Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Kidney Int ; 85(1): 134-41, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24088953

RESUMEN

Acute kidney injury (AKI) caused by ischemia-reperfusion is a major clinical problem in both native and transplanted kidneys. We had previously shown that deficiency of Nrf2, a potent bZIP transcription factor that binds to the antioxidant response element, enhances susceptibility to experimental ischemic AKI. Here we further explored the role of Nrf2 in AKI by amplifying Nrf2 activation in vivo and in vitro with the synthetic triterpenoid CDDO-imidazolide. Mice treated with CDDO-imidazolide and undergoing experimental bilateral ischemic AKI had improved survival and renal function. Treated mice had improved renal histology with a decrease in tubular injury, as well as a decrease in proinflammatory cytokine and chemokine production compared with vehicle-treated mice. In an exploration of protective mechanisms, we found an upregulation of Nrf2 target antioxidant genes in CDDO-imidazolide-treated mouse kidneys. Furthermore, Nrf2-deficient mice treated with CDDO-imidazolide had no significant improvement in mortality, renal function or histology, proinflammatory cytokine gene expression, and no significant increase in antioxidant gene expression. In vitro studies demonstrated that the renal epithelial cells were likely an important target of CDDO-imidazolide. Thus, activation of Nrf2 signaling with CDDO-imidazolide confers protection from AKI, and presents a new therapeutic opportunity for this common and serious condition.


Asunto(s)
Lesión Renal Aguda/prevención & control , Imidazoles/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Ácido Oleanólico/análogos & derivados , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/patología , Animales , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Células Epiteliales/efectos de los fármacos , Femenino , Hipoxia/prevención & control , Imidazoles/farmacología , Riñón/efectos de los fármacos , Riñón/patología , Pruebas de Función Renal , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Ácido Oleanólico/farmacología , Ácido Oleanólico/uso terapéutico , Daño por Reperfusión/patología , Activación Transcripcional/efectos de los fármacos
2.
Am J Physiol Renal Physiol ; 302(5): F519-25, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22114209

RESUMEN

In this study, we compared the traditional murine model with renal pedicle clamp with models that clamped the renal artery or vein alone as well as to a whole body ischemia-reperfusion injury (WBIRI) model. Male C57BL/6J mice underwent either clamping of the renal artery, vein, or both (whole pedicle) for 30 or 45 min followed by reperfusion, or 10 min of cardiac arrest followed by resuscitation up to 24 h. After 30 min of ischemia, the mice with renal vein clamping showed the mostly increased serum creatinine and the most severe renal tubule injury. After 45 min of ischemia, all mice with renal vasculature clamping had a comparable increase in serum creatinine but the renal tubule injury was most severe in renal artery-clamped mice. Renal arterial blood flow was most decreased in mice with a renal vein clamp compared with a renal artery or pedicle clamp. A 30-or 45-min renal ischemia time led to a significant increase in the protein level of interleukin-6, keratinocyte-derived chemokine (KC), and granular colony-stimulating factor in the ischemic kidney, but the KC was the highest in the renal pedicle-clamped kidney and the lowest in the renal vein-clamped kidney. Of note, 10 min of WBIRI led to kidney dysfunction and structural injury, although less than longer time clamping of isolated renal vasculature. Our data demonstrate important differences in ischemic AKI models. Understanding these differences is important in designing future experimental studies in mice as well as clinical trials in humans.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Isquemia/fisiopatología , Riñón/irrigación sanguínea , Obstrucción de la Arteria Renal/fisiopatología , Arteria Renal/fisiopatología , Venas Renales/fisiopatología , Lesión Renal Aguda/metabolismo , Animales , Velocidad del Flujo Sanguíneo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Isquemia/metabolismo , Riñón/metabolismo , Riñón/fisiopatología , Pruebas de Función Renal , Masculino , Ratones , Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Venas Renales/metabolismo
3.
Am J Physiol Renal Physiol ; 302(6): F762-73, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22160774

RESUMEN

Although T cells have been shown to play a direct role in kidney ischemia-reperfusion injury (IRI), little is known about the underlying mechanisms. We hypothesized that studying the transcriptional responses in kidney-infiltrating T cells would help elucidate novel therapeutic targets for kidney IRI. Unilateral renal pedicle clamping for 45 min was performed in male C57BL/6 mice, and CD3(+) T cells were isolated from the kidney and purified. Transcriptional activities of T cell were measured by array-based PCR compared between ischemic kidneys and contralateral nonischemic kidneys. Among total of 89 genes analyzed, 24, 22, 24, and 37 genes were significantly changed at 6 h, day 3, day 10, and day 28 after IRI. Genes associated with cytokines, chemokines, and costimulatory molecules were upregulated. Pathway analysis identified CC motif chemokine receptor 5 (CCR5) as a candidate pathophysiological pathway. CCR5 upregulation was validated at the protein level, and CCR5 blockade improved renal function after kidney IRI. Using discovery techniques to identify transcriptional responses in purified kidney-infiltrating cells enabled the elucidation of novel mechanisms and therapeutic targets for IRI.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Riñón/lesiones , Riñón/patología , Receptores CCR5/metabolismo , Daño por Reperfusión/fisiopatología , Linfocitos T/metabolismo , Animales , Anticuerpos , Complejo CD3/genética , Complejo CD3/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Análisis por Conglomerados , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Riñón/citología , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Masculino , Ratones , Receptores CCR5/genética , Daño por Reperfusión/metabolismo , Organismos Libres de Patógenos Específicos
4.
J Am Soc Nephrol ; 22(4): 732-42, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21436290

RESUMEN

Inflammation contributes to the pathogenesis of ischemic acute kidney injury (AKI), and T cells mediate the early phase of ischemia-reperfusion injury (IRI). The Fas/Fas ligand (FasL) pathway modulates the balance of T cell subsets in the peripheral circulation as well as multiple inflammatory responses, suggesting that FasL may mediate ischemic AKI. Here, we induced bilateral renal IRI in mice bearing a loss-of-function mutation of FasL (the gld mutation) and in wild-type mice. Compared with wild-type mice, serum creatinine was lower in gld mice (1.4 ± 0.9 mg/dl versus 2.6 ± 0.4) at 24 hours after IRI (P<0.05). In addition, gld mice had fewer TNF-α-producing T lymphocytes in the kidneys and renal lymph nodes. Furthermore, pharmacologic blockade of FasL protected the kidneys of wild-type mice from IRI. Analysis of bone marrow chimeric mice suggested that the pathogenic effect of FasL involves leukocytes; reconstitution of wild-type mice with gld splenocytes attenuated IRI. In contrast, reconstitution of gld mice with wild-type splenocytes enhanced IRI. These data demonstrate that FasL, particularly on leukocytes, mediates ischemic AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Proteína Ligando Fas/deficiencia , Leucocitos/metabolismo , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/metabolismo , Animales , Caspasa 3/metabolismo , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Peroxidasa/metabolismo , Daño por Reperfusión/metabolismo , Bazo/metabolismo , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Immunol ; 183(2): 984-92, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19561110

RESUMEN

T cells have been implicated in the early pathogenesis of ischemia reperfusion injury (IRI) of kidney, liver, lung, and brain. It is not known whether Ag-TCR engagement followed by Ag-specific T cell activation participates in IRI. T cell-deficient nu/nu mice are moderately resistant to renal IRI, which can be reversed upon reconstitution with syngeneic T cells. In this study, we found that nu/nu mice reconstituted with DO11.10 T cells, limited in their TCR repertoire, have significantly less kidney dysfunction and tubular injury after renal IRI compared with that in nu/nu mice reconstituted with wild-type T cells having a diverse TCR repertoire. CD4(+) T cells infiltrating ischemic kidneys of nu/nu mice reconstituted with DO11.10 T cells exhibited lower IFN-gamma production than that of wild-type controls. Frequency of regulatory T cells in kidneys of these mice was similar in both DO11.10 T cells and wild-type T cell recipient groups. DO11.10 mice immunized with OVA-CFA had significantly worse kidney function at 24 h after ischemia than those immunized with CFA alone. Thus, without T cell activation, diverse TCR repertoire was important for renal IRI in naive mice. However, once T cells were activated in an Ag-specific manner through TCR in DO11.10 mice, a restricted TCR repertoire no longer limited the extent of kidney injury. Thus, both TCR repertoire-dependent and -independent factors mediate T cell functions in kidney IRI.


Asunto(s)
Epítopos de Linfocito T/inmunología , Enfermedades Renales/patología , Daño por Reperfusión/inmunología , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/fisiología , Quimiotaxis de Leucocito , Activación de Linfocitos/inmunología , Ratones , Ratones Desnudos , Daño por Reperfusión/etiología , Linfocitos T/fisiología , Linfocitos T/trasplante , Linfocitos T Reguladores/fisiología
6.
Am J Physiol Renal Physiol ; 297(1): F125-37, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19403643

RESUMEN

Distant organ effects of acute kidney injury (AKI) are a leading cause of morbidity and mortality. While little is known about the underlying mechanisms, limited data suggest a role for inflammation and apoptosis. Utilizing a lung candidate gene discovery approach in a mouse model of ischemic AKI-induced lung dysfunction, we identified prominent lung activation of 66 apoptosis-related genes at 6 and/or 36 h following ischemia, of which 6 genes represent the tumor necrosis factor receptor (TNFR) superfamily, and another 23 genes are associated with the TNFR pathway. Given that pulmonary apoptosis is an important pathogenic mechanism of acute lung injury (ALI), we hypothesized that AKI leads to pulmonary proapoptotic pathways that facilitate lung injury and inflammation. Functional correlation with 1) terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling and 2) active caspase-3 (aC3) activity, immunoblotting, and immunohistochemistry (IHC) identified kidney IRI-induced pulmonary apoptosis at 24 h, and colocalization studies with CD34 identified predominantly endothelial apoptosis. Mice were treated with the caspase inhibitor Z-VAD-FMK (0.25 mg ip) or vehicle 1 h before and 8 h after sham or kidney IRI, and bronchoalveolar lavage fluid protein was measured at 36 h as a surrogate for lung leak. Caspase inhibition reduced lung microvascular changes after kidney IRI. The pulmonary apoptosis seen in wild-type control mice during AKI was absent in TNFR(-/-) mice. Using an initial genomic approach to discovery followed by a mechanistic approach to disease targeting, we demonstrate that pulmonary endothelial apoptosis is a direct mediator of the distant organ dysfunction during experimental AKI.


Asunto(s)
Lesión Renal Aguda/complicaciones , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/etiología , Apoptosis/fisiología , Caspasa 3/metabolismo , Daño por Reperfusión/complicaciones , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/genética , Inhibidores de Caspasas , Modelos Animales de Enfermedad , Endotelio/efectos de los fármacos , Endotelio/enzimología , Endotelio/patología , Perfilación de la Expresión Génica , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo
7.
Kidney Int ; 76(3): 277-85, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19436334

RESUMEN

Oxidative stress is involved in acute kidney injury due to ischemia-reperfusion and chemotherapy-induced nephrotoxicity. To investigate their basic mechanisms we studied the role of nuclear factor-erythroid 2-p45-related factor 2 (Nrf2), a redox-sensitive transcription factor that regulates expression of several antioxidant and cytoprotective genes. We compared the responses of Nrf2-knockout mice and their wild-type littermates in established mouse models of ischemia-reperfusion injury and cisplatin-induced nephrotoxicity. Several Nrf2-regulated genes encoding antioxidant enzymes/proteins were significantly upregulated in the kidneys of wild type but not Nrf2-knockout mice following renal ischemia. Renal function, histology, vascular permeability, and survival were each significantly worse in the Nrf2 knockout mice. Further, proinflammatory cytokine and chemokine expression tended to increase after ischemia in the knockout compared to the wild-type mice. Treatment of the knockout mice with the antioxidants N-acetyl-cysteine or glutathione improved renal function. The knockout mice were more susceptible to cisplatin-induced nephrotoxicity, and this was blunted by N-acetyl-cysteine pretreatment. Our study demonstrates that Nrf2-deficiency enhances susceptibility to both ischemic and nephrotoxic acute kidney injury, and identifies this transcription factor as a potential therapeutic target in these injuries.


Asunto(s)
Lesión Renal Aguda/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Daño por Reperfusión/metabolismo , Acetilcisteína/farmacología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Animales , Antineoplásicos/toxicidad , Permeabilidad Capilar , Cisplatino/toxicidad , Glutatión/farmacología , Mediadores de Inflamación/metabolismo , Riñón/patología , Pruebas de Función Renal , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Regulación hacia Arriba
8.
Microvasc Res ; 77(3): 340-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19323971

RESUMEN

Although previous studies have demonstrated that microvascular dysfunction and inflammation occur in ischemia-reperfusion injury (IRI), the underlying mechanisms are poorly understood. We hypothesized that T cells could mediate renal vascular permeability (RVP) during IRI. We evaluated renal vascular permeability by extravasation of Evans blue dye from the kidney in CD3, CD4 or CD8 T cell deficient mice as well as in TNF receptor knock out mice in our mouse model of kidney ischemia-reperfusion injury. In wild type mice, RVP was significantly increased at 3 h, peaked at 6 h and declined by 24 h after ischemia. Immunohistochemistry revealed that CD3(+) T cells trafficked into ischemic kidney at 1 h and peaked at 6 h. Gene microarray analysis demonstrated that endothelial-related genes including TNF-alpha were up-regulated in ischemic kidney. The production of TNF-alpha and IFN-gamma protein was increased in CD3 and CD4 T cells from the blood and kidney after ischemia. The rise in RVP after ischemia in wild type mice was attenuated in CD3, CD4 or CD8 T cell deficient mice as well as in TNF receptor knock out mice. The attenuation of RVP in CD3 T-cell deficient mice after ischemia was restored by adoptive transfer of T cells from WT mice. Our data demonstrate that T cells directly contribute to the increased RVP after kidney ischemia-reperfusion, potentially through T cell cytokine production.


Asunto(s)
Permeabilidad Capilar/inmunología , Riñón/inmunología , Daño por Reperfusión/inmunología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Permeabilidad Capilar/genética , Modelos Animales de Enfermedad , Técnicas para Inmunoenzimas , Interferón gamma/biosíntesis , Interferón gamma/genética , Riñón/patología , Riñón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Recuperación de la Función , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba
9.
Kidney Int ; 75(5): 526-35, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19092796

RESUMEN

It is well-established that significant ischemia-reperfusion injury during kidney transplantation results in increased incidence of long-term fibrosis and rejection. To test for a role of T cell infiltration and activation following ischemic injury, we induced both bilateral and unilateral renal ischemia in mice, followed by reperfusion, and then isolated mononuclear cells. Analysis of these cells by flow cytometry showed that 2 weeks after bilateral ischemia there was a significant increase of CD8(+) T cells. Furthermore, both CD4(+) and CD8(+) T cells infiltrated the injured kidney 6 weeks after unilateral ischemia. These T cells had increased expression of CD69(+) and CD44(hi)CD62L(-), markers of activation and effector-memory, respectively. CD4(+)NK1.1(+) and CD19(+) B cells were decreased in percentage both 6 and 11 weeks after bilateral or unilateral injury. There was a significant upregulation of IL-1beta, IL-6, TNF-alpha, IFN-gamma, MIP-2, and RANTES expression, measured by real-time PCR, 6 weeks after unilateral renal ischemia, further indicating T cell activation. Depletion of CD4(+) and CD8(+) T cells before ischemia caused less medullary damage and reduced kidney IFN-gamma expression, whereas their depletion following ischemia increased kidney IL-1beta; however, depletion of these cells had no effect on histological damage to the kidney. Our study demonstrates that moderate or severe kidney ischemia induces long-term T lymphocyte infiltration and cytokine/chemokine upregulation, leading to kidney structural changes.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Enfermedades Renales/inmunología , Daño por Reperfusión/inmunología , Linfocitos T/inmunología , Animales , Citocinas/biosíntesis , Citometría de Flujo , Memoria Inmunológica/inmunología , Inmunofenotipificación , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios , Ratones
10.
Am J Physiol Lung Cell Mol Physiol ; 296(1): L3-L11, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18849441

RESUMEN

We hypothesized that the influence of acute kidney injury (AKI) on the sensitivity of the lung to an injurious process varies with the severity of the injurious process. Thus, we thought that AKI would exacerbate lung injury from low degrees of lung trauma but attenuate lung injury from higher degrees of lung trauma. C57BL/6 mice underwent AKI (30-min kidney ischemia) or sham surgery, followed at 24 h by 4 h of spontaneous breathing (SB), mechanical ventilation with low tidal volume (7 ml/kg, LTV), or mechanical ventilation with high tidal volume (30 ml/kg, HTV). Compared with LTV, median bronchoalveolar lavage (BAL) protein leak was significantly lower with SB and greater with HTV in both sham and AKI mice. Compared with LTV, median Evans blue dye-labeled albumin extravasation in lungs (L-EBD) was also significantly lower with SB and greater with HTV. L-EBD showed a significant interaction between ventilatory mode and kidney health, such that AKI attenuated the L-EBD rise seen in HTV vs. LTV sham mice. An interaction between ventilatory mode and kidney health could also be seen in BAL neutrophil number (PMN). Thus, AKI attenuated the BAL PMN rise seen in HTV vs. LTV sham mice. These data support the presence of a complex interaction between mechanical ventilation and AKI in which the sensitivity of the lung to trauma varies with the magnitude of the trauma and may involve a modification of pulmonary neutrophil activity by AKI.


Asunto(s)
Lesión Pulmonar Aguda , Enfermedades Renales/complicaciones , Enfermedades Renales/fisiopatología , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Enfermedad Aguda , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/fisiopatología , Lesión Pulmonar Aguda/terapia , Resistencia de las Vías Respiratorias/fisiología , Animales , Presión Sanguínea/fisiología , Líquido del Lavado Bronquioalveolar/citología , Dióxido de Carbono/sangre , Colorantes/farmacocinética , Modelos Animales de Enfermedad , Azul de Evans/farmacocinética , Linfocitos/patología , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/sangre , Índice de Severidad de la Enfermedad , Organismos Libres de Patógenos Específicos , Volumen de Ventilación Pulmonar
11.
Kidney Int ; 74(7): 849-51, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18794816

RESUMEN

The high mortality rate of acute kidney injury (AKI) despite advances in dialysis led to a renewed appreciation of the impact of AKI on distant organ dysfunction. Mechanistic studies demonstrated that AKI induces increased lung vascular permeability, soluble and cellular inflammation, and dysregulated salt and water channels. AKI also affects the brain, heart, liver, bone marrow, and gastrointestinal tract. Klein et al. now demonstrate that interleukin-6 is a direct mediator of AKI-induced lung changes.


Asunto(s)
Lesión Renal Aguda/complicaciones , Enfermedades Pulmonares/etiología , Uremia/complicaciones , Lesión Renal Aguda/mortalidad , Animales , Citocinas , Humanos , Sistema Inmunológico , Inflamación , Enfermedades Pulmonares/patología
12.
J Am Soc Nephrol ; 19(7): 1360-70, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18385426

RESUMEN

Although neurologic sequelae of acute kidney injury (AKI) are well described, the pathogenesis of acute uremic encephalopathy is poorly understood. This study examined the short-term effect of ischemic AKI on inflammatory and functional changes of the brain in mice by inducing bilateral renal ischemia for 60 min and studying the brains 24 h later. Compared with sham mice, mice with AKI had increased neuronal pyknosis and microgliosis in the brain. AKI also led to increased levels of the proinflammatory chemokines keratinocyte-derived chemoattractant and G-CSF in the cerebral cortex and hippocampus and increased expression of glial fibrillary acidic protein in astrocytes in the cortex and corpus callosum. In addition, extravasation of Evans blue dye into the brain suggested that the blood-brain barrier was disrupted in mice with AKI. Because liver failure also leads to encephalopathy, ischemic liver injury was induced in mice with normal renal function; neuronal pyknosis and glial fibrillary acidic protein expression were not increased, suggesting differential effects on the brain depending on the organ injured. For evaluation of the effects of AKI on brain function, locomotor activity was studied using an open field test. Mice subjected to renal ischemia or bilateral nephrectomy had moderate to severe declines in locomotor activity compared with sham-operated mice. These data demonstrate that severe ischemic AKI induces inflammation and functional changes in the brain. Targeting these pathways could reduce morbidity and mortality in critically ill patients with severe AKI.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Encéfalo/fisiopatología , Permeabilidad Capilar/fisiología , Encefalitis/etiología , Agua/metabolismo , Lesión Renal Aguda/sangre , Lesión Renal Aguda/complicaciones , Animales , Encéfalo/metabolismo , Proteína C-Reactiva/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Factor Estimulante de Colonias de Granulocitos/sangre , Etiquetado Corte-Fin in Situ , Isquemia/fisiopatología , Fallo Hepático Agudo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Actividad Motora/fisiología , Neuronas/fisiología , Proteínas/metabolismo
13.
Am J Physiol Renal Physiol ; 294(4): F900-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18256314

RESUMEN

Acute lung injury (ALI) in combination with acute kidney injury carries a mortality approaching 80% in the intensive care unit. Recently, attention has focused on the interaction of the lung and kidney in the setting of ALI and mechanical ventilation (MV). Small animal models of ALI and MV have demonstrated changes in inflammatory mediators, water channels, apoptosis, and function in the kidney early in the course of injury. The purpose of this investigation was to test the hypothesis that ALI and injurious MV cause early, measurable changes in kidney structure and function in a canine HCl aspiration model of ALI when hemodynamics and arterial blood gas tensions are carefully controlled. Intratracheal HCl induced profound ALI as demonstrated by increased shunt fraction and airway pressures compared with sham injury. Sham-injured animals had similar mean arterial pressure and arterial Pco(2) and HCO(3) levels compared with injured animals. Measurements of renal function including renal blood flow, urine flow, serum creatinine, glomerular filtration rate, urine albumin-to-creatinine ratio, and kidney histology scores were not different between groups. With maintenance of hemodynamic parameters and alveolar ventilation, ALI and injurious MV do not alter kidney structure and function early in the course of injury in this acid aspiration canine model. Kidney injury in large animal models may be more similar to humans and may differ from results seen in small animal models.


Asunto(s)
Ácido Clorhídrico/toxicidad , Pruebas de Función Renal , Enfermedades Pulmonares/etiología , Lesión Pulmonar , Administración por Inhalación , Animales , Presión Sanguínea/efectos de los fármacos , Dióxido de Carbono/sangre , Gasto Cardíaco/efectos de los fármacos , Creatinina/sangre , Diuresis/efectos de los fármacos , Perros , Tasa de Filtración Glomerular/efectos de los fármacos , Humanos , Ácido Clorhídrico/administración & dosificación , Concentración de Iones de Hidrógeno , Pulmón/efectos de los fármacos , Pulmón/patología , Modelos Animales , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología
14.
J Am Soc Nephrol ; 19(3): 547-58, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18235097

RESUMEN

Studies in humans and animal models have demonstrated that acute kidney injury (AKI) has a significant effect on the function of extrarenal organs. The combination of AKI and lung dysfunction is associated with 80% mortality; the lung, because of its extensive capillary network, is a prime target for AKI-induced effects. The study presented here tested the hypothesis that AKI leads to a vigorous inflammatory response and produces distinct genomic signatures in the kidney and lung. In a murine model of ischemic AKI, prominent global transcriptomic changes and histologic injury in both kidney and lung tissues were identified. These changes were evident at both early (6 h) and late (36 h) timepoints after 60-min bilateral kidney ischemia and were more prominent than similar timepoints after sham surgery or 30 min of ischemia. The inflammatory transcriptome (109 genes) of both organs changed with marked similarity, including the innate immunity genes Cd14, Socs3, Saa3, Lcn2, and Il1r2. Functional genomic analysis of these genes suggested that IL-10 and IL-6 signaling was involved in the distant effects of local inflammation, and this was supported by increased serum levels of IL-10 and IL-6 after ischemia-reperfusion. In summary, this is the first comprehensive analysis of concomitant inflammation-associated transcriptional changes in the kidney and a remote organ during AKI. Functional genomic analysis identified potential mediators that connect local and systemic inflammation, suggesting that this type of analysis may be a useful discovery tool for novel biomarkers and therapeutic drug development.


Asunto(s)
Lesión Renal Aguda/metabolismo , Inflamación/metabolismo , Riñón/metabolismo , Pulmón/metabolismo , Daño por Reperfusión/metabolismo , Animales , Perfilación de la Expresión Génica , Genómica , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo , Transcripción Genética
15.
Am J Physiol Renal Physiol ; 293(1): F30-40, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17327501

RESUMEN

Acute kidney injury (AKI) is associated with significant mortality, which increases further when combined with acute lung injury. Experiments in rodents have shown that kidney ischemia-reperfusion injury (IRI) facilitates lung injury and inflammation. To identify potential ischemia-specific lung molecular pathways involved, we conducted global gene expression profiling of lung 6 or 36 h following 1) bilateral kidney IRI, 2) bilateral nephrectomy (BNx), and 3) sham laparotomy in C57BL/6J mice. Bronchoalveolar lavage fluid analysis revealed increased total protein, and lung histology revealed increased cellular inflammation following IRI, but not BNx, compared with sham controls. Total RNA from whole lung was isolated and hybridized to 430MOEA (22,626 genes) GeneChips (n = 3/group), which were analyzed by robust multichip average and significance analysis of microarrays and linked to gene ontology (GO) terms using MAPPFinder. The microarray power analysis predicted that the false discovery rate (q < 1%) and > or =50%-fold change compared with sham would represent significant changes in gene expression. Analysis identified 266 and 455 ischemia-specific, AKI-associated lung genes with increased expression and 615 and 204 with decreased expression at 6 and 36 h, respectively, compared with sham controls. Real-time PCR analysis validated select array changes in lung serum amyloid A3 and endothelin-1. GO analysis revealed significant activation (Z > 1.95) of several proinflammatory and proapoptotic biological processes. Ischemic AKI induces functional and transcriptional changes in the lung distinct from those induced by uremia alone. Further investigation using this lung molecular signature induced by kidney IRI will provide mechanistic insights and new therapies for critically ill patients with AKI.


Asunto(s)
Enfermedades Renales/patología , Nefrectomía , Daño por Reperfusión/patología , Enfermedad Aguda , Animales , Apoptosis/fisiología , Líquido del Lavado Bronquioalveolar/citología , Análisis por Conglomerados , Perfilación de la Expresión Génica , Enfermedades Renales/genética , Enfermedades Renales/fisiopatología , Pruebas de Función Renal , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/genética , Daño por Reperfusión/genética , Daño por Reperfusión/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tamaño de la Muestra , Transcripción Genética , Ubiquitina/metabolismo , Uremia/metabolismo
16.
J Immunol ; 177(5): 3380-7, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16920979

RESUMEN

T and B lymphocytes have been implicated in the pathogenesis of renal ischemia reperfusion injury (IRI). The trafficking of lymphocytes into kidneys during IRI has been postulated to underlie this effect, but has not been rigorously studied. We therefore characterized the lymphocyte populations infiltrating into mouse kidneys 3 and 24 h after renal IRI. Immunohistochemistry and flow cytometry staining of kidney lymphocytes showed increased trafficking of CD3+ T cells and CD19+ B cells in both sham-operated and IRI mice 3 h after renal IRI. In the IRI mice, increased infiltration of NK1.1+ and CD4+ NK1.1+ cells compared with normal and sham-operated mice was observed 3 and 24 h after renal IRI, respectively. After 24 h of renal IRI, the decreased percentages of CD3+, CD19+, and NK1.1+ populations in the IRI mice compared with control groups were observed. Increased TNF-alpha and IFN-gamma production of kidney infiltration CD3+ T cells in IRI mice but not sham-operated mice was found. Unexpectedly, isolation and transfer of kidney-infiltrating lymphocytes 24 h after renal IRI into T cell-deficient mice reduced their functional and histological injury after renal IRI, suggesting that kidney-infiltrating lymphocytes could have a protective function. These quantitative, qualitative, and functional changes in kidney lymphocytes provide mechanistic insight into how lymphocytes modulate IRI, as well as demonstrating that abdominal surgery alone leads to lymphocyte changes in kidney.


Asunto(s)
Riñón/inmunología , Riñón/patología , Linfocitos/inmunología , Fenotipo , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Complejo CD3/inmunología , Complejo CD3/metabolismo , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Movimiento Celular , Inmunohistoquímica , Interferón gamma/biosíntesis , Riñón/lesiones , Células Asesinas Naturales/inmunología , Lectinas Tipo C , Activación de Linfocitos , Linfocitos/citología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Am J Physiol Renal Physiol ; 291(5): F981-6, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16757731

RESUMEN

Severe ischemia-reperfusion injury (IRI) predisposes to long-term impairment in kidney function both in patients and experimentally through unknown mechanisms. Given emerging evidence implicating lymphocytes in the pathogenesis of early injury to kidney, liver, and lung after IRI, we hypothesized that kidney IRI would potentially release or expose normally sequestered antigens that would lead to proliferation of antigen-recognizing lymphocytes. This, in turn, would directly participate in progressive kidney injury. To test this hypothesis, we purified splenic lymphocytes from C57BL/6 mice with severe renal IRI or sham operation 6 wk postischemia and transferred these cells to normal mice. Donor mice with IRI had significant fibrosis and cellular inflammation. The recipient mice were followed for 6 or 12 wk. Donor lymphocytes were found to traffic into recipient kidney. Twelve weeks after transfer, kidneys from mice which received IRI-primed lymphocytes exhibited significantly increased urinary albumin excretion compared with lymphocytes from sham mice. Splenic CD3(+), CD4(+), CD3(+)CD25(+), and CD4(+)CD44(+) counts were significantly increased in mice after lymphocyte transfer from IRI mice vs. mice with lymphocytes from sham mice. These data demonstrate that lymphocytes from IRI mice can traffic to recipient kidney and directly mediate albuminuria. These data identify a novel mechanism by which initial kidney injury predisposes to long-term dysfunction and identify lymphocytes as potential therapeutic targets for progressive renal diseases.


Asunto(s)
Traslado Adoptivo , Albuminuria/inmunología , Linfocitos T CD4-Positivos/inmunología , Isquemia/inmunología , Insuficiencia Renal Crónica/inmunología , Albuminuria/patología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Fibrosis , Memoria Inmunológica , Isquemia/patología , Riñón/inmunología , Riñón/patología , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Nefritis Intersticial/inmunología , Nefritis Intersticial/patología , Insuficiencia Renal Crónica/patología , Bazo/inmunología
18.
J Immunol ; 176(11): 7015-20, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16709863

RESUMEN

Ischemic preconditioning (IP) is a well-established phenomenon, and the underlying mechanisms of IP are thought to involve adaptive changes within the injured tissue. Because one of the main functions of immune cells is to harbor memory, we hypothesized that circulating immune cells could mediate IP by responding to an initial ischemia reperfusion injury (IRI) and then mediate decreased injury after a second IRI event. C57BL/6 mice underwent 30 min of bilateral renal clamping or sham operation. At 5 days after ischemia, purified leukocytes from spleen were adoptively transferred into T cell-deficient (nu/nu) mice. After 1 wk, these mice underwent 30 min of renal IRI. The nu/nu mice receiving leukocytes from ischemic wild-type mice had significantly reduced renal injury compared with nu/nu mice receiving leukocytes from sham-operated, wild-type mice. Infiltration of neutrophil and macrophage in postischemic kidney did not correlate with the protection. No difference in kidney C3d or IgG deposition was detected between groups. Given that inducible NO synthase (iNOS) has been implicated in IP, leukocytes from ischemic or sham-operated, iNOS-deficient mice were transferred into nu/nu mice. Effects similar to those of wild-type transfer of ischemic leukocytes were demonstrated; thus, iNOS was not mediating the IP effect of leukocytes. This is the first evidence that immune cells are primed after renal IRI and thereby lose the capacity to cause kidney injury during a second episode of IRI. This finding may also be relevant for elucidating the mechanisms underlying cross-talk between injured kidney and distant organs.


Asunto(s)
Precondicionamiento Isquémico , Riñón/irrigación sanguínea , Riñón/inmunología , Linfopenia/genética , Linfopenia/inmunología , Traslado Adoptivo , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Complemento C3d/metabolismo , Hemoglobinas/metabolismo , Inmunoglobulina G/metabolismo , Riñón/enzimología , Transfusión de Leucocitos , Linfopenia/enzimología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Óxido Nítrico Sintasa de Tipo II/fisiología , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología , Daño por Reperfusión/prevención & control , Bazo/citología , Bazo/trasplante
19.
J Am Soc Nephrol ; 17(3): 765-74, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16481417

RESUMEN

Recent evidence supports a role for an inflammatory pathogenesis of cisplatin nephrotoxicity, but immune cell-mediated mechanisms in this disease are still largely unknown. The role for T lymphocytes on cisplatin-induced acute kidney injury was examined with C57BL/6 T cell-deficient (nu/nu) mice and CD4- or CD8-deficient mice and their wild-type (WT) littermates. All mice received a single dose of cisplatin at 40 mg/kg (intraperitoneally) and were followed up for 72 h. At 72 h after cisplatin administration, T cell-deficient mice had a marked attenuation in renal dysfunction (serum creatinine 3.2+/-0.5 versus 0.8+/-0.1 mg/dl; P=0.007), kidney tubular injury (scores 1.44+/-0.15 versus 0.22+/-0.08; P<0.0001), and survival. Adoptive transfer of T cells into nu/nu mice followed by cisplatin enhanced renal dysfunction and tubular injury. The increase in renal myeloperoxidase activity after cisplatin administration was blunted in nu/nu mice. Renal TNF-alpha, IL-1beta, and keratinocyte-derived chemokine protein expression was increased in WT mice but not in nu/nu mice after cisplatin administration. T cell levels significantly increased in kidneys of WT mice after cisplatin administration as early as at 1 h, peaked at 12 h, and declined by 24 h. CD4- and, to a lesser degree, CD8-deficient mice were relatively protected from cisplatin-induced mortality and renal dysfunction compared with WT mice. These data demonstrate that T lymphocytes are direct mediators of experimental cisplatin nephrotoxicity. Targeting T lymphocytes could lead to improved ways to administer cisplatin safely to cancer patients.


Asunto(s)
Cisplatino/toxicidad , Necrosis Tubular Aguda/inducido químicamente , Necrosis Tubular Aguda/patología , Linfocitos T/fisiología , Traslado Adoptivo , Animales , Biopsia con Aguja , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Inmunohistoquímica , Interleucina-1/metabolismo , Pruebas de Función Renal , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Especificidad de la Especie , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo
20.
Am J Physiol Renal Physiol ; 290(5): F1187-93, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16368740

RESUMEN

Renal ischemia-reperfusion injury (IRI) is the leading cause of acute kidney injury [AKI; acute renal failure (ARF)] in native kidneys and delayed graft function in deceased donor kidney transplants. Serum creatinine rises late after renal IRI, which results in delayed diagnosis. There is an important need to identify novel biomarkers for early diagnosis and prognosis in renal IRI. Given the inflammatory pathophysiology of renal IRI, we used a protein array to measure 18 cytokines and chemokines in a mouse model of renal IRI at 3, 24, and 72 h postischemia. A rise in renal keratinocyte-derived chemokine (KC) was the earliest and most consistent compared with other molecules, with 3-h postischemia values being 9- and 13-fold greater than sham and normal animals, respectively. Histological changes were evident within 1 h of IRI but serum creatinine only increased 24 h after IRI. With the use of an ELISA, KC levels in serum and urine were highest 3 h postischemia, well before a significant rise in serum creatinine. The human analog of KC, Gro-alpha, was markedly elevated in urine from humans who received deceased donor kidney transplants that required dialysis, compared with deceased donor kidney recipients with good graft function and live donor recipients with minimal ischemia. Measurement of KC and its human analog, Gro-alpha, could serve as a useful new biomarker for ischemic ARF.


Asunto(s)
Quimiocinas/análisis , Isquemia/diagnóstico , Queratinocitos/química , Riñón/irrigación sanguínea , Enfermedad Aguda , Lesión Renal Aguda , Animales , Biomarcadores , Citocinas/análisis , Diagnóstico Diferencial , Trasplante de Riñón , Masculino , Ratones , Ratones Endogámicos C57BL , Pronóstico , Daño por Reperfusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...