Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Kidney Int ; 96(2): 378-396, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31146971

RESUMEN

Prolyl hydroxylase domain enzyme inhibitors (PHDIs) stabilize hypoxia-inducible factors (HIFs), and are protective in models of acute ischemic and inflammatory kidney disease. Whether PHDIs also confer protection in chronic inflammatory kidney disease models remains unknown. Here we investigated long-term effects of PHDI treatment in adenine-induced nephropathy as a model for chronic tubulointerstitial nephritis. After three weeks, renal dysfunction and tubulointerstitial damage, including proximal and distal tubular injury, tubular dilation and renal crystal deposition were significantly attenuated in PHDI-treated (the isoquinoline derivative ICA and Roxadustat) compared to vehicle-treated mice with adenine-induced nephropathy. Crystal-induced renal fibrosis was only partially diminished by treatment with ICA. Renoprotective effects of ICA treatment could not be attributed to changes in adenine metabolism or urinary excretion of the metabolite 2,8-dihydroxyadenine. ICA treatment reduced inflammatory infiltrates of F4/80+ mononuclear phagocytes in the kidneys and supported a regulatory, anti-inflammatory immune response. Furthermore, interstitial deposition of complement C1q was decreased in ICA-treated mice fed an adenine-enriched diet. Tubular cell-specific HIF-1α and myeloid cell-specific HIF-1α and HIF-2α expression were not required for the renoprotective effects of ICA. In contrast, depletion of mononuclear phagocytes with clodronate largely abolished the nephroprotective effects of PHD inhibition. Thus, our findings indicate novel and potent systemic anti-inflammatory properties of PHDIs that confer preservation of kidney function and structure in chronic tubulointerstitial inflammation and might counteract kidney disease progression.


Asunto(s)
Nefritis Intersticial/tratamiento farmacológico , Fagocitos/efectos de los fármacos , Prolil Hidroxilasas/metabolismo , Inhibidores de Prolil-Hidroxilasa/farmacología , Insuficiencia Renal Crónica/prevención & control , Adenina/metabolismo , Adenina/toxicidad , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ácido Clodrónico/farmacología , Complemento C1q/inmunología , Complemento C1q/metabolismo , Modelos Animales de Enfermedad , Glicina/análogos & derivados , Glicina/farmacología , Glicina/uso terapéutico , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isoquinolinas/farmacología , Isoquinolinas/uso terapéutico , Túbulos Renales/citología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/inmunología , Túbulos Renales/patología , Masculino , Ratones , Ratones Transgénicos , Nefritis Intersticial/sangre , Nefritis Intersticial/inducido químicamente , Nefritis Intersticial/inmunología , Fagocitos/inmunología , Prolil Hidroxilasas/inmunología , Inhibidores de Prolil-Hidroxilasa/uso terapéutico , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Insuficiencia Renal Crónica/inmunología
2.
Kidney Int ; 94(5): 887-899, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30173898

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by mutations of the PKD1 gene and characterized by growth of bilateral renal cysts. Cyst growth is accompanied by regional hypoxia and induction of hypoxia-inducible factor (HIF)-1α in cyst-lining epithelial cells. To determine the relevance of HIF-1α for cyst growth in vivo we used an inducible kidney epithelium-specific knockout mouse to delete Pkd1 at postnatal day 20 or 35 to induce polycystic kidney disease of different severity and analyzed the effects of Hif-1α co-deletion and HIF-1α stabilization using a prolyl-hydroxylase inhibitor. HIF-1α expression was enhanced in kidneys with progressive cyst growth induced by early Pkd1 deletion, but unchanged in the milder phenotype induced by later Pkd1 deletion. Hif-1α co-deletion significantly attenuated cyst growth in the severe, but not in the mild, phenotype. Application of a prolyl-hydroxylase inhibitor resulted in severe aggravation of the mild phenotype with rapid loss of renal function. HIF-1α expression was associated with induction of genes that mediate calcium-activated chloride secretion. Thus, HIF-1α does not seem to play a role in early cyst formation, but accelerates cyst growth during progressive polycystic kidney disease. This novel mechanism of cyst growth may qualify as a therapeutic target.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Riñón Poliquístico Autosómico Dominante/etiología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones , Riñón Poliquístico Autosómico Dominante/terapia
3.
Kidney Blood Press Res ; 43(1): 1-11, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29393223

RESUMEN

BACKGROUND/AIMS: One potential pathomechanism how low nephron number leads to hypertension in later life is altered salt handling. We therefore evaluated changes in electrolyte and water content in wildtype (wt) and GDNF+/- mice with a 30% reduction of nephron number. METHODS: 32 GDNF+/- and 36 wt mice were fed with low salt (LSD, 0.03%, normal drinking water) or high salt (HSD, 4%, 0.9% drinking water) diet for 4 weeks. Blood pressure was continuously measured by telemetry in a subgroup. At the end of the experiment and after standardized ashing processes electrolyte- and water contents of the skin and the total body were determined. RESULTS: We found higher blood pressure in high salt treated GDNF+/-compared to wt mice. Of interest, we could not confirm an increase in total-body sodium as predicted by prevailing explanations, but found increased total body and skin chloride that interestingly correlated with relative kidney weight. CONCLUSION: We hereby firstly report significant total body and skin chloride retention in salt sensitive hypertension of GDNF+/-mice with genetically determined lower nephron number. Thus, in contrast to the prevailing opinion our data argue for the involvement of non-volume related mechanisms.


Asunto(s)
Cloruros/metabolismo , Hipertensión/etiología , Nefronas , Animales , Cloruros/análisis , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Riñón/fisiología , Ratones , Tamaño de los Órganos , Sodio/análisis , Cloruro de Sodio Dietético
4.
Kidney Int ; 91(3): 616-627, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27927598

RESUMEN

Chronic kidney disease (CKD) is associated with increased risk and worse prognosis of cardiovascular disease, including peripheral artery disease. An impaired angiogenic response to ischemia may contribute to poor outcomes of peripheral artery disease in patients with CKD. Hypoxia inducible factors (HIF) are master regulators of angiogenesis and therefore represent a promising target for therapeutic intervention. To test this we induced hind-limb ischemia in rats with CKD caused by 5/6 nephrectomy and administered two different treatments known to stabilize HIF protein in vivo: carbon monoxide and a pharmacological inhibitor of prolyl hydroxylation 2-(1-chloro-4- hydroxyisoquinoline-3-carboxamido) acetate (ICA). Expression levels of pro-angiogenic HIF target genes (Vegf, Vegf-r1, Vegf-r2, Ho-1) were measured by qRT-PCR. Capillary density was measured by CD31 immunofluorescence staining and HIF expression was evaluated by immunohistochemistry. Capillary density in ischemic skeletal muscle was significantly lower in CKD animals compared to sham controls. Rats with CKD showed significantly lower expression of HIF and all measured pro-angiogenic HIF target genes, including VEGF. Both HIF stabilizing treatments rescued HIF target gene expression in animals with CKD and led to significantly higher ischemia-induced capillary sprouting compared to untreated controls. ICA was effective regardless of whether it was administered before or after induction of ischemia and led to a HIF expression in skeletal muscle. Thus, impaired ischemia-induced angiogenesis in rats with CKD can be improved by HIF stabilization, even if started after onset of ischemia.


Asunto(s)
Capilares/efectos de los fármacos , Monóxido de Carbono/farmacología , Glicina/análogos & derivados , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isquemia/tratamiento farmacológico , Isoquinolinas/farmacología , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Capilares/metabolismo , Capilares/fisiopatología , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glicina/farmacología , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatología , Masculino , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Estabilidad Proteica , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/fisiopatología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Biochim Biophys Acta ; 1842(4): 558-65, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24418215

RESUMEN

Diabetes can disrupt endoplasmic reticulum (ER) homeostasis which leads to ER stress. ER stress-induced renal apoptosis seems to be involved in the development of diabetic nephropathy. The present study was designed to investigate the contribution of reduced ER stress to the beneficial effects of an angiotensin receptor blocker. Insulin-dependent diabetes mellitus was induced by streptozotocin injections to hypertensive mRen2-transgenic rats. After 2weeks animals were treated with 0.7mg/kg/day irbesartan. Blood glucose, blood pressure and protein excretion were assessed. Expression of ER stress markers was measured by real-time PCR. Immunohistochemistry was performed to detect markers of ER stress, renal damage and infiltrating cells. Glomerulosclerosis and apoptosis were evaluated. Diabetic mRen2-transgenic rats developed renal injury with proteinuria, tubulointerstitial cell proliferation as well as glomerulosclerosis and podocyte injury. Moreover, an increase in inflammation, podocyte ER stress and apoptosis was detected. Irbesartan somewhat lowered blood pressure and reduced proteinuria, tubulointerstitial cell proliferation and glomerulosclerosis. Podocyte damage was ameliorated but markers of ER stress (calnexin, grp78) and apoptosis were not reduced by irbesartan. On the other hand, inflammatory cell infiltration in the tubulointerstitium and the glomerulus was significantly attenuated. We conclude that irbesartan reduced renal damage even in a very low dose. The beneficial effects of low dose irbesartan were paralleled by a reduction of blood pressure and inflammation but not by a reduction of ER stress and apoptosis. Thus, sustained endoplasmic reticulum stress in the kidney does not necessarily lead to increased inflammation and tubulointerstitial or glomerular injury.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Nefropatías Diabéticas/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inflamación/prevención & control , Riñón/efectos de los fármacos , Tetrazoles/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Glucemia/análisis , Presión Sanguínea/efectos de los fármacos , Irbesartán , Masculino , Ratas , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta/fisiología
6.
Kidney Int ; 88(6): 1283-1292, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26200943

RESUMEN

Reduced nephron number predisposes to hypertension and kidney disease. Interaction of the branching ureteric bud and surrounding mesenchymal cells determines nephron number. Since oxygen supply may be critical for intrauterine development, we tested whether hypoxia and hypoxia-inducible factor-1α (HIF-1α) influence nephrogenesis. We found that HIF-1α is required for branching of MDCK cells. In addition, culture of metanephric mouse kidneys with ureteric bud cell-specific stabilization or knockout of HIF-1α revealed a positive impact of HIF-1α on nephrogenesis. In contrast, widespread stabilization of HIF-1α in metanephric kidneys through hypoxia or HIF stabilizers impaired nephrogenesis, and pharmacological HIF inhibition enhanced nephrogenesis. Several lines of evidence suggest an inhibitory effect through the hypoxia response of mesenchymal cells. HIF-1α was expressed in mesenchymal cells during nephrogenesis. Expression of the anti-branching factors Bmp4 and Vegfa, secreted by mesenchymal cells, was increased upon HIF stabilization. The conditioned medium from hypoxic metanephric kidneys inhibited MDCK branching, which was partially rescued by Vegfa antibodies. Thus, the effect of HIF-1α on nephrogenesis appears context dependent. While HIF-1α in the ureteric bud is of importance for proper branching morphogenesis, the net effect of hypoxia-induced HIF activation in the embryonic kidney appears to be mesenchymal cell-dependent inhibition of ureter branching.

7.
J Am Soc Nephrol ; 25(3): 465-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24203996

RESUMEN

Polycystic kidney diseases are characterized by numerous bilateral renal cysts that continuously enlarge and, through compression of intact nephrons, lead to a decline in kidney function over time. We previously showed that cyst enlargement is accompanied by regional hypoxia, which results in the stabilization of hypoxia-inducible transcription factor-1α (HIF-1α) in the cyst epithelium. Here we demonstrate a correlation between cyst size and the expression of the HIF-1α-target gene, glucose transporter 1, and report that HIF-1α promotes renal cyst growth in two in vitro cyst models-principal-like MDCK cells (plMDCKs) within a collagen matrix and cultured embryonic mouse kidneys stimulated with forskolin. In both models, augmenting HIF-1α levels with the prolyl hydroxylase inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate enhanced cyst growth. In addition, inhibition of HIF-1α degradation through tubule-specific knockdown of the von Hippel-Lindau tumor suppressor increased cyst size in the embryonic kidney cyst model. In contrast, inhibition of HIF-1α by chetomin and knockdown of HIF-1α both decreased cyst growth in these models. Consistent with previous reports, plMDCK cyst enlargement was driven largely by transepithelial chloride secretion, which consists, in part, of a calcium-activated chloride conductance. plMDCKs deficient for HIF-1α almost completely lacked calcium-activated chloride secretion. We conclude that regional hypoxia in renal cysts contributes to cyst growth, primarily due to HIF-1α-dependent calcium-activated chloride secretion. These findings identify the HIF system as a novel target for inhibition of cyst growth.


Asunto(s)
Cloruros/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Enfermedades Renales Poliquísticas/etiología , Animales , Canales de Cloruro/metabolismo , Perros , Femenino , Regulación de la Expresión Génica , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hipoxia/fisiopatología , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos C57BL , Enfermedades Renales Poliquísticas/metabolismo
8.
Mol Carcinog ; 53(12): 970-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23818324

RESUMEN

Hypoxia leads to the upregulation of a variety of genes mediated largely via the hypoxia inducible transcription factor (HIF). Prominent HIF-regulated target genes such as the vascular endothelial growth factor (VEGF), the glucose transporter 1 (Glut-1), or erythropoietin (EPO) help to assure survival of cells and organisms in a low oxygenated environment. Here, we are the first to report the hypoxic regulation of the sperm associated antigen 4 (SPAG4). SPAG4 is a member of the cancer testis (CT) gene family and to date little is known about its physiological function or its involvement in tumor biology. A number of CT family candidate genes are therefore currently being investigated as potential cancer markers, due to their predominant testicular expression pattern. We analyzed RNA and protein expression by RNAse protection assay, immunofluorescent as well as immunohistological stainings. To evaluate the influence of SPAG4 on migration and invasion capabilities, siRNA knockdown as well as transient overexpression was performed prior to scratch or invasion assay analysis. The hypoxic regulation of SPAG4 is clearly mediated in a HIF-1 and VHL dependent manner. We furthermore show upregulation of SPAG4 expression in human renal clear cell carcinoma (RCC) and co-localization within the nucleolus in physiological human testis tissue. SPAG4 knockdown reduces the invasion capability of RCC cells in vitro and overexpression leads to enhancement of tumor cell migration. Together, SPAG4 could possibly play a role in the invasion capability and growth of renal tumors and could represent an interesting target for clinical intervention.


Asunto(s)
Carcinoma de Células Renales/genética , Proteínas Portadoras/genética , Movimiento Celular/genética , Factor 1 Inducible por Hipoxia/genética , Hipoxia/genética , Neoplasias Renales/genética , Invasividad Neoplásica/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Células HeLa , Humanos , Regulación hacia Arriba/genética
9.
Am J Pathol ; 181(5): 1595-606, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22944601

RESUMEN

The role of proximal versus distal tubular injury in the pathogenesis of acute kidney injury (AKI) is debatable. Inhibition of prolyl hydroxylases that regulate the degradation of hypoxia-inducible transcription factors (HIFs) is a promising therapeutic approach to optimize energy preservation under hypoxia and has successfully been applied to protect kidney structure and function in AKI models. Presently used prolyl hydroxylase inhibitors are lipophilic 2-oxoglutarate analogues (2OGAs) that are widely taken up in cells of most organs. Given the selective expression of organic anion transporters (OATs) in renal proximal tubular cells, we hypothesized that hydrophilic 2OGAs can specifically target proximal tubular cells. We found that cellular hydrophilic 2OGAs uptake depended on OATs and largely confined to the kidney, where it resulted in activation of HIF target genes only in proximal tubular cells. When applied in ischemia-reperfusion experiments, systemically active 2OGA preserved kidney structure and function, but OAT1-transported 2OGA was not protective, suggesting that HIF stabilization in distal tubular rather than proximal tubular cells and/or nontubular cells mediates protective effects. This study provides proof of concept for selective drug targeting of proximal tubular cells on the basis of specific transporters, gives insights into the role of different nephron segments in AKI pathophysiology, and may offer options for long-term HIF stabilization in proximal tubules without confounding effects of erythropoietin induction in peritubular cells and unwarranted extrarenal effects.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/farmacología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Ácido Acético/química , Ácido Acético/farmacología , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Aminoácidos Dicarboxílicos/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Separación Celular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Isquemia/complicaciones , Isquemia/patología , Isquemia/fisiopatología , Precondicionamiento Isquémico , Pruebas de Función Renal , Túbulos Renales Proximales/efectos de los fármacos , Ratones , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Estabilidad Proteica/efectos de los fármacos , Piridinas/química , Piridinas/farmacología
10.
Biochim Biophys Acta ; 1813(1): 1-13, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21056597

RESUMEN

Hypoxia is a common pathogenic stress, which requires adaptive activation of the Hypoxia-inducible transcription factor (HIF). In concert transcriptional HIF targets enhance oxygen availability and simultaneously reduce oxygen demand, enabling survival in a hypoxic microenvironment. Here, we describe the characterization of a new HIF-1 target gene, Rab20, which is a member of the Rab family of small GTP-binding proteins, regulating intracellular trafficking and vesicle formation. Rab20 is directly regulated by HIF-1, resulting in rapid upregulation of Rab20 mRNA as well as protein under hypoxia. Furthermore, exogenous as well as endogenous Rab20 protein colocalizes with mitochondria. Knockdown studies reveal that Rab20 is involved in hypoxia induced apoptosis. Since mitochondria play a key role in the control of cell death, we suggest that regulating mitochondrial homeostasis in hypoxia is a key function of Rab20. Furthermore, our study implicates that cellular transport pathways play a role in oxygen homeostasis. Hypoxia-induced Rab20 may influence tissue homeostasis and repair during and after hypoxic stress.


Asunto(s)
Apoptosis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/patología , Mitocondrias/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Western Blotting , Caspasas/metabolismo , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Unión al GTP rab/antagonistas & inhibidores , Proteínas de Unión al GTP rab/genética
11.
J Am Soc Nephrol ; 22(11): 2004-15, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21921145

RESUMEN

Hypoxia-inducible transcription factors (HIF) protect cells against oxygen deprivation, and HIF stabilization before ischemia mitigates tissue injury. Because ischemic acute kidney injury (AKI) often involves the thick ascending limb (TAL), modulation of HIF in this segment may be protective. Here, we generated mice with targeted TAL deletion of the von Hippel-Lindau protein (Vhl), which mediates HIF degradation under normoxia, using Tamm-Horsfall protein (Thp)-driven Cre expression. These mice showed strong expression of HIF-1α in TALs but no changes in kidney morphology or function under control conditions. Deficiency of Vhl in the TAL markedly attenuated proximal tubular injury and preserved TAL function following ischemia-reperfusion, which may be partially a result of enhanced expression of glycolytic enzymes and lactate metabolism. These results highlight the importance of the thick ascending limb in the pathogenesis of AKI and suggest that pharmacologically targeting the HIF system may have potential to prevent and mitigate AKI.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Asa de la Nefrona/fisiología , Uromodulina/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Umbral Anaerobio/fisiología , Animales , Modelos Animales de Enfermedad , Eritropoyesis/fisiología , Glucólisis/fisiología , Integrasas/genética , Riñón/fisiología , Ratones , Ratones Mutantes , Nefritis/genética , Nefritis/metabolismo , Nefritis/fisiopatología , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Uromodulina/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
12.
Nephrol Dial Transplant ; 26(11): 3458-65, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21804086

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure. Aberrant epithelial cell proliferation is a major cause of progressive cyst enlargement in ADPKD. Since activation of the Ras/Raf signaling system has been detected in cyst-lining epithelia, inhibition of Raf kinase has been proposed as an approach to retard the progression of ADPKD. Methods and results. PLX5568, a novel selective small molecule inhibitor of Raf kinases, attenuated proliferation of human ADPKD cyst epithelial cells. It reduced in vitro cyst growth of Madin-Darby Canine Kidney cells and of human ADPKD cells within a collagen gel. In male cy/+ rats with polycystic kidneys, PLX5568 inhibited renal cyst growth along with a significant reduction in the number of proliferating cell nuclear antigen- and phosphorylated extracellular signal-regulated kinase-positive cyst-lining epithelial cells. Furthermore, treated animals showed increased capacity to concentrate urine. However, PLX5568 did not lead to a consistent improvement of renal function. Moreover, although relative cyst volume was decreased, total kidney-to-body weight ratio was not significantly reduced by PLX5568. Further analyses revealed a 2-fold increase of renal and hepatic fibrosis in animals treated with PLX5568. CONCLUSIONS: PLX5568 attenuated cyst enlargement in vitro and in a rat model of ADPKD without improving kidney function, presumably due to increased renal fibrosis. These data suggest that effective therapies for the treatment of ADPKD will need to target fibrosis as well as the growth of cysts.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Quistes/patología , Riñón/fisiopatología , Cirrosis Hepática/inducido químicamente , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Animales , Células Cultivadas , Quistes/tratamiento farmacológico , Perros , Células Epiteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular , Humanos , Immunoblotting , Técnicas para Inmunoenzimas , Riñón/efectos de los fármacos , Masculino , Proteínas Quinasas Activadas por Mitógenos , Fosforilación/efectos de los fármacos , Enfermedades Renales Poliquísticas/enzimología , Enfermedades Renales Poliquísticas/patología , Inhibidores de Proteínas Quinasas/toxicidad , Ratas , Ratas Sprague-Dawley
13.
Kidney Int ; 78(9): 857-67, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20720525

RESUMEN

The two hypoxia-inducible factors (HIF-1α and HIF-2α) are transcription factors that regulate the response to hypoxia. Recently, the factor inhibiting HIF (FIH1) was identified as a molecular oxygen-dependent dioxygenase that blunts the transcriptional activity of HIF and has also been implicated in HIF-dependent and -independent hypoxia responses. Interestingly, HIF accumulation in the kidney has been shown to confer renal protection and to also cause glomerular injury or enhance renal fibrosis. In order to better understand the regulation of hypoxia-inducible genes, we determined the expression of FIH1 in the kidney and its functional role in isolated renal cells. FIH1 was expressed only in distal tubules and in podocytes, thus showing a very distinct expression pattern, partially overlapping with sites of HIF-1α expression. In tubular cells, RNA silencing of FIH1 caused transcriptional activation of HIF target genes during hypoxia. In contrast, FIH1 silencing in podocytes enhanced transcription of hypoxia-inducible genes in an HIF-independent manner. Using the anti-Thy.1 rat model of glomerulonephritis, we found a gradual decrease of glomerular FIH1 expression during disease progression paralleled by an increase in hypoxia-inducible genes including CXCR4, a mediator of glomerular inflammation. Thus, FIH1 appears to be a suppressor of oxygen-dependent genes in the kidney, operating through HIF-dependent and -independent mechanisms.


Asunto(s)
Glomerulonefritis Membranoproliferativa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Túbulos Renales Distales/metabolismo , Oxigenasas de Función Mixta/metabolismo , Oxígeno/metabolismo , Podocitos/metabolismo , Proteínas Represoras/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glomerulonefritis Membranoproliferativa/genética , Glomerulonefritis Membranoproliferativa/inmunología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Isoanticuerpos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxigenasas de Función Mixta/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Factores de Tiempo
14.
Am J Pathol ; 174(5): 1663-74, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19349364

RESUMEN

Hypoxia-inducible transcription factors (HIFs) play important roles in the response of the kidney to systemic and regional hypoxia. Degradation of HIFs is mediated by three oxygen-dependent HIF-prolyl hydroxylases (PHDs), which have partially overlapping characteristics. Although PHD inhibitors, which can induce HIFs in the presence of oxygen, are already in clinical development, little is known about the expression and regulation of these enzymes in the kidney. Therefore, we investigated the expression levels of the three PHDs in both isolated tubular cells and rat kidneys. All three PHDs were present in the kidney and were expressed predominantly in three different cell populations: (a) in distal convoluted tubules and collecting ducts (PHD1,2,3), (b) in glomerular podocytes (PHD1,3), and (c) in interstitial fibroblasts (PHD1,3). Higher levels of PHDs were found in tubular segments of the inner medulla where oxygen tensions are known to be physiologically low. PHD expression levels were unchanged in HIF-positive tubular and interstitial cells after induction by systemic hypoxia. In rat models of acute renal injury, changes in PHD expression levels were variable; while cisplatin and ischemia/reperfusion led to significant decreases in PHD2 and 3 expression levels, no changes were seen in a model of contrast media-induced nephropathy. These results implicate the non-uniform expression of HIF-regulating enzymes that modify the hypoxic response in the kidney under both regional and temporal conditions.


Asunto(s)
Lesión Renal Aguda/enzimología , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Riñón/enzimología , Procolágeno-Prolina Dioxigenasa/metabolismo , Animales , Antineoplásicos/toxicidad , Western Blotting , Cisplatino/toxicidad , Medios de Contraste/farmacología , Proteínas de Unión al ADN/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Proteínas Inmediatas-Precoces/genética , Técnicas para Inmunoenzimas , Isquemia/metabolismo , Isquemia/patología , Riñón/efectos de los fármacos , Riñón/lesiones , Médula Renal/efectos de los fármacos , Médula Renal/enzimología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/enzimología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Podocitos/efectos de los fármacos , Podocitos/enzimología , Procolágeno-Prolina Dioxigenasa/genética , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Hypertens ; 27(2): 376-85, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19226708

RESUMEN

OBJECTIVE: We examined the effects of fluvastatin treatment on the development of kidney injury in experimental deoxycorticosterone-acetate (DOCA)-salt hypertension. METHODS: Male Sprague-Dawley rats underwent unilateral nephrectomy and received subcutaneous DOCA pellets as well as 1% NaCl for drinking. Simultaneously, rats were treated with 5 mg/kg per day fluvastatin, or solvent only for 6 weeks. Mean arterial pressure was measured intraarterially. Glomerulosclerosis, interstitial fibrosis, cell proliferation, inflammation and podocyte damage were evaluated on kidney sections. Inflammatory markers were measured by real-time PCR. RESULTS: Mean arterial pressure was elevated in DOCA-salt-treated rats but unaltered by fluvastatin. Serum cholesterol was markedly elevated in DOCA-salt-treated rats and tended to be lower in fluvastatin-treated animals. Fluvastatin treatment decreased the mortality of DOCA-salt-treated rats. Urinary protein excretion, glomerular proliferation and macrophage infiltration as well as glomerulosclerosis were reduced by fluvastatin. Fluvastatin alleviated podocyte damage and glomerular osteopontin protein expression, which was localized in podocytes. On the contrary, interstitial fibrosis, inflammation and interstitial cell proliferation of DOCA-salt-treated rat kidneys were not influenced by fluvastatin. CONCLUSION: Statin treatment reduces mortality and glomerular damage independent from blood pressure in a low-renin model of hypertensive nephrosclerosis. A reduction of podocyte damage and macrophage infiltration may explain the beneficial effects of fluvastatin.


Asunto(s)
Ácidos Grasos Monoinsaturados/uso terapéutico , Glomerulonefritis/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipertensión/tratamiento farmacológico , Indoles/uso terapéutico , Nefroesclerosis/prevención & control , Podocitos/efectos de los fármacos , Animales , Presión Sanguínea , Proliferación Celular , Desoxicorticosterona , Progresión de la Enfermedad , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Glomerulonefritis/etiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/patología , Indoles/farmacología , Riñón/patología , Masculino , Nefroesclerosis/etiología , Tamaño de los Órganos , Podocitos/patología , Ratas , Ratas Sprague-Dawley
16.
Biochem J ; 409(1): 233-42, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17822384

RESUMEN

BNP (brain-type natriuretic peptide) is a cardiac hormone with systemic haemodynamic effects as well as local cytoprotective and antiproliferative properties. It is induced under a variety of pathophysiological conditions, including decompensated heart failure and myocardial infarction. Since regional hypoxia is a potential common denominator of increased wall stretch and myocardial hypoperfusion, we investigated the direct effects of hypoxia on BNP expression, and the role of the HIF (hypoxia-inducible transcription factor) in BNP regulation. Using an RNase protection assay we found a strong hypoxic induction of BNP mRNA expression in different cell lines and in cultured adult rat cardiomyocytes. Systemic hypoxia and exposure to 0.1% CO induced BNP expression in the rodent myocardium in vivo, although this was at a lower amplitude. BNP promoter-driven luciferase expression increased 10-fold after hypoxic stimulation in transient transfections. Inactivation of four putative HREs (hypoxia-response elements) in the promoter by site-directed mutagenesis revealed that the HRE at -466 nt was responsible for hypoxic promoter activation. A functional CACAG motif was identified upstream of this HRE. The HIF-1 complex bound specifically and inducibly only to the HRE at -466 nt, as shown by EMSA (electrophoretic mobility-shift assay) and ChIP (chromatin immunoprecipitation). siRNA (small interfering RNA)-mediated knockdown of HIF-1alpha, but not HIF-2alpha, interfered with hypoxic BNP mRNA induction and BNP promoter activation, confirming that BNP is a specific HIF-1alpha target gene. In conclusion, BNP appears to be part of the protective program steered by HIF-1 in response to oxygen deprivation. Induction of BNP may therefore contribute to the potential benefits of pharmacological HIF inducers in the treatment of ischaemic heart disease and heart failure.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia , Péptido Natriurético Encefálico/metabolismo , Células 3T3-L1 , Secuencias de Aminoácidos , Animales , Humanos , Masculino , Ratones , Infarto del Miocardio/patología , Isquemia Miocárdica/patología , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Ratas Wistar
17.
J Am Soc Nephrol ; 19(3): 486-94, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18256363

RESUMEN

The contribution of hypoxia to cisplatin-induced renal tubular injury is controversial. Because the hypoxia-inducible factor (HIF) pathway is a master regulator of adaptation to hypoxia, we measured the effects of cisplatin on HIF accumulation in vitro and in vivo, and tested whether hypoxic preconditioning is protective against cisplatin-induced injury. We found that cisplatin did not stabilize HIF-1alpha protein in vitro or in vivo under normoxic conditions. However, hypoxic preconditioning of cisplatin-treated proximal tubular cells in culture reduced apoptosis in an HIF-1alpha-dependent fashion and increased cell proliferation as measured by BrdU incorporation. In vivo, rats preconditioned with carbon monoxide before cisplatin administration had significantly better renal function than rats kept in normoxic conditions throughout. Moreover, the histomorphological extent of renal damage and tubular apoptosis was reduced by the preconditional treatment. Therefore, development of pharmacologic agents to induce renal HIF might provide a new approach to ameliorate cisplatin-induced nephrotoxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia , Riñón/efectos de los fármacos , Lesión Renal Aguda/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Monóxido de Carbono/farmacología , Línea Celular , ADN/biosíntesis , Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Riñón/metabolismo , Riñón/patología , Pruebas de Función Renal , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
18.
Nephrol Dial Transplant ; 23(11): 3456-63, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18515791

RESUMEN

OBJECTIVE: We examined the contribution of high blood pressure versus direct mineralocorticoid effects to the progression of kidney inflammation and fibrosis in established experimental deoxycorticosterone-acetate (DOCA)-salt hypertension. METHODS: Male Sprague-Dawley rats underwent unilateral nephrectomy and received subcutaneous DOCA pellets as well as 1% NaCl for drinking. After 4 weeks of DOCA-salt hypertension, rats were either killed (n = 6), or treated with a non-hypotensive dose of spironolactone (n = 7) or triple therapy (hydrochlorothiazide, reserpine and hydralazine, n = 8) to normalize blood pressure or with vehicle (n = 19) for two further weeks. Mean arterial pressure (MAP) was measured intra-arterially. Glomerulosclerosis, interstitial fibrosis, macrophage infiltration and complement deposition were evaluated on kidney sections. Expression of collagens, chemokines and cytokines was measured by real-time PCR. RESULTS: MAP was elevated in DOCA rats, not affected by spironolactone and normalized by triple therapy. Glomerulosclerosis and interstitial fibrosis of DOCA rats were alleviated by spironolactone and triple therapy. Macrophage infiltration, complement C3 deposition and nitrotyrosine staining in the kidney were significantly reduced by spironolactone as well as triple therapy. The expression of collagens, chemokines, adhesion molecules and profibrotic cytokines in the kidney was elevated in hypertension and decreased by triple therapy but not significantly affected by spironolactone. CONCLUSION: Direct mineralocorticoid effects as well as high blood pressure per se contribute to inflammation and fibrosis of the kidney. Oxidative stress may mediate the direct mineralocorticoid effects on kidney inflammation.


Asunto(s)
Presión Sanguínea/fisiología , Glomerulonefritis/fisiopatología , Hipertensión/fisiopatología , Mineralocorticoides/efectos adversos , Nefroesclerosis/fisiopatología , Animales , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Desoxicorticosterona , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Diuréticos/farmacología , Relación Dosis-Respuesta a Droga , Glomerulonefritis/patología , Hidralazina/farmacología , Hidroclorotiazida/farmacología , Hipertensión/inducido químicamente , Hipertensión/patología , Masculino , Antagonistas de Receptores de Mineralocorticoides , Mineralocorticoides/farmacología , Nefroesclerosis/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reserpina/farmacología , Espironolactona/farmacología
19.
J Mol Med (Berl) ; 85(4): 343-50, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17333097

RESUMEN

Polymorphisms of the angiotensinogen (Agt) gene may affect blood pressure. We used a mouse model to test for the role of the Agt genotype in low-renin or high-renin forms of hypertension. Mice bearing one, two, three, or four copies of the Agt gene underwent renal artery clipping to induce high-renin two-kidney, one-clip renovascular hypertension (2K1C), or uninephrectomy, salt loading, and application of deoxycorticosterone-acetate (DOCA) pellets to induce low-renin mineralocorticoid hypertension. Appropriate control animals were also studied. Blood pressure was measured by tail cuff as well as by direct intra-arterial recordings. There was a small effect of the Agt genotype on baseline blood pressure before induction of hypertension. The extent of 2K1C hypertension was not affected by the genotype. In contrast, there was a marked gene-dose effect on DOCA-hypertension (21.2 mmHg over all genotypes). Treatment of DOCA mice with the angiotensin II type 1 receptor antagonist abolished the genotype effect on blood pressure and left ventricular hypertrophy. There was a trend towards less suppression of endogenous aldosterone by DOCA treatment with increasing number of Agt gene copies. We conclude that the Agt genotype exerts a marked effect on blood pressure in a low-renin form of hypertension but no effect in the face of stimulated renin, at least in mice.


Asunto(s)
Angiotensinógeno/genética , Hipertensión/genética , Sistema Renina-Angiotensina/genética , Angiotensinógeno/fisiología , Animales , Modelos Animales de Enfermedad , Genotipo , Hipertensión/sangre , Hipertensión/tratamiento farmacológico , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL
20.
J Mol Med (Berl) ; 93(8): 891-904, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25754172

RESUMEN

UNLABELLED: Hypoxia-inducible transcription factors (HIFs) control cellular adaptation to low oxygen. In the kidney, activation of HIF is beneficial during injury; however, the specific contribution of HIF-1α in renal endothelial cells (EC) remains elusive. Since EC display tissue-specific heterogeneity, we investigated how HIF-1α affects key functions of glomerular EC in vitro and its contribution to renal development and pathophysiological adaptation to acute or chronic renal injury in vivo. Loss of HIF-1α in glomerular EC induces hypoxic cell death and reduces hypoxic adhesion of macrophages in vitro. In vivo, HIF-1α expression in EC in mouse kidneys is detectable but limited. Accordingly, EC-specific ablation of HIF-1α does not lead to developmental or phenotypical abnormalities in the kidney. Renal function and expression of adhesion molecules during acute ischemic kidney injury is independent of HIF-1α in EC. Likewise, inflammation and development of fibrosis after unilateral ureteric obstruction is not influenced by endothelial HIF-1α. Taken together, although HIF-1α exerts effects on glomerular EC in vitro, endothelial HIF-1α does not influence renal development and pathophysiological adaptation to kidney injury in vivo. This implies a profound difference of the hypoxic response of the renal vascular bed compared to other organs, such as the heart. This has implications for the development of pharmacological strategies targeting the endothelial hypoxic response pathways. KEY MESSAGE: HIF-1α controls hypoxic survival and adhesion on endothelial cells (EC) in vitro. In vivo, HIF-1α expression in renal EC is low. Deletion of HIF-1α in EC does not affect kidney development and function in mice. Renal function after acute and chronic kidney injury is independent of HIF-1α in EC. Data suggest organ-specific regulation of HIF-1α function in EC.


Asunto(s)
Células Endoteliales/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/lesiones , Riñón/patología , Daño por Reperfusión/patología , Animales , Línea Celular , Células Endoteliales/metabolismo , Fibrosis , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Ratones , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA