Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Medicine (Baltimore) ; 100(11): e25069, 2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33725982

RESUMEN

RATIONALE: Atypical hemolytic uremic syndrome (aHUS) is an uncommon and serious disease that manifests hemolytic anemia, thrombocytopenia, and acute kidney injury. Genetic complement abnormalities have been shown to be responsible. Compared with the aHUS caused by other mutated genes, aHUS secondary to CFB mutation in adults is extremely rare. We report an adult with CFB mutation developing aHUS. PATIENT CONCERNS: A 56-year-old man was admitted for 4-day history of nausea and fatigue, anuria for 2 days, and unconsciousness for 10 hours. DIAGNOSES: The patient presented with life-threatening anemia, thrombocytopenia, acute kidney injury, and nervous system abnormalities. The patient had schistocytes on the peripheral blood smear, increased lactate dehydrogenase (LDH), and plasma-free hemoglobin levels. The patient was later found to harbor a pathogenic variant in the CFB gene (C.1598A>G), and was diagnosed with aHUS and acute kidney injury. INTERVENTION: The patient was treated by plasmapheresis, continuous renal replacement therapy, blood transfusion, and anti-infective and antihypertensive treatment. OUTCOMES: After the treatment, the patient's consciousness returned to normal, and the hemoglobin, platelet, and serum creatinine recovered. The disease activity remained quiescent during the follow-up. LESSONS: A rare heterozygous variant c.1598A>G p.Lys 533Arg in the CFB gene, which was associated with adult-onset aHUS, was described and successfully treated. This case can help in understanding the early diagnosis and effective therapies of this rare disease.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Factor B del Complemento/genética , Necrosis Tubular Aguda/genética , Mutación/genética , Humanos , Masculino , Persona de Mediana Edad
2.
Am J Physiol Renal Physiol ; 317(2): F229-F239, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091122

RESUMEN

Ischemia-reperfusion injury (IRI) is a clinically important cause of acute kidney injury leading to chronic kidney disease. Furthermore, IRI in renal transplantation still remains a risk factor for delayed graft function. Previous studies on IRI have had some limitations, and few of the studied therapies have been clinically applicable. Therefore, a new method for treating renal IRI is needed. We examined the effects of human mesothelial cell (MC) sheets and hepatocyte growth factor (HGF)-transgenic MC (tg MC) sheets transplanted under the renal capsule in an IRI rat model and compared these two treatments with the intravenous administration of HGF protein and no treatment through serum, histological, and mRNA analyses over 28 days. MC sheets and HGF-tg MC sheets produced HGF protein and significantly improved acute renal dysfunction, acute tubular necrosis, and survival rate. The improvement in necrosis was likely due to the cell sheets promoting the migration and proliferation of renal tubular cells, as observed in vitro. Expression of α-smooth muscle actin at day 14 and renal fibrosis at day 28 after IRI were significantly suppressed in MC sheet and HGF-tg MC sheet treatment groups compared with the other groups, and these effects tended to be reinforced by the HGF-tg MC sheets. These results suggest that the cell sheets locally and continuously affect renal paracrine factors, such as HGF, and support recovery from acute tubular necrosis and improvement of renal fibrosis in chronic disease.


Asunto(s)
Células Epiteliales/trasplante , Terapia Genética/métodos , Factor de Crecimiento de Hepatocito/metabolismo , Necrosis Tubular Aguda/terapia , Riñón/cirugía , Daño por Reperfusión/terapia , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Fibrosis , Factor de Crecimiento de Hepatocito/genética , Humanos , Riñón/metabolismo , Riñón/patología , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/metabolismo , Necrosis Tubular Aguda/patología , Masculino , Comunicación Paracrina , Ratas Endogámicas F344 , Ratas Desnudas , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal , Factores de Tiempo
3.
Am J Physiol Renal Physiol ; 312(6): F1166-F1183, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28331061

RESUMEN

Acute kidney injury (AKI) causes severe morbidity, mortality, and chronic kidney disease (CKD). Mortality is particularly marked in the elderly and with preexisting CKD. Oxidative stress is a common theme in models of AKI induced by ischemia-reperfusion (I-R) injury. We recently characterized an intracellular isoform of matrix metalloproteinase-2 (MMP-2) induced by oxidative stress-mediated activation of an alternate promoter in the first intron of the MMP-2 gene. This generates an NH2-terminal truncated MMP-2 (NTT-MMP-2) isoform that is intracellular and associated with mitochondria. The NTT-MMP-2 isoform is expressed in kidneys of 14-mo-old mice and in a mouse model of coronary atherosclerosis and heart failure with CKD. We recently determined that NTT-MMP-2 is induced in human renal transplants with delayed graft function and correlated with tubular cell necrosis. To determine mechanism(s) of action, we generated proximal tubule cell-specific NTT-MMP-2 transgenic mice. Although morphologically normal at the light microscopic level at 4 mo, ultrastructural studies revealed foci of tubular epithelial cell necrosis, the mitochondrial permeability transition, and mitophagy. To determine whether NTT-MMP-2 expression enhances sensitivity to I-R injury, we performed unilateral I-R to induce mild tubular injury in wild-type mice. In contrast, expression of the NTT-MMP-2 isoform resulted in a dramatic increase in tubular cell necrosis, inflammation, and fibrosis. NTT-MMP-2 mice had enhanced expression of innate immunity genes and release of danger-associated molecular pattern molecules. We conclude that NTT-MMP-2 "primes" the kidney to enhanced susceptibility to I-R injury via induction of mitochondrial dysfunction. NTT-MMP-2 may be a novel AKI treatment target.


Asunto(s)
Lesión Renal Aguda/enzimología , Necrosis Tubular Aguda/enzimología , Túbulos Renales Proximales/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Daño por Reperfusión/enzimología , Lesión Renal Aguda/genética , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/patología , Factores de Edad , Animales , Enfermedad de la Arteria Coronaria/enzimología , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Humanos , Inmunidad Innata , Isoenzimas , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/inmunología , Necrosis Tubular Aguda/patología , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/ultraestructura , Metaloproteinasa 2 de la Matriz/genética , Potencial de la Membrana Mitocondrial , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/enzimología , Mitocondrias/ultraestructura , Mitofagia , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Necrosis , Estrés Oxidativo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Transducción de Señal
4.
Kidney Int ; 91(2): 387-401, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27789056

RESUMEN

Acute kidney injury is a devastating disease with high morbidity in hospitalized patients and contributes to the pathogenesis of chronic kidney disease. An underlying mechanism of acute kidney injury involves ischemia-reperfusion injury which, in turn, induces oxidative stress and provokes organ damage. Nrf2 is a master transcription factor that regulates the cellular response to oxidative stress. Here, we examined the role of Nrf2 in the progression of ischemia-reperfusion injury-induced kidney damage in mice using genetic and pharmacological approaches. Both global and tubular-specific Nrf2 activation enhanced gene expression of antioxidant and NADPH synthesis enzymes, including glucose-6-phosphate dehydrogenase, and ameliorated both the initiation of injury in the outer medulla and the progression of tubular damage in the cortex. Myeloid-specific Nrf2 activation was ineffective. Short-term administration of the Nrf2 inducer CDDO during the initial phase of injury ameliorated the late phase of tubular damage. This inducer effectively protected the human proximal tubular cell line HK-2 from oxidative stress-mediated cell death while glucose-6-phosphate dehydrogenase knockdown increased intracellular reactive oxygen species. These findings demonstrate that tubular hyperactivation of Nrf2 in the initial phase of injury prevents the progression of reactive oxygen species-mediated tubular damage by inducing antioxidant enzymes and NADPH synthesis. Thus, Nrf2 may be a promising therapeutic target for preventing acute kidney injury to chronic kidney disease transition.


Asunto(s)
Necrosis Tubular Aguda/prevención & control , Túbulos Renales/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Daño por Reperfusión/prevención & control , Animales , Antioxidantes/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Enzimológica de la Expresión Génica , Genotipo , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Glutatión/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/metabolismo , Necrosis Tubular Aguda/patología , Túbulos Renales/patología , Ratones Noqueados , NADP/metabolismo , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Fenotipo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal , Factores de Tiempo , Transfección
5.
Kidney Int ; 91(2): 352-364, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27692564

RESUMEN

Acute kidney injury is often the result of ischemia reperfusion injury, which leads to activation of coagulation and inflammation, resulting in necrosis of renal tubular epithelial cells. Platelets play a central role in coagulation and inflammatory processes, and it has been shown that platelet activation exacerbates acute kidney injury. However, the mechanism of platelet activation during ischemia reperfusion injury and how platelet activation leads to tissue injury are largely unknown. Here we found that renal ischemia reperfusion injury in mice leads to increased platelet activation in immediate proximity of necrotic cell casts. Furthermore, platelet inhibition by clopidogrel decreased cell necrosis and inflammation, indicating a link between platelet activation and renal tissue damage. Necrotic tubular epithelial cells were found to release extracellular DNA, which, in turn, activated platelets, leading to platelet-granulocyte interaction and formation of neutrophil extracellular traps ex vivo. Renal ischemia reperfusion injury resulted in increased DNA-platelet and DNA-platelet-granulocyte colocalization in tissue and elevated levels of circulating extracellular DNA and platelet factor 4 in mice. After renal ischemia reperfusion injury, neutrophil extracellular traps were formed within renal tissue, which decreased when mice were treated with the platelet inhibitor clopidogrel. Thus, during renal ischemia reperfusion injury, necrotic cell-derived DNA leads to platelet activation, platelet-granulocyte interaction, and subsequent neutrophil extracellular trap formation, leading to renal inflammation and further increase in tissue injury.


Asunto(s)
Plaquetas/metabolismo , ADN/metabolismo , Células Epiteliales/metabolismo , Trampas Extracelulares/metabolismo , Necrosis Tubular Aguda/metabolismo , Túbulos Renales/metabolismo , Activación Plaquetaria , Daño por Reperfusión/metabolismo , Animales , Plaquetas/efectos de los fármacos , Línea Celular , Clopidogrel , ADN/genética , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/patología , Necrosis Tubular Aguda/prevención & control , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones Endogámicos C57BL , Nefritis/genética , Nefritis/metabolismo , Nefritis/patología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Factor Plaquetario 4/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Transducción de Señal , Ticlopidina/análogos & derivados , Ticlopidina/farmacología , Factores de Tiempo
7.
Am J Physiol Renal Physiol ; 300(4): F999-1007, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21228114

RESUMEN

Tamm-Horsfall protein (THP) is a glycoprotein expressed exclusively in thick ascending limbs (TAL) of the kidney. We recently described a novel protective role of THP against acute kidney injury (AKI) via downregulation of inflammation in the outer medulla. Our current study investigates the mechanistic relationships among the status of THP, inflammation, and tubular injury. Using an ischemia-reperfusion model in wild-type and THP-/- mice, we demonstrate that it is the S3 proximal segments but not the THP-deficient TAL that are the main targets of tubular injury during AKI. The injured S3 segments that are surrounded by neutrophils in THP-/- mice have marked overexpression of neutrophil chemoattractant MIP-2 compared with wild-type counterparts. Neutralizing macrophage inflammatory protein-2 (MIP-2) antibody rescues S3 segments from injury, decreases neutrophil infiltration, and improves kidney function in THP-/- mice. Furthermore, using immunofluorescence volumetric imaging of wild-type mouse kidneys, we show that ischemia alters the intracellular translocation of THP in the TAL cells by partially shifting it from its default apical surface domain to the basolateral domain, the latter being contiguous to the basolateral surface of S3 segments. Concomitant with this is the upregulation, in the basolateral surface of S3 segments, of the scavenger receptor SRB-1, a putative receptor for THP. We conclude that TAL affects the susceptibility of S3 segments to injury at least in part by regulating MIP-2 expression in a THP-dependent manner. Our findings raise the interesting possibility of a direct role of basolaterally released THP on regulating inflammation in S3 segments.


Asunto(s)
Quimiocina CXCL2/metabolismo , Necrosis Tubular Aguda/metabolismo , Asa de la Nefrona/metabolismo , Daño por Reperfusión/metabolismo , Uromodulina/metabolismo , Animales , Quimiocina CXCL2/genética , Técnica del Anticuerpo Fluorescente , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/patología , Asa de la Nefrona/patología , Ratones , Ratones Noqueados , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Regulación hacia Arriba , Uromodulina/genética
8.
J Immunol ; 181(12): 8670-6, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19050287

RESUMEN

Neutrophils and macrophages rapidly infiltrate the kidney after renal ischemia-reperfusion injury, however specific molecular recruitment mechanisms have not been fully delineated for these cell types. Here we provide genetic and pharmacologic evidence supporting a positive role for the chemokine receptor CCR1 in macrophage and neutrophil infiltration in a 7 day mouse model of renal ischemia-reperfusion injury. By day 7, injured kidneys from mice lacking CCR1 contained 35% fewer neutrophils and 45% fewer macrophages than injured kidneys from wild-type control mice. Pretreatment of wild-type mice with the specific CCR1 antagonist BX471 also suppressed neutrophil and macrophage infiltration in the model. Injured kidneys from mice lacking CCR1 also had reduced content of the CCR1 ligands CCL3 (MIP-1alpha) and CCL5 (RANTES) compared with injured kidneys from wild-type controls, suggesting a leukocyte source for these inflammatory chemokines and existence of a CCR1-dependent positive feedback loop for leukocyte infiltration in the model. Local leukocyte proliferation and apoptosis were detected after injury, but were not dependent on CCR1. Also, the extent of necrotic and fibrotic damage and decline in renal function in injured kidneys was similar in wild-type and CCR1-deficient mice. Thus, CCR1 appears to regulate trafficking of macrophages and neutrophils to kidney in a mouse model of renal ischemia-reperfusion injury, however this activity does not appear to affect tissue injury.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Mediadores de Inflamación/fisiología , Necrosis Tubular Aguda/inmunología , Necrosis Tubular Aguda/patología , Receptores CCR1/fisiología , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Animales , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR1/deficiencia , Receptores CCR1/genética , Daño por Reperfusión/genética , Daño por Reperfusión/fisiopatología
9.
Nephrol Dial Transplant ; 23(1): 101-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17984101

RESUMEN

BACKGROUND: Acute tubular necrosis (ATN) is characterized by an initiation phase, followed by an extension phase, and a maintenance and recovery phase, the latter of which involves increased regeneration of tubular cells. Nephronectin (NPNT), a ligand for alpha8beta1 integrin, is expressed in the ureteric bud epithelium during kidney morphogenesis. However, little is known about the potential involvement of NPNT in the regeneration phase of ATN. METHODS: cDNA microarray, real-time polymerase chain reaction, in situ hybridization, immunohistochemistry, immuno-electron microscopy and immunoassay (for urine) were used to identify the time-course NPNT expression in a murine model of ATN. RESULTS: The gene transcript of NPNT was examined during a 14-day course of ATN by a cursory cDNA microarray analysis. Although NPNT was observed focally in normal renal tubular epithelium, it was greatly expressed in regenerating tubular cells during the maintenance and recovery phases of ATN. As early as day 1 following onset of ATN, NPNT was already present in the urine. Importantly, NPNT expression preceded proliferating cell nuclear antigen protein expression in regenerating renal tubular epithelial cells, as demonstrated by double immunohistochemistry. CONCLUSION: The present study was the first to identify an enhanced expression of NPNT in regenerating tubular epithelium in an experimental model of ATN. NPNT may play a crucial role in the regenerating process of nephrotoxic ATN. Our data also suggest that NPNT may provide a useful tissue and urine biomarker for both the development and evolution of nephrotoxic acute renal injury.


Asunto(s)
Proteínas de la Matriz Extracelular/biosíntesis , Necrosis Tubular Aguda/metabolismo , Animales , Proteínas de la Matriz Extracelular/genética , Femenino , Necrosis Tubular Aguda/genética , Túbulos Renales/citología , Túbulos Renales/fisiología , Ratones , ARN Mensajero/análisis , Regeneración
10.
Drug Discov Today ; 11(7-8): 364-70, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16580979

RESUMEN

Acute renal failure (ARF) is a common cause of mortality and morbidity in hospitalized patients. Ischemia is an important cause of ARF, and ARF caused by ischemic injury is referred to as ischemic acute tubular necrosis (ATN). There is growing evidence from models that ischemic ATN is associated with intrarenal inflammation. Consequently, intrarenal inflammation is an attractive target for the development of novel drug therapies for ARF. This review outlines ischemic ATN models, the pathophysiological roles of inflammatory cells such as T and B cells in ischemic ATN models, and effective T and B cell therapeutic reagents.


Asunto(s)
Antiinflamatorios/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inflamación/tratamiento farmacológico , Isquemia/tratamiento farmacológico , Necrosis Tubular Aguda/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inflamación/metabolismo , Isquemia/genética , Isquemia/metabolismo , Riñón/irrigación sanguínea , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/metabolismo , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología , Ácido Micofenólico/uso terapéutico , Análisis de Secuencia por Matrices de Oligonucleótidos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
12.
Toxicol Pathol ; 32(5): 577-90, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15603542

RESUMEN

Kidney slices represent an in vitro model that has the cellular complexity of in vivo tissue to provide insights into mechanisms of organ injury, as shown in this study with the model nephrotoxicant cisplatin. Cell pathways altered by cisplatin exposure are assessed by gene expression analysis, cell function, and morphology in human and rat kidney slices in comparison to rat kidney from an in vivo study. The acute nephrosis of the tubular epithelium induced by cisplatin in vivo was reproduced in both human and rat kidney slices, while the glomerulus appeared resistant even at high concentrations. Kidney gene expression changes of in vivo and in vitro samples were indicative of transcription, DNA damage, cell cycle, proliferation, and apoptosis that are in agreement with the mechanism of cisplatin causing DNA damage, growth arrest, and apoptosis; while genes indicative of protein damage, the disruption of transport and calcium homeostasis, cellular metabolism, and oxidative stress are pathways linked with cisplatin binding to various cellular proteins and macromolecules. Both concentration and time-dependent gene expression changes evident in the in vitro model preceded a change in tissue morphology. Functional assays confirming cell dysfunction and increased apoptosis revealed the rat kidney to be more sensitive to the effects of cisplatin than human kidney as demonstrated by significant decreases in slice ATP and GSH levels, significant increases in caspase 9 and 3 activity, p53 protein levels, and increased DNA laddering. The regional markers of proximal and distal tubular injury, alpha- and pi-glutathione S-transferases, were shown for the human kidney slices to be significantly increased by cisplatin. In this study, cisplatin-induced nephrotoxicity was demonstrated morphologically in rat and human kidney slices, and the associated gene expression and functional changes characterized the cellular pathways involved.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Corteza Renal/efectos de los fármacos , Necrosis Tubular Aguda/inducido químicamente , Riñón/efectos de los fármacos , Adulto , Animales , Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Medios de Cultivo/química , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Humanos , Inyecciones Intravenosas , Riñón/metabolismo , Riñón/patología , Corteza Renal/metabolismo , Corteza Renal/patología , Pruebas de Función Renal , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/patología , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar
13.
J Immunol ; 169(5): 2648-52, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12193737

RESUMEN

Ischemia reperfusion injury (IRI) is a major cause of delayed graft function. Recent studies have shown that selectins play an important role in IRI. Selectins bind to sialylated and fucosylated sLe(x) receptors, and two enzymes, fucosyltransferase IV (FucT-IV) and VII (FucT-VII), are important in the function of these receptors. We hypothesized that fucosyltransferase (FucT) enzymes were important pathophysiologic mediators of renal IRI. We therefore evaluated renal IRI in mice deficient in FucT-IV, FucT-VII, and both FucT-IV and FucT-VII and compared their renal function, tubular injury, selectin ligand expression, and neutrophil infiltration to those in wild-type control mice. Bilateral 30-min renal IRI was performed, and the results demonstrated that mice deficient in both FucT-IV/FucT-VII were significantly protected from renal IRI at 24 and 48 h compared with wild-type control mice. FucT-IV-deficient mice showed only modest protection from renal injury at 24 h. However, FucT-VII-deficient mice had similar injury as wild-type mice. Histological analysis of kidney tissue postischemia revealed that mice deficient in both FucT-IV and FucT-VII had significantly reduced tubular injury compared with wild-type mice. Selectin ligand expression increased postischemia in wild-type, but not FucT-IV/FucT-VII-deficient, mice. Neutrophil infiltration in postischemic kidneys of FucT-IV/FucT-VII-deficient mice was also attenuated. These data demonstrate that fucosyltransferases are important in the pathogenesis of renal IRI and are potential therapeutic targets.


Asunto(s)
Fucosiltransferasas/fisiología , Riñón/enzimología , Riñón/fisiopatología , Daño por Reperfusión/enzimología , Daño por Reperfusión/fisiopatología , Animales , Creatinina/sangre , Selectina E/análisis , Selectina E/biosíntesis , Selectina E/genética , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Humanos , Inmunoglobulina M/genética , Riñón/irrigación sanguínea , Riñón/patología , Pruebas de Función Renal , Necrosis Tubular Aguda/enzimología , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/prevención & control , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/genética , Neutrófilos/patología , Proteínas Recombinantes de Fusión/análisis , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Coloración y Etiquetado
14.
J Am Soc Nephrol ; 12(3): 531-540, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11181801

RESUMEN

In acute tubular necrosis, there are early transient increases in circulating and local bioactive hepatocyte growth factor (HGF) levels and renal HGF receptor (c-MET) gene expression. It has therefore been suggested that endogenous HGF may play a role in initiating renal repair. To test this hypothesis, changes in the levels, activity, and anatomic distribution of c-MET protein were characterized in relation to the onset and localization of DNA synthesis in kidneys of rats with ischemia-induced acute tubular necrosis. Whole-kidney c-MET protein levels were significantly increased in the injured kidneys 12 h after injury and rose to a maximum after 1 d, exceeding the control values by sevenfold. Eight days after injury, c-MET levels, although decreasing, were still elevated above control values. An increase in the levels of activated c-MET, i.e., tyrosine-phosphorylated c-MET, was also evident as early as 12 h after injury. Histologic analyses demonstrated that the increase in c-MET immunoreactivity was most marked in the most severely damaged nephron segments in the outer medulla. In injured proximal tubules, the receptor was redistributed from an apical location to an intracellular location. DNA synthesis was increased in the injured kidneys, especially in the outer medulla, where the increase in c-MET protein levels was most prominent. The increase in DNA synthesis was first detected 12 h after the initial increase in activated c-MET levels. It is concluded that the early increases in the levels of c-MET protein and activated receptor support the hypothesis that HGF participates in the initiation of renal regeneration. In addition, the persistent elevation of c-Met protein levels suggests that prolonged and even late treatment with HGF may be of therapeutic value


Asunto(s)
Necrosis Tubular Aguda/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Secuencia de Bases , ADN/biosíntesis , Cartilla de ADN/genética , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/patología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Proteínas Proto-Oncogénicas c-met/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Kidney Int ; 55(3): 1011-8, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10027938

RESUMEN

BACKGROUND: Endothelin (ET), a potent vasoconstrictor, is known to play a role in ischemic acute renal failure. Although preproET-1 (ppET-1) mRNA is known to be up-regulated following ischemia/reperfusion injury, it has not been determined which component of the injury (ischemia or reperfusion) leads to initial gene up-regulation. Likewise, although ET-1 peptide expression has been localized in the normal kidney, its expression pattern in the ischemic kidney has not been determined. Therefore, the purpose of this study was twofold: (a) to determine whether ischemia alone or ischemia plus reperfusion is required for the up-regulation of ppET-1 mRNA to occur, and (b) to localize ET-1 peptide expression following ischemia in the rat kidney to clarify better the role of ET in the pathophysiology of ischemia-induced acute renal failure. METHODS: Male Lewis rats underwent clamping of the right renal vascular pedicle for either 30 minutes of ischemia (group 1), 60 minutes of ischemia (group 2), 30 minutes of ischemia followed by 30 minutes of reperfusion (group 3), or 60 minutes of ischemia followed by three hours of reperfusion (group 4). The contralateral kidney acted as a control. ppET-1 mRNA up-regulation and ET-1 peptide expression were examined using the reverse transcription-polymerase chain reaction and immunohistochemistry, respectively. RESULTS: Reverse transcription-polymerase chain reaction yielded a control (nonischemic) value of 0.6 +/- 0.2 densitometric units (DU) of ppET-1 mRNA in the kidney. Group 1 levels (30 min of ischemia alone) were 1.8 +/- 0.4 DU, a threefold increase (P < 0.05). Group 2 levels (60 min of ischemia alone) increased almost six times above baseline, 3.5 +/- 0.2 DU (P < 0.01), whereas both group 3 and group 4 (ischemia plus reperfusion) did not experience any further significant increases in mRNA levels (1.9 +/- 0.4 DU and 2.8 +/- 0.6 DU, respectively) beyond levels in group 1 or 2 animals subjected to similar ischemic periods. ET-1 peptide expression in the ischemic kidneys was significantly increased over controls and was clearly localized to the endothelium of the peritubular capillary network of the kidney. CONCLUSIONS: Initial ET-1 gene up-regulation in the kidney occurs secondary to ischemia, but reperfusion most likely contributes to sustaining this up-regulation. The marked increase of ET-1 in the peritubular capillary network suggests that ET-induced vasoconstriction may have a pathophysiological role in ischemic acute tubular necrosis.


Asunto(s)
Endotelina-1/genética , Endotelina-1/metabolismo , Riñón/irrigación sanguínea , Riñón/lesiones , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Animales , Secuencia de Bases , Capilares/metabolismo , Cartilla de ADN/genética , Endotelinas/genética , Endotelio Vascular/metabolismo , Inmunohistoquímica , Riñón/metabolismo , Necrosis Tubular Aguda/etiología , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/metabolismo , Masculino , Precursores de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
16.
Transpl Immunol ; 1(2): 109-13, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-7521737

RESUMEN

Expression of the cellular adhesion molecules ICAM-1, VCAM-1, E-selectin and PECAM in human kidney allografts was assessed by immunoperoxidase labelling of cryostat sections. Biopsies from 10 kidneys immediately prior to transplantation and 58 biopsies from 51 kidney transplants with graft dysfunction were studied. Allograft dysfunction was due to acute tubular necrosis (n = 5), acute rejection (n = 30), cyclosporin A (CyA) nephrotoxicity (n = 6), acute pyelonephritis (n = 3), recurrent glomerulonephritis (n = 4) and chronic rejection (n = 10). There was variability in the distribution of ICAM-1, VCAM-1 and E-selectin expression in pretransplant kidneys but the principal observation was a marked increase in the expression of ICAM-1 and VCAM-1 by the renal vasculature and the proximal tubules during acute rejection. By contrast, grafts with dysfunction not attributed to rejection showed a pattern of ICAM-1 and VCAM-1 expression similar to that observed prior to transplantation. E-selectin was expressed only weakly by occasional intertubular capillaries during acute rejection but the three grafts with pyelonephritis displayed strong expression of E-selectin on intertubular capillaries. There was no change in the pattern of PECAM expression following transplantation. The induction of ICAM-1 and VCAM-1 during rejection may contribute to the recruitment of mononuclear cells and render endothelial and tubular renal cells more susceptible to cell-mediated injury.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Rechazo de Injerto/metabolismo , Enfermedades Renales/metabolismo , Trasplante de Riñón , Complicaciones Posoperatorias/metabolismo , Adolescente , Adulto , Anciano , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Antígenos de Diferenciación Mielomonocítica/genética , Moléculas de Adhesión Celular/genética , Ciclosporina/efectos adversos , Selectina E , Femenino , Regulación de la Expresión Génica , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Rechazo de Injerto/genética , Humanos , Molécula 1 de Adhesión Intercelular , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Trasplante de Riñón/inmunología , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/metabolismo , Masculino , Persona de Mediana Edad , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Pielonefritis/genética , Pielonefritis/metabolismo , Recurrencia , Molécula 1 de Adhesión Celular Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...