Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Kidney Int ; 101(4): 720-732, 2022 04.
Article in English | MEDLINE | ID: mdl-35090878

ABSTRACT

To guide the development of therapeutic interventions for acute kidney injury, elucidating the deleterious pathways of this global health problem is highly warranted. Emerging evidence has indicated a pivotal role of endothelial dysfunction in the etiology of this disease. We found that the class III semaphorin SEMA3C was ectopically upregulated with full length protein excreted into the blood and truncated protein secreted into the urine upon kidney injury and hypothesized a role for SEAM3C in acute kidney injury. Sema3c was genetically abrogated during acute kidney injury and subsequent kidney morphological and functional defects in two well-characterized models of acute kidney injury; warm ischemia/reperfusion and folic acid injection were analyzed. Employing a beta actin-dependent, inducible knockout of Sema3c, we demonstrate that in acute kidney injury SEMA3C promotes interstitial edema, leucocyte infiltration and tubular injury. Additionally, intravital microscopy combined with Evans Blue dye extravasation and primary culture of magnetically sorted peritubular endothelial cells identified a novel role for SEMA3C in promoting vascular permeability. Thus, our study points to microvascular permeability as an important driver of injury in acute kidney injury, and to SEMA3C as a novel permeability factor and potential target for therapeutic intervention.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Semaphorins , Acute Kidney Injury/genetics , Acute Kidney Injury/prevention & control , Animals , Capillary Permeability , Endothelial Cells/metabolism , Female , Humans , Kidney/metabolism , Male , Mice , Reperfusion Injury/complications , Reperfusion Injury/genetics , Reperfusion Injury/prevention & control , Semaphorins/genetics , Semaphorins/metabolism
2.
J Am Soc Nephrol ; 31(1): 85-100, 2020 01.
Article in English | MEDLINE | ID: mdl-31690575

ABSTRACT

BACKGROUND: The matricellular protein periostin has been associated with CKD progression in animal models and human biopsy specimens. Periostin functions by interacting with extracellular matrix components to drive collagen fibrillogenesis and remodeling or by signaling through cell-surface integrin receptors to promote cell adhesion, migration, and proliferation. However, its role in AKI is unknown. METHODS: We used mice with conditional tubule-specific overexpression of periostin or knockout mice lacking periostin expression in the renal ischemia-reperfusion injury model, and primary cultures of isolated tubular cells in a hypoxia-reoxygenation model. RESULTS: Tubular epithelial cells showed strong production of periostin during the repair phase of ischemia reperfusion. Periostin overexpression protected mice from renal injury compared with controls, whereas knockout mice showed increased tubular injury and deteriorated renal function. Periostin interacted with its receptor, integrin-ß1, to inhibit tubular cell cycle arrest and apoptosis in in vivo and in vitro models. After ischemia-reperfusion injury, periostin-overexpressing mice exhibited diminished expression of proinflammatory molecules and had more F4/80+ macrophages compared with knockout mice. Macrophages from periostin-overexpressing mice showed increased proliferation and expression of proregenerative factors after ischemia-reperfusion injury, whereas knockout mice exhibited the opposite. Coculturing a macrophage cell line with hypoxia-treated primary tubules overexpressing periostin, or treating such macrophages with recombinant periostin, directly induced macrophage proliferation and expression of proregenerative molecules. CONCLUSIONS: In contrast to the detrimental role of periostin in CKD, we discovered a protective role of periostin in AKI. Our findings suggest periostin may be a novel and important mediator of mechanisms controlling renal repair after AKI.


Subject(s)
Acute Kidney Injury , Cell Adhesion Molecules/physiology , Cell Proliferation , Macrophages/physiology , Acute Kidney Injury/etiology , Animals , Disease Models, Animal , Kidney/blood supply , Male , Mice , Mice, Knockout , Reperfusion Injury/complications , Reperfusion Injury/pathology
3.
Kidney Int ; 94(1): 126-138, 2018 07.
Article in English | MEDLINE | ID: mdl-29751972

ABSTRACT

Acute kidney injury is a major risk factor for subsequent chronic renal and/or cardiovascular complications. Previous studies have shown that Notch3 was de novo expressed in the injured renal epithelium in the early phases of chronic kidney disease. Here we examined whether Notch3 is involved in the inflammatory response and the epithelial cell damage that typifies ischemic kidneys using Notch3 knockout mice and mice with short-term activated Notch3 signaling (N3ICD) in renal epithelial cells. After ischemia/reperfusion, N3ICD mice showed exacerbated infiltration of inflammatory cells and severe tubular damage compared to control mice. Inversely, Notch3 knockout mice were protected against ischemia/reperfusion injury. Renal macrophages derived from Notch3 knockout mice failed to activate proinflammatory cytokines. Chromatin immunoprecipitation analysis of the Notch3 promoter identified NF-κB as the principal inducer of Notch3 in ischemia/reperfusion. Thus, Notch3 induced by NF-κB in the injured epithelium sustains a proinflammatory environment attracting activated macrophages to the site of injury leading to a rapid deterioration of renal function and structure. Hence, targeting Notch3 may provide a novel therapeutic strategy against ischemia/reperfusion and acute kidney injury by preservation of epithelial structure and disruption of proinflammatory signaling.


Subject(s)
Acute Kidney Injury/pathology , Kidney Tubules/pathology , Receptor, Notch3/metabolism , Reperfusion Injury/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Animals , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/metabolism , Epithelium/pathology , Humans , Kidney Tubules/immunology , Kidney Tubules/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Receptor, Notch3/genetics
4.
J Am Soc Nephrol ; 28(5): 1475-1490, 2017 May.
Article in English | MEDLINE | ID: mdl-27920156

ABSTRACT

De novo expression in the kidney of periostin, a protein involved in odontogenesis and osteogenesis, has been suggested as a biomarker of renal disease. In this study, we investigated the mechanism(s) of induction and the role of periostin in renal disease. Using a combination of bioinformatics, reporter assay, and chromatin immunoprecipitation analyses, we found that NFκB and other proinflammatory transcription factors induce periostin expression in vitro and that binding of these factors on the periostin promoter is enriched in glomeruli during experimental GN. Mice lacking expression of periostin displayed preserved renal function and structure during GN. Furthermore, delayed administration of periostin antisense oligonucleotides in wild-type animals with GN reversed already established proteinuria, diminished tissue inflammation, and improved renal structure. Lack of periostin expression also blunted the de novo renal expression of integrin-ß3 and phosphorylation of focal adhesion kinase and AKT, known mediators of integrin-ß3 signaling that affect cell motility and survival, observed during GN in wild-type animals. In vitro, recombinant periostin increased the expression of integrin-ß3 and the concomitant phosphorylation of focal adhesion kinase and AKT in podocytes. Notably, periostin and integrin-ß3 were highly colocalized in biopsy specimens from patients with inflammatory GN. These results demonstrate that interplay between periostin and renal inflammation orchestrates inflammatory and fibrotic responses, driving podocyte damage through downstream activation of integrin-ß3 signaling. Targeting periostin may be a novel therapeutic strategy for treating CKD.


Subject(s)
Cell Adhesion Molecules/physiology , Integrin beta3/physiology , Kidney Diseases/etiology , NF-kappa B/physiology , Animals , Female , Glomerulonephritis/complications , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction
5.
J Am Soc Nephrol ; 28(10): 2915-2930, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28667079

ABSTRACT

GN refers to a variety of renal pathologies that often progress to ESRD, but the molecular mechanisms underlying this progression remain incompletely characterized. Here, we determined whether dysregulated expression of the gap junction protein connexin 43, which has been observed in the progression of renal disease, contributes to GN progression. Immunostaining revealed de novo expression of connexin 43 in damaged glomeruli in patients with glomerular diseases as well as in mice after induction of experimental GN. Notably, 2 weeks after the induction of GN with nephrotoxic serum, mice with a heterozygous deletion of the connexin 43 gene (connexin 43+/-) had proteinuria, BUN, and serum creatinine levels significantly lower than those of wild-type animals. Additionally, the connexin 43+/- mice showed less crescent formation, tubular dilation, monocyte infiltration, and interstitial renal fibrosis. Treatment of cultured podocytes with connexin 43-specific blocking peptides attenuated TGF-ß-induced cytoskeletal and morphologic changes and apoptosis as did treatment with the purinergic blocker suramin. Finally, therapeutic treatment of GN mice with connexin 43-specific antisense oligodeoxynucleotide improved functional and structural renal parameters. These findings suggest that crosstalk between connexin 43 and purinergic signaling contributes to podocyte damage in GN. Given that this protein is highly induced in individuals with glomerular diseases, connexin 43 may be a novel target for therapeutic treatment of GN.


Subject(s)
Connexin 43/metabolism , Glomerulonephritis/metabolism , Animals , Apoptosis , Cell Dedifferentiation , Cell Line , Disease Models, Animal , Disease Progression , Female , Fibrosis , Glomerulonephritis/pathology , Humans , Kidney/pathology , Mice , Podocytes/metabolism , Receptors, Purinergic/metabolism
6.
J Am Soc Nephrol ; 26(7): 1561-75, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25421557

ABSTRACT

Notch3 expression is found in the glomerular podocytes of patients with lupus nephritis or focal segmental GN but not in normal kidneys. Here, we show that activation of the Notch3 receptor in the glomeruli is a turning point inducing phenotypic changes in podocytes promoting renal inflammation and fibrosis and leading to disease progression. In a model of rapidly progressive GN, Notch3 expression was induced by several-fold in podocytes concurrently with disease progression. By contrast, mice lacking Notch3 expression were protected because they exhibited less proteinuria, uremia, and inflammatory infiltration. Podocyte outgrowth from glomeruli isolated from wild-type mice during the early phase of the disease was higher than outgrowth from glomeruli of mice lacking Notch3. In vitro studies confirmed that podocytes expressing active Notch3 reorganize their cytoskeleton toward a proliferative/migratory and inflammatory phenotype. We then administered antisense oligodeoxynucleotides targeting Notch3 or scramble control oligodeoxynucleotides in wild-type mice concomitant to disease induction. Both groups developed chronic renal disease, but mice injected with Notch3 antisense had lower values of plasma urea and proteinuria and inflammatory infiltration. The improvement of renal function was accompanied by fewer deposits of fibrin within the glomeruli and by decreased peritubular inflammation. Finally, abnormal Notch3 staining was observed in biopsy samples of patients with crescentic GN. These results demonstrate that abnormal activation of Notch3 may be involved in the progression of renal disease by promoting migratory and proinflammatory pathways. Inhibiting Notch3 activation could be a novel, promising approach to treat GN.


Subject(s)
Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/genetics , Kidney Glomerulus/pathology , Podocytes/pathology , Receptors, Notch/genetics , Transcriptional Activation/genetics , Analysis of Variance , Animals , Blood Urea Nitrogen , Cells, Cultured , Disease Models, Animal , Disease Progression , Enzyme-Linked Immunosorbent Assay , Glomerulosclerosis, Focal Segmental/pathology , Humans , Immunohistochemistry , Kidney Glomerulus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Podocytes/metabolism , Proteinuria/physiopathology , RNA, Messenger/analysis , Random Allocation , Real-Time Polymerase Chain Reaction/methods , Receptor, Notch3 , Reference Values , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Sheep , Up-Regulation
7.
Nephrol Dial Transplant ; 30(12): 1965-71, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25829327

ABSTRACT

The incidence and prevalence of chronic kidney disease represents an important problem for public health. In renal diseases, the main histologic alterations derive from the development of renal fibrosis which results from the loss of the balance between pro- and anti-fibrotic factors. Tyrosine kinase receptors (RTKs) and matricellular proteins (MPs) are nowadays studied as potential modulators of renal injury. RTKs regulate cell cycle, migration, metabolism and cellular differentiation. Discoidin domain receptor-1 (DDR-1) is an RTK that has been extensively studied in cancer, and lung and renal diseases. It modulates inflammatory recruitment, extracellular matrix deposition and fibrosis; in renal diseases, it appears to act independently of the underlying disease. MPs regulate cell-matrix interactions and matrix accumulation, cellular adhesion and migration, and expression of inflammatory cells. Periostin is an MP, mainly studied in bone, heart, lung and cancer. Several studies demonstrated that it mediates cell-matrix interactions, migration of inflammatory cells and development of fibrosis. Recently, it has been reported in several nephropathies. In this review, we discuss the potential pathological roles of DDR-1 and periostin focussing on the kidney in both experimental models and human diseases.


Subject(s)
Cell Adhesion Molecules/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Renal Insufficiency, Chronic/metabolism , Discoidin Domain Receptor 1 , Humans
8.
J Am Soc Nephrol ; 25(8): 1724-36, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24578131

ABSTRACT

Increased renal expression of periostin, a protein normally involved in embryonic and dental development, correlates with the decline of renal function in experimental models and patient biopsies. Because periostin has been reported to induce cell differentiation, we investigated whether it is also involved in the development of renal disease and whether blocking its abnormal expression improves renal function and/or structure. After unilateral ureteral obstruction in wild-type mice, we observed a progressive increase in the expression and synthesis of periostin in the obstructed kidney that associated with the progression of renal lesions. In contrast, mice lacking the periostin gene showed less injury-induced interstitial fibrosis and inflammation and were protected against structural alterations. This protection was associated with a preservation of the renal epithelial phenotype. In vitro, administration of TGF-ß to renal epithelial cells increased the expression of periostin several-fold, leading to subsequent loss of the epithelial phenotype. Furthermore, treatment of these cells with periostin increased the expression of collagen I and stimulated the phosphorylation of FAK, p38, and ERK 42/44. In vivo delivery of antisense oligonucleotides to inhibit periostin expression protected animals from L-NAME-induced renal injury. These data strongly suggest that periostin mediates renal disease in response to TGF-ß and that blocking periostin may be a promising therapeutic strategy against the development of CKD.


Subject(s)
Cell Adhesion Molecules/physiology , Nephritis/etiology , Nephrosclerosis/etiology , Animals , Cell Culture Techniques , Disease Models, Animal , Female , Gene Silencing , Male , Mice, Inbred C57BL , Nephritis/metabolism , Nephritis/prevention & control , Nephrosclerosis/metabolism , Nephrosclerosis/prevention & control , Podocytes/physiology , Rats, Sprague-Dawley , Transforming Growth Factor beta/physiology , Ureteral Obstruction/complications , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
9.
Kidney Int ; 86(4): 768-79, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24850151

ABSTRACT

Excessive recruitment of monocytes and progression of fibrosis are hallmarks of chronic kidney disease (CKD). Recently we reported that the expression of connexin 43 (Cx43) was upregulated in the kidney during experimental nephropathy. To investigate the role of Cx43 in the progression of CKD, we interbred RenTg mice, a genetic model of hypertension-induced CKD, with Cx43+/- mice. The renal cortex of 5-month-old RenTgCx43+/- mice showed a marked decrease of cell adhesion markers leading to reduced monocyte infiltration and interstitial renal fibrosis compared with their littermates. In addition, functional and histological parameters such as albuminuria and glomerulosclerosis were ameliorated in RenTgCx43+/- mice. Interestingly, treatment with Cx43 antisense produced remarkable improvement of renal function and structure in 1-year-old RenTg mice. Similar results were found in Cx43+/- or wild-type mice treated with Cx43 antisense after obstructive nephropathy. Furthermore, in these mice, Cx43 antisense attenuated E-cadherin downregulation and phosphorylation of the transcription factor Sp1 by the ERK pathway resulting in decreased transcription of type I collagen gene. Interestingly, Cx43-specific blocking peptide inhibited monocyte adhesion in activated endothelium and profibrotic pathways in tubular cells. Cx43 was highly increased in biopsies of patients with CKD. Thus, Cx43 may represent a new therapeutic target against the progression of CKD.


Subject(s)
Connexin 43/genetics , Oligonucleotides, Antisense/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Albuminuria/drug therapy , Albuminuria/genetics , Animals , Cadherins/metabolism , Cell Adhesion , Collagen Type I/genetics , Connexin 43/analysis , Disease Progression , Down-Regulation/drug effects , Fibrosis , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/genetics , Humans , MAP Kinase Signaling System , Mice , Monocytes/physiology , Oligonucleotides, Antisense/pharmacology , Phosphorylation/drug effects , Renal Insufficiency, Chronic/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic
10.
FASEB J ; 26(10): 4079-91, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22751008

ABSTRACT

This study investigated the role of discoidin domain receptor 1 (DDR1), a collagen receptor that displays tyrosine-kinase activity, in the development of glomerulonephritis. Crescentic glomerulonephritis was induced in DDR1-deficient mice and their wild-type (WT) littermates as controls, by injection of alloimmune sheep nephrotoxic serum (NTS). Histological, functional and transcriptomic studies were performed. Glomerulonephritis produced a 17-fold increase of DDR1 expression, predominantly in glomeruli. DDR1 deletion protected NTS-treated mice against glomerular disease (proteinuria/creatininuria 5.5±1.1 vs. 13.2±0.8 g/mmol in WT, crescents 12±2 vs. 24±2% of glomeruli, urea 16±2 vs. 28±5 mM), hypertension (123±11 vs. 157±8 mmHg), and premature death (70 vs. 10% survival) (all P<0.05). Reciprocal stimulation between DDR1 and interleukin-1b expression in vivo and in cultured podocytes suggested a positive feed-back loop between DDR1 and inflammation. In NTS-treated WT mice, administration of DDR1-specific antisense oligodeoxynucleotides decreased DDR1 expression (-56%) and protected renal function and structure, including nephrin expression (4.2±1.4 vs. 0.9±0.4 arbitrary units, P<0.05), compared to control mice receiving scrambled oligodeoxynucleotides. The therapeutic potential of this approach was reinforced by the observation of increased DDR1 expression in glomeruli of patients with lupus nephritis and Goodpasture's syndrome. These results prompt further interest in DDR1 blockade strategies, especially in the treatment of glomerulonephritis.


Subject(s)
Glomerulonephritis/prevention & control , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Animals , Blood Pressure/genetics , Blood Pressure/physiology , Blood Urea Nitrogen , Blotting, Western , Discoidin Domain Receptors , Female , Fluorescent Antibody Technique , Glomerulonephritis/genetics , Glomerulonephritis/urine , Humans , Immunohistochemistry , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Mutant Strains , Mice, Transgenic , Proteinuria/genetics , Proteinuria/urine , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
J Pathol ; 228(3): 286-99, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22806125

ABSTRACT

Kidney diseases impart a vast burden on affected individuals and the overall health care system. Progressive loss of renal parenchymal cells and functional decline following injury are often observed. Notch-1 and -2 receptors are crucially involved in nephron development and contribute to inflammatory kidney diseases. We specifically determined the participation of receptor Notch-3 following tubulointerstitial injury and in inflammatory responses. Here we show by heat map analyses that Notch-3 transcripts are up-regulated in human kidney diseases. A similar response was corroborated with kidney cells following TGF-ß exposure in vitro. The murine unilateral ureteral obstruction (UUO) model mirrors hallmarks of tubulointerstitial injury and damage. A subset of tubular and interstitial cells demonstrated up-regulated Notch-3 receptor expression in diseased animals. We hypothesized a relevance of Notch-3 receptors for the chemotactic response. To address this question, animals with genetic ablation of receptor Notch-3 were analysed following UUO. As a result, we found that Notch-3-deficient animals are protected from tubular injury and cell loss with significantly reduced interstitial collagen deposition. Monocytic cell infiltration was significantly reduced and retarded, likely due to abrogated chemokine synthesis. A cell model was set up that mimics enhanced receptor Notch-3 expression and activation. Here a pro-mitogenic response was seen with activated signalling in tubular cells and fibroblasts. In conclusion, Notch-3 receptor fulfils non-redundant roles in the inflamed kidney that may not be replaced by other Notch receptor family members. Thus, specific blockade of this receptor may be suitable as therapeutic option to delay progression of kidney disease.


Subject(s)
Inflammation/physiopathology , Kidney/physiopathology , Nephritis, Interstitial/physiopathology , Receptors, Notch/physiology , Ureteral Obstruction/physiopathology , Animals , Biopsy , Cell Line , Cell Proliferation , Disease Models, Animal , Female , Fibrosis , Humans , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-2 Protein , Kidney/metabolism , Kidney/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Rats , Receptor, Notch1/physiology , Receptor, Notch3 , Receptors, Notch/deficiency , Receptors, Notch/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
12.
Kidney Int ; 81(12): 1226-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22418977

ABSTRACT

Thrombospondin-1 (TSP-1) is an endogenous activator of transforming growth factor-ß (TGF-ß), and an anti-angiogenic factor, which may prevent kidney repair. Here we investigated whether TSP-1 is involved in the development of chronic kidney disease using rats with unilateral ureteral obstruction, a well-known model to study renal fibrosis. Obstruction of 10 days duration induced inflammation, tubular cell atrophy, dilation, apoptosis, and proliferation, leading to interstitial fibrosis. TSP-1 expression was increased in parallel to that of collagen III and TGF-ß. Relief of the obstruction at day 10 produced a gradual improvement in renal structure and function, the reappearance of peritubular capillaries, and restoration of renal VEGF content over a 7- to 15-day post-relief period. TSP-1 expression decreased in parallel with that of TGF-ß1 and collagen III. Mice in which the TSP-1 gene was knocked out displayed less inflammation and had better preservation of renal tissue and the peritubular capillary network compared to wild-type mice. Additional studies showed that the inflammatory effect of TSP-1 was mediated, at least in part, by monocyte chemoattractant protein-1 and activation of the Th17 pathway. Thus, TSP-1 is an important profibrotic and inflammatory mediator of renal disease. Blockade of its action may be a treatment against the development of chronic kidney disease.


Subject(s)
Inflammation Mediators/metabolism , Kidney Diseases/etiology , Kidney/metabolism , Nephritis/etiology , Thrombospondin 1/metabolism , Ureteral Obstruction/complications , Animals , Apoptosis , Atrophy , Capillaries/metabolism , Capillaries/pathology , Cell Proliferation , Chemokine CCL2/metabolism , Chronic Disease , Collagen Type III/metabolism , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Kidney/blood supply , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Knockout , Nephrectomy , Nephritis/genetics , Nephritis/metabolism , Nephritis/pathology , Nephritis/prevention & control , Rats , Rats, Sprague-Dawley , Signal Transduction , Thrombospondin 1/deficiency , Thrombospondin 1/genetics , Time Factors , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Vascular Endothelial Growth Factor A/metabolism
13.
Am J Pathol ; 179(1): 83-91, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21640971

ABSTRACT

The interactions between tubulointerstitial infiltrating cells and the extracellular matrix play an important role in regulating renal fibrosis. Discoidin domain receptor 1 (DDR1) is a nonintegrin tyrosine kinase receptor for collagen implicated in cell adhesion, proliferation, and extracellular matrix remodeling. We have previously demonstrated that transgenic mice lacking DDR1 are protected from hypertension-associated renal fibrosis. The purpose of this study was to determine the role of DDR1 in renal inflammation and fibrosis related to primitive tubulointerstitial injury. After 12 days of unilateral ureteral obstruction (UUO), kidney histopathologic and real-time quantitative PCR analyses were performed in DDR1(-/-) and wild-type mice. DDR1 expression was strongly increased in the obstructed kidney. Wild-type mice developed important perivascular and interstitial inflammation and fibrosis. In comparison, DDR1(-/-) mice displayed reduced accumulation of fibrillar collagen and transforming growth factor ß expression. F4/80(+) cell count and proinflammatory cytokines were remarkably blunted in DDR1(-/-) obstructed kidneys. Leukocyte rolling and adhesion evaluated by intravital microscopy were not different between DDR1(-/-) and wild-type mice. Importantly, macrophages isolated from DDR1(-/-) mice presented similar M1/M2 polarization but displayed impaired migration in response to monocyte chemoattractant protein-1. Together, these data suggest that DDR1 plays an important role in the pathogenesis of renal disease via enhanced inflammation. Inhibition of DDR1 expression or activity may represent a novel therapeutic target against the progression of renal diseases.


Subject(s)
Cell Movement , Fibrosis/etiology , Inflammation/etiology , Kidney Diseases/complications , Receptor Protein-Tyrosine Kinases/physiology , Animals , Blotting, Western , Cell Adhesion , Cell Proliferation , Cells, Cultured , Discoidin Domain Receptor 1 , Fibrosis/pathology , Humans , Inflammation/pathology , Kidney Diseases/pathology , Leukocytes/pathology , Macrophages/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic
14.
J Pathol ; 223(1): 37-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20845411

ABSTRACT

Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase (NOS) inhibitor, is accumulated in plasma during chronic kidney disease (CKD). It is considered an independent mortality and cardiovascular risk factor in CKD patients. To test the involvement of ADMA in CKD progression, we investigated the effects of chronic ADMA administration on renal structure and compared these effects with NG-nitro-L-arginine methyl ester (L-NAME) treatment, a widely used exogenous inhibitor of NOS that induces CKD. Three groups of uninephrectomized mice were studied: ADMA (60 mg/kg per day), L-NAME (60 mg/kg per day), and isotonic saline (control) were infused through osmotic mini-pumps for 8 weeks. ADMA and L-NAME induced hypertension (PAS 167 ± 16 and 168 ± 10 versus 100 ± 4 mmHg, p < 0.01, respectively). High level of ADMA was associated with increased renal oxidative stress. ADMA treatment induced glomerular and vascular fibrosis as evidenced by the elevated deposits of collagen I, III, and fibronectin (p < 0.01). A similar profile was observed in the L-NAME group. Mice treated with ADMA had reduced peritubular capillaries versus controls (p < 0.01). Collagen I mRNA expression and renal TGF-ß1 concentrations were higher in the ADMA and L-NAME groups. Increased level of TGF-ß1 was associated with a significant rise of HIF-1α and endothelin-1 expression. These results demonstrate for the first time that elevated concentrations of ADMA are associated with the development of renal fibrosis. These data suggest that in pathophysiological conditions of endothelial dysfunction, the exaggerated endogenous synthesis of ADMA could contribute to CKD progression by favouring hypertension, extracellular matrix synthesis, and rarefaction of peritubular capillaries.


Subject(s)
Arginine/analogs & derivatives , Collagen/biosynthesis , Enzyme Inhibitors/toxicity , Renal Insufficiency, Chronic/chemically induced , Transforming Growth Factor beta1/biosynthesis , Animals , Arginine/blood , Arginine/toxicity , Collagen/genetics , Endothelin-1/biosynthesis , Endothelin-1/genetics , Enzyme Inhibitors/blood , Fibrosis , Gene Expression Regulation/drug effects , Hypertension/chemically induced , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney/pathology , Male , Mice , Mice, Inbred Strains , NG-Nitroarginine Methyl Ester/toxicity , Nitric Oxide Synthase/antagonists & inhibitors , Oxidative Stress/drug effects , RNA, Messenger/genetics , Renal Insufficiency, Chronic/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transforming Growth Factor beta1/genetics
15.
Rev Prat ; 62(1): 72-5, 2012 Jan.
Article in French | MEDLINE | ID: mdl-22335071

ABSTRACT

Despite the use of angiotensin blockers, chronic kidney diseases still progress. New therapeutic approaches aim to strengthen and to complete angiotensin blocker effects. Endothelin receptor antagonists, in addition to angiotensin blockers reduce blood pressure and urinary albumin excretion in diabetic nephropathies but can induce fluid overload. A second therapeutic approach consists in preventing the development of interstitial renal fibrosis which is a prognostic factor of CKD. Transforming growth factor-beta (TGF-beta) plays a major role in this process. Several molecules such as pirfenidone, microARN are in development to block TGF-beta or its downstream signaling pathways. Another approach aims to promote resolution of inflammation and renal repair Interesting experimental results were obtained with tyrosine kinase inhibitors and with methyl of bardoxolone in humans.


Subject(s)
Renal Insufficiency, Chronic/drug therapy , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Disease Progression , Drug Therapy, Combination , Endothelin Receptor Antagonists , Humans , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/therapeutic use , Prognosis , Protein Kinase Inhibitors/therapeutic use , Renal Insufficiency, Chronic/diagnosis , Treatment Outcome
16.
Am J Physiol Renal Physiol ; 301(1): F24-32, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21429966

ABSTRACT

Chronic kidney disease is promoted by a variety of factors that induce chronic inflammation and fibrosis. Inflammation and excessive scaring have been recently associated with disruptions of the gap junction-mediated intercellular communication. Nevertheless, little is known about alterations of the expression of gap junction proteins such as connexin (Cx) 43 and 37 in chronic renal disease. In this study, we investigated the expression of these two Cxs in the hypertensive RenTg mice, the anti-glomerular basement membrane glomerulonephritis, and the unilateral ureteral obstruction models, all leading to the development of chronic kidney disease in mice. Expression of Cx43 was almost negligible in the renal cortex of control mice. In contrast, Cx43 was markedly increased in the endothelium of peritubular and glomerular capillaries of the 3-mo-old RenTg mice, in the glomeruli of mice suffering from glomerulonephritis, and in the tubules after obstructive nephropathy. The Cx43 expression pattern was paralleled closely by that of the adhesion markers such as vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 as well as the inflammatory biomarker monocyte chemoattractant protein-1. In contrast, Cx37 that was abundantly expressed in the renal cortex of healthy mice was markedly decreased in the three experimental models. Interestingly, Cx43+/- mice showed restricted expression of VCAM-1 after 2 wk of obstructive nephropathy. These findings suggest the importance of Cxs as markers of chronic renal disease and indicate that these proteins may participate in the inflammatory process during the development of this pathology.


Subject(s)
Connexins/biosynthesis , Kidney Diseases/metabolism , Animals , Anti-Glomerular Basement Membrane Disease/genetics , Anti-Glomerular Basement Membrane Disease/metabolism , Biomarkers , Blood Pressure/physiology , Blood Urea Nitrogen , Cell Adhesion Molecules/biosynthesis , Chemokine CCL2/biosynthesis , Chronic Disease , Connexin 43/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Inflammation/pathology , Liver/metabolism , Mice , Mice, Knockout , Proteinuria/genetics , Proteinuria/metabolism , RNA/biosynthesis , RNA/isolation & purification , Renin/biosynthesis , Renin/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
J Clin Invest ; 118(5): 1924-33, 2008 May.
Article in English | MEDLINE | ID: mdl-18382768

ABSTRACT

Patients with sickle-cell disease (SCD) suffer from tissue damage and life-threatening complications caused by vasoocclusive crisis (VOC). Endothelin receptors (ETRs) are mediators of one of the most potent vasoconstrictor pathways in mammals, but the relationship between vasoconstriction and VOC is not well understood. We report here that pharmacological inhibition of ETRs prevented hypoxia-induced acute VOC and organ damage in a mouse model of SCD. An in vivo ultrasonographic study of renal hemodynamics showed a substantial increase in endothelin-mediated vascular resistance during hypoxia/reoxygenation-induced VOC. This increase was reversed by administration of the dual ETR antagonist (ETRA) bosentan, which had pleiotropic beneficial effects in vivo. It prevented renal and pulmonary microvascular congestion, systemic inflammation, dense rbc formation, and infiltration of activated neutrophils into tissues with subsequent nitrative stress. Bosentan also prevented death of sickle-cell mice exposed to a severe hypoxic challenge. These findings in mice suggest that ETRA could be a potential new therapy for SCD, as it may prevent acute VOC and limit organ damage in sickle-cell patients.


Subject(s)
Anemia, Sickle Cell , Antihypertensive Agents/therapeutic use , Endothelin Receptor Antagonists , Hypoxia , Receptors, Endothelin/metabolism , Sulfonamides/therapeutic use , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/mortality , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/physiopathology , Animals , Bosentan , Disease Models, Animal , Endothelin-1/genetics , Endothelin-1/metabolism , Hemodynamics , Humans , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Kidney/physiology , Lung/cytology , Lung/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Receptors, Endothelin/genetics , Regional Blood Flow , Renal Circulation/physiology , Vasoconstriction/physiology
18.
Int J Exp Pathol ; 92(3): 151-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21314743

ABSTRACT

The need for novel insights into the mechanisms of progression of renal disease has become urgent during the last several years because of the increasing incidence of chronic renal disease worldwide. Independent of the underlying disease, the subsequent progression of renal fibrosis is characterized mainly by both an exaggerated synthesis and abnormal accumulation of extracellular matrix proteins produced by mesenchymal cells within the kidney. These cells are mainly myofibroblasts deriving from a variety of renal cells such as vascular smooth muscle, mesangial, resident stem, tubular epithelial, vascular endothelial cells or pericytes. The appearance of myofibroblasts is a reversible process, as suggested by studies in experimental models showing regression of renal fibrosis during therapy with antagonists and/or blockers of the renin-angiotensin system. An additional factor that can also affect the mechanisms of progression/regression of fibrosis is the plasticity of podocytes controlling glomerular filtration.


Subject(s)
Kidney Glomerulus/pathology , Kidney Tubules/pathology , Kidney/pathology , Muscle, Smooth, Vascular/pathology , Animals , Disease Models, Animal , Disease Progression , Fibrosis , Humans , Kidney/physiopathology , Kidney Glomerulus/physiopathology , Kidney Tubules/physiopathology , Mesoderm/pathology , Mesoderm/physiopathology , Mice , Muscle, Smooth, Vascular/physiopathology
19.
Front Physiol ; 12: 624052, 2021.
Article in English | MEDLINE | ID: mdl-33692701

ABSTRACT

Since December 2019, the coronavirus 2019 (COVID-19) pandemic has rapidly spread and overwhelmed healthcare systems worldwide, urging physicians to understand how to manage this novel infection. Early in the pandemic, more severe forms of COVID-19 have been observed in patients with cardiovascular comorbidities, who are often treated with renin-angiotensin aldosterone system (RAAS)-blockers, such as angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs), but whether these are indeed independent risk factors is unknown. The cellular receptor for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the membrane-bound angiotensin converting enzyme 2 (ACE2), as for SARS-CoV(-1). Experimental data suggest that expression of ACE2 may be increased by RAAS-blockers, raising concerns that these drugs may facilitate viral cell entry. On the other hand, ACE2 is a key counter-regulator of the RAAS, by degrading angiotensin II into angiotensin (1-7), and may thereby mediate beneficial effects in COVID-19. These considerations have raised concerns about the management of these drugs, and early comments shed vivid controversy among physicians. This review will describe the homeostatic balance between ACE-angiotensin II and ACE2-angiotensin (1-7) and summarize the pathophysiological rationale underlying the debated role of the RAAS and its modulators in the context of the pandemic. In addition, we will review available evidence investigating the impact of RAAS blockers on the course and prognosis of COVID-19 and discuss why retrospective observational studies should be interpreted with caution. These considerations highlight the importance of solid evidence-based data in order to guide physicians in the management of RAAS-interfering drugs in the general population as well as in patients with more or less severe forms of SARS-CoV-2 infection.

20.
Am J Physiol Renal Physiol ; 297(1): F191-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19420116

ABSTRACT

The present study investigated mechanisms of regression of renal disease after severe proteinuria by focusing on the interaction among EGF receptors, renal hemodynamics, and structural lesions. The nitric oxide (NO) inhibitor N(G)-nitro-l-arginine-methyl ester (l-NAME) was administered chronically in Sprague-Dawley rats. When proteinuria exceeded 2 g/mmol creatinine, animals were divided into three groups for an experimental period of therapy of 2 wk; in one group, l-NAME was removed to allow reactivation of endogenous NO synthesis; in the two other groups, l-NAME removal was combined with EGF or angiotensin receptor type 1 (AT(1)) antagonism. l-NAME removal partially reduced mean arterial pressure and proteinuria and increased renal blood flow (RBF), but not microvascular hypertrophy. Progression of structural damage was stopped, but not reversed. The administration of an EGF receptor antagonist did not have an additional effect on lowering blood pressure or on renal inflammation but did normalize RBF and afferent arteriole hypertrophy; the administration of an AT(1) antagonist normalized all measured functional and structural parameters. Staining with a specific marker of endothelial integrity indicated loss of functional endothelial cells in the l-NAME removal group; in contrast, in the animals treated with an EGF or AT(1) receptor antagonist, functional endothelial cells reappeared at levels equal to control animals. In addition, afferent arterioles freshly isolated from the l-NAME removal group showed an exaggerated constrictor response to endothelin; this response was blunted in the vessels isolated from the EGF or AT(1) receptor antagonist groups. The EGF receptor is an important mediator of endothelial dysfunction and contributes to the decline of RBF in the chronic kidney disease induced by NO deficiency. The EGF receptor antagonist-induced improvement of RBF is important but not sufficient for a complete reversal of renal disease, because it has little effect on renal inflammation. To achieve full recovery, it is necessary to apply AT(1) receptor antagonism.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Hypertension/complications , Hypertension/physiopathology , Kidney Diseases/etiology , Kidney Diseases/physiopathology , Kidney/blood supply , Regional Blood Flow/physiology , Animals , Arterioles/drug effects , Arterioles/pathology , Arterioles/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Chronic Disease , Disease Models, Animal , Enzyme Inhibitors/pharmacology , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Gefitinib , Kidney Diseases/pathology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Vasoconstriction/drug effects , Vasoconstriction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL