Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Article in English | MEDLINE | ID: mdl-32900697

ABSTRACT

INTRODUCTION: Diabetic nephropathy (DN) is the leading cause of chronic kidney disease worldwide. The Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway participates in the development and progression of DN. Among the different mechanisms involved in JAK/STAT negative regulation, the family of suppressor of cytokine signaling (SOCS) proteins has been proposed as a new target for DN. Our aim was to evaluate the effect of SOCS1 mimetic peptide in a mouse model of obesity and type 2 diabetes (T2D) with progressive DN. RESEARCH DESIGN AND METHODS: Six-week-old BTBR (black and tan brachyuric) mice with the ob/ob (obese/obese) leptin-deficiency mutation were treated for 7 weeks with two different doses of active SOCS1 peptide (MiS1 2 and 4 µg/g body weight), using inactive mutant peptide (Mut 4 µg) and vehicle as control groups. At the end of the study, the animals were sacrificed to obtain blood, urine and kidney tissue for further analysis. RESULTS: Treatment of diabetic mice with active peptide significantly decreased urine albumin to creatinine ratio by up to 50%, reduced renal weight, glomerular and tubulointerstitial damage, and restored podocyte numbers. Kidneys from treated mice exhibited lower inflammatory infiltrate, proinflammatory gene expression and STAT activation. Concomitantly, active peptide administration modulated redox balance markers and reduced lipid peroxidation and cholesterol transporter gene expression in diabetic kidneys. CONCLUSION: Targeting SOCS proteins by mimetic peptides to control JAK/STAT signaling pathway ameliorates albuminuria, morphological renal lesions, inflammation, oxidative stress and lipotoxicity, and could be a therapeutic approach to T2D kidney disease.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Animals , Anti-Inflammatory Agents , Antioxidants/pharmacology , Antioxidants/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Mice , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling Proteins
2.
Int J Mol Sci ; 21(12)2020 Jun 13.
Article in English | MEDLINE | ID: mdl-32545818

ABSTRACT

Diabetic nephropathy (DN) is a multifactorial disease characterized by hyperglycemia and close interaction of hemodynamic, metabolic and inflammatory factors. Nuclear factor-κB (NF-κB) is a principal matchmaker linking hyperglycemia and inflammation. The present work investigates the cell-permeable peptide containing the inhibitor of kappa B kinase γ (IKKγ)/NF-κB essential modulator (NEMO)-binding domain (NBD) as therapeutic option to modulate inflammation in a preclinical model of type 2 diabetes (T2D) with DN. Black and tan, brachyuric obese/obese mice were randomized into 4 interventions groups: Active NBD peptide (10 and 6 µg/g body weight); Inactive mutant peptide (10 µg/g); and vehicle control. In vivo/ex vivo fluorescence imaging revealed efficient delivery of NBD peptide, systemic biodistribution and selective renal metabolization. In vivo administration of active NBD peptide improved albuminuria (>40% reduction on average) and kidney damage, decreased podocyte loss and basement membrane thickness, and modulated the expression of proinflammatory and oxidative stress markers. In vitro, NBD blocked IKK-mediated NF-κB induction and target gene expression in mesangial cells exposed to diabetic-like milieu. These results constitute the first nephroprotective effect of NBD peptide in a T2D mouse model that recapitulates the kidney lesions observed in DN patients. Targeting IKK-dependent NF-κB activation could be a therapeutic strategy to combat kidney inflammation in DN.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/drug therapy , Intracellular Signaling Peptides and Proteins/chemistry , Serum Albumin/drug effects , Signal Transduction/drug effects , Animals , Binding Sites , Cell Line , Cell-Penetrating Peptides/pharmacology , Diabetes Mellitus, Type 2/diagnostic imaging , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/diagnostic imaging , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , NF-kappa B/metabolism , RAW 264.7 Cells , Random Allocation , Tissue Distribution , Treatment Outcome
3.
J Clin Med ; 9(1)2020 Jan 19.
Article in English | MEDLINE | ID: mdl-31963845

ABSTRACT

Chronic kidney disease has become a major medical issue in recent years due to its high prevalence worldwide, its association with premature mortality, and its social and economic implications. A number of patients gradually progress to end-stage renal disease (ESRD), requiring then dialysis and kidney transplantation. Currently, approximately 40% of patients with diabetes develop kidney disease, making it the most prevalent cause of ESRD. Thus, more effective therapies for diabetic nephropathy are needed. In preclinical studies of diabetes, anti-inflammatory therapeutic strategies have been used to protect the kidneys. Recent evidence supports that immune cells play an active role in the pathogenesis of diabetic nephropathy. Th17 immune cells and their effector cytokine IL-17A have recently emerged as promising targets in several clinical conditions, including renal diseases. Here, we review current knowledge regarding the involvement of Th17/IL-17A in the genesis of diabetic renal injury, as well as the rationale behind targeting IL-17A as an additional therapy in patients with diabetic nephropathy.

4.
Sci Rep ; 9(1): 6867, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31053735

ABSTRACT

Gremlin renal overexpression has been reported in diabetic nephropathy, pauci-immune crescentic glomerulonephritis and chronic allograft nephropathy and has been implicated in the pathophysiology of the progression of renal damage. However, it is unknown whether urinary Gremlin can be associated with renal functional status, renal biopsy findings and outcome. To examine these associations we studied 20 patients with ANCA+ renal vasculitis and very high urinary Gremlin (354 ± 76 ug/gCr), 86 patients with other glomerular diseases and moderately elevated urinary Gremlin (83 ± 14 ug/gCr) and 11 healthy controls (urinary Gremlin 11.3 ± 2.4 ug/gCr). Urinary Gremlin was significantly correlated with renal expression of Gremlin (r = 0.64, p = 0.013) observed in cellular glomerular crescents, tubular epithelial cells and interstitial inflammatory cells. Moreover, urinary Gremlin levels were correlated with the number of glomerular crescents (r = 0.53; p < 0.001), renal CD68 positive cells (r = 0.71; p < 0.005), tubulointerstitial fibrosis (r = 0.50; p < 0.05), and serum creatinine levels (r = 0.60; p < 0.001). Interestingly, Gremlin expression was colocalized with CD68, CD163 (monocyte/macrophage markers) and CCL18 positive cells. ROC curve analysis showed that the cutoff value of urinary Gremlin in glomerular diseases as 43 ug/gCr with 72% of sensitivity and 100% of specificity [AUC: 0.96 (CI 95% 0.92-0.99] (p < 0.001). For ANCA+ renal vasculitis the value of urinary Gremlin of 241 ug/gCr had 55% of sensitivity and 100% of specificity [AUC: 0.81 (CI 95% 0.68-0.94) (p < 0.001]. Based on these results we propose that urinary Gremlin represents a non-invasive biomarker in ANCA+ renal vasculitis, and suggest a role of Gremlin in the formation of crescents.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Glomerulonephritis/pathology , Glomerulonephritis/urine , Intercellular Signaling Peptides and Proteins/urine , Kidney Glomerulus/pathology , Adult , Aged , Aged, 80 and over , Biomarkers/urine , Case-Control Studies , Female , Glomerulonephritis/immunology , Humans , Male , Middle Aged
5.
Kidney Int ; 95(6): 1418-1432, 2019 06.
Article in English | MEDLINE | ID: mdl-30982673

ABSTRACT

Diabetic nephropathy (DN) is one of the most common complications of diabetes, and currently the first end-stage renal disease worldwide. New strategies to treat DN using agents that target inflammatory pathways have attracted special interest. Recent pieces of evidences suggest a promising effect of IL-17A, the Th17 effector cytokine. Among experimental DN models, mouse strain BTBR ob/ob (leptin deficiency mutation) develops histological features similar to human DN, which means an opportunity to study mechanisms and novel therapies aimed at DN regression. We found that BTBR ob/ob mice presented renal activation of the factors controlling Th17 differentiation. The presence of IL-17A-expressing cells, mainly CD4+ and γδ lymphocytes, was associated with upregulation of proinflammatory factors, macrophage infiltration and the beginning of renal damage. To study IL-17A involvement in experimental DN pathogenesis, treatment with an IL-17A neutralizing antibody was carried out starting when the renal damage had already appeared. IL-17A blockade ameliorated renal dysfunction and disease progression in BTBR ob/ob mice. These beneficial effects correlated to podocyte number restoration and inhibition of NF-κB/proinflammatory factors linked to a decrease in renal inflammatory-cell infiltration. These data demonstrate that IL-17A takes part in diabetes-mediated renal damage and could be a promising therapeutic target to improve DN.


Subject(s)
Albuminuria/drug therapy , Antibodies, Neutralizing/administration & dosage , Diabetic Nephropathies/drug therapy , Interleukin-17/antagonists & inhibitors , Albuminuria/genetics , Albuminuria/immunology , Albuminuria/pathology , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Diabetic Nephropathies/urine , Disease Progression , Humans , Interleukin-17/immunology , Interleukin-17/metabolism , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Leptin/genetics , Male , Mice , Mice, Transgenic , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
6.
Nephrol Dial Transplant ; 33(5): 735-741, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28992340

ABSTRACT

The current therapeutic strategy for the treatment of chronic kidney diseases only ameliorates disease progression. During renal injury, developmental genes are re-expressed and could be potential therapeutic targets. Among those genes reactivated in the adult damaged kidney, Gremlin is of particular relevance since recent data suggest that it could be a mediator of diabetic nephropathy and other progressive renal diseases. Earlier studies have shown that Gremlin is upregulated in trans-differentiated renal proximal tubular cells and in several chronic kidney diseases associated with fibrosis. However, not much was known about the mechanisms by which Gremlin acts in renal pathophysiology. The role of Gremlin as a bone morphogenetic protein antagonist has clearly been demonstrated in organogenesis and in fibrotic-related disorders. Gremlin binds to vascular endothelial growth factor receptor 2 (VEGFR2) in endothelial and tubular epithelial cells. Activation of the Gremlin-VEGFR2 axis was found in several human nephropathies. We have recently described that Gremlin activates the VEGFR2 signaling pathway in the kidney, eliciting a downstream mechanism linked to renal inflammatory response. Gremlin deletion improves experimental renal damage, diminishing fibrosis. Overall, the available data identify the Gremlin-VEGFR2 axis as a novel therapeutic target for kidney inflammation and fibrosis and provide a rationale for unveiling new concepts to investigate in several clinical conditions.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Kidney Diseases/pathology , Kidney Diseases/therapy , Vascular Endothelial Growth Factor A/metabolism , Humans , Kidney Diseases/metabolism , Signal Transduction
7.
Am J Physiol Renal Physiol ; 309(6): F559-68, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26155842

ABSTRACT

Diabetic nephropathy (DN) is currently a leading cause of end-stage renal failure worldwide. Gremlin was identified as a gene differentially expressed in mesangial cells exposed to high glucose and in experimental diabetic kidneys. We have described that Gremlin is highly expressed in biopsies from patients with diabetic nephropathy, predominantly in areas of tubulointerstitial fibrosis. In streptozotocin (STZ)-induced experimental diabetes, Gremlin deletion using Grem1 heterozygous knockout mice or by gene silencing, ameliorates renal damage. To study the in vivo role of Gremlin in renal damage, we developed a diabetic model induced by STZ in transgenic (TG) mice expressing human Gremlin in proximal tubular epithelial cells. The albuminuria/creatinuria ratio, determined at week 20 after treatment, was significantly increased in diabetic mice but with no significant differences between transgenic (TG/STZ) and wild-type mice (WT/STZ). To assess the level of renal damage, kidney tissue was analyzed by light microscopy (periodic acid-Schiff and Masson staining), electron microscopy, and quantitative PCR. TG/STZ mice had significantly greater thickening of the glomerular basement membrane, increased mesangial matrix, and podocytopenia vs. WT/STZ. At the tubulointerstitial level, TG/STZ showed increased cell infiltration and mild interstitial fibrosis. In addition, we observed a decreased expression of podocin and overexpression of monocyte chemoattractant protein-1 and fibrotic-related markers, including transforming growth factor-ß1, Col1a1, and α-smooth muscle actin. Together, these results show that TG mice overexpressing Gremlin in renal tubules develop greater glomerular and tubulointerstitial injury in response to diabetic-mediated damage and support the involvement of Gremlin in diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Kidney Tubules/metabolism , Kidney Tubules/pathology , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Fibrosis/genetics , Fibrosis/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Kidney Glomerulus/pathology , Mice, Inbred C57BL , Mice, Transgenic , Nephritis, Interstitial/pathology , Podocytes/pathology
8.
PLoS One ; 9(7): e101879, 2014.
Article in English | MEDLINE | ID: mdl-25036148

ABSTRACT

A growing number of patients are recognized worldwide to have chronic kidney disease. Glomerular and interstitial fibrosis are hallmarks of renal progression. However, fibrosis of the kidney remains an unresolved challenge, and its molecular mechanisms are still not fully understood. Gremlin is an embryogenic gene that has been shown to play a key role in nephrogenesis, and its expression is generally low in the normal adult kidney. However, gremlin expression is elevated in many human renal diseases, including diabetic nephropathy, pauci-immune glomerulonephritis and chronic allograft nephropathy. Several studies have proposed that gremlin may be involved in renal damage by acting as a downstream mediator of TGF-ß. To examine the in vivo role of gremlin in kidney pathophysiology, we generated seven viable transgenic mouse lines expressing human gremlin (GREM1) specifically in renal proximal tubular epithelial cells under the control of an androgen-regulated promoter. These lines demonstrated 1.2- to 200-fold increased GREM1 expression. GREM1 transgenic mice presented a normal phenotype and were without proteinuria and renal function involvement. In response to the acute renal damage cause by folic acid nephrotoxicity, tubule-specific GREM1 transgenic mice developed increased proteinuria after 7 and 14 days compared with wild-type treated mice. At 14 days tubular lesions, such as dilatation, epithelium flattening and hyaline casts, with interstitial cell infiltration and mild fibrosis were significantly more prominent in transgenic mice than wild-type mice. Tubular GREM1 overexpression was correlated with the renal upregulation of profibrotic factors, such as TGF-ß and αSMA, and with increased numbers of monocytes/macrophages and lymphocytes compared to wild-type mice. Taken together, our results suggest that GREM1-overexpressing mice have an increased susceptibility to renal damage, supporting the involvement of gremlin in renal damage progression. This transgenic mouse model could be used as a new tool for enhancing the knowledge of renal disease progression.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Kidney Tubules/injuries , Kidney Tubules/metabolism , Animals , Cell Line , Disease Susceptibility , Folic Acid/adverse effects , Gene Expression , Humans , Kidney Tubules/drug effects , Mice , Mice, Transgenic , Phenotype
9.
Biomed Res Int ; 2014: 802841, 2014.
Article in English | MEDLINE | ID: mdl-24949470

ABSTRACT

Gremlin is a developmental gene upregulated in human chronic kidney disease and in renal cells in response to transforming growth factor-ß (TGF-ß). Epithelial mesenchymal transition (EMT) is one process involved in renal fibrosis. In tubular epithelial cells we have recently described that Gremlin induces EMT and acts as a downstream TGF-ß mediator. Our aim was to investigate whether Gremlin participates in EMT by the regulation of the Smad pathway. Stimulation of human tubular epithelial cells (HK2) with Gremlin caused an early activation of the Smad signaling pathway (Smad 2/3 phosphorylation, nuclear translocation, and Smad-dependent gene transcription). The blockade of TGF-ß, by a neutralizing antibody against active TGF-ß, did not modify Gremlin-induced early Smad activation. These data show that Gremlin directly, by a TGF-ß independent process, activates the Smad pathway. In tubular epithelial cells long-term incubation with Gremlin increased TGF-ß production and caused a sustained Smad activation and a phenotype conversion into myofibroblasts-like cells. Smad 7 overexpression, which blocks Smad 2/3 activation, diminished EMT changes observed in Gremlin-transfected tubuloepithelial cells. TGF-ß neutralization also diminished Gremlin-induced EMT changes. In conclusion, we propose that Gremlin could participate in renal fibrosis by inducing EMT in tubular epithelial cells through activation of Smad pathway and induction of TGF-ß.


Subject(s)
Cell Transdifferentiation/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Smad Proteins/biosynthesis , Transforming Growth Factor beta1/biosynthesis , Cell Line , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Humans , Intercellular Signaling Peptides and Proteins/genetics , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Signal Transduction/genetics , Smad Proteins/genetics
10.
Nefrología (Madr.) ; 34(3): 369-376, mayo-jun. 2014. ilus
Article in Spanish | IBECS | ID: ibc-126608

ABSTRACT

Introducción: La ruta de señalización de Notch está activada en una gran variedad de patologías renales humanas. Recientemente hemos demostrado que la activación de esta ruta no estaría implicada en la fibrosis renal experimental inducida por angiotensina II o hipertensión. Objetivos: Evaluar si la vía Notch está activada en la fibrosis renal asociada a nefroesclerosis hipertensiva. Para validar la hipótesis se estudiaron varias patologías glomerulares caracterizadas por fibrosis túbulo-intersticial. Métodos: Se utilizaron biopsias renales de pacientes con nefroesclerosis hipertensiva, en comparación con nefropatía diabética y nefropatía membranosa en diferentes etapas de progresión. La expresión génica y proteica se evaluó por hibridación in situ e inmunohistoquímica, respectivamente. Resultados: En nefroesclerosis hipertensiva se observó baja expresión renal de proteínas de la vía Notch, no existiendo asociación entre la fibrosis túbulo-intersticial y los niveles de estas proteínas. Por el contrario, en las patologías glomerulares estudiadas se observó una elevada expresión de los transcritos Jagged-1, HES-1 y TGF-β, y de las proteínas Jagged-1 y Notch-1, localizados principalmente en células túbulo-epiteliales. Los niveles de expresión de los componentes de la vía Notch se relacionaron con el grado de fibrosis túbulo-intersticial, lo que confirma la activación de esta vía en nefropatías progresivas. Conclusiones: Nuestros datos muestran que la vía Notch no está activada en el riñón de pacientes con nefropatía hipertensiva, ampliando los resultados de los modelos experimentales de daño renal asociado a hipertensión a la patología humana. Nuestros estudios aportan nueva información sobre la compleja regulación del sistema Notch en el riñón (AU)


Introduction: The Notch signalling pathway is activated in a wide variety of human renal diseases. We have recently demonstrated that the activation of this pathway is not involved in experimental renal fibrosis induced by angiotensin II or hypertension. Objectives: To assess whether the Notch pathway is activated in renal fibrosis related to hypertensive nephrosclerosis. To test the hypothesis, various glomerular diseases characterised by tubulointerstitial fibrosis were analysed. Method: Renal biopsies were performed on patients with hypertensive nephrosclerosis, in comparison with diabetic nephropathy and membranous nephropathy at various stages. Gene and protein expression were evaluated by in-situ hybridisation and immunohistochemistry respectively. Results: In hypertensive nephrosclerosis low renal expression of notch-related proteins was observed. There was no link between tubulointerstitial fibrosis and the levels of these proteins. By contrast, in the glomerular diseases studied we observed high expression of the transcripts Jagged-1, HES-1 and TGF-β and the proteins Jagged-1 y Notch-1, localised primarily in tubuloepithelial cells. The levels of expression of the components of the Notch pathway correlate to the degree of tubulointerstitial fibrosis, which confirms the activation of this pathway in progressive nephropathies. Conclusions: Our data demonstrate that the Notch pathway is not activated in the kidneys of patients with hypertensive nephropathy, which extends the results of experimental models of kidney damage related to hypertension to the realm of human pathology. Our studies provide new information on the complex regulation of the Notch pathway in the kidney (AU)


Subject(s)
Humans , Kidney Diseases/physiopathology , Hypertension/complications , Receptors, Notch/biosynthesis , Translational Research, Biomedical , Angiotensins/physiology , Fibrosis/physiopathology , Nephrosclerosis/physiopathology , Kidney Tubules/physiopathology
11.
Nefrologia ; 34(3): 369-76, 2014 May 21.
Article in English, Spanish | MEDLINE | ID: mdl-24798563

ABSTRACT

INTRODUCTION: The Notch signalling pathway is activated in a wide variety of human renal diseases. We have recently demonstrated that the activation of this pathway is not involved in experimental renal fibrosis induced by angiotensin II or hypertension. OBJECTIVES: To assess whether the Notch pathway is activated in renal fibrosis related to hypertensive nephrosclerosis. To test the hypothesis, various glomerular diseases characterised by tubulointerstitial fibrosis were analysed. METHOD: Renal biopsies were performed on patients with hypertensive nephrosclerosis, in comparison with diabetic nephropathy and membranous nephropathy at various stages. Gene and protein expression were evaluated by in-situ hybridisation and immunohistochemistry respectively. RESULTS: In hypertensive nephrosclerosis low renal expression of notch-related proteins was observed. There was no link between tubulointerstitial fibrosis and the levels of these proteins. By contrast, in the glomerular diseases studied we observed high expression of the transcripts Jagged-1, HES-1 and TGF-ß and the proteins Jagged-1 y Notch-1, localised primarily in tubuloepithelial cells. The levels of expression of the components of the Notch pathway correlate to the degree of tubulointerstitial fibrosis, which confirms the activation of this pathway in progressive nephropathies. CONCLUSIONS: Our data demonstrate that the Notch pathway is not activated in the kidneys of patients with hypertensive nephropathy, which extends the results of experimental models of kidney damage related to hypertension to the realm of human pathology. Our studies provide new information on the complex regulation of the Notch pathway in the kidney.


Subject(s)
Hypertension, Renal/physiopathology , Kidney/pathology , Kidney/physiopathology , Nephritis/physiopathology , Receptors, Notch/physiology , Signal Transduction , Female , Fibrosis/physiopathology , Humans , Hypertension, Renal/genetics , Male , Middle Aged , Nephritis/genetics , Protein Biosynthesis , Receptors, Notch/genetics , Retrospective Studies
12.
Am J Physiol Renal Physiol ; 304(12): F1399-410, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23552867

ABSTRACT

The albumin overload model induces proteinuria and tubulointersitial damage, followed by hypertension when rats are exposed to a hypersodic diet. To understand the effect of kinin system stimulation on salt-sensitive hypertension and to explore its potential renoprotective effects, the model was induced in Sprague-Dawley rats that had previously received a high-potassium diet to enhance activity of the kinin pathway, followed with/without administration of icatibant to block the kinin B2 receptor (B2R). A disease control group received albumin but not potassium or icatibant, and all groups were exposed to a hypersodic diet to induce salt-sensitive hypertension. Potassium treatment increased the synthesis and excretion of tissue kallikrein (Klk1/rKLK1) accompanied by a significant reduction in blood pressure and renal fibrosis and with downregulation of renal transforming growth factor-ß (TGF-ß) mRNA and protein compared with rats that did not receive potassium. Participation of the B2R was evidenced by the fact that all beneficial effects were lost in the presence of the B2R antagonist. In vitro experiments using the HK-2 proximal tubule cell line showed that treatment of tubular cells with 10 nM bradykinin reduced the epithelial-mesenchymal transdifferentiation and albumin-induced production of TGF-ß, and the effects produced by bradykinin were prevented by pretreatment with the B2R antagonist. These experiments support not only the pathogenic role of the kinin pathway in salt sensitivity but also sustain its role as a renoprotective, antifibrotic paracrine system that modulates renal levels of TGF-ß.


Subject(s)
Bradykinin/analogs & derivatives , Fibrosis/prevention & control , Hypertension/drug therapy , Kidney Diseases/prevention & control , Kinins/physiology , Potassium, Dietary/pharmacology , Proteinuria/physiopathology , Transforming Growth Factor beta/physiology , Animals , Bradykinin/pharmacology , Bradykinin B2 Receptor Antagonists , Cell Line , Female , Humans , Hypertension/physiopathology , Kidney Diseases/pathology , Kidney Tubules/pathology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Proteinuria/chemically induced , Rats , Rats, Sprague-Dawley , Serum Albumin, Bovine , Sodium Chloride, Dietary/adverse effects , Tissue Kallikreins/urine , Transforming Growth Factor beta/biosynthesis
13.
FEBS J ; 280(14): 3232-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23617393

ABSTRACT

Diabetic nephropathy (DN) is a progressive fibrotic condition that may lead to end-stage renal disease and kidney failure. Transforming growth factor-ß1 and bone morphogenetic protein-7 (BMP7) have been shown to induce DN-like changes in the kidney and protect the kidney from such changes, respectively. Recent data identified insulin action at the level of the nephron as a crucial factor in the development and progression of DN. Insulin requires a family of insulin receptor substrate (IRS) proteins for its physiological effects, and many reports have highlighted the role of insulin and IRS proteins in kidney physiology and disease. Here, we observed IRS2 expression predominantly in the developing and adult kidney epithelium in mouse and human. BMP7 treatment of human kidney proximal tubule epithelial cells (HK-2 cells) increases IRS2 transcription. In addition, BMP7 treatment of HK-2 cells induces an electrophoretic shift in IRS2 migration on SDS/PAGE, and increased association with phosphatidylinositol-3-kinase, probably due to increased tyrosine/serine phosphorylation. In a cohort of DN patients with a range of chronic kidney disease severity, IRS2 mRNA levels were elevated approximately ninefold, with the majority of IRS2 staining evident in the kidney tubules in DN patients. These data show that IRS2 is expressed in the kidney epithelium and may play a role in the downstream protective events triggered by BMP7 in the kidney. The specific up-regulation of IRS2 in the kidney tubules of DN patients also indicates a novel role for IRS2 as a marker and/or mediator of human DN progression.


Subject(s)
Diabetic Nephropathies/metabolism , Gene Expression , Insulin Receptor Substrate Proteins/metabolism , Kidney Tubules/metabolism , Adolescent , Adult , Animals , Base Sequence , Binding Sites , Bone Morphogenetic Protein 7/physiology , Case-Control Studies , Cell Line , Child , Epithelium/metabolism , Female , Humans , Insulin Receptor Substrate Proteins/genetics , Kidney Tubules/pathology , Male , Mice , Middle Aged , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Smad4 Protein/genetics , Transcriptional Activation , Young Adult
14.
Nephrol Dial Transplant ; 24(4): 1121-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19028757

ABSTRACT

BACKGROUND: Vascular calcification has been widely recognized as a significant contributor to cardiovascular risk in patients with chronic kidney disease. Recent evidence suggests that BMP-7 decreases the vascular calcification observed in uraemic rats, while BMP-2 could also be participating in this process. Gremlin, a bone morphogenetic protein antagonist, has been detected in rat aortic vascular smooth muscle cells (VSMCs), and since the role of the VSMCs into vascular calcification in uraemia is considered critical in this process, we hypothesized that gremlin could be participating in its pathogenesis. With this aim, we studied its expression in aorta from uraemic rats with calcitriol-induced vascular calcification and in 16-vessel biopsies of uraemic patients undergoing kidney transplantation. METHODS: Gremlin was detected by in situ hybridization (ISH) and immunohistochemistry (IMH). BMP-7, BMP-2 and BMP-2 receptor (BMPR2) were detected by IMH. Vascular calcification was assessed by the von Kossa staining method. Sham-operated and 5/6 nephrectomized rats (NFX) (1.2%P) were treated with vehicle or calcitriol (80 ng/kg, intraperitoneally every other day). Rats were killed after 4 weeks of treatment, and abdominal aorta was dissected for assessment of gremlin expression and vascular calcification. Epigastric arteries were obtained from dialysis patients during kidney transplantation procedure. Arteries from kidney donors were also studied. RESULTS: NFX rats developed a mild vascular calcification, whereas NFX-calcitriol rats developed a severe vascular and tissue calcification. A marked overexpression of gremlin was observed in the vascular media of aorta from NFX-calcitriol rats as compared with NFX and sham-calcitriol groups (4.8 +/- 1.3 versus 0.59 +/- 0.17 versus 0.19 +/- 0.07 percentage/mm(2), P < 0.01), and correlated with the BMP-2 and BMPR2 expression. Sham rats showed minimal or null gremlin expression. BMP-7 was not found in sham or calcified arteries. In human studies, we observed strong expression of gremlin mRNA and protein in the media layer of vessels from uraemic patients as compared with those from normal humans (staining score 3.72 +/- 0.95 versus 0.91 +/- 0.08 percentage/mm(2), P < 0.05). CONCLUSION: We observed a marked gremlin overexpression in the media layer of vessels in uraemic rats and patients in association with vascular calcification and BMP-2 expression. We postulate that gremlin may play a role in the vascular calcification process in uraemia, and its interaction with BMP-7 or BMP-2 remains to be elucidated.


Subject(s)
Aortic Diseases/physiopathology , Bone Morphogenetic Proteins/antagonists & inhibitors , Calcinosis/physiopathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Uremia/complications , Animals , Aortic Diseases/pathology , Disease Models, Animal , Kidney Transplantation , Male , Rats , Rats, Sprague-Dawley , Uremia/surgery
15.
J Am Soc Nephrol ; 19(9): 1672-80, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18508967

ABSTRACT

Induced in high glucose-1 (IHG-1) is an evolutionarily conserved gene transcript upregulated by high extracellular glucose concentrations, but its function is unknown. Here, it is reported that the abundance of IHG-1 mRNA is nearly 10-fold higher in microdissected, tubule-rich renal biopsies from patients with diabetic nephropathy compared with control subjects. In the diabetic nephropathy specimens, in situ hybridization localized IHG-1 to tubular epithelial cells along with TGF-beta1 and activated Smad3, suggesting a possible role in the development of tubulointerstitial fibrosis. Supporting this possibility, IHG-1 mRNA and protein expression also increased with unilateral ureteral obstruction. In the HK-2 proximal tubule cell line, overexpression of IHG-1 increased TGF-beta1-stimulated expression of connective tissue growth factor and fibronectin. IHG-1 was found to amplify TGF-beta1-mediated transcriptional activity by increasing and prolonging phosphorylation of Smad3. Conversely, inhibition of endogenous IHG-1 with small interference RNA suppressed transcriptional responses to TGF-beta1. In summary, IHG-1, which increases in diabetic nephropathy, may enhance the actions of TGF-beta1 and contribute to the development of tubulointerstitial fibrosis.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney Tubules/metabolism , Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/metabolism , Amino Acid Sequence , Cell Line , Connective Tissue Growth Factor , Conserved Sequence , Extracellular Fluid/metabolism , Fibronectins/metabolism , Fibrosis , Glucose/metabolism , Humans , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Tubules/pathology , Molecular Sequence Data , Phosphorylation , Proteins/genetics , Sequence Alignment , Signal Transduction , Smad3 Protein/metabolism
16.
Nephrol Dial Transplant ; 22(7): 1882-90, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17403698

ABSTRACT

BACKGROUND: Recent evidence in vitro and in vivo suggests that gremlin, a bone morphogenetic protein antagonist, is participating in tubular epithelial mesenchymal transition (EMT) in diabetic nephropathy as a downstream mediator of TGF-beta. Since EMT also occurs in parietal epithelial glomerular cells (PECs) leading to crescent formation, we hypothesized that gremlin could participate in this process. With this aim we studied its expression in 30 renal biopsies of patients with pauci-immune crescentic nephritis. METHODS: Gremlin was detected by in situ hybridization (ISH) and immunohistochemistry (IMH) and TGF-beta by ISH and Smads by southwestern histochemistry (SWH). Phosphorylated Smad2, CTGF, BMP-7, PCNA, alpha-SMA, synaptopodin, CD-68, and phenotypic markers of PECs (cytokeratin, E-cadherin), were detected by IMH. In cultured human monocytes, gremlin and CTGF induction by TGF-beta was studied by western blot. RESULTS: We observed strong expression of gremlin mRNA and protein in cellular and fibrocellular crescents corresponding to proliferating PECs and monocytes, in co-localization with TGF-beta. A marked over-expression of gremlin was also observed in tubular and infiltrating interstitial cells, correlating with tubulointerstitial fibrosis (r=0.59; P<0.01). A nuclear Smad activation in the same tubular cells, that are expressing TGF-beta and gremlin, was detected. In human cultured monocytes, TGF-beta induced gremlin production while CTGF expression was not detected. CONCLUSION: We postulate that gremlin may play a role in the fibrous process in crescentic nephritis, both in glomerular crescentic and tubular epithelial cells. The co-localization of gremlin and TGF-beta expression found in glomeruli and tubular cells suggest that gremlin may be important in mediating some of the pathological effects of TGF-beta.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Glomerulus/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Fibrosis , Glomerulonephritis/pathology , Histocytochemistry , Humans , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Kidney Glomerulus/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Monocytes/drug effects , Monocytes/metabolism , RNA, Messenger/metabolism , Tissue Distribution , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
17.
Am J Kidney Dis ; 45(6): 1034-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15957132

ABSTRACT

BACKGROUND: We report the induction of gremlin, a bone morphogenetic protein antagonist, in cultured human mesangial cells exposed to high glucose and transforming growth factor beta (TGF-beta) levels in vitro and kidneys from diabetic rats in vivo. METHODS: Gremlin expression was assessed in human diabetic nephropathy by means of in situ hybridization, immunohistochemistry, and real-time polymerase chain reaction and correlated with clinical and pathological indices of disease. RESULTS: Gremlin was not expressed in normal human adult kidneys. Conversely, abundant gremlin expression was observed in human diabetic nephropathy. Although some gremlin expression was observed in occasional glomeruli, gremlin expression was most prominent in areas of tubulointerstitial fibrosis, where it colocalized with TGF-beta expression. Gremlin messenger RNA levels correlated directly with renal dysfunction, determined by means of serum creatinine level, but not with proteinuria level. There was a strong correlation between gremlin expression and tubulointerstitial fibrosis score. CONCLUSION: In aggregate, these results indicate that the developmental gene gremlin reemerges in the context of tubulointerstitial fibrosis in diabetic nephropathy and suggests a role for TFG-beta as an inducer of gremlin expression in this context.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Diabetic Nephropathies/metabolism , Gene Expression Regulation , Glomerular Mesangium/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Transforming Growth Factor beta/physiology , Bone Morphogenetic Proteins/genetics , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cytokines , Diabetic Nephropathies/pathology , Fibrosis , Gene Expression Regulation/drug effects , Glomerular Mesangium/pathology , Glucose/pharmacology , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Kidney Neoplasms/metabolism , Nephritis, Interstitial/metabolism , Proteins , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
18.
Nephrol Dial Transplant ; 19(10): 2505-12, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15280531

ABSTRACT

BACKGROUND: Nuclear factor-kappaB (NF-kappaB) regulates genes involved in renal disease progression, such as the chemokines monocyte chemoattractant protein-1 (MCP-1) and RANTES. NF-kappaB is activated in experimental models of renal injury, and in vitro studies also suggest that proteinuria and angiotensin II could be important NF-kappaB activators. It has been proposed that locally produced MCP-1 may be involved in the development of diabetic nephropathy (DN). We examined the hypothesis that NF-kappaB could be an indicator of renal damage progression in DN. METHODS: Biopsy specimens from 11 patients with type 2 diabeties and overt nephropathy were studied by southwestern histochemistry for the in situ detection of activated NF-kappaB. In addition, by immunohistochemistry and/or in situ hybridization, we studied the expression of MCP-1 and RANTES, whose genes are regulated by NF-kappaB. RESULTS: NF-kappaB was detected mainly in cortical tubular epithelial cells and, to a lesser extent, in some glomerular and interstitial cells. A strong upregulation of MCP-1 and RANTES was observed in all the cases, mainly in tubular cells, and there was a strong correlation between the expression of these chemokines and NF-kappaB activation in the same cells, as observed in serial sections (r = 0.7; P = 0.01). In addition, the tubular expression of these chemokines was correlated mainly with the magnitude of the proteinuria (P = 0.002) and with interstitial cell infiltration (P<0.05). CONCLUSIONS: The activation of NF-kappaB and the transcription of certain pro-inflammatory chemokines in tubular epithelial cells are markers of progressive DN. Proteinuria might be one of the main factors inducing the observed pro-inflammatory phenotype.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Diabetic Nephropathies/metabolism , NF-kappa B/metabolism , Adult , Biomarkers/metabolism , Chemokine CCL2/genetics , Chemokine CCL5/genetics , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Progression , Female , Gene Expression , Gene Expression Regulation , Humans , Immunohistochemistry , In Situ Hybridization , Inflammation/pathology , Kidney/metabolism , Kidney/pathology , Male , Middle Aged , NF-kappa B p50 Subunit , RNA, Messenger/metabolism , Transcription Factor RelA , Up-Regulation
19.
Kidney Int Suppl ; (86): S39-45, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12969126

ABSTRACT

BACKGROUND: The molecular mechanisms of renal injury and fibrosis in proteinuric nephropathies are not completely elucidated but the renin-angiotensin system (RAS) is involved. Idiopathic membranous nephropathy (MN), a proteinuric disease, may progress to renal failure. Our aim was to investigate the localization of RAS components in MN and their correlation with profibrotic parameters and renal injury. METHODS: Renal biopsies from 20 patients with MN (11 with progressive disease) were studied for the expression of RAS components [angiotensin-converting enzyme (ACE) and angiotensin II (Ang II)] by immunohistochemistry. Transforming growth factor-beta (TGF-beta) and platelet-derived growth factor (PDGF)-BB were studied by by in situ hybridization, and myofibroblast transdifferentiation by alpha-smooth muscle actin (alpha-SMA) staining. RESULTS: ACE immunostaining was elevated in tubular cells and appeared in interstitial cells colocalized in alpha-actin-positive cells in progressive disease. Elevated levels of Ang II were observed in tubules and infiltrating interstitial cells. TGF-beta and PDGF mRNAs were up-regulated mainly in cortical tubular epithelial cells in progressive disease (P < 0.01) and correlated with the myofibroblast transdifferentiation (r = 0.8, P < 0.01 for TGF-beta; r = 0.6, P < 0.01 for PDGF). Moreover, in serial sections of progressive cases, the ACE and Ang II over-expression was associated with the tubular expression of these pro-fibrogenic factors, and with the interstitial infiltration and myofibroblast activation. CONCLUSION: Intrarenal RAS is selectively activated in progressive MN. De novo expression of ACE at sites of tubulointerstitial injury suggests that the in situ Ang II generation could participate in tubular TGF-beta up-regulation, epithelial-myofibroblast transdifferentiation, and disease progression. These results suggest a novel role of Ang II in human tubulointerstitial injury.


Subject(s)
Angiotensin II/metabolism , Fibroblasts , Glomerulonephritis, Membranous/physiopathology , Kidney/metabolism , Myocytes, Smooth Muscle , Becaplermin , Fibroblasts/pathology , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/pathology , Humans , Myocytes, Smooth Muscle/pathology , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Renin-Angiotensin System , Transforming Growth Factor beta/metabolism , Up-Regulation
20.
Kidney Int Suppl ; (86): S64-70, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12969130

ABSTRACT

BACKGROUND: The molecular mechanisms of renal injury in diabetic nephropathy (DN) are not completely understood, although inflammatory cells play a key role. The renin-angiotensin system (RAS) is involved in kidney damage; however, few studies have examined the localization of RAS components in human DN. Our aim was to investigate in renal biopsies the expression of RAS and their correlation with proinflammatory parameters and renal injury. METHODS: The biopsies from 10 patients with type 2 diabetes mellitus and overt nephropathy were studied for the expression of RAS components by immunohistochemistry (IH). In addition, by Southwestern histochemistry we studied the in situ detection of the activated nuclear factor kappa B (NFkappaB), and by IH and/or in situ hybridization (ISH), the expression of monocyte chemoattractant protein-1 (MCP-1) and regulated upon activation, normal T cell expressed and secreted (RANTES), whose genes are regulated by NFkappaB. RESULTS: Angiotensin-converting enzyme (ACE) immunostaining was elevated in tubular cells and appeared in interstitial cells. Elevated levels of angiotensin II (Ang II) immunostaining were observed in tubular and infiltrating interstitial cells. There was also a down-regulation of AT1 and up-regulation of AT2 receptors. An activation of NFkappaB and a marked up-regulation of NFkappaB-dependent chemokines mainly in tubular cells was observed. Elevated levels of NFkappaB, chemokines, and Ang II in tubules were correlated with proteinuria and interstitial cell infiltration. CONCLUSIONS: Our results show that in human DN, RAS components are modified in renal compartments, showing elevated local Ang II production, activation of tubular cells, and induction of proinflammatory parameters. These data suggest that Ang II contributes to the renal inflammatory process, and may explain the molecular mechanisms of the beneficial effect of RAS blockade.


Subject(s)
Diabetic Nephropathies/complications , Diabetic Nephropathies/metabolism , Nephritis/etiology , Renin-Angiotensin System , Chemokines/metabolism , Humans , In Vitro Techniques , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney/pathology , NF-kappa B/metabolism , Nephritis/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...