Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Am J Physiol Renal Physiol ; 327(3): F412-F425, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38961845

RESUMEN

There are diverse pathophysiological mechanisms involved in acute kidney injury (AKI). Among them, overactivity of the renin-angiotensin system (RAS) has been described. Angiotensin-converting enzyme 2 (ACE2) is a tissue RAS enzyme expressed in the apical border of proximal tubules. Given the important role of ACE2 in the metabolism of angiotensin II, this study aimed to characterize kidney and urinary ACE2 in a mouse model of AKI. Ischemia-reperfusion injury (IRI) was induced in C57BL/6 mice by clamping of the left renal artery followed by removal of the right kidney. In kidneys harvested 48 h after IRI, immunostaining revealed a striking maldistribution of ACE2 including spillage into the tubular lumen and the presence of ACE2-positive luminal casts in the medulla. In cortical membranes, ACE2 protein and enzymatic activity were both markedly reduced (37 ± 4 vs. 100 ± 6 ACE2/ß-actin, P = 0.0004, and 96 ± 14 vs. 152 ± 6 RFU/µg protein/h, P = 0.006). In urine, full-length membrane-bound ACE2 protein (100 kDa) was markedly increased (1,120 ± 405 vs. 100 ± 46 ACE2/µg creatinine, P = 0.04), and casts stained for ACE2 were recovered in the urine sediment. In conclusion, in AKI caused by IRI, there is a marked loss of ACE2 from the apical tubular border with deposition of ACE2-positive material in the medulla and increased urinary excretion of full-length membrane-bound ACE2 protein. The deficiency of tubular ACE2 in AKI suggests that provision of this enzyme could have therapeutic applications and that its excretion in the urine may also serve as a diagnostic marker of severe proximal tubular injury.NEW & NOTEWORTHY This study provides novel insights into the distribution of kidney ACE2 in a model of AKI by IRI showing a striking detachment of apical ACE2 from proximal tubules and its loss in urine and urine sediment. The observed deficiency of kidney ACE2 protein and enzymatic activity in severe AKI suggests that administration of forms of this enzyme may mitigate AKI and that urinary ACE2 may serve as a potential biomarker for tubular injury.


Asunto(s)
Lesión Renal Aguda , Enzima Convertidora de Angiotensina 2 , Riñón , Daño por Reperfusión , Animales , Masculino , Ratones , Lesión Renal Aguda/orina , Lesión Renal Aguda/patología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/enzimología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/orina , Biomarcadores/orina , Modelos Animales de Enfermedad , Riñón/metabolismo , Riñón/patología , Riñón/enzimología , Ratones Endogámicos C57BL , Peptidil-Dipeptidasa A/orina , Peptidil-Dipeptidasa A/metabolismo , Sistema Renina-Angiotensina , Daño por Reperfusión/orina , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/enzimología
2.
Cell Mol Neurobiol ; 44(1): 49, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38836960

RESUMEN

Mild hypothermia (MH) is an effective measure to alleviate cerebral ischemia-reperfusion (I/R) injury. However, the underlying biological mechanisms remain unclear. This study set out to investigate dynamic changes in urinary proteome due to MH in rats with cerebral I/R injury and explore the neuroprotective mechanisms of MH. A Pulsinelli's four-vessel occlusion (4-VO) rat model was used to mimic global cerebral I/R injury. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was employed to profile the urinary proteome of rats with/without MH (32 °C) treatment after I/R injury. Representative differentially expressed proteins (DEPs) associated with MH were validated by western blotting in hippocampus. A total of 597 urinary proteins were identified, among which 119 demonstrated significant changes associated with MH. Gene Ontology (GO) annotation of the DEPs revealed that MH significantly enriched in endopeptidase activity, inflammatory response, aging, response to oxidative stress and reactive oxygen species, blood coagulation, and cell adhesion. Notably, changes in 12 DEPs were significantly reversed by MH treatment. Among them, 8 differential urinary proteins were previously reported to be closely associated with brain disease, including NP, FZD1, B2M, EPCR, ATRN, MB, CA1and VPS4A. Two representative proteins (FZD1, B2M) were further validated by western blotting in the hippocampus and the results were shown to be consistent with urinary proteomic analysis. Overall, this study strengthens the idea that urinary proteome can sensitively reflect pathophysiological changes in the brain, and appears to be the first study to explore the neuroprotective effects of MH by urinary proteomic analysis. FZD1 and B2M may be involved in the most fundamental molecular biological mechanisms of MH neuroprotection.


Asunto(s)
Isquemia Encefálica , Hipotermia Inducida , Proteómica , Ratas Sprague-Dawley , Daño por Reperfusión , Animales , Daño por Reperfusión/metabolismo , Daño por Reperfusión/orina , Proteómica/métodos , Masculino , Hipotermia Inducida/métodos , Isquemia Encefálica/metabolismo , Isquemia Encefálica/orina , Proteoma/metabolismo , Ratas , Hipocampo/metabolismo
3.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-34768879

RESUMEN

Oxidative stress plays an important role in the pathophysiology of acute kidney injury (AKI). Previously, we reported that vanin-1, which is involved in oxidative stress, is associated with renal tubular injury. This study was aimed to determine whether urinary vanin-1 is a biomarker for the early diagnosis of AKI in two experimental models: in vivo and in vitro. In a rat model of AKI, ischemic AKI was induced in uninephrectomized rats by clamping the left renal artery for 45 min and then reperfusing the kidney. On Day 1 after renal ischemia/reperfusion (I/R), serum creatinine (SCr) in I/R rats was higher than in sham-operated rats, but this did not reach significance. Urinary N-acetyl-ß-D-glucosaminidase (NAG) exhibited a significant increase but decreased on Day 2 in I/R rats. In contrast, urinary vanin-1 significantly increased on Day 1 and remained at a significant high level on Day 2 in I/R rats. Renal vanin-1 protein decreased on Days 1 and 3. In line with these findings, immunofluorescence staining demonstrated that vanin-1 was attenuated in the renal proximal tubules of I/R rats. Our in vitro results confirmed that the supernatant from HK-2 cells under hypoxia/reoxygenation included significantly higher levels of vanin-1 as well as KIM-1 and NGAL. In conclusion, our results suggest that urinary vanin-1 might be a potential novel biomarker of AKI induced by I/R.


Asunto(s)
Lesión Renal Aguda/metabolismo , Amidohidrolasas/metabolismo , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/orina , Amidohidrolasas/orina , Animales , Biomarcadores/orina , Creatinina/análisis , Creatinina/sangre , Diagnóstico Precoz , Hexosaminidasas/metabolismo , Hexosaminidasas/orina , Isquemia/metabolismo , Riñón/metabolismo , Masculino , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Reperfusión , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/orina , Sistema Urinario/metabolismo
4.
Int J Mol Sci ; 22(3)2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-33573145

RESUMEN

Renal ischemia and reperfusion (I/R) injury is the most common cause of acute kidney injury (AKI). Pathogenesis of postischemic AKI involves hemodynamic changes, oxidative stress, inflammation process, calcium ion overloading, apoptosis and necrosis. Up to date, therapeutic approaches to treat AKI are extremely limited. Thus, the aim of this study was to evaluate the effects of hyperbaric oxygen (HBO) preconditioning on citoprotective enzyme, heme oxygenase-1 (HO-1), pro-apoptotic Bax and anti-apoptotic Bcl-2 proteins expression, in postischemic AKI induced in normotensive Wistar and spontaneously hypertensive rats (SHR). The animals were randomly divided into six experimental groups: SHAM-operated Wistar rats (W-SHAM), Wistar rats with induced postischemic AKI (W-AKI) and Wistar group with HBO preconditioning before AKI induction (W-AKI + HBO). On the other hand, SHR rats were also divided into same three groups: SHR-SHAM, SHR-AKI and SHR-AKI + HBO. We demonstrated that HBO preconditioning upregulated HO-1 and anti-apoptotic Bcl-2 protein expression, in both Wistar and SH rats. In addition, HBO preconditioning improved glomerular filtration rate, supporting by significant increase in creatinine, urea and phosphate clearances in both rat strains. Considering our results, we can also say that even in hypertensive conditions, we can expect protective effects of HBO preconditioning in experimental model of AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Hemo Oxigenasa (Desciclizante)/metabolismo , Oxigenoterapia Hiperbárica/métodos , Hipertensión/complicaciones , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/orina , Animales , Creatinina/metabolismo , Creatinina/orina , Modelos Animales de Enfermedad , Humanos , Hipertensión/fisiopatología , Hipertensión/terapia , Riñón/irrigación sanguínea , Riñón/patología , Riñón/fisiopatología , Masculino , Oxígeno/administración & dosificación , Fosfatos/metabolismo , Fosfatos/orina , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Eliminación Renal/fisiología , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Daño por Reperfusión/orina , Regulación hacia Arriba , Urea/metabolismo , Urea/orina
5.
J Surg Res ; 247: 429-437, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31662185

RESUMEN

BACKGROUND: Renal ischemia/reperfusion injury (IRI) can result in impaired ability of urine concentration and increased sodium fractional excretion. Apelin, a (neuro) vasoactive peptide, enhances diuresis by increasing the renal microcirculation and by counteracting the antidiuretic effect of arginine vasopressin on the tubules. However, changes in renal apelin expression in renal IRI rat model have not been elucidated. Remote ischemic perconditioning (RIPerC) improves renal sodium and water handling after IRI. Here, we investigated whether RIPerC prevents dysregulation of renal sodium and water handling in response to IRI by apelin signaling pathway in rats. MATERIALS AND METHODS: Renal IRI was induced by 45-min clamping of renal arteries followed by 24 h reperfusion. RIPerC was created by applying four cycles of 2-min ischemia of the left femoral artery followed by 3-min reperfusion at the start of renal ischemia. Rats were randomly divided into sham, ischemia/reperfusion, and RIPerC + ischemia/reperfusion groups. Urine and blood were sampled after reperfusion period. The kidney was harvested for mRNA isolation and histopathological study. RESULTS: IRI resulted in decreased clearance of creatinine, increased sodium fractional excretion, and reduced urine osmolality compared with sham animals. This occurred with an increase in mRNA expression levels of apelin and histological damages in both cortical and medullary regions of kidney tissues. RIPerC treatment ameliorated all these changes. CONCLUSIONS: This study showed that RIPerC has protective effects against dysregulation of renal sodium and water handling after renal IRI, which might be related with inhibition of apelin signaling pathway.


Asunto(s)
Lesión Renal Aguda/prevención & control , Apelina/metabolismo , Poscondicionamiento Isquémico/métodos , Riñón/patología , Daño por Reperfusión/patología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/orina , Animales , Apelina/genética , Creatinina/sangre , Creatinina/metabolismo , Creatinina/orina , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Riñón/irrigación sanguínea , Riñón/fisiopatología , Masculino , Concentración Osmolar , ARN Mensajero/metabolismo , Ratas , Eliminación Renal/fisiología , Daño por Reperfusión/sangre , Daño por Reperfusión/complicaciones , Daño por Reperfusión/orina , Transducción de Señal/fisiología , Sodio/sangre , Sodio/metabolismo , Sodio/orina , Orina/química , Agua/metabolismo
6.
Int J Mol Sci ; 21(18)2020 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-32937906

RESUMEN

Acute kidney injury (AKI), caused mainly by ischemia-reperfusion, sepsis, or nephrotoxins (such as contrast medium), is identified by an abrupt decline in kidney function and is associated with high morbidity and mortality. Despite decades of efforts, the pathogenesis of AKI remains poorly understood, and effective therapies are lacking. MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level to control cell differentiation, development, and homeostasis. Additionally, extracellular miRNAs might mediate cell-cell communication during various physiological and pathological processes. Recently, mounting evidence indicates that miRNAs play a role in the pathogenesis of AKI. Moreover, emerging research suggests that because of their remarkable stability in body fluids, microRNAs can potentially serve as novel diagnostic biomarkers of AKI. Of note, our previous finding that miR-494 is rapidly elevated in urine but not in serum provides insight into the ultimate role of urine miRNAs in AKI. Additionally, exosomal miRNAs derived from stem cells, known as the stem cell secretome, might be a potential innovative therapeutic strategy for AKI. This review aims to provide new data obtained in this field of research. It is hoped that new studies on this topic will not only generate new insights into the pathophysiology of urine miRNAs in AKI but also might lead to the precise management of this fatal disease.


Asunto(s)
Lesión Renal Aguda/genética , Biomarcadores/orina , Inflamación/genética , MicroARNs/genética , MicroARNs/orina , Daño por Reperfusión/genética , Lesión Renal Aguda/orina , Animales , Humanos , Inflamación/orina , Daño por Reperfusión/orina
7.
Med Mol Morphol ; 53(2): 115-123, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31802235

RESUMEN

There is a clinical need for sensitive acute kidney injury (AKI) biomarkers that enable early therapeutic interventions and prediction of disease prognosis. In this study, we monitored interleukin (IL)-24 expressed in kidneys with severe AKI that progresses to atrophic kidney in a mouse model of ischemia-reperfusion injury (IRI). Therefore, we evaluated IL-24 as a potential biomarker not only for early diagnosis of AKI, but also for predicting progression to chronic kidney disease (CKD). Serum IL-24 was detected earlier than the elevation of serum creatinine levels and urinary IL-24 was detected as early as neutrophil gelatinase associated lipocalin (NGAL) in severe AKI (60 min of IRI). In addition, serum and urine IL-24 levels tended to increase in relation to ischemia duration. In such kidneys, vascular smooth muscle cells expressed IL-24 in response to the injury in the renal tubular epithelial cell and its target was the renal tubular epithelial cell itself. IL-24 may play a pivotal role in the communication between tubular epithelial cells and vascular smooth muscle cells and, in conclusion, IL-24 can be used as a sensitive biomarker for AKI.


Asunto(s)
Lesión Renal Aguda/diagnóstico , Citocinas/metabolismo , Túbulos Renales/patología , Daño por Reperfusión/diagnóstico , Lesión Renal Aguda/sangre , Lesión Renal Aguda/patología , Lesión Renal Aguda/orina , Animales , Atrofia/sangre , Atrofia/diagnóstico , Atrofia/patología , Atrofia/orina , Biomarcadores/sangre , Biomarcadores/metabolismo , Biomarcadores/orina , Comunicación Celular , Células Cultivadas , Citocinas/sangre , Citocinas/orina , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales/patología , Humanos , Túbulos Renales/irrigación sanguínea , Túbulos Renales/citología , Lipocalina 2/sangre , Masculino , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Cultivo Primario de Células , Pronóstico , Daño por Reperfusión/sangre , Daño por Reperfusión/patología , Daño por Reperfusión/orina , Índice de Severidad de la Enfermedad
8.
J Cell Mol Med ; 23(6): 3867-3877, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30968541

RESUMEN

Macrophage migration inhibitory factor (MIF) is pleiotropic cytokine that has multiple effects in many inflammatory and immune diseases. This study reveals a potential role of MIF in acute kidney injury (AKI) in patients and in kidney ischemic reperfusion injury (IRI) mouse model in MIF wild-type (WT) and MIF knockout (KO) mice. Clinically, plasma and urinary MIF levels were largely elevated at the onset of AKI, declined to normal levels when AKI was resolved and correlated tightly with serum creatinine independent of disease causes. Experimentally, MIF levels in plasma and urine were rapidly elevated after IRI-AKI and associated with the elevation of serum creatinine and the severity of tubular necrosis, which were suppressed in MIF KO mice. It was possible that MIF may mediate AKI via CD74/TLR4-NF-κB signalling as mice lacking MIF were protected from AKI by largely suppressing CD74/TLR-4-NF-κB associated renal inflammation, including the expression of MCP-1, TNF-α, IL-1ß, IL-6, iNOS, CXCL15(IL-8 in human) and infiltration of macrophages, neutrophil, and T cells. In conclusion, our study suggests that MIF may be pathogenic in AKI and levels of plasma and urinary MIF may correlate with the progression and regression of AKI.


Asunto(s)
Lesión Renal Aguda/sangre , Oxidorreductasas Intramoleculares/sangre , Riñón/patología , Factores Inhibidores de la Migración de Macrófagos/sangre , Daño por Reperfusión/sangre , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/orina , Adulto , Anciano , Animales , Antígenos de Diferenciación de Linfocitos B/metabolismo , Quimiocina CCL2/metabolismo , Creatinina/sangre , Citocinas/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/orina , Riñón/inmunología , Riñón/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/orina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , FN-kappa B/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/orina , Receptor Toll-Like 4/metabolismo
9.
Am J Physiol Renal Physiol ; 317(5): F1098-F1110, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31390267

RESUMEN

Numerous candidate biomarkers in urine extracellular vesicles (EVs) have been described for kidney diseases, but none are yet in clinical use, possibly due to a lack of proper normalization. Proper normalization corrects for normal biological variation in urine flow rate or concentration, which can vary by over one order of magnitude. Here, we observed inter- and intra-animal variation in urine excretion rates of small EVs (<200 nm in diameter) in healthy rats as a series of six 4-h fractions. To visualize intra-animal variation, we normalized a small EV excretion rate to a peak excretion rate, revealing a circadian pattern for each rat. This circadian pattern was distinct from urine volume, urine albumin, urine creatinine, and urine albumin-to-creatinine ratio. Furthermore, urine small EV excretion was not significantly altered by sex, food/water deprivation, or ischemic acute kidney injury. Urine excretion of the exosomal/small EV marker protein tumor susceptibility gene 101 (TSG101) displayed a similar circadian pattern to urine small EV excretion; both measurements were highly correlated (R2 = 0.85), with an average stoichiometry of 10.0 molecules of TSG101/vesicle in healthy rats. The observed stoichiometry of TSG101/vesicle in rat urine translated to human spot urine samples (10.2 molecules/vesicle) and cultured kidney-derived cell lines (human embryonic kidney-293 and normal rat kidney 52E cells). Small EV number and its surrogate, TSG101 protein, can normalize for circadian variation when testing candidate biomarkers in small EVs. Just as creatinine has emerged as the customary normalization factor for liquid-phase urine biomarkers, vesicle number and its surrogate, molecules of exosome/small EV-associated TSG101, should be considered as viable, normalizing factors for small EV biomarkers.


Asunto(s)
Ritmo Circadiano/fisiología , Vesículas Extracelulares/fisiología , Daño por Reperfusión/orina , Animales , Biomarcadores/orina , Línea Celular , Femenino , Privación de Alimentos , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Privación de Agua
10.
Am J Physiol Renal Physiol ; 316(5): F957-F965, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30864839

RESUMEN

Ischemia-reperfusion injury represents one of the most common causes of acute kidney injury, a serious and often deadly condition that affects up to 20% of all hospitalized patients in the United States. However, the current standard assay used universally for the diagnosis of acute kidney injury, serum creatinine, does not detect renal damage early in its course. Serendipitously, we found that the immunofluorescent signal of the constitutive podocyte marker podoplanin fades in the glomerulus and intensifies in the tubulointerstitial compartment of the kidney shortly after ischemia-reperfusion injury in 8- to 10-wk-old male C57Bl/6j mice. Therefore, we sought to define the appearance and course of the podoplanin-positive signal in the kidney after ischemia-reperfusion injury. The tubulointerstitial podoplanin-positive signal increased as early as 2 h but persisted for 7 days after ischemia-reperfusion injury. In addition, the strength of this tubulointerstitial signal was directly proportional to the severity of ischemia, and its location shifted from the tubules to interstitial cells over time. Finally, we detected podoplanin in the urine of mice after ischemia, and we observed that an increase in the urine podoplanin-to-creatinine ratio correlated strongly with the onset of renal ischemia-reperfusion injury. Our findings indicate that the measurement of urine podoplanin harbors promising potential for use as a novel biomarker for the early detection of ischemia-reperfusion injury of the kidney.


Asunto(s)
Lesión Renal Aguda/orina , Glicoproteínas de Membrana/orina , Podocitos/metabolismo , Daño por Reperfusión/orina , Lesión Renal Aguda/patología , Animales , Biomarcadores/orina , Creatinina/orina , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Podocitos/patología , Daño por Reperfusión/patología , Índice de Severidad de la Enfermedad , Factores de Tiempo , Regulación hacia Arriba
11.
Am J Physiol Renal Physiol ; 317(3): F650-F657, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31313951

RESUMEN

Experiments in mouse models have shown that the complement cascade is activated within the kidney after ischemia-reperfusion and that complement activation contributes to tubular injury in this setting. Less is known, however, about complement activation in human kidneys after ischemia or whether complement activation in the tubulointerstitium can be detected by measurement of complement fragments in the urine. We hypothesized that urine biomarkers of complement activation would rapidly increase in patients who develop ischemic acute kidney injury, signaling complement activation within the kidney. We confirmed that the alternative pathway of complement is activated in the kidneys of mice after ischemia-reperfusion, and we found that levels of factor B fragments (generated during alternative pathway activation) rapidly increase in the urine. We next performed a case-control study in which we measured complement fragments in human urine samples from patients undergoing cardiac surgery using ELISAs. The level of Ba increased after cardiac surgery and was significantly higher in patients who developed acute kidney injury. The increase in Ba also correlated with magnitude of the subsequent rise in serum creatinine and with the need for hemodialysis during the hospitalization. These findings demonstrate that the alternative pathway of complement is activated in patients who develop acute kidney injury after cardiac surgery and that increases in the level of urine Ba may be a predictive and functional biomarker of severe kidney injury.


Asunto(s)
Lesión Renal Aguda/orina , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Factor B del Complemento/orina , Vía Alternativa del Complemento , Fragmentos de Péptidos/orina , Daño por Reperfusión/orina , Lesión Renal Aguda/etiología , Lesión Renal Aguda/inmunología , Anciano , Animales , Biomarcadores/orina , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , América del Norte , Estudios Prospectivos , Daño por Reperfusión/etiología , Daño por Reperfusión/inmunología , Regulación hacia Arriba , Urinálisis
12.
Am J Physiol Renal Physiol ; 314(4): F584-F601, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357442

RESUMEN

Acute kidney injury (AKI) is an important risk factor for the development of chronic kidney disease (CKD), and an alteration in renal water handling has been observed during the transition of AKI to CKD. Urinary exosomal release of aquaporin-1 (AQP1) and AQP2, important proteins for renal water handling, has recently been reported to predict their levels of renal expression. Therefore, we examined the patterns of urinary exosomal release of AQP1 and AQP2, and the exosomal marker proteins tumor susceptibility 101 protein (TSG101) and ALG-2 interacting protein X (Alix), in the acute and chronic phases following induction of AKI by renal bilateral ischemia/reperfusion (I/R) in rats. Blood tests and histological examinations indicated that AKI occurred before at 7 days after renal I/R ( day 7) and that renal fibrosis developed progressively thereafter. Immunoblotting demonstrated significant decreases in the urinary exosomal release of AQP1 and AQP2 during severe AKI. Urinary exosomal release of Alix and TSG101 was significantly increased on day 7. These data were also confirmed in rats with unilateral renal I/R causing more serious AKI. Urinary exosomal release of either the Ser-256- or Ser-269-phosphorylated form of AQP2, both of which are involved in apical trafficking of AQP2, was positively correlated with that of total AQP2. These results suggest that urinary exosomal release of AQP1 and AQP2 is reduced in I/R-induced AKI, whereas that of Alix and TSG101 is increased in the initial phase of renal fibrosis. Furthermore, apical trafficking of AQP2 appears to be related to urinary exosomal release of AQP2.


Asunto(s)
Lesión Renal Aguda/orina , Acuaporina 1/orina , Acuaporina 2/orina , Exosomas/metabolismo , Riñón/metabolismo , Eliminación Renal , Daño por Reperfusión/orina , Lesión Renal Aguda/patología , Animales , Proteínas de Unión al Calcio/orina , Proteínas de Unión al ADN/orina , Modelos Animales de Enfermedad , Complejos de Clasificación Endosomal Requeridos para el Transporte/orina , Fibrosis , Riñón/patología , Masculino , Fosforilación , Transporte de Proteínas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Factores de Tiempo , Factores de Transcripción/orina
13.
Am J Physiol Renal Physiol ; 315(5): F1329-F1335, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30066587

RESUMEN

p21 is upregulated in renal tubules in response to acute kidney injury ( AKI). and localizes in the nucleus, where it induces cell cycle arrest (CCA). These events can mitigate early injury but can also facilitate the onset of the degenerative cell senescence/"aging" process. Hence, we asked the following: 1) can AKI-induced p21 upregulation be gauged by plasma and/or urinary p21 assay; 2) might p21 serve as an AKI/CCA biomarker; and 3) does p21 accumulate during normal renal aging, and might plasma p21 reflect this process? Mice were subjected to either ischemia-reperfusion (I/R) or nephotoxic (maleate) AKI. Renal cortical p21 expression (protein, mRNA) was assessed 2-18 h later and contrasted with plasma/urine p21 concentrations (ELISA). p21 mRNA/protein levels were also measured in aging mice (2, 12, 24 mo). AKI induced marked, progressive, increases in renal cortical p21 mRNA and protein levels. These changes were marked by acute (within 2-4 h) and profound increases (up to 200×) in both plasma and urine p21 concentrations. Renal I/R also activated p21 gene expression in extrarenal organs (heart, brain), consistent with so-called "organ cross talk". p21 efflux from damaged cells was confirmed with studies of hypoxia-injured, isolated proximal tubules. Aging was associated with progressive renal cortical p21 expression, which correlated ( r, 0.83) with rising plasma p21 concentrations. We concluded that 1) during AKI, renal p21 increases can be gauged by either plasma or urine p21 assay, serving as potentially useful AKI/CCA biomarkers; 2) AKI can activate p21 in extrarenal organs; and 3) plasma p21 levels may provide an index of the renal/systemic aging process.


Asunto(s)
Lesión Renal Aguda/sangre , Lesión Renal Aguda/orina , Envejecimiento/sangre , Envejecimiento/orina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/sangre , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/orina , Riñón/metabolismo , Daño por Reperfusión/sangre , Daño por Reperfusión/orina , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Factores de Edad , Envejecimiento/genética , Animales , Biomarcadores/sangre , Biomarcadores/orina , Encéfalo/metabolismo , Muerte Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Riñón/patología , Masculino , Ratones , Miocardio/metabolismo , Daño por Reperfusión/genética , Factores de Tiempo , Regulación hacia Arriba
14.
Clin Sci (Lond) ; 132(19): 2121-2133, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30224346

RESUMEN

One of the major obstacles to prevent AKI-CKD transition is the lack of effective methods to follow and predict the ongoing kidney injury after an AKI episode. In the present study, we test the utility of urinary angiotensinogen (UAGT) for dynamically evaluating renal structural changes and predicting AKI-CKD progression by using both mild and severe bilateral renal ischemia/reperfusion injury mice. UAGT returns to pre-ischemic levels 14 days after mild AKI followed by kidney architecture restoration, whereas sustained increase in UAGT accompanies by ongoing renal fibrosis after severe AKI. UAGT at day 14-42 correlates with renal fibrosis 84 days after AKI. For predicting fibrosis at day 84, the area under receiver operating characteristics curve of UAGT at day 14 is 0.81. Persistent elevation in UAGT correlates with sustained activation of intrarenal renin-angiotensin system (RAS) during AKI-CKD transition. Abrogating RAS activation post AKI markedly reduced renal fibrosis, with early RAS intervention (from 14 days after IRI) more beneficial than late intervention (from 42 days after IRI) in alleviating fibrosis. Importantly, UAGT decreases after RAS intervention, and its level at day 14-28 correlates with the extent of renal fibrosis at day 42 post RAS blockade. A pilot study conducted in patients with acute tubular necrosis finds that compared with those recovered, patients with AKI-CKD progression exhibits elevated UAGT during the 3-month follow-up after biopsy. Our study suggests that UAGT enables the dynamical monitoring of renal structural recovery after an AKI episode and may serve as an early predictor for AKI-CKD progression and treatment response.


Asunto(s)
Lesión Renal Aguda/orina , Angiotensinógeno/orina , Biomarcadores/orina , Riñón/patología , Insuficiencia Renal Crónica/orina , Lesión Renal Aguda/complicaciones , Animales , Progresión de la Enfermedad , Fibrosis , Humanos , Riñón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Valor Predictivo de las Pruebas , Curva ROC , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/etiología , Sistema Renina-Angiotensina/fisiología , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/orina , Factores de Tiempo
15.
Artif Organs ; 42(7): 723-727, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29611214

RESUMEN

In renal transplantation, ischemia reperfusion injury impairs early graft function and can reduce long term graft survival. Hydrogen has antioxidant and anti-inflammatory properties that can reduce the effects of ischemic injury. The aim of this study was to examine the effects of hydrogen gas administered during reperfusion in a preclinical model of kidney ischemia reperfusion injury. Porcine kidneys underwent 15 min of warm ischemia followed by 22 h of cold ischemia. They were then reperfused for 6 h with whole autologous blood on an ex vivo reperfusion circuit. Paired kidneys were randomized to control (n = 6) (25% oxygen, 5% carbon dioxide, 70% nitrogen) or hydrogen (n = 6) (2% hydrogen, 25% oxygen, 5% carbon dioxide, 68% nitrogen) groups. Tissue, urine, and blood samples were collected at baseline and hourly throughout the reperfusion period. Baseline measurements were similar across groups. Following perfusion, there was no significant difference between control and hydrogen groups in urine output (693 mL vs. 608 mL, P = 0.86), renal blood flow (105.9 vs. 108 mL/min/100g, P = 0.89), acid-base homeostasis, or creatinine clearance. There was a significant increase in cytokine levels from baseline to 6 h in both groups (IL-1ß P = 0.002; IL-6 P = 0.004; IL-8 P = 0.002). However, there were no significant differences in levels of inflammatory cytokines (IL1ß, IL-6, and IL-8) between the groups. The administration of hydrogen gas did not improve renal function, reduce oxidative damage, or inflammation during the reperfusion of ischemically damaged kidneys.


Asunto(s)
Hidrógeno/uso terapéutico , Daño por Reperfusión/terapia , Animales , Isquemia Fría , Modelos Animales de Enfermedad , Riñón/fisiopatología , Trasplante de Riñón , Estrés Oxidativo , Circulación Renal , Daño por Reperfusión/sangre , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/orina , Porcinos , Isquemia Tibia
16.
Nephrology (Carlton) ; 23(4): 308-316, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28063188

RESUMEN

AIM: The aim of this study was to elucidate whether urinary tubular markers during the chronic phase of acute kidney injury (AKI) are associated with chronic tubulointerstitial damage. METHODS: Male human L-type fatty acid binding protein (L-FABP) chromosomal transgenic (Tg) mice underwent ischaemic reperfusion (I/R) injury via renal pedicle clamping for either 10 min or 20 min. Contralateral nephrectomy was performed at the time of tissue reperfusion. The kidneys were analyzed 20 days after the last I/R. RESULTS: Serum creatinine levels 20 days post-I/R were significantly higher in the 20 min I/R than in the 10 min I/R and control groups and were similar between the 10 min I/R and control groups. The degree of tubulointerstitial damage 20 days post-I/R was significantly more severe in the 20 min I/R than in the 10 min I/R and control groups, as well as in the 10 min I/R than in the control group. Urinary levels of human L-FABP, albumin, and kidney injury molecule-1 (KIM-1) 20 days post-I/R were significantly higher in the 20 min I/R than in the control group, whereas urinary L-FABP was significantly higher in the 10 min I/R than in the control group. Conversely, urinary neutrophil gelatinase-associated lipocalin levels did not significantly differ between the three groups. Finally, the urinary levels of human L-FABP, albumin, and KIM-1 levels 20 days post-I/R were significantly correlated with the degree of renal damage. CONCLUSIONS: Urinary levels of human L-FABP, albumin and, KIM-1 may be useful for monitoring AKI-to-CKD transition in clinical practice.


Asunto(s)
Lesión Renal Aguda/orina , Albuminuria/orina , Proteínas de Unión a Ácidos Grasos/orina , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Túbulos Renales/metabolismo , Daño por Reperfusión/orina , Lesión Renal Aguda/patología , Albuminuria/patología , Animales , Biomarcadores/sangre , Biomarcadores/orina , Creatinina/sangre , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Fibrosis , Túbulos Renales/patología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Transgénicos , Valor Predictivo de las Pruebas , Daño por Reperfusión/patología , Índice de Severidad de la Enfermedad , Factores de Tiempo , Urinálisis
17.
Acta Biochim Biophys Sin (Shanghai) ; 49(4): 338-348, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28338909

RESUMEN

Mesenchymal stem cell (MSC) transplantation is a promising therapy for acute kidney injury; however, the efficacy is limited due to poor survival after transplantation. In this study, we investigated how MSC transplantation timing affected the survival and therapeutic potential of MSCs in the kidney ischemia-reperfusion (I/R) injury model. After kidney I/R injury, the inflammatory process and tissue damage were characterized over 1 week post-I/R, we found that inflammation peaked at 12-24 h post-I/R (h.p.i.), and urine  neutrophil gelatinase-associated lipocalin (NGAL) measurements correlated highly with measures of inflammation. We cultured MSCs with supernatants from I/R injured kidney tissue homogenates collected at different time points and found that kidney homogenates from 12 and 24 h.p.i. were most toxic to MSCs, whereas homogenates from 1 h.p.i. were not as cytotoxic as those from 12 and 24 h.p.i. Compared with MSCs administered at 12, or 24 h.p.i., cells administered immediately after ischemia or 1 h.p.i. yielded the highest renoprotective and anti-inflammatory effects. Our findings indicate that MSC treatment for acute kidney injury is most effective when applied prior to the development of a potent inflammatory microenvironment, and urine NGAL may be helpful for detecting inflammation and selecting MSC transplantation timing in I/R kidney injury.


Asunto(s)
Lesión Renal Aguda/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Daño por Reperfusión/terapia , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/orina , Animales , Supervivencia Celular/inmunología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica , Inflamación/diagnóstico , Inflamación/inmunología , Inflamación/orina , Mediadores de Inflamación/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Lipocalina 2/orina , Masculino , Células Madre Mesenquimatosas/inmunología , Ratas Sprague-Dawley , Daño por Reperfusión/inmunología , Daño por Reperfusión/orina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
18.
Ren Fail ; 39(1): 484-490, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28494217

RESUMEN

In this study, we examined whether the IL-8 content of urine sampled on day 1 and day 14 after renal transplantation is a marker of early and long-term renal function. Moreover, we assessed whether its concentration is positively correlated with the matrix metalloproteinase-9 (MMP-9) content of urine sampled on day 1 and day 30 and 12 months after renal transplantation. Our analysis covered 87 patients who underwent a kidney transplant. The patients were observed for an average of 30 months (12-60 months). The IL-8 concentration determined on day 1 was significantly negatively correlated with creatinine clearance early after renal transplantation (on days 1, 7, 14 and 30), as well as during long-term observations. IL-8 concentration in urine sampled on day 1 and day 14 was higher in patients demonstrating DGF than in those without DGF. No relationship was found between IL-8 content and cold ischaemia time. MMP-9 activity determined on day 1 and month 3 after renal transplantation was positively correlated with the IL-8 content determined in urine sampled on day 1, Rs = +0.32, p < .05 and Rs = +0.31, p < .05, respectively. The results of this study suggest that a high IL-8 content in urine sampled on day 1 after renal transplantation is an unfavourable marker of early and long-term (years-long) graft function. A high IL-8 content in urine sampled on day 1 after renal transplantation was positively correlated with the activity of metalloproteinase-9 in urine. This proves that both of these chemokines cooperate in ischaemia-reperfusion injuries in transplanted kidneys.


Asunto(s)
Funcionamiento Retardado del Injerto/orina , Interleucina-8/orina , Fallo Renal Crónico/cirugía , Trasplante de Riñón/efectos adversos , Metaloproteinasa 9 de la Matriz/orina , Daño por Reperfusión/orina , Adulto , Anciano , Anciano de 80 o más Años , Aloinjertos/patología , Biomarcadores/orina , Biopsia , Isquemia Fría/efectos adversos , Creatinina/sangre , Creatinina/orina , Estudios de Seguimiento , Tasa de Filtración Glomerular , Rechazo de Injerto/orina , Supervivencia de Injerto , Humanos , Riñón/patología , Persona de Mediana Edad , Daño por Reperfusión/etiología , Adulto Joven
19.
Med Mol Morphol ; 50(3): 161-169, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28382533

RESUMEN

The purpose is to evaluate quantified kidney echogenicity as a biomarker for the early diagnosis of acute kidney injury (AKI) and predicting progression to chronic kidney disease (CKD) in a mouse model of ischemia-reperfusion injury (IRI). Two separate protocols of murine models of IRI were used: (1) 10, 30, and 40 min of bilateral ischemia duration and (2) 45 and 60 min of unilateral ischemia duration. Renal echogenicity was measured with ultrasound and compared with serum creatinine or urine neutrophil gelatinase-associated lipocalin (NGAL) at various timepoints after IRI. In mice subjected to 10, 30, and 40 min of bilateral ischemia, renal echogenicity increased about 2 h after IRI for all ischemia times, earlier than serum creatinine or urine NGAL. In those subjected to 45 and 60 min of unilateral ischemia, 60 min of unilateral ischemia, which represents atrophic changes 28 days after IRI, resulted in a sustained high level of echogenicity and was significantly different 24 h after IRI, while 45 min of unilateral ischemia resulted in trivial levels of histological damage 28 days after IRI. Renal echogenicity might have the potential to be a biomarker for the early diagnosis of AKI and the prognosis of CKD.


Asunto(s)
Lesión Renal Aguda/diagnóstico , Biomarcadores/análisis , Riñón/diagnóstico por imagen , Daño por Reperfusión/patología , Lesión Renal Aguda/sangre , Lesión Renal Aguda/orina , Animales , Creatinina/sangre , Modelos Animales de Enfermedad , Riñón/patología , Lipocalina 2/orina , Masculino , Ratones , Daño por Reperfusión/sangre , Daño por Reperfusión/orina
20.
Am J Nephrol ; 41(4-5): 383-91, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26111494

RESUMEN

BACKGROUND/AIMS: Ischemia-reperfusion injury (IRI) and innate immune response augment adaptive immunity and may also trigger repair processes that lead to uncontrolled fibrosis and atherosclerosis as seen in chronic allograft injury. Simvastatin has been shown to protect from renal IRI in several experimental studies. The aim of this study was to examine the effect of donor simvastatin pretreatment and early initiation of recipient simvastatin treatment on chronic kidney allograft injury. METHODS: A rat renal transplantation model was used. Simvastatin was administered perorally for donor (5 mg/kg) and/or for recipient (2 mg/kg) 2 hours before transplantation and/or as daily treatment starting on the first postoperative day (2 mg/kg/day). The study included 5 groups: (1) no simvastatin, (2) donor pretreatment, (3) daily recipient treatment, (4) donor pretreatment + daily recipient treatment and (5) donor pretreatment + recipient pretreatment + daily recipient treatment. The grafts were recovered at day 90 for histopathological and immunohistochemical analysis. Kidney function was followed weekly with serum creatinine, and 24-hour urine protein was measured 60 and 90 days after transplantation. RESULTS: We found that donor and recipient simvastatin pretreatment combined with daily recipient treatment reduced graft inflammation and chronic allograft injury. Treatment using only statins started after transplantation reduced inflammation to some extent, but did not affect chronic kidney allograft injury. Pretreatment using only donor statins impaired graft function and increased proteinuria. CONCLUSION: Our data suggest that perioperative recipient statin treatment reduces inflammation and may protect the graft in the long term.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Trasplante de Riñón/métodos , Atención Perioperativa/métodos , Proteinuria/prevención & control , Insuficiencia Renal Crónica/prevención & control , Daño por Reperfusión/prevención & control , Simvastatina/uso terapéutico , Aloinjertos , Animales , Creatinina/sangre , Ratas , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/orina , Daño por Reperfusión/sangre , Daño por Reperfusión/orina , Donantes de Tejidos , Receptores de Trasplantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA