Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
PLoS One ; 12(11): e0188204, 2017.
Article in English | MEDLINE | ID: mdl-29145462

ABSTRACT

Inflammation is associated with every health condition, and is an important component of many pathologies such as cardiovascular diseases. Circulating levels of soluble endoglin have been shown to be higher in the serum of patients with cardiovascular diseases with a significant inflammatory component. The aim of this study was to evaluate the implication of circulating soluble endoglin in the inflammatory response. For this purpose, a transgenic mouse expressing human soluble endoglin (sEng+) was employed, and three different inflammatory approaches were used to mimic inflammatory conditions in different tissues. This study shows that control sEng+ mice have a normal inflammatory state. The lung and kidney injury induced by the inflammatory agents was reduced in sEng+ mice, especially the intra-alveolar and kidney infiltrates, suggesting a possible reduction in inflammation induced by soluble endoglin. To deepen into this possible effect, the leukocyte number in the bronchoalveolar lavage and air pouch lavage was evaluated and a significant reduction of neutrophil infiltration in LPS-treated lungs and ischemic kidneys from sEng+ with respect to WT mice was observed. Additionally, the mechanisms through which soluble endoglin prevents inflammation were studied. We found that in sEng+ animals the increment of proinflammatory cytokines, TNFα, IL1ß and IL6, induced by the inflammatory stimulus was reduced. Soluble endoglin also prevents the augmented adhesion molecules, ICAM, VCAM and E-selectin induced by the inflammatory stimulus. In addition, vascular permeability increased by inflammatory agents was also reduced by soluble endoglin. These results suggest that soluble endoglin modulates inflammatory-related diseases and open new perspectives leading to the development of novel and targeted approaches for the prevention and treatment of cardiovascular diseases.


Subject(s)
Endoglin/blood , Inflammation/blood , Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid , Capillary Permeability , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic
2.
Biochim Biophys Acta ; 1862(9): 1801-14, 2016 09.
Article in English | MEDLINE | ID: mdl-27321931

ABSTRACT

Transforming growth factor beta 1 (TGF-ß1) is one of the most studied cytokines involved in renal tubulo-interstitial fibrosis, which is characterized by myofibroblast abundance and proliferation, and high buildup of extracellular matrix in the tubular interstitium leading to organ failure. Endoglin (Eng) is a 180-kDa homodimeric transmembrane protein that regulates a great number of TGF-ß1 actions in different biological processes, including ECM synthesis. High levels of Eng have been observed in experimental models of renal fibrosis or in biopsies from patients with chronic kidney disease. In humans and mice, two Eng isoforms are generated by alternative splicing, L-Eng and S-Eng that differ in the length and composition of their cytoplasmic domains. We have previously described that L-Eng overexpression promotes renal fibrosis after unilateral ureteral obstruction (UUO). However, the role of S-Eng in renal fibrosis is unknown and its study would let us analyze the possible function of the cytoplasmic domain of Eng in this process. For this purpose, we have generated a mice strain that overexpresses S-Eng (S-ENG(+)) and we have performed an UUO in S-ENG(+) and their wild type (WT) control mice. Our results indicate that obstructed kidney of S-ENG(+) mice shows lower levels of tubulo-interstitial fibrosis, less inflammation and less interstitial cell proliferation than WT littermates. Moreover, S-ENG(+) mice show less activation of Smad1 and Smad2/3 pathways. Thus, S-Eng overexpression reduces UUO-induced renal fibrosis and some associated mechanisms. As L-Eng overexpression provokes renal fibrosis we conclude that Eng-mediated induction of renal fibrosis in this model is dependent on its cytoplasmic domain.


Subject(s)
Endoglin/genetics , Endoglin/metabolism , Kidney/metabolism , Kidney/pathology , Nephritis/prevention & control , Ureteral Obstruction/metabolism , Animals , Cell Proliferation , Collagen Type I/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Biological , Myofibroblasts/metabolism , Myofibroblasts/pathology , Nephritis/metabolism , Nephritis/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
3.
Dis Model Mech ; 8(11): 1427-39, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26398936

ABSTRACT

The activin receptor-like kinase 1 (ALK-1) is a type I cell-surface receptor for the transforming growth factor-ß (TGF-ß) family of proteins. Hypertension is related to TGF-ß1, because increased TGF-ß1 expression is correlated with an elevation in arterial pressure (AP) and TGF-ß expression is upregulated by the renin-angiotensin-aldosterone system. The purpose of this study was to assess the role of ALK-1 in regulation of AP using Alk1 haploinsufficient mice (Alk1(+/-)). We observed that systolic and diastolic AP were significantly higher in Alk1(+/-) than in Alk1(+/+) mice, and all functional and structural cardiac parameters (echocardiography and electrocardiography) were similar in both groups. Alk1(+/-) mice showed alterations in the circadian rhythm of AP, with higher AP than Alk1(+/+) mice during most of the light period. Higher AP in Alk1(+/-) mice is not a result of a reduction in the NO-dependent vasodilator response or of overactivation of the peripheral renin-angiotensin system. However, intracerebroventricular administration of losartan had a hypotensive effect in Alk1(+/-) and not in Alk1(+/+) mice. Alk1(+/-) mice showed a greater hypotensive response to the ß-adrenergic antagonist atenolol and higher concentrations of epinephrine and norepinephrine in plasma than Alk1(+/+) mice. The number of brain cholinergic neurons in the anterior basal forebrain was reduced in Alk1(+/-) mice. Thus, we concluded that the ALK-1 receptor is involved in the control of AP, and the high AP of Alk1(+/-) mice is explained mainly by the sympathetic overactivation shown by these animals, which is probably related to the decreased number of cholinergic neurons.


Subject(s)
Activin Receptors, Type I/deficiency , Arterial Pressure , Heterozygote , Hypertension/enzymology , Activin Receptors, Type I/genetics , Activin Receptors, Type II , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Arterial Pressure/genetics , Central Nervous System/pathology , Central Nervous System/physiopathology , Cholinergic Neurons/pathology , Circadian Rhythm , Dose-Response Relationship, Drug , Genetic Predisposition to Disease , Haploinsufficiency , Hypertension/drug therapy , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Renin-Angiotensin System/drug effects , Sympathetic Nervous System/enzymology , Sympathetic Nervous System/physiopathology , Time Factors , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
4.
PLoS One ; 9(10): e110365, 2014.
Article in English | MEDLINE | ID: mdl-25313562

ABSTRACT

Transforming growth factor-ß (TGF-ß) plays a pivotal role in renal fibrosis. Endoglin, a 180 KDa membrane glycoprotein, is a TGF-ß co-receptor overexpressed in several models of chronic kidney disease, but its function in renal fibrosis remains uncertain. Two membrane isoforms generated by alternative splicing have been described, L-Endoglin (long) and S-Endoglin (short) that differ from each other in their cytoplasmic tails, being L-Endoglin the most abundant isoform. The aim of this study was to assess the effect of L-Endoglin overexpression in renal tubulo-interstitial fibrosis. For this purpose, a transgenic mouse which ubiquitously overexpresses human L-Endoglin (L-ENG+) was generated and unilateral ureteral obstruction (UUO) was performed in L-ENG+ mice and their wild type (WT) littermates. Obstructed kidneys from L-ENG+ mice showed higher amounts of type I collagen and fibronectin but similar levels of α-smooth muscle actin (α-SMA) than obstructed kidneys from WT mice. Smad1 and Smad3 phosphorylation were significantly higher in obstructed kidneys from L-ENG+ than in WT mice. Our results suggest that the higher increase of renal fibrosis observed in L-ENG+ mice is not due to a major abundance of myofibroblasts, as similar levels of α-SMA were observed in both L-ENG+ and WT mice, but to the higher collagen and fibronectin synthesis by these fibroblasts. Furthermore, in vivo L-Endoglin overexpression potentiates Smad1 and Smad3 pathways and this effect is associated with higher renal fibrosis development.


Subject(s)
Antigens, CD/genetics , Gene Expression , Kidney Diseases/etiology , Kidney Diseases/pathology , Receptors, Cell Surface/genetics , Ureteral Obstruction/complications , Animals , Collagen/metabolism , Disease Models, Animal , Endoglin , Extracellular Matrix/metabolism , Fibronectins , Fibrosis , Humans , Kidney Diseases/metabolism , Mice , Mice, Transgenic , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
5.
Biomed Res Int ; 2014: 124902, 2014.
Article in English | MEDLINE | ID: mdl-25101263

ABSTRACT

Tubulointerstitial fibrosis is a major feature of chronic kidney disease. Unilateral ureteral obstruction (UUO) in rodents leads to the development of renal tubulointerstitial fibrosis consistent with histopathological changes observed in advanced chronic kidney disease in humans. The purpose of this study was to assess the effect of inhibiting angiotensin II receptors or Ras activation on early renal fibrotic changes induced by UUO. Animals either received angiotensin II or underwent UUO. UUO animals received either losartan, atorvastatin, and farnesyl transferase inhibitor (FTI) L-744,832, or chaetomellic acid A (ChA). Levels of activated Ras, phospho-ERK1/2, phospho-Akt, fibronectin, and α-smooth muscle actin were subsequently quantified in renal tissue by ELISA, Western blot, and/or immunohistochemistry. Our results demonstrate that administration of angiotensin II induces activation of the small GTPase Ras/Erk/Akt signaling system, suggesting an involvement of angiotensin II in the early obstruction-induced activation of renal Ras. Furthermore, upstream inhibition of Ras signalling by blocking either angiotensin AT1 type receptor or by inhibiting Ras prenylation (atorvastatin, FTI o ChA) reduced the activation of the Ras/Erk/Akt signaling system and decreased the early fibrotic response in the obstructed kidney. This study points out that pharmacological inhibition of Ras activation may hold promise as a future strategy in the prevention of renal fibrosis.


Subject(s)
Angiotensin II/administration & dosage , Fibrosis/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Monomeric GTP-Binding Proteins/metabolism , Angiotensin II/metabolism , Animals , Atorvastatin , Disease Models, Animal , Fibrosis/drug therapy , Fibrosis/physiopathology , Heptanoic Acids/administration & dosage , Humans , Kidney Diseases/drug therapy , Kidney Diseases/physiopathology , Mice , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Pyrroles/administration & dosage , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Ureteral Obstruction/diet therapy , Ureteral Obstruction/metabolism , Ureteral Obstruction/physiopathology
6.
J Invest Dermatol ; 134(1): 247-255, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23765132

ABSTRACT

Endoglin (Eng) is a transmembrane glycoprotein that is mainly expressed in endothelial cells, but it is also present in the epidermis and skin appendages. To address the role of Eng in cutaneous wound healing, we compared the kinetics of reepithelialization in Eng heterozygous null (Eng(+/-)) mice and their normal littermates (Eng(+/+)) following skin wounds. The wound area was significantly larger in Eng(+/-) than in Eng(+/+) mice from 2 to 8 days after injury; overall wound closure was delayed by 1 to 2 days. In Eng(+/-) mice, keratinocytes at the wound edges exhibited impaired proliferation but were more migratory, as shown by their elongated morphology and increased keratin 17 expression. Inhibition of nitric oxide (NO) synthesis delayed healing in Eng(+/+) but not in Eng(+/-) mice. Administration of the NO donor LA-803 accelerated wound closure in Eng(+/-) mice, with no effect on normal littermates. The acute stimulation with 12-O-tetradecanoylphorbol-13-acetate (TPA) enhanced Eng expression in mouse epidermal keratinocytes in vivo and in vitro associated with hyperproliferation. Similarly, the skin of Eng(+/-) mice failed to mount a hyperplastic response to acute stimulation with TPA. These results demonstrate an important involvement of Eng in wound healing that is associated with NO bioavailability.


Subject(s)
Epidermis/injuries , Intracellular Signaling Peptides and Proteins/genetics , Nitric Oxide/metabolism , Wound Healing/physiology , Age Factors , Animals , Carcinogens/pharmacology , Cell Proliferation , Endoglin , Epidermis/metabolism , Epidermis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Heterozygote , Hyperplasia , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Tetradecanoylphorbol Acetate/pharmacology
7.
Nephrol Dial Transplant ; 26(11): 3484-95, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21602180

ABSTRACT

BACKGROUND: Nephrotoxicity is the major limitation for the clinical use of cisplatin as an anti-tumoural drug. Our aim was to investigate the protective effect of quercetin on cisplatin nephrotoxicity in a rat tumour model in vivo and to examine the mechanisms of renal protection. METHODS: Breast adenocarcinoma (13762 Mat B-III) cells were inoculated subcutaneously in male Fischer rats and 7 days later, the rats were administered daily with quercetin [50 mg/kg/day, intraperitoneally (i.p.)] or vehicle. Four days after that, the rats were given a single dose of cisplatin (4 mg/kg, i.p.) or vehicle. Tumour growth and renal function were monitored throughout the experiment. Two or 6 days after cisplatin administration, the rats were killed and the kidneys and tumours were removed to examine renal function and toxicity markers in both tissues. RESULTS: In the kidney, cisplatin treatment induced: (i) a decrease in renal blood flow and glomerular filtration rate, (ii) tubular necrosis/apoptosis, (iii) increased lipid peroxidation and decreased endogenous antioxidant systems, (iv) increased expression of inflammation markers and (v) increased activity of the apoptosis executioner caspase-3. Cisplatin effectively reduced tumour size and weight. CONCLUSIONS: Co-treatment with quercetin partially prevented all the renal effects of cisplatin, whereas it did not impair its anti-tumour activity. In conclusion, in a model of tumour-bearing rats, quercetin prevents the nephrotoxic effect of cisplatin without affecting its anti-tumour activity.


Subject(s)
Acute Kidney Injury/prevention & control , Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Antioxidants/therapeutic use , Cisplatin/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Quercetin/therapeutic use , Acute Kidney Injury/chemically induced , Adenocarcinoma/complications , Animals , Antineoplastic Agents/toxicity , Antioxidants/metabolism , Apoptosis/drug effects , Caspase 3 , Cisplatin/toxicity , Creatinine/urine , Drug Therapy, Combination , Female , Glomerular Filtration Rate , Inflammation/chemically induced , Inflammation/prevention & control , Inflammation Mediators/metabolism , Kidney Function Tests , Lipid Peroxidation , Mammary Neoplasms, Experimental/complications , Oxidative Stress/drug effects , Rats , Rats, Wistar
8.
Toxicol Sci ; 118(2): 324-47, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20554698

ABSTRACT

As in the case of other heavy metals, a considerable body of evidence suggests that overexposure to uranium may cause pathological alterations to the kidneys in both humans and animals. In the present work, our aim was to analyze the available data from a critical perspective that should provide a view of the real danger of the nephrotoxicity of this metal for human beings. A further aim was to elaborate a comparative compilation of the renal pathophysiological data obtained in humans and experimental animals with a view to gaining more insight into our knowledge of the mechanisms of action and renal damage. Finally, we address the existing perspectives for the improvement of diagnostic methods and the treatment of intoxications by uranium, performing an integrated analysis of all these aspects.


Subject(s)
Environmental Pollutants/toxicity , Kidney Diseases/chemically induced , Kidney/drug effects , Uranium Compounds/toxicity , Acute Disease , Animals , Disease Models, Animal , Female , Humans , Kidney/metabolism , Kidney/physiopathology , Kidney Diseases/physiopathology , Kidney Diseases/therapy , Male , Oxidative Stress/drug effects , Toxicity Tests
9.
J Inflamm (Lond) ; 7: 19, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20412564

ABSTRACT

Obstructive nephropathy is characterized by an inflammatory state in the kidney, that is promoted by cytokines and growth factors produced by damaged tubular cells, infiltrated macrophages and accumulated myofibroblasts. This inflammatory state contributes to tubular atrophy and interstitial fibrosis characteristic of obstructive nephropathy. Accumulation of leukocytes, especially macrophages and T lymphocytes, in the renal interstitium is strongly associated to the progression of renal injury. Proinflammatory cytokines, NF-kappaB activation, adhesion molecules, chemokines, growth factors, NO and oxidative stress contribute in different ways to progressive renal damage induced by obstructive nephropathy, as they induce leukocytes recruitment, tubular cell apoptosis and interstitial fibrosis. Increased angiotensin II production, increased oxidative stress and high levels of proinflammatory cytokines contribute to NF-kappaB activation which in turn induce the expression of adhesion molecules and chemokines responsible for leukocyte recruitment and iNOS and cytokines overexpression, which aggravates the inflammatory response in the damaged kidney. In this manuscript we revise the different events and regulatory mechanisms involved in inflammation associated to obstructive nephropathy.

10.
Kidney Int ; 75(2): 205-13, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18818685

ABSTRACT

E-cadherin expression in the kidney is used as a surrogate marker of epithelial mesenchymal transition for the testing of various antifibrotic strategies. Here we reexamined E-cadherin expression in the kidneys of rats with unilateral ureteric obstruction, which was previously reported to decrease in parallel with the development of tubulointerstitial disease in this widely used experimental model of renal fibrosis and epithelial mesenchymal transition. E-cadherin mRNA expression was consistently increased both acutely (hours) and chronically (days) in the ligated kidney compared to the cognate non-ligated kidney. Increased E-cadherin protein levels were also found in the ligated kidney particularly in dilated tubular segments. Simulation of early pressure changes in the ligated kidney by mechanical stretch of human renal epithelial cells in culture did not alter E-cadherin expression. Porcine LLCPK-1 cells subjected to hypotonic stretch, however, did have increased E-cadherin mRNA and protein levels, responses that were not prevented by transforming growth factor-beta, a cytokine that promotes epithelial mesenchymal transition. Our findings question the utility of E-cadherin as a marker of epithelial mesenchymal transition in this model of renal fibrosis.


Subject(s)
Cadherins/genetics , Fibrosis/pathology , Kidney Diseases/pathology , Ureteral Obstruction , Animals , Biomarkers , Cell Line , Cell Shape , Cell Transdifferentiation , Disease Models, Animal , Epithelial Cells/cytology , Fibrosis/metabolism , Gene Expression Regulation , Kidney Diseases/metabolism , Mesenchymal Stem Cells/cytology , Pressure , RNA, Messenger/analysis , Rats , Swine
11.
Kidney Blood Press Res ; 30(2): 67-80, 2007.
Article in English | MEDLINE | ID: mdl-17268203

ABSTRACT

Female gender is associated with better renal function and resistance to renal injury, suggesting that an oestrogen-based effect or increased androgenic effects are responsible. Studies in rodents have confirmed a biological basis for this, based on the differential effects of androgens and oestrogens on the normal and diseased kidney. Many researchers in the field believe that the pre-menopausal levels of oestrogen are key to the protection observed in females. The key pressor effects of the renin-angiotensin (RA) system are due to both direct vasoconstrictory properties and alterations in renal control of extracellular fluid volume. Additionally, the RA has been shown to promote diverse aspects of renal injury. RA activity is positively modulated by androgens and antagonized by oestrogens. Nitric oxide (NO) is a potent vasorelaxant with a key role in renal control of extracellular fluid homeostasis. NO can variously have both protective and deleterious effects on renal injury. Endogenous oestrogen has an anti-hypertensive effect as well as protective effects against cell and organ damage, many of which are mediated via increases in NO generation. We examine how the RA- and NO-generating systems may underpin key aspects of gender differences in normal renal function and renal disease.


Subject(s)
Kidney Diseases/physiopathology , Kidney/physiology , Nitric Oxide/physiology , Renin-Angiotensin System/physiology , Sex Characteristics , Androgens/physiology , Disease Progression , Estrogens/physiology , Female , Homeostasis/physiology , Humans , Male , Vasodilation/physiology
12.
J Cell Physiol ; 210(2): 456-68, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17058229

ABSTRACT

The endothelial nitric oxide synthase (eNOS) is a critical regulator of cardiovascular homeostasis, whose dysregulation leads to different vascular pathologies. Endoglin is a component of the transforming growth factor beta (TGF-beta) receptor complex present in endothelial cells that is involved in angiogenesis, cardiovascular development, and vascular homeostasis. Haploinsufficient expression of endoglin has been shown to downregulate endothelium-derived nitric oxide in endoglin(+/-) (Eng(+/-)) mice and cultured endothelial cells. Here, we find that TGF-beta1 leads to an increased vasodilatation in Eng(+/+) mice that is severely impaired in Eng(+/-) mice, suggesting the involvement of endoglin in the TGF-beta regulated vascular homeostasis. The endoglin-dependent induction of eNOS occurs at the transcriptional level and is mediated by the type I TGF-beta receptor ALK5 and its downstream substrate Smad2. In addition, Smad2-specific signaling is upregulated in endoglin-induced endothelial cells, whereas it is downregulated upon endoglin gene suppression with small interference RNA (siRNA). The endoglin-dependent upregulation of Smad2 was confirmed using eNOS and pARE promoters, whose activities are known to be Smad2 dependent, as well as with the interference of Smad2 with siRNA, Smurf2, or a dominant negative form of Smad2. Furthermore, increased expression of endoglin in endoglin-inducible endothelial cells or in transfectants resulted in increased levels of Smad2 protein without affecting the levels of Smad2 mRNA. The increased levels of Smad2 appear to be due to a decreased ubiquitination and proteasome-dependent degradation leading to stabilization of Smad2. These results suggest that endoglin enhances Smad2 protein levels potentiating TGF-beta signaling, and leading to an increased eNOS expression in endothelial cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Signal Transduction/physiology , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation/genetics , Animals , COS Cells , Cattle , Chlorocebus aethiops , Endoglin , Endothelial Cells/metabolism , Gene Expression Regulation, Enzymologic/physiology , Homeostasis/physiology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Nitric Oxide/biosynthesis , RNA Interference , RNA, Messenger/metabolism , Rats , Smad2 Protein/genetics , Vasodilation/genetics
13.
Circ Res ; 99(3): 248-56, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16840721

ABSTRACT

The endoglin heterozygous (Eng(+/-)) mouse, which serves as a model of hereditary hemorrhagic telangiectasia (HHT), was shown to express reduced levels of endothelial NO synthase (eNOS) with impaired activity. Because of intricate changes in vasomotor function in the Eng(+/-) mice and the potential interactions between the NO- and prostaglandin-producing pathways, we assessed the expression and function of cyclooxygenase (COX) isoforms. A specific upregulation of COX-2 in the vascular endothelium and increased urinary excretion of prostaglandin E(2) were observed in the Eng(+/-) mice. Specific COX-2 inhibition with parecoxib transiently increased arterial pressure in Eng(+/-) but not in Eng(+/+) mice. Transfection of endoglin in L6E9 myoblasts, shown previously to stimulate eNOS expression, led to downregulation of COX-2 with no change in COX-1. In addition, COX-2 promoter activity and protein levels were inversely correlated with endoglin levels, in doxycyclin-inducible endothelial cells. Chronic NO synthesis inhibition with N(omega)-nitro-l-arginine methyl ester induced a marked increase in COX-2 only in the normal Eng(+/+) mice. N(omega)-nitro-l-arginine methyl ester also increased COX-2 expression and promoter activity in doxycyclin-inducible endoglin expressing endothelial cells, but not in control cells. The level of COX-2 expression following transforming growth factor-beta1 treatment was less in endoglin than in mock transfected L6E9 myoblasts and was higher in human endothelial cells silenced for endoglin expression. Our results indicate that endoglin is involved in the regulation of COX-2 activity. Furthermore, reduced endoglin levels and associated impaired NO production may be responsible, at least in part, for augmented COX-2 expression and activity in the Eng(+/-) mice.


Subject(s)
Cyclooxygenase 2/genetics , Gene Expression Regulation, Enzymologic , Intracellular Signaling Peptides and Proteins/physiology , Animals , Antigens, CD/physiology , Cyclooxygenase 2/metabolism , Dinoprostone/urine , Endoglin , Endothelium, Vascular/metabolism , Heterozygote , Humans , Mice , Mice, Knockout , Nitric Oxide/biosynthesis , Promoter Regions, Genetic , Receptors, Cell Surface/physiology , Telangiectasia, Hereditary Hemorrhagic/etiology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
14.
Nephrol Dial Transplant ; 21(8): 2106-19, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16751653

ABSTRACT

BACKGROUND: Renal ischaemia-reperfusion (I-R) can cause acute tubular necrosis and chronic renal deterioration. Endoglin, an accessory receptor for Transforming Growth Factor-beta1 (TGF-beta1), is expressed on activated endothelium during macrophage maturation and implicated in the control of fibrosis, angiogenesis and inflammation. METHODS: Endoglin expression was monitored over 14 days after renal I-R in rats. As endoglin-null mice are not viable, the role of endoglin in I-R was studied by comparing renal I-R injury in haploinsufficient mice (Eng(+/-)) and their wild-type littermates (Eng(+/+)). Renal function, morphology and molecular markers of acute renal injury and inflammation were compared. RESULTS: Endoglin mRNA up-regulation in the post-ischaemic kidneys of rats occurred at 12 h after I-R; endoglin protein levels were elevated throughout the study period. Expression was initially localized to the vascular endothelium, then extended to fibrotic and inflamed areas of the interstitium. Two days after I-R, plasma creatinine elevation and acute tubular necrosis were less marked in Eng(+/-) than in Eng(+/+) mice. Significant up-regulation of endoglin protein was found only in the post-ischaemic kidneys of Eng(+/+) mice and coincided with an increased mRNA expression of the TGF-beta1 and collagen IV (alpha1) chain genes. Significant increases in vascular cell adhesion molecule-1 (VCAM-1) and inducible nitric oxide synthase (iNOS) expression, nitrosative stress, myeloperoxidase activity and CD68 staining for macrophages were evident in post-ischaemic kidneys of Eng(+/+), but not Eng(+/-) mice, suggesting that impaired endothelial activation and macrophage maturation may account for the reduced injury in post-ischaemic kidneys of Eng(+/-) mice. CONCLUSIONS: Endoglin is up-regulated in the post-ischaemic kidney and endoglin-haploinsufficient mice are protected from renal I-R injury. Endoglin may play a primary role in promoting inflammatory responses following renal I-R.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Kidney Tubular Necrosis, Acute/physiopathology , Kidney/blood supply , Reperfusion Injury/physiopathology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/genetics , Collagen Type IV/biosynthesis , Collagen Type IV/genetics , Creatinine/blood , Endoglin , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme Induction , Fibrosis , Gene Expression Regulation , Heterozygote , Inflammation , Intracellular Signaling Peptides and Proteins/genetics , Kidney/metabolism , Kidney/pathology , Kidney Tubular Necrosis, Acute/etiology , Macrophages/enzymology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/enzymology , Monocytes/pathology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Peroxidase/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reperfusion Injury/complications , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
15.
Cardiovasc Res ; 69(4): 845-54, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16405930

ABSTRACT

OBJECTIVE: To determine if angiogenesis is altered in adult Endoglin heterozygous (Eng(+/-)) mice, the animal model for the vascular disorder hereditary hemorrhagic telangiectasia type 1 (HHT1). METHODS: Primary cultures of endothelial cells were generated from Eng(+/-) and Eng(+/+) mice and analyzed for proliferation, migration, and ability to form capillary-like tubes. Endothelial cells derived from umbilical veins of newborns (HUVEC) with an HHT1 genotype were also tested for capillary formation. Two in vivo models of angiogenesis were tested in the Eng(+/-) and Eng(+/+) mice: Matrigel implant-dependent angiogenesis and reperfusion following hindlimb ischemia. RESULTS: The Eng(+/-) endothelial cells displayed significantly reduced proliferation and migration, increased collagen production, and decreased NO synthase expression and vascular endothelial growth factor (VEGF) secretion. They also showed impaired capillary tube formation in vitro, as did the HHT1 HUVEC. These endothelial cell-specific abnormalities were associated with impaired Matrigel-dependent capillary tube formation in vivo and delayed reperfusion following hindlimb ischemia. CONCLUSIONS: Although vascular development is normal in Eng(+/-) mice, angiogenic abnormalities were observed in the adult mice and their isolated endothelial cells. These results suggest that a normal level of endoglin is required for full angiogenic activity.


Subject(s)
Endothelial Cells/physiology , Endothelium, Vascular/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Telangiectasia, Hereditary Hemorrhagic/metabolism , Animals , Blotting, Northern/methods , Blotting, Western/methods , Cell Movement , Cell Proliferation , Collagen/metabolism , Endoglin , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Heterozygote , Hindlimb/blood supply , Intracellular Signaling Peptides and Proteins/genetics , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Models, Animal , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/metabolism
16.
Life Sci ; 78(20): 2373-7, 2006 Apr 11.
Article in English | MEDLINE | ID: mdl-16310224

ABSTRACT

Gentamicin is an aminoglycoside antibiotic that is very effective in treating different gram negative infections, however, one of its main side effects is nephrotoxicity. Gentamicin-induced decreases in glomerular filtration rate could be mediated by mesangial cell contraction. Resveratrol, a natural hydroxystilbene, has been identified to be a potent antioxidant with many biological activities including protection against kidney diseases. As we have previously demonstrated that gentamicin induced a reduction of planar surface area of cultured rat mesangial cells, and that resveratrol has a protective effect on gentamicin-induced nephrotoxicity in vivo, the aim of this study was to investigate the effect of resveratrol on gentamicin-induced mesangial cell contraction. This study demonstrates that the contractile effect of gentamicin on mesangial cells can be prevented by incubation with resveratrol at an optimal dose of 10 microM, as it blunted the gentamicin-induced reduction in planar cell surface area and the number of contracted cells. Besides, the preincubation with 10(-5)M diphenylene iodinium (DPI), an inhibitor of the NADP(H) oxidase, also blunted gentamicin-induced cell contraction. This preventive effect was higher when cells were incubated with both substances together. These results strongly suggest that the protective effect resveratrol against gentamicin-induced reduction in renal function in vivo could be mediated by inhibiting gentamicin-induced mesangial cells contraction.


Subject(s)
Anti-Bacterial Agents/antagonists & inhibitors , Antioxidants/pharmacology , Gentamicins/antagonists & inhibitors , Glomerular Mesangium/drug effects , Stilbenes/pharmacology , Animals , Cell Size/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glomerular Mesangium/cytology , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , NADP/metabolism , NADPH Oxidases/antagonists & inhibitors , Rats , Resveratrol
17.
Toxicol Appl Pharmacol ; 210(1-2): 128-35, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16226777

ABSTRACT

Inflammation can play a key role in Cd-induced dysfunctions. Quercetin is a potent oxygen free radical scavenger and a metal chelator. Our aim was to study the effect of quercetin on Cd-induced kidney damage and metallothionein expression. The study was performed in Wistar rats that were administered during 9 weeks with either cadmium (1.2 mg Cd/kg/day, s.c.), quercetin (50 mg/kg/day, i.p.) or cadmium + quercetin. Renal toxicity was evaluated by measuring blood urea nitrogen concentration and urinary excretion of enzymes marker of tubular damage. Endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) renal expression were assessed by Western blot. Renal expression of metallothionein 1 and 2 (MT-1, MT-2) and eNOS mRNA was assessed by Northern blot. Our data demonstrated that Cd-induced renal toxicity was markedly reduced in rats that also received quercetin. MT-1 and MT-2 mRNA levels in kidney were substantially increased during treatment with Cd, being even higher when the animals received Cd and quercetin. Renal eNOS expression was significantly higher in rats receiving Cd and quercetin than in animals receiving Cd alone or in control rats. In the group that received Cd, COX-2 and iNOS expression was markedly higher than in control rats. In the group Cd+quercetin, no changes in COX-2 and iNOS expression were observed compared with the control group. Our results demonstrate that quercetin treatment prevents Cd-induced overexpression of iNOS and COX-2, and increases MT expression. These effects can explain the protection by quercetin of Cd-induced nephrotoxicity.


Subject(s)
Cadmium Compounds/toxicity , Cyclooxygenase 2/biosynthesis , Kidney Diseases/chemically induced , Metallothionein/biosynthesis , Nitric Oxide Synthase/biosynthesis , Protective Agents/therapeutic use , Quercetin/therapeutic use , Animals , Chronic Disease , Disease Models, Animal , Enzyme Induction , Kidney Diseases/enzymology , Kidney Diseases/metabolism , Kidney Diseases/prevention & control , Male , Rats , Rats, Wistar
18.
Kidney Blood Press Res ; 28(1): 32-40, 2005.
Article in English | MEDLINE | ID: mdl-15475654

ABSTRACT

BACKGROUND: Endoglin is a membrane glycoprotein that regulates TGF-beta1 signaling. Previous studies have revealed that endoglin is upregulated in several models of experimental fibrosis, and that endoglin expression can counteract the fibrogenic effects of TGF-beta1. As treatment with angiotensin converting enzyme (ACE) inhibitors reduces renal fibrosis by mechanisms that are, in part, not dependent on angiotensin II blockade, we have assessed the hypothesis that this effect could be mediated by endoglin upregulation. METHODS: We have used the 5/6-nephrectomy renal mass reduction (RMR) model of renal fibrosis in rats treated (RMR+T) or not treated with the ACE inhibitor trandolapril (0.7 mg/kg/day). One, 3 and 5 months after RMR, mean arterial pressure and renal function were measured. In addition, renal fibrosis was evaluated quantitatively and endoglin, TGF-beta1, collagen type I and collagen type IV expression was assessed by Northern blot and immunohistochemistry. RESULTS: RMR induced a progressive increase in mean arterial pressure, urinary protein excretion and glomerular and tubulointerstitial fibrosis, which is accompanied by an increased expression of TGF-beta1, endoglin and collagen types I and IV. Trandolapril treatment reduced systemic blood pressure and lessened proteinuria after RMR, as well as expression of TGF-beta1, endoglin and collagens. CONCLUSION: The present study demonstrates an increased TGF-beta1, endoglin, collagen type I and collagen type IV expression in rats with severe hypertension and renal damage. The effect of trandolapril to decrease renal fibrosis seems to be based in a reduced TGF-beta1 expression but not in an increased expression of endoglin.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Diseases/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Animals , Down-Regulation/drug effects , Endoglin , Fibrosis/drug therapy , Fibrosis/metabolism , Fibrosis/pathology , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Male , Rats , Rats, Wistar , Time Factors , Transforming Growth Factor beta/biosynthesis
19.
Kidney Int ; 65(6): 2161-71, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15149329

ABSTRACT

BACKGROUND: Gentamicin (G)-induced acute renal failure is characterized by an impairment of glomerular function without apparent changes in glomerular structure. However, G stimulates reactive oxygen species (ROS)-mediated mesangial cell proliferation in vitro. We studied whether G promotes mesangial cell apoptosis in vitro, and if apoptosis and proliferation in parallel may occur in glomerular cells in vivo after a renal damage induced by G treatment. METHODS: For in vivo studies, rats were treated with G (100 mg/kg body weight/day) for 6 days, and functional and histologic studies were performed. For in vitro studies, mesangial cell proliferation and apoptosis were evaluated after 24, 48, and 72 hours of 10(-5) mol/L G incubation. RESULTS: After G injections, the number of nuclei per glomerulus did not change, whereas proliferating and apoptotic cell numbers increased. G increases DNA synthesis and cell number in cultured mesangial cells, and increases markedly the apoptotic cell number. ROS scavengers superoxide dismutase and catalase reduce G-induced mesangial cell apoptosis, whereas the incubation with the ROS donor system xanthine plus xanthine oxidase increases apoptosis to levels similar to G. G-induced cellular proliferation and apoptosis either in vitro or in vivo is associated to an early increase in the pro-apoptotic protein Bax and a delayed increase in the survival protein Bcl-2. CONCLUSION: G simultaneously induces proliferation and apoptosis of mesangial cells in vitro and glomerular mesangial cells in vivo. ROS may mediate G-induced mesangial apoptosis in vitro. The equilibrium proliferation/apoptosis may maintain mesangial cell number within normal limits after a G-induced glomerular insult.


Subject(s)
Gentamicins/toxicity , Glomerular Mesangium/drug effects , Glomerular Mesangium/pathology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cells, Cultured , Glomerular Mesangium/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Time Factors
20.
FASEB J ; 18(3): 609-11, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14734648

ABSTRACT

Endoglin is a membrane glycoprotein that plays an important role in cardiovascular development and angiogenesis. We examined the role of endoglin in the control of vascular tone by measuring nitric oxide (NO)-dependent vasodilation in haploinsufficient mice (Eng+/-) and their Eng+/+ littermates. The vasodilatory effect of acetylcholine, bradykinin, and sodium nitroprusside was assessed in anesthetized mice; in isolated, perfused hindlimbs; and in aortic rings. The substantial hypotensive and vasodilatory response induced by acetylcholine and bradykinin in Eng+/+ was markedly reduced in Eng+/- mice. Both kinds of animals had similar responses to sodium nitroprusside, suggesting that the deficient vasodilatory effect is not due to a NO response impairment. Urinary and plasma concentrations of nitrites, a NO metabolite, were lower in Eng+/- than in Eng+/+ mice. The levels of endothelial nitric oxide synthase (eNOS) in kidneys and femoral arteries were about half in Eng+/- than in Eng+/+ mice and were also reduced in primary cultures of aortic endothelial cells from Eng+/- compared with those from Eng+/+ mice. Furthermore, overexpression or suppression of endoglin in cultured cells induced a marked increase or decrease in the protein levels of eNOS, respectively. Thus, our results in vivo and in vitro demonstrate a relationship between endoglin and NO-dependent vasodilation mediated by the regulation of eNOS expression.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/physiology , Vascular Cell Adhesion Molecule-1/physiology , Vasodilation/physiology , Acetylcholine/pharmacology , Acetylcholine/toxicity , Animals , Antigens, CD , Blood Pressure/drug effects , Bradykinin/toxicity , Cell Line , Endoglin , Endothelial Cells/metabolism , Enzyme Induction/physiology , Hypotension/chemically induced , Kidney/metabolism , Lung/metabolism , Mice , Mice, Knockout , Myoblasts/cytology , Myoblasts/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitroprusside/pharmacology , Receptors, Cell Surface , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Transfection , Transforming Growth Factor beta/deficiency , Transforming Growth Factor beta1 , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...