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

3.
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
4.
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
5.
Nephron ; 137(3): 212-220, 2017.
Article in English | MEDLINE | ID: mdl-28743124

ABSTRACT

Discoidin domain receptors (DDRs) are a family of 2 non-integrin collagen receptors, DDR1 and DDR2, which display a tyrosine kinase activity. They are mainly expressed during embryonic development and their role during adulthood is very limited. DDR1 has been widely studied in several types of cancers, in atherosclerosis and fibrosis, but also in chronic kidney disease (CKD). This review focuses on the role of DDR1 in chronic nephropathies and on the effect of its deletion in the pathological processes involved in renal disease progression. DDR1 was shown to be de novo expressed in several models of experimental CKD. Its genetic or pharmaco-genetic inhibition led to the preservation of renal structure and function, and to decreased inflammatory influx and fibrosis. Furthermore, delayed pharmaco-genetic inhibition of DDR1 led to significant protection in models of renal disease. These results demonstrate the involvement of DDR1 in inflammatory and fibrotic processes occurring during CKD and the beneficial effect of its inhibition. Thus, DDR1 could be an interesting therapeutic target to treat renal pathologies.


Subject(s)
Discoidin Domain Receptor 1/physiology , Fibrosis/physiopathology , Inflammation/physiopathology , Kidney Diseases/physiopathology , Animals , Discoidin Domain Receptor 1/genetics , Humans , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/physiopathology , Neoplasms/genetics , Neoplasms/physiopathology
6.
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
7.
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
8.
Physiol Rep ; 4(11)2016 Jun.
Article in English | MEDLINE | ID: mdl-27255359

ABSTRACT

Low-energy extracorporeal shock wave therapy (SWT) has been shown to improve myocardial dysfunction, hind limb ischemia, erectile function, and to facilitate cell therapy and healing process. These therapeutic effects were mainly due to promoting angiogenesis. Since chronic kidney diseases are characterized by renal fibrosis and capillaries rarefaction, they may benefit from a proangiogenic treatment. The objective of our study was to determine whether SWT could ameliorate renal repair and favor angiogenesis in L-NAME-induced hypertensive nephropathy in rats. SWT was started when proteinuria exceeded 1 g/mmol of creatinine and 1 week after L-NAME removal. SWT consisted of implying 0.09 mJ/mm(2) (400 shots), 3 times per week. After 4 weeks of SWT, blood pressure, renal function and urinary protein excretion did not differ between treated (LN + SWT) and untreated rats (LN). Histological lesions including glomerulosclerosis and arteriolosclerosis scores, tubular dilatation and interstitial fibrosis were similar in both groups. In addition, peritubular capillaries and eNOS, VEGF, VEGF-R, SDF-1 gene expressions did not increase in SWT-treated compared to untreated animals. No procedural complications or adverse effects were observed in control (C + SWT) and hypertensive rats (LN + SWT). These results suggest that extracorporeal kidney shock wave therapy does not induce angiogenesis and does not improve renal function and structure, at least in the model of hypertensive nephropathy although the treatment is well tolerated.


Subject(s)
Hypertension, Renal/therapy , Kidney/pathology , Lithotripsy , Nephritis/therapy , Animals , Disease Models, Animal , Hypertension, Renal/chemically induced , Hypertension, Renal/pathology , Male , NG-Nitroarginine Methyl Ester , Nephritis/chemically induced , Nephritis/pathology , Rats , Treatment Outcome
9.
Sci Rep ; 6: 21262, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26880216

ABSTRACT

Chronic kidney disease is a progressive incurable pathology affecting millions of people. Intensive investigations aim to identify targets for therapy. We have previously demonstrated that abnormal expression of the Discoidin Domain Receptor 1 (DDR1) is a key factor of renal disease by promoting inflammation and fibrosis. The present study investigates whether blocking the expression of DDR1 after the initiation of renal disease can delay or arrest the progression of this pathology. Severe renal disease was induced by either injecting nephrotoxic serum (NTS) or performing unilateral ureteral obstruction in mice, and the expression of DDR1 was inhibited by administering antisense oligodeoxynucleotides either at 4 or 8 days after NTS (corresponding to early or more established phases of disease, respectively), or at day 2 after ligation. DDR1 antisense administration at day 4 stopped the increase of proteinuria and protected animals against the progression of glomeruloneprhitis, as evidenced by functional, structural and cellular indexes. Antisense administration at day 8 delayed progression -but to a smaller degree- of renal disease. Similar beneficial effects on renal structure and inflammation were observed with the antisense administration of DDR1 after ureteral ligation. Thus, targeting DDR1 can be a promising strategy in the treatment of chronic kidney disease.


Subject(s)
Discoidin Domain Receptor 1/genetics , Kidney Diseases/genetics , Animals , Cytokines/metabolism , DNA, Antisense/administration & dosage , Discoidin Domain Receptor 1/metabolism , Disease Models, Animal , Disease Progression , Fibrosis , Gene Expression , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Mice
10.
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
11.
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
12.
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
13.
Article in English | MEDLINE | ID: mdl-24720461

ABSTRACT

The renal endothelium plays a critical role in kidney physiopathology as it is implicated in various processes such as the regulation of vasomotor tone, the control of tissue inflammation and thrombosis. Recent evidence highlights direct implication of renal endothelial dysfunction in the progression of chronic kidney disease. Renal endothelial dysfunction is a multifaceted process in which chemokines, cytokines, prothrombotic factors and adhesion molecules are known to play a crucial role. Apart from paracrine cell-to-cell signaling, the role for gap junction-mediated intercellular communication in renal physiopathology has been recently suggested. Gap junction channels are formed by the hexameric assembly of connexins and directly connect the cytoplasm of adjacent cells. Due to their ability to regulate multiple physiological and pathological signals connexins are currently taking an important place in the list of actors involved in renal endothelial function and dysfunction. In this review we will focus on possible implications of connexins in the physiopathological processes associated with renal vascular endothelium.


Subject(s)
Connexins/metabolism , Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Kidney/blood supply , Kidney/physiopathology , Animals , Connexins/immunology , Gap Junctions/immunology , Gap Junctions/metabolism , Gap Junctions/pathology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/physiopathology , Kidney/physiology , Oxidative Stress
14.
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
15.
Kidney Int Suppl (2011) ; 4(1): 53-57, 2014 Nov.
Article in English | MEDLINE | ID: mdl-26312150

ABSTRACT

Over the last decade, identification and characterization of novel markers of progression and targets for therapy of chronic kidney disease (CKD) have been challenging for the research community. Several promising candidates have emerged, mainly from experimental models of CKD that are yet to be investigated in clinical studies. The authors identified two candidate genes: periostin, an extracellular matrix protein involved in bone and dental development, and the discoidin domain receptor 1 (DDR1), a collagen-binding membrane receptor with tyrosine kinase activity. Both genes are inactive in adulthood under normal conditions but have been shown to be highly inducible following injury to glomerular or tubular epithelial cells. The objective of this review is to summarize recent evidence supporting the role of periostin and DDR1 as potential novel biomarkers and therapeutic targets in CKD.

16.
Hypertension ; 61(4): 901-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23438929

ABSTRACT

Aliskiren, a direct renin inhibitor, is a novel antihypertensive drug. To study whether aliskiren can reverse chronic kidney disease, we administered it to renin transgenic mice, a strain characterized by elevated blood pressure and a slow decline of renal function, mimicking well the progression of hypertensive chronic kidney disease. Ten-month-old transgenic mice were treated either with aliskiren or placebo for 28 days. Age-matched wild-type mice treated or not with aliskiren were considered as normotensive controls. Aliskiren reduced blood pressure to wild-type levels from as early as day 14. Proteinuria and cardiac hypertrophy and fibrosis were also normalized. Renal interstitial fibrosis and inflammation were significantly ameliorated in aliskiren-treated mice (shown by the decrease of proinflammatory and profibrotic markers), and the phenotypes of tubular epithelial cells and podocytes were restored as evidenced by the reappearance of cellular proteins characteristic of normal phenotype of these cells. Profibrotic p38 and Erk mitogen-activated protein kinases were highly activated in placebo-treated transgenic animals. Aliskiren treatment cancelled this activation. This nephroprotection was not attributed to the antihypertensive activity of aliskiren, because blood pressure normalization after treatment with hydralazine failed to induce the regression of renal fibrosis. Direct inhibition of renin can restore renal function and structure in aged hypertensive animals with existing proteinuria. This finding suggests that, in addition to antihypertensive action, aliskiren can be also used to treat chronic kidney disease.


Subject(s)
Amides/pharmacology , Fumarates/pharmacology , Hypertension/drug therapy , Kidney Diseases/drug therapy , Kidney/pathology , Renin/antagonists & inhibitors , Animals , Disease Models, Animal , Fibrosis/pathology , Fibrosis/prevention & control , Hypertension/complications , Hypertension/pathology , Kidney/metabolism , Kidney Diseases/complications , Kidney Diseases/pathology , Mice , Mice, Transgenic
17.
Fibrogenesis Tissue Repair ; 5(Suppl 1): S15, 2012.
Article in English | MEDLINE | ID: mdl-23259724

ABSTRACT

The vasculature of the kidney is a heterogeneous structure, whose functional integrity is essential for the regulation of renal function. Owing to the importance of the endothelium in vascular biology, chronic endothelial alterations are therefore susceptible to impair multiple aspects of renal physiology and, in turn, to contribute to renal fibrosis. Although systemic endothelial dysfunction is undoubtedly associated with chronic kidney disease, the role of the renal endothelium in the initiation and the progression of renal fibrosis remains largely elusive. In this article, we critically review recent evidence supporting direct and indirect contributions of renal endothelial alterations to fibrosis in the kidney. Specifically, the potential implications of renal endothelial dysfunction and endothelial paucity in parenchymal hypoxia, in the regulation of local inflammation, and in the generation of renal mesenchymal cells are reviewed. We thereafter discuss therapeutic perspectives targeting renal endothelial alterations during the initiation and the progression of renal fibrogenesis.

18.
Neurology ; 79(18): 1898-907, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23077007

ABSTRACT

OBJECTIVE: Fragile X-associated tremor ataxia syndrome (FXTAS) is defined by FMR1 premutation, cerebellar ataxia, intentional tremor, and middle cerebellar peduncle (MCP) hyperintensities. We delineate the clinical, neurophysiologic, and morphologic characteristics of FXTAS. METHODS: Clinical, morphologic (brain MRI, (123)I-ioflupane SPECT), and neurophysiologic (tremor recording, nerve conduction studies) study in 22 patients with FXTAS, including 4 women. RESULTS: A total of 43% of patients had no family history of fragile X syndrome (FXS), which contrasts with previous FXTAS series. A total of 86% of patients had tremor and 81% peripheral neuropathy. We identified 3 electroclinical tremor patterns: essential-like (35%), cerebellar (29%), and parkinsonian (12%). Two electrophysiologic patterns evocative of non-length-dependent (56%) and length-dependent sensory neuropathy (25%) were identified. Corpus callosum splenium (CCS) hyperintensity was as frequent (68%) as MCP hyperintensities (64%). Sixty percent of patients had parkinsonism and 47% abnormal (123)I-ioflupane SPECT. Unified Parkinson's Disease Rating Scale motor score was correlated to abnormal (123)I-ioflupane SPECT (p = 0.02) and to CGG repeat number (p = 0.0004). Scale for the assessment and rating of ataxia correlated with dentate nuclei hyperintensities (p = 0.03) and CCS hyperintensity was a marker of severe disease progression (p = 0.04). CONCLUSIONS: We recommend to include in the FXTAS testing guidelines both CCS hyperintensity and peripheral neuropathy and to consider them as new major radiologic and minor clinical criterion, respectively, for the diagnosis of FXTAS. FXTAS should also be considered in women or when tremor, MCP hyperintensities, or family history of FXS are lacking. Our study broadens the spectrum of tremor, peripheral neuropathy, and MRI abnormalities in FXTAS, hence revealing the need for revised criteria.


Subject(s)
Ataxia/diagnosis , Fragile X Syndrome/diagnosis , Parkinsonian Disorders/diagnosis , Peripheral Nervous System Diseases/diagnosis , Tremor/diagnosis , Adult , Aged , Ataxia/genetics , Ataxia/physiopathology , Female , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Humans , Male , Middle Aged , Neurology/standards , Parkinsonian Disorders/genetics , Parkinsonian Disorders/physiopathology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/physiopathology , Practice Guidelines as Topic/standards , Tremor/genetics , Tremor/physiopathology
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...