang II in vascular diseases.


Subject(s)
Angiotensin II/pharmacology , Muscle, Smooth, Vascular/cytology , Smad Proteins/metabolism , Transforming Growth Factor beta/pharmacology , Active Transport, Cell Nucleus , Angiotensin II/administration & dosage , Animals , Connective Tissue Growth Factor , Extracellular Matrix Proteins , Fibrosis/etiology , Immediate-Early Proteins , Intercellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Smad Proteins/genetics , Smad2 Protein/genetics , Smad4 Protein/genetics , Up-Regulation/drug effects
12.
J Am Soc Nephrol ; 16(6): 1673-83, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15829701

ABSTRACT

Suppressors of cytokine signaling (SOCS) family is constituted by cytokine-inducible proteins that modulate receptor signal transduction via tyrosine kinases, mainly the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway. Differential SOCS expression was noted in renal cells that were incubated with inflammatory stimuli, but the role of SOCS in the pathogenesis of renal diseases is not yet well defined. Because angiotensin II (Ang II) plays a key role in renal disease, SOCS proteins were studied as a novel mechanism involved in the negative regulation of Ang II-mediated processes. Systemic Ang II infusion for 3 d increased the renal mRNA expression of SOCS-3 and SOCS-1. SOCS protein synthesis was found in glomerular mesangial area and tubules. In cultured mesangial cells and tubular epithelial cells, Ang II induced a rapid and transient SOCS-3 and SOCS-1 expression in parallel with JAK2 and STAT1 activation. In both cell types, overexpression of SOCS proteins prevented the STAT activation in response to Ang II. SOCS expression observed in Ang II-infused rats and in Ang II-stimulated cells was significantly inhibited by treatment with AT(1) but not AT(2) receptor antagonist and was attenuated in mesangial cells from AT(1a)-deficient mice, demonstrating the implication of AT(1) in those responses. In SOCS-3 knockdown studies, antisense oligonucleotides inhibited the expression of SOCS-3 and increased the Ang II-induced STAT activation and c-Fos/c-Jun expression, then resulting in a more severe renal damage. These results suggest that SOCS proteins may act as negative regulators of Ang II signaling in renal cells and implicate SOCS as important modulators of renal damage.


Subject(s)
Angiotensin II/metabolism , DNA-Binding Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Kidney/drug effects , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cells, Cultured , Cytokines/antagonists & inhibitors , Epithelial Cells/drug effects , Humans , Janus Kinase 2 , Losartan/pharmacology , Mice , Rats , Repressor Proteins/physiology , STAT1 Transcription Factor , Signal Transduction/drug effects , Transcription Factors/physiology , Transcriptional Activation/drug effects
13.
Circ Res ; 96(9): 965-73, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15831814

ABSTRACT

Inflammation is a key event in the development of atherosclerosis. Nuclear factor-kappaB (NF-kappaB) is important in the inflammatory response regulation. The effector peptide of the renin angiotensin system Angiotensin II (Ang II) activates NF-kappaB and upregulates some related proinflammatory genes. Our aim was to investigate whether other angiotensin-related peptides, as the N-terminal degradation peptide Ang IV, could regulate proinflammatory factors (activation of NF-kappaB and related genes) in cultured vascular smooth muscle cells (VSMCs). In these cells, Ang IV increased NF-kappaB DNA binding activity, caused nuclear translocation of p50/p65 subunits, cytosolic IkappaB degradation and induced NF-kappaB-dependent gene transcription. Ang II activates NF-kappaB via AT1 and AT2 receptors, but AT1 or AT2 antagonists did not inhibit NF-kappaB activation caused by Ang IV. In VSMC from AT1a receptor knockout mice, Ang IV also activated NF-kappaB pathway. In those cells, the AT4 antagonist divalinal diminished dose-dependently Ang IV-induced NF-kappaB activation and prevented IkappaB degradation, but had no effect on the Ang II response, indicating that Ang IV activates the NF-kappaB pathway via AT4 receptors. Ang IV also increased the expression of proinflammatory factors under NF-kappaB control, such as MCP-1, IL-6, TNF-alpha, ICAM-1, and PAI-1, which were blocked by the AT4 antagonist. Our results reveal that Ang IV, via AT4 receptors, activates NF-kappaB pathway and increases proinflammatory genes. These data indicate that Ang IV possesses proinflammatory properties, suggesting that this Ang degradation peptide could participate in the pathogenesis of cardiovascular diseases.


Subject(s)
Angiotensin II/analogs & derivatives , Inflammation Mediators/metabolism , Muscle, Smooth, Vascular/metabolism , NF-kappa B/metabolism , Transcriptional Activation , Angiotensin II/pharmacology , Animals , Cells, Cultured , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Rats , Rats, Inbred WKY , Receptor, Angiotensin, Type 1/genetics , Receptors, Angiotensin/metabolism
14.
J Am Soc Nephrol ; 15(6): 1514-29, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15153562

ABSTRACT

Inflammatory cell infiltration plays a key role in the onset and progression of renal injury. The NF-kappaB participates in the inflammatory response, regulating many proinflammatory genes. Angiotensin II (Ang II), via AT(1) and AT(2) receptors, activates NF-kappaB. Although the contribution of Ang II to kidney damage progression is already established, the receptor subtype involved in the inflammatory cell recruitment is not clear. For investigating this issue, the unilateral ureteral obstruction (UUO) model was used in mice, blocking Ang II production/receptors and NF-kappaB pathway. Two days after UUO, obstructed kidneys of wild-type mice presented a marked interstitial inflammatory cell infiltration and increased NF-kappaB activity. Treatment with AT(1) or AT(2) antagonists partially decreased NF-kappaB activation, whereas only the AT(2) blockade diminished monocyte infiltration. Obstructed kidneys of AT(1)-knockout mice showed interstitial monocyte infiltration and NF-kappaB activation; both processes were abolished by an AT(2) antagonist, suggesting AT(2)/NF-kappaB involvement in monocyte recruitment. In wild-type mice, only angiotensin-converting enzyme inhibition or combined therapy with AT(1) plus AT(2) antagonists blocked monocyte infiltration, NF-kappaB activation, and upregulation of NF-kappaB-related proinflammatory genes. Therefore, AT(1) and AT(2) blockade is necessary to arrest completely the inflammatory process. Treatment with two different NF-kappaB inhibitors, pirrolidin-dithiocarbamate and parthenolide, diminished monocyte infiltration and gene overexpression. These data show that Ang II, via AT(1) and AT(2) receptors and NF-kappaB pathway, participates in the regulation of renal monocyte recruitment and may provide a rationale to investigate further the role of AT(2) in human kidney diseases.


Subject(s)
Angiotensin II/metabolism , Inflammation , NF-kappa B/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Ureteral Obstruction/metabolism , Animals , Disease Progression , Gene Expression Regulation , Genotype , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Kidney Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Up-Regulation
15.
Am J Pathol ; 163(5): 1937-47, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14578193

ABSTRACT

Connective tissue growth factor (CTGF) has been described as a novel fibrotic mediator. CTGF is overexpressed in several kidney diseases and is induced by different factors involved in renal injury. Angiotensin II (AngII) participates in the pathogenesis of kidney damage, contributing to fibrosis; however, whether AngII regulates CTGF in the kidney has not been explored. Systemic infusion of AngII into normal rats for 3 days increased renal CTGF mRNA and protein levels. At day 7, AngII-infused rats presented overexpression of CTGF in glomeruli, tubuli, and renal arteries, as well as tubular injury and elevated fibronectin deposition. Only treatment with an AT(1) receptor antagonist, but not an AT(2), diminished CTGF and fibronectin overexpression and ameliorated tubular damage. In rats with immune complex nephritis, renal overexpression of CTGF was diminished by the ACE inhibitor quinapril, correlated with a diminution in fibrosis. In cultured renal cells (mesangial and tubular epithelial cells) AngII, via AT(1), increased CTGF mRNA and protein production, and a CTGF antisense oligonucleotide decreased AngII-induced fibronectin synthesis. Our data show that AngII regulates CTGF in the kidney and cultured in mesangial and tubular cells. This novel finding suggests that CTGF could be a mediator of the profibrogenic effects of AngII in the kidney.


Subject(s)
Angiotensin II/pharmacology , Immediate-Early Proteins/drug effects , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/drug effects , Vasoconstrictor Agents/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Blotting, Western , Cells, Cultured , Connective Tissue Growth Factor , Female , Immune Complex Diseases/drug therapy , Immune Complex Diseases/pathology , Immunohistochemistry , In Situ Hybridization , Kidney/pathology , Nephritis/drug therapy , Nephritis/immunology , Quinapril , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tetrahydroisoquinolines/pharmacology , Up-Regulation
16.
Kidney Int Suppl ; (86): S21-6, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12969123

ABSTRACT

BACKGROUND: Activation of the renin angiotensin system has been described in pathologic conditions, including kidney damage. Angiotensin II (Ang II) acts through two receptors, AT1 and AT2. Most of the known actions of Ang II, including vasoconstriction and fibrosis, are due to AT1 activation. Recent data suggest that AT2 participates in the regulation of cell growth and renal inflammatory infiltration. Therefore, we investigated the renal expression of AT2 receptors in several models of renal injury. METHODS: Investigations were done in the following experimental models of kidney damage: systemic infusion of Ang II (inflammation), folic acid nephropathy (tubular cell death), and protein overload proteinuria. AT2 expression was determined by immunohistochemistry (protein) and reverse transcription-polymerase chain reaction (RT-PCR) (gene). RESULTS: In control animals, low levels of renal expression of AT2 were found. Ang II infusion resulted in an up-regulation of AT2 in tubular cells and de novo AT2 expression in glomeruli and vessels, associated with the presence of inflammatory cells. Acute tubular injury induced by folic acid was characterized by AT2 overexpression and apoptosis in tubular cells. Protein overload caused heavy proteinuria and tubular AT2 up-regulation. CONCLUSION: AT2 is re-expressed in pathologic conditions of kidney damage, such as inflammation, apoptosis, and proteinuria, suggesting a potential role of this receptor during renal injury.


Subject(s)
Kidney Diseases/metabolism , Kidney/metabolism , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II , Animals , Folic Acid , Immunohistochemistry , Kidney Diseases/chemically induced , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proteinuria/metabolism , Rats , Rats, Wistar , Up-Regulation
17.
Kidney Int Suppl ; (86): S33-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12969125

ABSTRACT

BACKGROUND: Angiotensin II (Ang II) is a cytokine that participates in the inflammatory response. The nuclear factor kappa B (NFkappaB) is involved in the regulation of many immune and inflammatory factors. Different works have shown that both angiotensin II receptor type 1 (AT1) and type 2 (AT2) receptors are involved in the NFkappaB pathway; however, some aspects remain mysterious. AT1 antagonists increased plasma Ang II levels that could bind to AT2, so understanding the clinical importance of AT2 stimulation or inhibition is an interesting unresolved point. METHODS: Experiments were done in wild-type (WT) and AT1a receptor knockout mice that received subcutaneous Ang II infusions (1000 ng/kg/min) for 3 days. Specific blockers of AT1 (losartan 10 mg/kg/day) and AT2 (PD123319 30 mg/kg/day) receptors were administered 1 day before and during Ang II infusion. NFkappaB activity was examined by electrophoretic mobility assay and inflammatory (monocyte/macrophage) cell infiltration by immunohistochemistry RESULTS: In WT mice, Ang II infusion caused renal NFkappaB activation that was partially diminished by either AT1 or AT2 antagonists. In AT1 knockout mice, Ang II also activated renal NFkappaB, which was only blocked by the AT2 antagonist. Both Ang II-infused WT and AT1 knockout mice showed inflammatory infiltration in tubulointerstitial areas that were suppressed by the AT2, but not AT1, antagonist. Combined therapy of both AT1 and AT2 antagonists blocked renal NFkappaB activation and inflammatory cell infiltration, both in WT and in AT1 knockout mice. CONCLUSION: Ang II, via AT1 and AT2 stimulation, leads to NFkappaB activation that was only blocked by combined therapy with both antagonists. The participation of AT2 receptors in the recruitment of inflammatory cells underscores the need of future studies that evaluate the clinical usefulness of this strategy.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Angiotensin II Type 2 Receptor Blockers , NF-kappa B/metabolism , Nephritis/metabolism , Angiotensin II/pharmacology , Animals , Imidazoles/pharmacology , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis/chemically induced , Nephritis/pathology , Pyridines/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism
18.
Circulation ; 108(12): 1499-505, 2003 Sep 23.
Article in English | MEDLINE | ID: mdl-12952842

ABSTRACT

BACKGROUND: Angiotensin II (Ang II) participates in the development of fibrosis during vascular damage. Connective tissue growth factor (CTGF) is a novel fibrotic mediator. However, the potential link between CTGF and Ang II has not been investigated. METHODS AND RESULTS: In vivo Ang II effects were studied by systemic infusion into normal rats to evaluate CTGF and extracellular matrix protein (ECM) expression by immunohistochemistry. In aorta of Ang II-infused rats, CTGF staining was markedly increased and ECM overexpression was observed. An AT1 antagonist diminished CTGF and ECM. In growth-arrested vascular smooth muscle cells, Ang II induced CTGF mRNA expression after 1 hour, remained elevated up to 24 hours, and increased CTGF protein production, which was increased up to 72 hours. The AT1 antagonist blocked Ang II-induced CTGF gene and protein expression. Early CTGF upregulation is independent of new protein synthesis. Several intracellular signals elicited by Ang II are involved in CTGF synthesis, including protein kinase C activation, reactive oxygen species, and transforming growth factor-beta endogenous production. Incubation with a CTGF antisense oligonucleotide decreased CTGF and fibronectin upregulation caused by Ang II. CONCLUSIONS: Our results show that Ang II, via AT1, increases CTGF in vascular cells both in vivo and in vitro. This novel finding suggests that CTGF may be a mediator of the profibrogenic effects of Ang II in vascular diseases.


Subject(s)
Angiotensin II/pharmacology , Fibrosis/chemically induced , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Angiotensin Receptor Antagonists , Animals , Aorta/drug effects , Aorta/pathology , Cells, Cultured , Connective Tissue Growth Factor , Drug Administration Routes , Drug Administration Schedule , Extracellular Matrix Proteins/metabolism , Female , Fibrosis/metabolism , Fibrosis/pathology , Gene Expression/drug effects , Imidazoles/pharmacology , Immediate-Early Proteins/genetics , Immunohistochemistry , Infusion Pumps, Implantable , Intercellular Signaling Peptides and Proteins/genetics , Losartan/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Pyridines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2
19.
Int J Biochem Cell Biol ; 35(6): 881-900, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12676174

ABSTRACT

Angiotensin II (AngII), the major effector peptide of renin-angiotensin system (RAS), is now recognized as a growth factor that regulates cell growth and fibrosis, besides being a physiological mediator restoring circulatory integrity. In the last few years, a large number of experimental studies has further demonstrated that AngII is involved in key events of the inflammatory process. Here, we summarize the wide variety of AngII functions and discuss them in relation with the inflammatory cascade. AngII increases vascular permeability (via the release of prostaglandins and vascular endothelial cell growth factor or rearrangement of cytoskeletal proteins) that initiates the inflammatory process. AngII could contribute to the recruitment of inflammatory cells into the tissue through the regulation of adhesion molecules and chemokines by resident cells. Moreover, AngII could directly activate infiltrating immunocompetent cells, including chemotaxis, differentiation and proliferation. Recent data also suggest that RAS activation could play a certain role even in immunologically-induced inflammation. Transcriptional regulation, predominantly via nuclear factor-kappaB (NF-kappaB) and AP-1 activation, and second mediator systems, such as endothelin-1, the small G protein (Rho) and redox-pathways are shown to be involved in the molecular mechanism by which AngII exerts those functions. Finally, AngII participates in tissue repair and remodeling, through the regulation of cell growth and matrix synthesis. In summary, recent data support the hypothesis that RAS is key mediator of inflammation. Further understanding of the role of the RAS in this process may provide important opportunities for clinical research and treatment of inflammatory diseases.


Subject(s)
Angiotensin II/physiology , Inflammation/physiopathology , Animals , Capillary Permeability , Cell Adhesion/immunology , Chemokines/biosynthesis , Chemokines/immunology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/physiology , Endothelium, Vascular/physiology , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/physiology , Lymphokines/biosynthesis , Lymphokines/physiology , Neutrophil Infiltration/immunology , Peptidyl-Dipeptidase A , Prostaglandins/biosynthesis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Curr Hypertens Rep ; 5(1): 73-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12530939

ABSTRACT

Blockers of the renin-angiotensin system are used in the treatment of several cardiovascular and renal diseases, including hypertension, atherosclerosis, and cardiac failure. Angiotensin II plays an essential role in the pathogenesis of these diseases through the regulation of cell growth, inflammation, and fibrosis. There are two main angiotensin II receptors, AT(1) and AT(2). The AT(1) receptor is responsible for most of the pathophysiologic actions of angiotensin II, including cell proliferation, production of growth factors and cytokines, and fibrosis. AT(2) causes antiproliferation and counteracts the cell growth induced by AT(1) activation. We review the mechanisms whereby AT(1) and AT(2) receptors elicit their respective actions. We discuss the current understanding of the signaling mechanisms involved in angiotensin II-induced vascular damage, describing the mediators (growth factors and cytokines) and intracellular signals (activation of protein kinases, transcription factors, and redox pathways) implicated in these processes, with special emphasis on novel information and open questions.


Subject(s)
Angiotensin II/physiology , Arteries/pathology , Muscle, Smooth, Vascular/physiology , Animals , Arteries/physiology , Arteries/physiopathology , Cell Division/physiology , Extracellular Matrix/metabolism , Fibrosis , Growth Substances/physiology , Humans , Inflammation Mediators/metabolism , Mitogen-Activated Protein Kinase Kinases/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , NF-kappa B/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...