Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(2): e0229036, 2020.
Article in English | MEDLINE | ID: mdl-32084172

ABSTRACT

LAT molecules defective in ubiquitination have an increased half-life and induce enhanced signaling when expressed in T cells. In this study, we have examined the role of ubiquitination in regulating LAT endocytosis, recycling, and degradation in resting and stimulated T cells. By tracking and comparing plasma membrane-labeled wild type and ubiquitination-resistant 2KR LAT, we find that ubiquitination promotes the degradation of surface LAT in T cells. Activation of T cells increases LAT ubiquitination and promotes trafficking of internalized LAT to lysosomes for degradation. Ubiquitination of LAT does not change internalization rates from the cell surface, but prevents efficient recycling of LAT to the surface of T cells. Our study demonstrates that surface LAT levels are tightly controlled by ubiquitination. LAT in unstimulated cells lacks ubiquitin allowing for increased LAT stability and efficient T cell activation upon TCR triggering; ubiquitination leads to efficient removal of LAT after activation.


Subject(s)
Lymphocyte Activation/physiology , Ubiquitination/physiology , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Endocytosis/physiology , Humans , Immunoblotting , Lysosomes/metabolism , Microscopy, Confocal , Phosphorylation/physiology , Signal Transduction/physiology
2.
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
3.
Breast Cancer Res ; 16(3): R53, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24886537

ABSTRACT

INTRODUCTION: Endocrine therapies targeting cell proliferation and survival mediated by estrogen receptor α (ERα) are among the most effective systemic treatments for ERα-positive breast cancer. However, most tumors initially responsive to these therapies acquire resistance through mechanisms that involve ERα transcriptional regulatory plasticity. Herein we identify VAV3 as a critical component in this process. METHODS: A cell-based chemical compound screen was carried out to identify therapeutic strategies against resistance to endocrine therapy. Binding to ERα was evaluated by molecular docking analyses, an agonist fluoligand assay and short hairpin (sh)RNA-mediated protein depletion. Microarray analyses were performed to identify altered gene expression. Western blot analysis of signaling and proliferation markers, and shRNA-mediated protein depletion in viability and clonogenic assays, were performed to delineate the role of VAV3. Genetic variation in VAV3 was assessed for association with the response to tamoxifen. Immunohistochemical analyses of VAV3 were carried out to determine its association with therapeutic response and different tumor markers. An analysis of gene expression association with drug sensitivity was carried out to identify a potential therapeutic approach based on differential VAV3 expression. RESULTS: The compound YC-1 was found to comparatively reduce the viability of cell models of acquired resistance. This effect was probably not due to activation of its canonical target (soluble guanylyl cyclase), but instead was likely a result of binding to ERα. VAV3 was selectively reduced upon exposure to YC-1 or ERα depletion, and, accordingly, VAV3 depletion comparatively reduced the viability of cell models of acquired resistance. In the clinical scenario, germline variation in VAV3 was associated with the response to tamoxifen in Japanese breast cancer patients (rs10494071 combined P value = 8.4 × 10-4). The allele association combined with gene expression analyses indicated that low VAV3 expression predicts better clinical outcome. Conversely, high nuclear VAV3 expression in tumor cells was associated with poorer endocrine therapy response. Based on VAV3 expression levels and the response to erlotinib in cancer cell lines, targeting EGFR signaling may be a promising therapeutic strategy. CONCLUSIONS: This study proposes VAV3 as a biomarker and a rationale for its use as a signaling target to prevent and/or overcome resistance to endocrine therapy in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/metabolism , Indazoles/pharmacology , Proto-Oncogene Proteins c-vav/genetics , Androstadienes/therapeutic use , Antineoplastic Agents, Hormonal/pharmacology , Aromatase Inhibitors/therapeutic use , Biomarkers, Tumor/genetics , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activators/pharmacology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Variation , Humans , Letrozole , MCF-7 Cells , Nitriles/therapeutic use , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , RNA Interference , RNA, Small Interfering , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Toremifene/pharmacology , Toremifene/therapeutic use , Triazoles/therapeutic use
4.
J Transl Med ; 9: 13, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21251296

ABSTRACT

Chronic kidney disease (CKD) comprises a group of pathologies in which the renal excretory function is chronically compromised. Most, but not all, forms of CKD are progressive and irreversible, pathological syndromes that start silently (i.e. no functional alterations are evident), continue through renal dysfunction and ends up in renal failure. At this point, kidney transplant or dialysis (renal replacement therapy, RRT) becomes necessary to prevent death derived from the inability of the kidneys to cleanse the blood and achieve hydroelectrolytic balance. Worldwide, nearly 1.5 million people need RRT, and the incidence of CKD has increased significantly over the last decades. Diabetes and hypertension are among the leading causes of end stage renal disease, although autoimmunity, renal atherosclerosis, certain infections, drugs and toxins, obstruction of the urinary tract, genetic alterations, and other insults may initiate the disease by damaging the glomerular, tubular, vascular or interstitial compartments of the kidneys. In all cases, CKD eventually compromises all these structures and gives rise to a similar phenotype regardless of etiology. This review describes with an integrative approach the pathophysiological process of tubulointerstitial, glomerular and renovascular diseases, and makes emphasis on the key cellular and molecular events involved. It further analyses the key mechanisms leading to a merging phenotype and pathophysiological scenario as etiologically distinct diseases progress. Finally clinical implications and future experimental and therapeutic perspectives are discussed.


Subject(s)
Kidney Diseases/complications , Kidney Diseases/etiology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Renal Artery/pathology , Animals , Chronic Disease , Fibrosis/etiology , Humans , Kidney Diseases/pathology , Models, Biological
5.
Pharmacol Ther ; 128(1): 61-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20600306

ABSTRACT

It is estimated that over 10% of the adult population in developed countries have some degree of chronic kidney disease (CKD). CKD is a progressive and irreversible deterioration of the renal excretory function that results in implementation of renal replacement therapy in the form of dialysis or renal transplant, which may also lead to death. CKD poses a growing problem to society as the incidence of the disease increases at an annual rate of 8%, and consumes up to 2% of the global health expenditure. CKD is caused by a variety of factors including diabetes, hypertension, infection, reduced blood supply to the kidneys, obstruction of the urinary tract and genetic alterations. The nephropathies associated with some of these conditions have been modeled in animals, this being crucial to understanding their pathophysiological mechanism and assessing prospective treatments at the preclinical level. This article reviews and updates the pathophysiological knowledge acquired primarily from experimental models and human studies of CKD. It also highlights the common mechanism(s) underlying the most relevant chronic nephropathies which lead to the appearance of a progressive, common renal phenotype regardless of aetiology. Based on this knowledge, a therapeutic horizon for the treatment of CKD is described. Present therapy primarily based upon renin-angiotensin inhibition, future diagnostics and therapeutic perspectives based upon anti-inflammatory, anti-fibrotic and hemodynamic approaches, new drugs targeting specific signaling pathways, and advances in gene and cell therapies, are all elaborated.


Subject(s)
Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/physiopathology , Animals , Calcium Channel Blockers/therapeutic use , Diabetic Nephropathies/complications , Diabetic Nephropathies/physiopathology , Disease Progression , Humans , Hypertension/complications , Hypertension/physiopathology , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/physiopathology , Nephrons/physiopathology , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/etiology
6.
Kidney Int ; 74(2): 196-209, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18449171

ABSTRACT

Chronic unilateral ureteral obstruction is a well characterized model of renal injury leading to tubulointerstitial fibrosis and distinct patterns of cell proliferation and apoptosis in the obstructed kidney. In this study we assessed the contribution of the mitogen activated protein kinase (MAPK)-ERK1/2 and the phosphatidylinositol 3 kinase (PI3K)-Akt pathways to early renal changes following unilateral obstruction. Increased activation of small Ras GTPase and its downstream effectors ERK1/2 and Akt was detected in ligated kidneys. The use of specific pharmacological inhibitors to either ERK1/2 or Akt activation led to decreased levels of fibroblast-myofibroblast markers in the interstitium while inhibition of PI3K reduced the number of proliferating cells and the amount of interstitial extracellular matrix deposition. Treatment with an ERK1/2 inhibitor diminished the number of apoptotic tubule and interstitial cells. Our results suggest a role for the MAPK-ERK1/2 and PI3K-Akt systems in early changes induced by ureteral obstruction and that inhibition of these signaling pathways may provide a novel approach to prevent progression of renal fibrosis.


Subject(s)
Kidney/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nephritis, Interstitial/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Ureteral Obstruction/enzymology , Animals , Apoptosis , Enzyme Activation , Fibrosis , Kidney/enzymology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nephritis, Interstitial/etiology , Nephritis, Interstitial/pathology , Nephritis, Interstitial/prevention & control , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
7.
J Hypertens ; 25(12): 2486-96, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17984671

ABSTRACT

OBJECTIVES: D/L-Nebivolol is a lypophilic beta1-adrenergic antagonist which is devoid of intrinsic sympathomimetic activity and can increase nitric oxide (NO) bioavailability with its subsequent vasodilating properties. The purpose of the present work was to assess the effect of long-term nebivolol administration on both renal damage and endothelial dysfunction induced by renal mass reduction (RMR) in rats. Atenolol, which does not increase NO bioavailability, was included in the study as a comparative beta-adrenoceptor antagonist. METHODS: Rats were subjected to both right nephrectomy and surgical removal of two-thirds of the left kidney in order to retain approximately one-sixth of the total renal mass. One week after ablation, rats were distributed randomly according to the following experimental groups: control group containing RMR rats without treatment; RMR rats treated daily with nebivolol for 6 months (drinking water, 8 mg/kg per day); and RMR rats treated daily with atenolol for 6 months (drinking water, 80 mg/kg per day). A group of sham-operated animals was also included. RESULTS: Administration of either nebivolol or atenolol similarly reduced arterial pressure in comparison with RMR untreated animals; however, animals receiving nebivolol presented lower levels of collagen type I expression as well as lower glomerular and interstitial fibrosis than those receiving atenolol. Urinary excretion of oxidative stress markers were also lower in animals receiving nebivolol than in rats treated with atenolol. Furthermore, nebivolol prevented RMR-induced endothelial dysfunction more efficiently than atenolol. CONCLUSIONS: Nebivolol protects against renal fibrosis, oxidative stress and endothelial dysfunction better than equivalent doses, in terms of arterial pressure reduction, of atenolol in a hypertensive model of renal damage induced by RMR.


Subject(s)
Adrenergic beta-Antagonists/administration & dosage , Benzopyrans/administration & dosage , Ethanolamines/administration & dosage , Hypertension, Renal/drug therapy , Animals , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Fibrosis , Hypertension, Renal/pathology , Hypertension, Renal/physiopathology , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Male , Nebivolol , Oxidative Stress/drug effects , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
8.
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
9.
J Nephrol ; 18(3): 341-9, 2005.
Article in English | MEDLINE | ID: mdl-16013027

ABSTRACT

Although mechanisms responsible for the initiation and development of renal fibrosis have been extensively studied, the intracellular signals involved in this phenomenon are still poorly understood. Ras proteins are the prototype members of a large family of small GTPases bound to membrane that control signalling pathways implicated in cellular growth, differentiation, proliferation and apoptosis. The purpose of the present manuscript is to review Ras signalling studies focusing on the possible role of Ras activation in renal fibrosis. A cell-specific expression of the three Ras isoforms (K-Ras, H-Ras and N-Ras) has been found in both normal and injured kidneys. Ras activation has been described in cultured mesangial cells or renal fibroblasts when challenged with several cytokines, high glucose medium or advanced glycation end-products. A role for K-Ras has been demonstrated in renal fibroblast proliferation. Mesangial cell proliferation induced by high glucose can be reverted by 3-hydroxy-3-methylglutaryl CoA reductase inhibitor which blocks the synthesis of prenyl groups and consequently Ras activation. In addition, increased Ras activation measured by Ras-GTP/total Ras ratio has been found in an experimental model of tubulointerstitial fibrosis induced by unilateral ureteral ligation. In overall, these data give enough evidence of a role for Ras activation in renal fibrosis.


Subject(s)
Kidney/pathology , ras GTPase-Activating Proteins/physiology , Animals , Apoptosis , Cell Proliferation , Enzyme Activation , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Signal Transduction
10.
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
11.
Am J Transplant ; 4(10): 1605-13, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367215

ABSTRACT

Ischemia reperfusion (I-R)-induced renal damage is reduced by systemic administration of the NO-dependent vasodilator molsidomine. The aim of this study was to estimate the effect of direct intrarenal molsidomine administration on renal dysfunction and inflammatory reaction after experimental I-R in rats, in order to assess only renal NO effects and to obviate its systemic hemodynamic action. Ischemia was induced by renal pedicle ligation (60 min) followed by reperfusion and contralateral nephrectomy. Molsidomine (4 mg/kg) was infused into the renal artery 15 min before reperfusion and its effects were compared with those of the NO-independent vasodilator hydralazine (2 mg/kg). Survival rates after 7 days were 100% in the sham-operated group and 75% in the I-R rats. Molsidomine treatment almost completely prevented the I-R-induced renal dysfunction, and survival reached 100%. Molsidomine prevented an I-R-induced increase in superoxide anion and reduced plasma levels of pro-inflammatory cytokines (TNF-alpha, IL-1beta and IFN-gamma), whereas it enhanced anti-inflammatory cytokines (IL-6 and IL-10). Inflammatory cell infiltration and cell-adhesion molecules (ICAM-1, PECAM-1, VCAM-1 and P-selectin) were lower in the molsidomine-treated kidneys than in the untreated animals. All these protective effects were not observed after hydralazine administration. In conclusion, intrarenal administration of molsidomine before reperfusion improved renal function and decreased inflammatory responses after I-R.


Subject(s)
Kidney/drug effects , Kidney/pathology , Molsidomine/pharmacology , Nitric Oxide Donors/pharmacology , Reperfusion Injury/drug therapy , Animals , Catalase/metabolism , Creatinine/metabolism , Cytokines/blood , Glutathione Peroxidase/metabolism , Intercellular Adhesion Molecule-1/metabolism , Kidney/metabolism , Nitric Oxide/metabolism , Rats , Reperfusion Injury/mortality , Superoxide Dismutase/metabolism , Superoxides/metabolism , Survival
12.
Hypertension ; 40(5): 713-20, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12411467

ABSTRACT

The goal of the present study was to evaluate the role of endoglin, a transforming growth factor-beta1 (TGF-beta1) accessory receptor, in the pathogenesis of renal fibrosis. This was achieved by testing a model of tubulo-interstitial fibrosis induced by unilateral ureteral obstruction in endoglin heterozygous (Eng(+/-)) mice. Northern and Western blot analysis revealed that endoglin expression in kidneys of these mice was significantly reduced compared with Eng(+/+) littermates. Pronounced interstitial fibrosis induced by ureteral obstruction was confirmed histologically by Masson's trichromic staining and by increased immunostaining for fibronectin and laminin without significant differences between Eng(+/-) and Eng(+/+) mice. Ureteral obstruction induced significant increases in alpha2(I) and alpha1(IV) collagen, fibronectin, and TGF-beta1 mRNA levels, as well as in total kidney collagen but changes were similar in Eng(+/-) and Eng(+/+) mouse kidneys. Ureteral obstruction also induced a 2-fold increase in endoglin mRNA levels in both Eng(+/+) mice and Eng(+/-) mice, which was confirmed by Western blot analysis. Thus, the present study provides clear evidence that endoglin is upregulated in the kidneys of mice with interstitial fibrosis induced by unilateral ureteral ligation. However, Eng(+/-) mice do not show any changes in the severity of renal disease induced in this model when compared with normal mice, suggesting that the absolute level of endoglin is not critical for the effects of TGF-beta1 in the renal fibrosis process.


Subject(s)
Fibrosis/metabolism , Kidney Diseases/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Antigens, CD , Blotting, Northern , Blotting, Western , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Endoglin , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis/complications , Fibrosis/pathology , Heterozygote , Immunohistochemistry , Kidney/chemistry , Kidney/metabolism , Kidney/pathology , Kidney Diseases/complications , Kidney Diseases/pathology , Ligation , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Cell Surface , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Up-Regulation , Ureter , Vascular Cell Adhesion Molecule-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...