Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pflugers Arch ; 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39042141

RESUMEN

Advanced glycation endproducts (AGEs) contribute to cellular damage of various pathologies, including kidney diseases. Acute kidney injury (AKI) represents a syndrome seldom characterized by a single, distinct pathophysiological cause. Rhabdomyolysis-induced acute kidney injury (RIAKI) constitutes roughly 15% of AKI cases, yet its underlying pathophysiology remains poorly understood. Using a murine model of RIAKI induced by muscular glycerol injection, we observed elevated levels of AGEs and the AGE receptor galectin-3 (LGALS3) in the kidney. Immunofluorescence localized LGALS3 to distal nephron segments. According to transcriptomic profiling via next-generation sequencing, RIAKI led to profound changes in kidney metabolism, oxidative stress, and inflammation. Cellular stress was evident in both proximal and distal tubules, as shown by kidney injury markers KIM-1 and NGAL. However, only proximal tubules exhibited overt damage and apoptosis, as detected by routine morphology, active Caspase-3, and TUNEL assay, respectively. In vitro, distal convoluted tubule (DCT) cells challenged with AGEs underwent apoptosis, which was markedly enhanced by Lgals3 siRNA treatment. Thus, in RIAKI, the upregulation of LGALS3 may protect the distal nephron from AGE-mediated damage, while proximal tubules lacking LGALS3 stay at risk. Thus, stimulating LGALS3 in the proximal nephron, if achievable, may attenuate RIAKI.

2.
Kidney Int ; 104(1): 90-107, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37121432

RESUMEN

The polyamines spermidine and spermine and their common precursor molecule putrescine are involved in tissue injury and repair. Here, we test the hypothesis that impaired polyamine homeostasis contributes to various kidney pathologies in mice during experimental models of ischemia-reperfusion, transplantation, rhabdomyolysis, cyclosporine treatment, arterial hypertension, diabetes, unilateral ureteral obstruction, high oxalate feeding, and adenine-induced injuries. We found a remarkably similar pattern in most kidney pathologies with reduced expression of enzymes involved in polyamine synthesis together with increased expression of polyamine degrading enzymes. Transcript levels of amine oxidase copper-containing 1 (Aoc1), an enzyme which catalyzes the breakdown of putrescine, were barely detectable by in situ mRNA hybridization in healthy kidneys. Aoc1 was highly expressed upon various experimental kidney injuries resulting in a significant reduction of kidney putrescine content. Kidney levels of spermine were also significantly reduced, whereas spermidine was increased in response to ischemia-reperfusion injury. Increased Aoc1 expression in injured kidneys was mainly accounted for by an Aoc1 isoform that harbors 22 additional amino acids at its N-terminus and shows increased secretion. Mice with germline deletion of Aoc1 and injured kidneys showed no decrease of kidney putrescine content; although they displayed no overt phenotype, they had fewer tubular casts upon ischemia-reperfusion injury. Hyperosmotic stress stimulated AOC1 expression at the transcriptional and post-transcription levels in metanephric explants and kidney cell lines. AOC1 expression was also significantly enhanced after kidney transplantation in humans. These data demonstrate that the kidneys respond to various forms of injury with down-regulation of polyamine synthesis and activation of the polyamine breakdown pathway. Thus, an imbalance in kidney polyamines may contribute to various etiologies of kidney injury.


Asunto(s)
Amina Oxidasa (conteniendo Cobre) , Daño por Reperfusión , Humanos , Ratones , Animales , Poliaminas/metabolismo , Espermidina/metabolismo , Putrescina/metabolismo , Espermina/metabolismo , Espermina/farmacología , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Riñón/patología , Amina Oxidasa (conteniendo Cobre)/metabolismo , Daño por Reperfusión/patología , Expresión Génica
3.
Kidney Int ; 101(4): 670-673, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35314049

RESUMEN

Class 3 semaphorins (SEMA3) are secreted glycoproteins with established roles in the developing brain, heart, and kidney. In this issue of Kidney International, Cai et al. show that in acutely injured kidneys, semaphorin isoform SEMA3C is expressed de novo in glomeruli and the nephron, secreted into the circulation, and excreted into the urine. Compelling evidence is provided for SEMA3C promoting microvascular permeability, kidney swelling, and acute injury. SEMA3C antagonism may be a treatment option for acute kidney injury.


Asunto(s)
Permeabilidad Capilar , Semaforinas , Glomérulos Renales/metabolismo , Semaforinas/metabolismo
4.
Kidney Int ; 102(4): 750-765, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35643373

RESUMEN

Chronic Cyclosporine-A treatment is associated with serious side effects, including kidney toxicity and anemia. Although pathophysiology of Cyclosporine-A-induced kidney injury remains incompletely understood, hypoxia is likely involved. Here, we investigated the effect of the hypoxia inducible factor activator daprodustat on Cyclosporine-A -induced kidney toxicity. As Cyclosporine-A profoundly alters protein phosphorylation by inhibiting the phosphatase calcineurin, special attention was directed towards the kidney phospho-proteome. Mice received Cyclosporine-A with or without daprodustat for up to eight weeks. In kidney homogenates, 1360 selected proteins were analyzed at expression and phosphorylation levels. Of these, Cyclosporine-A changed the expression of 79 and the phosphorylation of 86 proteins. However, when Cyclosporine-A treatment was combined with daprodustat, the expression of 95 proteins and phosphorylation of only six proteins was altered suggesting that daprodustat prevented most protein phosphorylation brought about by Cyclosporine-A. Although daprodustat showed only marginal effect on its own, angiogenesis-related pathways were among the most profoundly impacted by daprodustat when given on top of Cyclosporine-A. Additionally, Cyclosporine-A lowered the blood hemoglobin concentration and caused kidney capillary rarefaction, which daprodustat prevented. Thus, combined daprodustat/Cyclosporine-A treatment prevented deleterious Cyclosporine-A effects on microcirculation and hemoglobin, and the protective action of daprodustat involves suppression of broad protein phosphorylation changes caused by Cyclosporine-A.


Asunto(s)
Anemia , Ciclosporina , Anemia/inducido químicamente , Anemia/prevención & control , Animales , Barbitúricos , Calcineurina , Ciclosporina/toxicidad , Glicina/análogos & derivados , Hemoglobinas/metabolismo , Hipoxia/complicaciones , Ratones , Proteoma
5.
Am J Physiol Renal Physiol ; 318(5): F1086-F1099, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32174143

RESUMEN

Unilateral ischemia-reperfusion (UIR) injury leads to progressive renal atrophy and tubulointerstitial fibrosis (TIF) and is commonly used to investigate the pathogenesis of the acute kidney injury-chronic kidney disease transition. Although it is well known that contralateral nephrectomy (CNX), even 2 wk post-UIR injury, can improve recovery, the physiological mechanisms and tubular signaling pathways mediating such improved recovery remain poorly defined. Here, we examined the renal hemodynamic and tubular signaling pathways associated with UIR injury and its reversal by CNX. Male Sprague-Dawley rats underwent left UIR or sham UIR and 2 wk later CNX or sham CNX. Blood pressure, left renal blood flow (RBF), and total glomerular filtration rate were assessed in conscious rats for 3 days before and over 2 wk after CNX or sham CNX. In the presence of a contralateral uninjured kidney, left RBF was lower (P < 0.05) from 2 to 4 wk following UIR (3.6 ± 0.3 mL/min) versus sham UIR (9.6 ± 0.3 mL/min). Without CNX, extensive renal atrophy, TIF, and tubule dedifferentiation, but minimal pimonidazole and hypoxia-inducible factor-1α positivity in tubules, were present at 4 wk post-UIR injury. Conversely, CNX led (P < 0.05) to sustained increases in left RBF (6.2 ± 0.6 mL/min) that preceded the increases in glomerular filtration rate. The CNX-induced improvement in renal function was associated with renal hypertrophy, more redifferentiated tubules, less TIF, and robust pimonidazole and hypoxia-inducible factor-1α staining in UIR injured kidneys. Thus, contrary to expectations, indexes of hypoxia are not observed with the extensive TIF at 4 wk post-UIR injury in the absence of CNX but are rather associated with the improved recovery of renal function and structure following CNX.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Riñón/irrigación sanguínea , Circulación Renal , Insuficiencia Renal Crónica/etiología , Daño por Reperfusión/fisiopatología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Animales , Atrofia , Hipoxia de la Célula , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Hemodinámica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/metabolismo , Riñón/patología , Masculino , Nefrectomía , Ratas Sprague-Dawley , Recuperación de la Función , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Nephrol Dial Transplant ; 35(2): 206-212, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30768198

RESUMEN

Concepts regarding hypoxic acute kidney injury (AKI) are derived from widely used warm ischemia-reflow (WIR) models, characterized by extensive proximal tubular injury and associated with profound inflammation. However, there is ample clinical and experimental data indicating that hypoxic AKI may develop without total cessation of renal blood flow, with a different injury pattern that principally affects medullary thick limbs in the outer medulla. This injury pattern likely reflects an imbalance between blood and oxygen supply and oxygen expenditure, principally for tubular transport. Experimental models of hypoxic AKI other than WIR are based on mismatched oxygen delivery and consumption, particularly within the physiologically hypoxic outer medulla. However, evidence for such circumstances in human AKI is lacking. Recent analysis of the clinical course and laboratory findings of patients following near-drowning (ND) provides a rare glimpse into such a scenario. This observation supports the role of renal hypoxia in the evolution of AKI, as renal impairment could be predicted by the degree of whole-body hypoxia (reflected by lactic acidosis). Furthermore, there was a close association of renal functional impairment with indices of reduced oxygen delivery (respiratory failure and features of intense sympathetic activity) and of enhanced oxygen consumption for active tubular transport (extrapolated from the calculated volume of consumed hypertonic seawater). This unique study in humans supports the concept of renal oxygenation imbalance in hypoxic AKI. The drowning scenario, particularly in seawater, may serve as an archetype of this disorder, resulting from reduced oxygen delivery, combined with intensified oxygen consumption for tubular transport.


Asunto(s)
Lesión Renal Aguda/etiología , Hipoxia/complicaciones , Oxígeno/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Humanos , Consumo de Oxígeno , Circulación Renal
7.
Cell Physiol Biochem ; 52(5): 1075-1091, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30977989

RESUMEN

BACKGROUND/AIMS: Recently, we have demonstrated that episodic hypoxia occurs in kidneys of mice challenged repetitively with the immunosuppressant cyclosporine A (CsA), in analogy to humans on CsA treatment. However, the molecular consequences of episodic hypoxia remain poorly defined, as is its impact on cell survival. Here, we systematically study cell response to episodic, as compared to single course hypoxia. METHODS: In vivo, kidneys of mice challenged daily with CsA for one week were analyzed by microarray analysis, gene ontology analysis, and qPCR. In vitro, renal cells were subjected to hypoxia (1 % O2) which was either episodic (4 h for 6 consecutive days), short-term (4 h), or sustained (24 h). Western blot analysis quantified hypoxia-inducible factor-1α (HIF-1α). 2',7'-dichlorofluorescein diacetate detected intracellular ROS. After re-oxygenation, staurosporine served to induce apoptosis, quantified by active caspase-3. RESULTS: In vivo, HIF target gene expression was suppressed by daily CsA treatment. Yet, we found up-regulation of genes involved in defence against cellular stress, notably against ROS. Renal cells in vitro behaved largely different under episodic and sustained hypoxia, while their response to short-term hypoxia oscillated between the previous two. Episodic hypoxia exhibited the highest total HIF-1α protein level, lowest nucleus-to-cytoplasm ratio, and lowest HIF target gene expression. When compared with normoxia, re-oxygenation after sustained hypoxia increased ROS by 3.04 ± 1.04 fold (p<0.001), and re-oxygenation after episodic hypoxia by 1.26 ± 0.16 fold (p<0.01). Staurosporine-induced active caspase-3 was highest after sustained, and lowest after episodic hypoxia. CONCLUSION: In vitro episodic hypoxia mimics the largely HIF-independent transcriptome observed after repetitive CsA treatment in vivo. In vitro preconditioning with episodic hypoxia protects against stress-induced apoptosis. Despite of its long-term adverse effects, CsA derived episodic hypoxia induces a unique renal hypoxia response that provides adaptation to re-oxygenation mediated ROS damage.


Asunto(s)
Adaptación Fisiológica , Apoptosis , Hipoxia , Riñón , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclosporina/farmacología , Hipoxia/metabolismo , Hipoxia/patología , Hipoxia/fisiopatología , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Ratones , Ratones Transgénicos
8.
Kidney Int ; 87(4): 761-70, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25469848

RESUMEN

Renal endothelin-converting enzyme (ECE)-1 is induced in experimental diabetes and following radiocontrast administration, conditions characterized by renal hypoxia, hypoxia-inducible factor (HIF) stabilization, and enhanced endothelin synthesis. Here we tested whether ECE-1 might be a HIF-target gene in vitro and in vivo. ECE-1 transcription and expression increased in cultured vascular endothelial and proximal tubular cell lines, subject to hypoxia, to mimosine or cobalt chloride. These interventions are known to stabilize HIF signaling by inhibition of HIF-prolyl hydroxylases. In rats, HIF-prolyl-hydroxylase inhibition by mimosine or FG-4497 increased HIF-1α immunostaining in renal tubules, principally in distal nephron segments. This was associated with markedly enhanced ECE-1 protein expression, predominantly in the renal medulla. A progressive and dramatic increase in ECE-1 immunostaining over time, in parallel with enhanced HIF expression, was also noted in conditional von Hippel-Lindau knockout mice. Since HIF and STAT3 are cross-stimulated, we triggered HIF expression by STAT3 activation in mice, transfected by or injected with a chimeric IL-6/IL-6-receptor protein, and found a similar pattern of enhanced ECE-1 expression. Chromatin immunoprecipitation sequence (ChIP-seq) and PCR analysis in hypoxic endothelial cells identified HIF binding at the ECE-1 promoter and intron regions. Thus, our findings suggest that ECE-1 may be a novel HIF-target gene.


Asunto(s)
Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Animales , Hipoxia de la Célula/fisiología , Células Cultivadas , Cobalto/farmacología , Dioxigenasas/antagonistas & inhibidores , Enzimas Convertidoras de Endotelina , Células Endoteliales de la Vena Umbilical Humana , Humanos , Intrones , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mimosina/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidores de Prolil-Hidroxilasa/farmacología , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transcripción Genética , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
9.
World J Urol ; 32(6): 1485-92, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24378824

RESUMEN

PURPOSE: Recently, a proteomic study of sera from patients with bladder cancer identified S100A8 and S100A9 as tumor-associated proteins. The present cross-sectional study investigates whether calprotectin, the heterodimer of S100A8/S100A9 may serve as a urinary biomarker for the detection of urothelial bladder cancer. METHODS: Urinary calprotectin concentrations were assessed in a population of 181 subjects including 46 cases of bladder cancer. 41 cases of renal cell cancer, 54 cases of prostate cancer, and 40 healthy subjects served as control. Acute kidney injury, urinary tract infection, previous BCG-treatment and secondary transurethral resection of the bladder tumor were defined as exclusion criteria. Assessment was performed by enzyme-linked immunosorbent assay and immunohistochemistry detecting calprotectin. RESULTS: Median calprotectin concentrations (ng/ml) were significantly higher in patients with bladder cancer than in healthy controls (522.3 vs. 51.0, p < 0.001), renal cell cancer (90.4, p < 0.001), and prostate cancer (71.8, p < 0.001). In urothelial carcinoma prominent immunostaining occurred in a subset of tumor cells and in infiltrating myeloid cells. Receiver operating characteristic analysis provided an area under the curve of 0.88 for the differentiation of bladder cancer and healthy control. A cut-off value of 140 ng/ml (determined by Youden's index) resulted in sensitivity and specificity values of 80.4 and 92.5 %. Low grade tumors were associated with significantly lower calprotectin concentrations than high grade tumors (351.9 vs. 1635.2 ng/ml, p = 0.004). CONCLUSIONS: Urothelial malignancies are associated with highly increased concentrations of calprotecin in the urine. In absence of renal failure and pyuria, calprotectin constitutes a promising biomarker for the detection of bladder cancer.


Asunto(s)
Biomarcadores de Tumor/orina , Carcinoma/diagnóstico , Complejo de Antígeno L1 de Leucocito/orina , Neoplasias de la Vejiga Urinaria/diagnóstico , Anciano , Carcinoma/orina , Estudios Transversales , Femenino , Humanos , Neoplasias Renales/orina , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Neoplasias de la Próstata/orina , Curva ROC , Neoplasias de la Vejiga Urinaria/orina , Urotelio
10.
J Am Soc Nephrol ; 24(11): 1806-19, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23970125

RESUMEN

Renal hypoxia occurs in AKI of various etiologies, but adaptation to hypoxia, mediated by hypoxia-inducible factor (HIF), is incomplete in these conditions. Preconditional HIF activation protects against renal ischemia-reperfusion injury, yet the mechanisms involved are largely unknown, and HIF-mediated renoprotection has not been examined in other causes of AKI. Here, we show that selective activation of HIF in renal tubules, through Pax8-rtTA-based inducible knockout of von Hippel-Lindau protein (VHL-KO), protects from rhabdomyolysis-induced AKI. In this model, HIF activation correlated inversely with tubular injury. Specifically, VHL deletion attenuated the increased levels of serum creatinine/urea, caspase-3 protein, and tubular necrosis induced by rhabdomyolysis in wild-type mice. Moreover, HIF activation in nephron segments at risk for injury occurred only in VHL-KO animals. At day 1 after rhabdomyolysis, when tubular injury may be reversible, the HIF-mediated renoprotection in VHL-KO mice was associated with activated glycolysis, cellular glucose uptake and utilization, autophagy, vasodilation, and proton removal, as demonstrated by quantitative PCR, pathway enrichment analysis, and immunohistochemistry. In conclusion, a HIF-mediated shift toward improved energy supply may protect against acute tubular injury in various forms of AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Rabdomiólisis/complicaciones , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Caspasa 3/análisis , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Riñón/patología , Masculino , Ratones , Ratones Noqueados
11.
Clin Kidney J ; 17(8): sfae205, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39135937

RESUMEN

Anaemia is common in chronic kidney disease (CKD) and has a significant impact on quality of life (QoL), work productivity and outcomes. Current management includes oral or intravenous iron and erythropoiesis-stimulating agents (ESAs), to which hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PHIs) have been recently added, increasing the available therapeutic options. In randomised controlled trials, only intravenous iron improved cardiovascular outcome, while some ESAs were associated with increased adverse cardiovascular events. Despite therapeutic advances, several challenges and unmet needs remain in the current management of anaemia of CKD. In particular, clinical practice does not include an assessment of QoL, which prompted a group of European nephrologists and representatives of patient advocacy groups to revisit the current approach. In this consensus document, the authors propose a move towards a more holistic, personalised and long-term approach, based on existing evidence. The focus of treatment should be on improving QoL without increasing the risk of adverse cardiovascular events, and tailoring management strategies to the needs of the individual. In addition, the authors discuss the suitability of a currently available anaemia of CKD-specific health-related QoL measure for inclusion in the routine clinical management of anaemia of CKD. The authors also outline the logistics and challenges of incorporating such a measure into electronic health records and how it may be used to improve QoL for people with anaemia of CKD.

12.
Clin Exp Pharmacol Physiol ; 40(2): 148-57, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22905709

RESUMEN

Hypoxia plays a crucial role in the pathophysiology of acute kidney injury (AKI) and presumably also chronic kidney disease (CKD). Hypoxia-inducible factor (HIF) is the master transcription factor that regulates adaptive responses against hypoxia. Under hypoxic conditions, HIF activates target genes with hypoxia-responsive elements in their regulatory regions. The HIF isoforms and regulators of HIF (i.e. prolyl hydroxylases) show cell type-specific distributions. Hypoxia is observed in both ischaemic and so-called non-ischaemic forms of AKI. In addition to the acute phase, hypoxia may ensue during the recovery phase of AKI, possibly due to the oxygen-consuming processes of cell growth and proliferation for repair. Although HIF protects the kidney against AKI, intrinsic HIF activation is submaximal in AKI and further augmentation of HIF ameliorates disease manifestations. The kidney in CKD also suffers from hypoxia caused by multiple mechanisms, including sustained oxygen demands in the remaining nephrons due to maladaptive tubuloglomerular feedback. Whether HIF is chronically upregulated in CKD is contentious. Hypoxia-inducible factor activation is a promising therapeutic approach to CKD, but excessive activation of HIF may be deleterious. It is likely that there is a therapeutic window of HIF activation in chronic conditions. Under certain circumstances, animals with CKD are protected against AKI and this may be explained by non-physiological hypoxia of the kidney and subsequent HIF expression. In addition, an acute hypoxic insult may induce long-lasting changes, possibly including epigenetic modifications induced by HIF. These observations suggest a complex interaction between AKI and CKD via hypoxia and HIF activation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Factor 1 Inducible por Hipoxia/fisiología , Enfermedades Renales/metabolismo , Animales , Humanos , Enfermedades Renales/patología , Enfermedades Renales/terapia
13.
Clin Exp Pharmacol Physiol ; 40(4): 262-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23384058

RESUMEN

In vitro studies suggest that combined activation of hypoxia-inducible factor (HIF) and signal transducer and activator of transcription 3 (STAT3) promotes the hypoxia response. However, their interrelationship in vivo remains poorly defined. The present study investigated the possible relationship between HIF-1 upregulation and STAT3 activation in the rodent kidney in vivo. Activation of HIF-1 and STAT3 was analysed by immunohistochemical staining and western blot analysis in: (i) models of hypoxia-associated kidney injury induced by radiocontrast media or rhabdomyolysis; (ii) following activation of STAT3 by the interleukin (IL)-6-soluble IL-6 receptor complex; or (iii) following HIF-1α stabilization using hypoxic and non-hypoxic stimuli (mimosine, FG-4497, CO, CoCl(2)) and in targeted von Hippel-Lindau-knockout mice. Western blot analysis and immunostaining revealed marked induction of both transcription factors under all conditions tested, suggesting that in vivo STAT3 can trigger HIF and vice versa. Colocalization of HIF-1α and phosphorylated STAT3 was detected in some, but not all, renal cell types, suggesting that in some cells a paracrine mechanism may be responsible for the reciprocal activation of the two transcription factors. Nevertheless, in several cell types spatial concordance was observed under the majority of conditions tested, suggesting that HIF-1 and STAT3 may act as cotranscription factors. These in vivo studies suggest that, in response to renal hypoxic-stress, upregulation of HIF-1 and activation of STAT3 may be both reciprocal and cell type dependent.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Riñón/metabolismo , Factor de Transcripción STAT3/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Animales , Hipoxia/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Oxígeno/farmacología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
14.
J Am Soc Nephrol ; 23(3): 483-93, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22266667

RESUMEN

Nephron loss in a diseased kidney invokes adaptations in the remaining nephrons. Whether and how these adaptations condition the response of the kidney to injury is not known. We examined the susceptibility of the kidney after subtotal (5/6th) nephrectomy (STN) to ischemic injury in rats. GFR in STN kidneys did not significantly change after ischemia reperfusion (IR), whereas GFR fell by 70% after IR in unilateral nephrectomy controls. In micropuncture experiments, single-nephron GFR responses mirrored the whole-kidney responses: in STN, single-nephron GFR decreased by 7% after IR compared with 28% in controls. Furthermore, we found that tubuloglomerular feedback, a mechanism that links proximal tubular injury to a fall in GFR, was inoperative in STN but was normal in controls. Restoration of normal feedback in STN attenuated the functional resistance to IR. In addition to the functional resilience, the morphology of the kidney was better preserved in STN. In STN kidneys, the S3 segment of the proximal tubule, normally injured after ischemia, constitutively expressed hypoxia-inducible factor-1α (HIF-1α), which is cytoprotective in ischemia. Inducing HIF before IR improved GFR in control animals, and inhibiting the HIF target heme-oxygenase-1 before IR reduced GFR in STN animals. Taken together, these data suggest that fewer functioning nephrons in a diseased kidney do not increase the susceptibility to injury, but rather, hemodynamic and molecular adaptations in the remnant nephrons precondition them against ischemic injury.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Glomérulos Renales/fisiología , Túbulos Renales Proximales/fisiología , Nefrectomía , Daño por Reperfusión/prevención & control , Daño por Reperfusión/fisiopatología , Adaptación Fisiológica/fisiología , Animales , Tasa de Filtración Glomerular/fisiología , Riñón/patología , Riñón/cirugía , Glomérulos Renales/patología , Túbulos Renales Proximales/patología , Masculino , Modelos Animales , Nefronas/fisiología , Punciones , Ratas , Ratas Wistar , Transducción de Señal/fisiología
15.
Nephrol Dial Transplant ; 27(5): 1721-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22547749

RESUMEN

Hypoxia plays a role in ischemic, toxic and sepsis-induced acute kidney injury. Evolving hypoxia triggers renal adaptive responses that may mitigate the insult, leading to sublethal forms of cell injury. The unique capability of the kidney to downregulate oxygen consumption for tubular transport could represent one such adaptive response which promotes maintenance of renal oxygenation, thereby preserving cellular integrity. Tran et al. recently explored a novel mechanism that might prevent tubular damage by downregulation of mitochondrial biogenesis and oxygen consumption. Using expression profiling of kidney RNA in endotoxemic rodents and complementary studies in vitro and in PGC-1α knockout mice, they found a sepsis-related decline in PPARγ coactivator-1α (PGC-1α) expression and of PGC-1α-dependent genes involved in oxidative phosphorylation. This response may explain their observation of a paradoxical preservation of kidney oxygenation and structural integrity in sepsis, despite reduced renal blood flow and oxygen delivery. Thus, resetting of mitochondrial respiration and oxygen consumption during sepsis might be added to the growing list of adaptive responses that occur during hypoxic stress. This review will focus on these mechanisms that mitigate evolving hypoxic injury, even at the expense of transient renal dysfunction.


Asunto(s)
Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Adaptación Fisiológica/fisiología , Hipoxia/fisiopatología , Animales , Humanos , Ratones , Ratones Noqueados , Microcirculación/fisiología , Mitocondrias/fisiología , Modelos Animales , Consumo de Oxígeno/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transactivadores/fisiología , Factores de Transcripción
16.
Proc Natl Acad Sci U S A ; 105(1): 282-7, 2008 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-18162550

RESUMEN

A key energy-saving adaptation to chronic hypoxia that enables cardiomyocytes to withstand severe ischemic insults is hibernation, i.e., a reversible arrest of contractile function. Whereas hibernating cardiomyocytes represent the critical reserve of dysfunctional cells that can be potentially rescued, a lack of a suitable animal model has hampered insights on this medically important condition. We developed a transgenic mouse system for conditional induction of long-term hibernation and a system to rescue hibernating cardiomyocytes at will. Via myocardium-specific induction (and, in turn, deinduction) of a VEGF-sequestering soluble receptor, we show that VEGF is indispensable for adjusting the coronary vasculature to match increased oxygen consumption and exploit this finding to generate a hypoperfused heart. Importantly, ensuing ischemia is tunable to a level at which large cohorts of cardiomyocytes are driven to enter a hibernation mode, without cardiac cell death. Relieving the VEGF blockade even months later resulted in rapid revascularization and full recovery of contractile function. Furthermore, we show that left ventricular remodeling associated with hibernation is also fully reversible. The unique opportunity to uncouple hibernation from other ischemic heart phenotypes (e.g., infarction) was used to determine the genetic program of hibernation; uncovering hypoxia-inducible factor target genes associated with metabolic adjustments and induced expression of several cardioprotective genes. Autophagy, specifically self-digestion of mitochondria, was identified as a key prosurvival mechanism in hibernating cardiomyocytes. This system may lend itself for examining the potential utility of treatments to rescue dysfunctional cardiomyocytes and reverse maladaptive remodeling.


Asunto(s)
Hibernación , Modelos Genéticos , Miocardio/patología , Transgenes , Animales , Perfilación de la Expresión Génica , Corazón/fisiología , Hipoxia , Isquemia/patología , Ratones , Ratones Transgénicos , Isquemia Miocárdica , Neovascularización Fisiológica , Oxígeno/metabolismo , Fenotipo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Kidney Int ; 77(1): 9-16, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19759527

RESUMEN

Although the understanding of processes associated with hypoxic tubular cell injury has remarkably improved, controversies remain regarding the appropriateness of various animal models to the human syndrome of acute kidney injury (AKI). We herein compare available experimental models of hypoxic acute kidney damage, which differ both conceptually and morphologically in the distribution of tubular cell injury. Tubular segment types differ in their capacity to mount hypoxia-adaptive responses, mediated by hypoxia-inducible factors (HIFs), and in cell type-specific molecules shed into the urine, which may serve as early biomarkers for renal damage. These differences may be of value in the perception of the human AKI, its detection, and prevention.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Renales/etiología , Daño por Reperfusión/patología , Animales , Humanos , Hipoxia , Enfermedades Renales/patología , Necrosis Tubular Aguda/etiología , Necrosis Tubular Aguda/patología , Túbulos Renales Distales/patología , Túbulos Renales Proximales/patología
18.
Kidney Int ; 77(4): 312-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20016470

RESUMEN

The adaptation of erythropoietin production to oxygen supply is determined by the abundance of hypoxia-inducible factor (HIF), a regulation that is induced by a prolyl hydroxylase. To identify cells that express HIF subunits (HIF-1alpha and HIF-2alpha) and erythropoietin, we treated Sprague-Dawley rats with the prolyl hydroxylase inhibitor FG-4497 for 6 h to induce HIF-dependent erythropoietin transcription. The kidneys were analyzed for colocalization of erythropoietin mRNA with HIF-1alpha and/or HIF-2alpha protein along with cell-specific identification markers. FG-4497 treatment strongly induced erythropoietin mRNA exclusively in cortical interstitial fibroblasts. Accumulation of HIF-2alpha was observed in these fibroblasts and in endothelial and glomerular cells, whereas HIF-1alpha was induced only in tubular epithelia. A large proportion (over 90% in the juxtamedullary cortex) of erythropoietin-expressing cells coexpressed HIF-2alpha. No colocalization of erythropoietin and HIF-1alpha was found. Hence, we conclude that in the adult kidney, HIF-2alpha and erythropoietin mRNA colocalize only in cortical interstitial fibroblasts, which makes them the key cell type for renal erythropoietin synthesis as regulated by HIF-2alpha.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Eritropoyetina/biosíntesis , Fibroblastos/metabolismo , Riñón/citología , Riñón/metabolismo , Animales , Ratas , Ratas Sprague-Dawley
20.
Methods Mol Biol ; 466: 161-74, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19148611

RESUMEN

Immunohistochemistry for pimonidazole adducts serves to define hypoxia within tissues. For this purpose, pimonidazole is delivered in vivo, binds to thiol groups at oxygen tensions below 10 mmHg, and is visualized with help of commercially available anti-pimonidazole antibodies. Renal parenchymal oxygen distribution is highly variable under normal conditions and during acute renal failure and chronic renal disorders. Pimonidazole immunostaining clearly helps in delineating hypoxic regions within the kidneys, but technical pitfalls should be taken into account. In particular, tissue fixation by in vivo perfusion is strongly recommended in order to eliminate artificial staining, because immersion fixation per se can promote a hypoxic environment within kidney tissue.


Asunto(s)
Hipoxia/patología , Inmunohistoquímica/métodos , Riñón , Nitroimidazoles , Animales , Riñón/citología , Riñón/metabolismo , Riñón/patología , Estructura Molecular , Nitroimidazoles/química , Nitroimidazoles/metabolismo , Ratas , Fijación del Tejido/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA