Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Am J Physiol Renal Physiol ; 319(6): F1073-F1080, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33103444

RESUMEN

Cisplatin, a commonly used anticancer drug, has been shown to induce acute kidney injury, which limits its clinical use in cancer treatment. Emerging evidence has suggested that AMP-activated protein kinase (AMPK), which functions as a cellular energy sensor, is activated by various cellular stresses that deplete cellular ATP. However, the potential role of AMPK in cisplatin-induced apoptosis of renal tubular epithelial cells has not been studied. In this study, we demonstrated that cisplatin activates AMPK (Thr172 phosphorylation) in cultured renal tubular epithelial cells in a time-dependent manner, which was associated with p53 phosphorylation. Compound C, a selective AMPK inhibitor, suppressed cisplatin-induced AMPK activation, p53 phosphorylation, Bax induction, and caspase 3 activation. Furthermore, silencing AMPK expression by siRNA attenuated cisplatin-induced p53 phosphorylation, Bax induction, and caspase 3 activation. In a mouse model of cisplatin-induced kidney injury, compound C inhibited p53 phosphorylation, Bax expression, caspase 3 activation, and apoptosis, protecting the kidney from injury and dysfunction. Taken together, these results suggest that the AMPK-p53-Bax signaling pathway plays a crucial role in cisplatin-induced tubular epithelial cell apoptosis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Lesión Renal Aguda/inducido químicamente , Apoptosis/efectos de los fármacos , Cisplatino/toxicidad , Células Epiteliales/efectos de los fármacos , Túbulos Renales/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/patología , Animales , Caspasa 3/metabolismo , Línea Celular , Células Epiteliales/enzimología , Células Epiteliales/patología , Túbulos Renales/enzimología , Túbulos Renales/patología , Ratones , Fosforilación , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
2.
Am J Physiol Renal Physiol ; 318(1): F209-F215, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31813254

RESUMEN

Cisplatin can cause acute kidney injury (AKI), but the molecular mechanisms are not well understood. The objective of the present study was to examine the role of transforming growth factor-ß-activated kinase-1 (TAK1) in the pathogenesis of cisplatin-induced AKI. Wild-type mice and proximal tubule TAK1-deficient mice were treated with vehicle or cisplatin. Compared with wild-type control mice, proximal tubule TAK1-deficient mice had less severe kidney dysfunction, tubular damage, and apoptosis after cisplatin-induced AKI. Furthermore, conditional disruption of TAK1 in proximal tubular epithelial cells reduced caspase-3 activation, proinflammatory molecule expression, and JNK phosphorylation in the kidney in cisplatin-induced AKI. Taken together, cisplatin activates TAK1-JNK signaling pathway to promote tubular epithelial cell apoptosis and inflammation in cisplatin-induced AKI. Targeting TAK1 could be a novel therapeutic strategy against cisplatin-induced AKI.


Asunto(s)
Lesión Renal Aguda/genética , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Riñón/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis/genética , Células Epiteliales/patología , Riñón/patología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Noqueados , Fosforilación
3.
Sci Rep ; 13(1): 22255, 2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-38097707

RESUMEN

Cisplatin (CP) induces acute kidney injury (AKI) whereby proximal tubules undergo regulated necrosis. Repair is almost complete after a single dose. We now demonstrate a role for Apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (Apobec-1) that is prominently expressed at the interface between acute and chronic kidney injury (CKD), in the recovery from AKI. Apobec-1 knockout (KO) mice exhibited greater mortality than in wild type (WT) and more severe AKI in both CP- and unilateral ischemia reperfusion (IR) with nephrectomy. Specifically, plasma creatinine (pCr) 2.6 ± 0.70 mg/dL for KO, n = 10 and 0.16 ± 0.02 for WT, n = 6, p < 0.0001 in CP model and 1.34 ± 0.22 mg/dL vs 0.75 ± 0.06, n = 5, p < 0.05 in IR model. The kidneys of Apobec-1 KO mice showed increased necrosis, increased expression of KIM-1, NGAL, RIPK1, ASCL4 and increased lipid accumulation compared to WT kidneys (p < 0.01). Neutrophils and activated T cells were both increased, while macrophages were reduced in kidneys of Apobec-1 KO animals. Overexpression of Apobec-1 in mouse proximal tubule cells protected against CP-induced cytotoxicity. These findings suggest that Apobec-1 mediates critical pro-survival responses to renal injury and increasing Apobec-1 expression could be an effective strategy to mitigate AKI.


Asunto(s)
Lesión Renal Aguda , Daño por Reperfusión , Ratones , Animales , Desaminasas APOBEC-1/metabolismo , Cisplatino/efectos adversos , Cisplatino/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Riñón/metabolismo , Necrosis/metabolismo , Ratones Noqueados , Daño por Reperfusión/metabolismo , Ratones Endogámicos C57BL
4.
Am J Physiol Renal Physiol ; 300(5): F1171-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325496

RESUMEN

Cisplatin cytotoxicity is dependent on cyclin-dependent kinase 2 (Cdk2) activity in vivo and in vitro. We found that an 18-kDa protein identified by mass spectrometry as p21(WAF1/Cip1) was phosphorylated by Cdk2 starting 12 h after cisplatin exposure. The analysis showed it was phosphorylated at serine 78, a site not previously identified. The adenoviral transduction of p21 before cisplatin exposure protects from cytotoxicity by inhibiting Cdk2. Although cisplatin causes induction of endogenous p21, the protection is inefficient. We hypothesized that phosphorylation of p21 at serine 78 could affect its role as a Cdk inhibitor, and thereby lessen its ability to protect from cisplatin cytotoxicity. To investigate the effect of serine 78 phosphorylation on p21 activity, we replaced serine 78 with aspartic acid, creating the phosphomimic p21(S78D). Mutant p21(S78D) was an inefficient inhibitor of Cdk2 and was inefficient at protecting TKPTS cells from cisplatin-induced cell death. We conclude that phosphorylation of p21 by Cdk2 limits the effectiveness of p21 to inhibit Cdk2, which is the mechanism for continued cisplatin cytotoxicity even after the induction of a protective protein.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/patología , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Ciclina A/genética , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Células HEK293 , Humanos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Ratones , Ratones de la Cepa 129 , Datos de Secuencia Molecular , Mutación , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Serina , Espectrometría de Masas en Tándem , Factores de Tiempo , Transducción Genética , Transfección
5.
Kidney Int ; 79(2): 149-50, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21191389

RESUMEN

Experimental acute kidney injury (AKI) is accompanied by the death of renal tubule epithelial cells, necrosis and apoptosis of the terminal portion of the proximal tubule, and apoptosis in the distal nephron. While immune competent cells invading the kidney play a role in such cell death, intervention in these processes only partially ameliorates the extent of cell death. Given the results of Linkermann et al. in this issue of KI, an epithelium-derived component of immune mediated cell death must now be strongly considered.


Asunto(s)
Lesión Renal Aguda/patología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/inmunología , Animales , Apoptosis , Cisplatino/toxicidad , Células Epiteliales/inmunología , Células Epiteliales/patología , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/patología , Ratones , Necrosis
6.
Am J Physiol Renal Physiol ; 299(1): F112-20, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20444741

RESUMEN

Cisplatin cytotoxicity is dependent on cyclin-dependent kinase 2 (Cdk2) activity in vivo and in vitro. A Cdk2 mutant (Cdk2-F80G) was designed in which the ATP-binding pocket was altered. When expressed in mouse kidney cells, this protein was kinase inactive, did not inhibit endogenous Cdk2, but protected from cisplatin. The mutant was localized in the cytoplasm, but when coexpressed with cyclin A, it was activated, localized to the nucleus, and no longer protected from cisplatin cytotoxicity. Cells exposed to cisplatin in the presence of the activated mutant had an apoptotic phenotype, and endonuclease G was released from mitochondria similar to that mediated by endogenous Cdk2. But unlike apoptosis mediated by wild-type Cdk2, cisplatin exposure of cells expressing the activated mutant did not cause cytochrome c release or significant caspase-3 activation. We conclude that cisplatin likely activates both caspase-dependent and -independent cell death, and Cdk2 is required for both pathways. The mutant-inactive Cdk2 protected from both death pathways, but after activation by excess cyclin A, caspase-independent cell death predominated.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Quinasa 2 Dependiente de la Ciclina/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Transporte Activo de Núcleo Celular , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Caspasa 3/metabolismo , Núcleo Celular/enzimología , Células Cultivadas , Ciclina A/genética , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Citocromos c/metabolismo , Citoplasma/enzimología , Citoprotección , Endodesoxirribonucleasas/metabolismo , Humanos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mutación , Proteínas Recombinantes de Fusión/metabolismo , Transfección
7.
Kidney Int ; 76(6): 604-13, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19536080

RESUMEN

Acute kidney injury (AKI) activates pathways of cell death and cell proliferation. Although seemingly discrete and unrelated mechanisms, these pathways can now be shown to be connected and even to be controlled by similar pathways. The dependence of the severity of renal-cell injury on cell cycle pathways can be used to control and perhaps to prevent acute kidney injury. This review is written to address the correlation between cellular life and death in kidney tubules, especially in acute kidney injury.


Asunto(s)
Ciclo Celular , Riñón/patología , Enfermedad Aguda , Animales , Apoptosis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Quinasas Ciclina-Dependientes/fisiología , Daño del ADN , Humanos
8.
Kidney Int ; 76(5): 521-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19553911

RESUMEN

Angiotensin II upregulates the expression of LOX-1, a recently identified oxidized low-density lipoprotein receptor controlled by redox state which in turn upregulates angiotensin II activity on its activation. To test whether interruption of this positive feedback loop might reduce angiotensin II-induced hypertension and subsequent renal injury, we studied LOX-1 knockout mice. After infusion with angiotensin II for 4 weeks systolic blood pressure gradually increased in the wild-type mice; this rise was significantly attenuated in the LOX-1 knockout mice. Along with the rise in systolic blood pressure, renal function (blood urea nitrogen and creatinine) decreased in the wild-type mice, but the deterioration of function was significantly less in the LOX-1 knockout mice. Glomerulosclerosis, arteriolar sclerosis, tubulointerstitial damage, and renal collagen accumulation were all significantly less in the LOX-1 knockout mice. The reduction in collagen formation was accompanied by a decrease in connective tissue growth factor mRNA, angiotensin type 1 receptor expression, and phosphorylation of p38 and p44/42 mitogen-activated protein kinases. Expression of endothelial nitric oxide synthase was increased in the kidneys of the LOX-1 knockout mice compared to the wild-type mice. Overall, our study suggests that LOX-1 is a key modulator in the development of angiotensin II-induced hypertension and subsequent renal damage.


Asunto(s)
Angiotensina III/toxicidad , Riñón/patología , Receptores Depuradores de Clase E/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/genética , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Fibrosis , Hipertensión/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/genética , Receptor de Angiotensina Tipo 1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
10.
Semin Nephrol ; 23(5): 460-4, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-13680535

RESUMEN

Cisplatin remains a major antineoplastic drug for the treatment of solid tumors. Its chief dose-limiting side effect is nephrotoxicity, which evolves slowly and predictably after initial and repeated exposure. The kidney accumulates cisplatin to a higher degree than other organs perhaps via mediated transport. Functionally, reduced renal perfusion and a concentrating defect characterize its nephrotoxicity, whereas morphologically necrosis of the terminal portion of the proximal tubule and apoptosis predominantly in the distal nephron characterize its effects on cell fate. Among the earliest reactions of the kidney to cisplatin is the activation of the MAPK cascade and molecular responses typical of the stress response. Repression of genes characteristic of the mature phenotype of the kidney, especially those serving transport function of the kidney, is also prominent. Metabolic responses, cell cycle events and the inflammatory cascade seem to be important determinants of the degree of renal failure induced by cisplatin. Manipulation of these responses may be exploited to reduce its toxicity clinically.


Asunto(s)
Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Riñón/efectos de los fármacos , Insuficiencia Renal/inducido químicamente , Caspasas/fisiología , Ciclo Celular/fisiología , Muerte Celular/fisiología , Citocinas/inmunología , Diuresis/fisiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inducido químicamente , Fluidoterapia/métodos , Humanos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Insuficiencia Renal/prevención & control
11.
Semin Nephrol ; 23(5): 449-59, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-13680534

RESUMEN

Research into mechanisms of acute renal failure has begun to reveal molecular targets for possible therapeutic intervention. Much useful knowledge into the causes and prevention of this syndrome has been gained by the study of animal models. Most recently, investigation of the effects on acute renal failure of selected gene knock-outs in mice has contributed to our recognition of many previously unappreciated molecular pathways. Particularly, experiments have revealed the protective nature of 2 highly induced genes whose functions are to inhibit and control the cell cycle after acute renal failure. By use of these models we have started to understand the role of increased cell cycle activity after renal stress and the role of proteins induced by these stresses that limit this proliferation.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Proteínas de Ciclo Celular/fisiología , Ciclo Celular/fisiología , Riñón/fisiología , Regeneración/fisiología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/etiología , Lesión Renal Aguda/genética , Animales , Antineoplásicos/efectos adversos , Ciclo Celular/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Muerte Celular/fisiología , División Celular/fisiología , Cisplatino/efectos adversos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Modelos Animales , Recuperación de la Función/fisiología
12.
Kidney Int Suppl ; (91): S62-6, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15461706

RESUMEN

Acute renal failure (ARF) is defined as an abrupt fall in glomerular filtration rate. Fully 5% of all patients admitted to the hospital undergo ARF with an attendant increase in morbidity and mortality. We have studied murine models of ischemia/reperfusion and cisplatin-induced renal failure in detail to determine the physiologic and molecular events that are responsible for the syndrome. Both forms of treatment induce necrosis of the proximal tubules, as well as more subtle changes in distal nephron viability, including apoptosis. Both forms are characterized by reduced renal blood, reduced glomerular filtration rate, and a urine-concentrating defect. Simultaneous with the onset of these morphologic and functional features is the commitment of cells to DNA synthesis and cell division, which is preceded by activation of signal transduction pathways and gene transcription that presumably underlie the morphologic and functional changes responsible for the syndrome. We describe a functional genomic approach using microarray data and available database searches to attempt to predict new targets for investigation of the pathogenesis and treatment of this disease.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Riñón/fisiología , Lesión Renal Aguda/inducido químicamente , Animales , Cisplatino/toxicidad , Tasa de Filtración Glomerular , Humanos , Ratones , Daño por Reperfusión/metabolismo , Transducción de Señal , Transcripción Genética
13.
Cell Metab ; 11(6): 451-2, 2010 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-20519117

RESUMEN

Genome-wide association studies (GWAS) have been applied to complex diseases such as diabetes and hypertension, successfully uncovering strong gene associations of potential pathophysiologic significance. Recently, two studies (Köttgen et al., 2010; Chambers et al., 2010) have been applied to uncover genes relevant to the pathophysiology of chronic kidney disease (CKD).

14.
Am J Physiol Renal Physiol ; 294(3): F577-81, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18094030

RESUMEN

We have shown that mouse proximal tubule cells (TKPTS) survive H(2)O(2) stress by activating the cAMP-responsive element binding protein (CREB)-mediated transcription via the canonical EGFR-Ras/ERK pathway. By contrast, cisplatin activates EGFR/Ras/ERK signaling in TKPTS cells yet promotes cell death rather than survival. We now demonstrate that the cisplatin-induced activated EGFR/Ras/ERK signaling cascade fails to activate CREB-mediated transcription even in the presence of phosphorylated CREB. CREB-mediated transcription as well as survival was restored by the histone deacetylase (HDAC) inhibitor trichostatine A (TSA), an effective chemotherapeutic agent. Similar to severe oxidant stress, TSA-mediated survival could be abrogated by inhibition of CREB-mediated transcription. These studies confirm the importance of CREB-mediated transcription in the survival of renal cells subjected to either oxidant- or cisplatin-induced stress. The use of cisplatin and TSA in combined chemotherapy protocols may be an effective strategy to enhance cancer cell death and limit nephrotoxicity.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inhibidores de Histona Desacetilasas , Túbulos Renales Proximales/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Hidroxámicos/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Ratones , Fosforilación , Estrés Fisiológico/metabolismo , Activación Transcripcional/efectos de los fármacos
15.
J Biol Chem ; 283(10): 6110-7, 2008 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-18174162

RESUMEN

The fully executed epidermal growth factor receptor (EGFR)/Ras/MEK/ERK pathway serves a pro-survival role in renal epithelia under moderate oxidative stress. We and others have demonstrated that during severe oxidative stress, however, the activated EGFR is disconnected from ERK activation in cultured renal proximal tubule cells and also in renal proximal tubules after ischemia/reperfusion injury, resulting in necrotic death. Studies have shown that the tyrosine-phosphorylated p46/52 isoforms of the ShcA family of adaptor proteins connect the activated EGFR to activation of Ras and ERK, whereas the p66(shc) isoform can inhibit this p46/52(shc) function. Here, we determined that severe oxidative stress (after a brief period of activation) terminates activation of the Ras/MEK/ERK pathway, which coincides with ERK/JNK-dependent Ser(36) phosphorylation of p66(shc). Isoform-specific knockdown of p66(shc) or mutation of Ser(36) to Ala, but not to Asp, attenuated severe oxidative stress-mediated ERK inhibition and cell death in vitro. Also, severe oxidative stress (unlike ligand stimulation and moderate oxidative stress, both of which support survival) increased binding of p66(shc) to the activated EGFR and Grb2. This binding dissociated the SOS1 adaptor protein from the EGFR-recruited signaling complex, leading to termination of Ras/MEK/ERK activation. Notably, Ser(36) phosphorylation of p66(shc) and its increased binding to the EGFR also occurred in the kidney after ischemia/reperfusion injury in vivo. At the same time, SOS1 binding to the EGFR declined, similar to the in vitro findings. Thus, the mechanism we propose in vitro offers a means to ameliorate oxidative stress-induced cell injury by either inhibiting Ser(36) phosphorylation of p66(shc) or knocking down p66(shc) expression in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Túbulos Renales Proximales/metabolismo , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Animales , Línea Celular Transformada , Supervivencia Celular , Activación Enzimática , Epitelio/metabolismo , Proteína Adaptadora GRB2/metabolismo , Túbulos Renales Proximales/patología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Proteína Oncogénica p21(ras)/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Proteína SOS1/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
16.
Am J Physiol Renal Physiol ; 293(1): F52-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17459956

RESUMEN

E2F1 is a key regulator that links cell cycle progression and cell death. E2F1 activity is controlled by Cdk2-cyclin complexes via several mechanisms, such as phosphorylation of retinoblastoma protein (pRb) to release E2F1, direct phosphorylation, and stable physical interaction. We have demonstrated that cisplatin cytotoxicity depends on Cdk2 activity, and Cdk2 inhibition protects kidney cells from cisplatin-induced cell death in vitro and in vivo. Now we show that E2F1 is an important downstream effector of Cdk2 that accumulates in mouse kidneys and in cultured mouse proximal tubular cells (TKPTS) after cisplatin exposure by a Cdk2-dependent mechanism. Direct inhibition of E2F1 by transduction with adenoviruses expressing an E2F1-binding protein (TopBP1) protected TKPTS cells from cisplatin-induced apoptosis, whereas overexpression of E2F1 caused cell death. Moreover, E2F1 knockout mice were markedly protected against cisplatin nephrotoxicity by both functional and histological criteria. Collectively, cisplatin-induced cell death is dependent on Cdk2 activity, which is at least partly through the Cdk2-E2F1 pathway both in vitro and in vivo.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Quinasa 2 Dependiente de la Ciclina/fisiología , Factor de Transcripción E2F1/fisiología , Adenoviridae/genética , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Muerte Celular/efectos de los fármacos , Células Cultivadas , Colorantes , Quinasa 2 Dependiente de la Ciclina/genética , Factor de Transcripción E2F1/deficiencia , Factor de Transcripción E2F1/genética , Citometría de Flujo , Indoles , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Azul de Tripano
17.
J Am Soc Nephrol ; 17(9): 2434-42, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16914540

RESUMEN

Cisplatin is one of the most effective chemotherapeutics, but its usefulness is limited by its toxicity to normal tissues, including cells of the kidney proximal tubule. The purpose of these studies was to determine the mechanism of cisplatin cytotoxicity. It was shown in vivo that cisplatin administration induces upregulation of the gene for the p21 cyclin-dependent kinase (cdk) inhibitor in kidney cells. This protein is a positive effector on the fate of cisplatin-exposed renal tubule cells in vivo and in vitro; adenoviral transduction of p21 completely protected proximal tubule cells from cisplatin toxicity. Herein is reported that cdk2 inhibitory drugs protect kidney cells in vivo and in vitro, that transduction of kidney cells in vitro with dominant-negative cdk2 also protected, and that cdk2 knockout cells were resistant to cisplatin. The cdk2 knockout cells regained cisplatin sensitivity after transduction with wild-type cdk2. It is concluded that cisplatin cytotoxicity depends on cdk2 activation and that the mechanism of p21 protection is by direct inhibition of cdk2. This demonstrated the involvement of a protein that previously was associated with cell-cycle progression with pathways of apoptosis. It also was demonstrated that this pathway of cisplatin-induced cell death can be interceded in vivo to prevent nephrotoxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Quinasa 2 Dependiente de la Ciclina/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Células Cultivadas , Creatinina/sangre , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Ratones , Regulación hacia Arriba
18.
Am J Physiol Renal Physiol ; 289(3): F514-20, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15840769

RESUMEN

The p21 cyclin-dependent kinase (cdk) inhibitor protects cells from cisplatin cytotoxicity in vivo and in vitro. However, the mechanism of protection is not known. Separate p21 domains are known to interact with several different proteins having proapoptotic functions. To investigate the mechanism of protection by p21, we have constructed adenoviruses encoding the different domains of p21. We were able to localize the protective activity to a region of 54 amino acids containing the cyclin-cdk interacting moiety. Other protein binding domains of p21, including the NH2-terminal procaspase-3 interactive region and the COOH-terminal region containing the proliferating cell nuclear antigen binding domain and the nuclear localization signal, had little protective effect on cisplatin cytotoxicity. The dependence of cisplatin cytotoxicity on cdk2 activity was also demonstrated because 1) cisplatin caused a marked increase in cdk2 activity, which was prevented by the p21 expression adenovirus, and 2) a cdk2 dominant-negative adenovirus also protected cells from cisplatin-induced apoptosis. Thus the data suggest that the mechanism of p21 protection is by direct inhibition of cdk2 activity and that cisplatin-induced apoptosis is caused by a cdk2-dependent pathway.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antineoplásicos/toxicidad , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cisplatino/toxicidad , Lesión Renal Aguda/fisiopatología , Adenoviridae/genética , Apoptosis/fisiología , Quinasas CDC2-CDC28/genética , Quinasas CDC2-CDC28/metabolismo , Proteínas de Ciclo Celular/química , Línea Celular , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Humanos , Riñón/citología , Unión Proteica , Estructura Terciaria de Proteína
19.
Kidney Int ; 68(4): 1573-82, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16164634

RESUMEN

BACKGROUND: We showed that extracellular signal-regulated protein kinase (ERK) is prosurvival during oxidant stress both in the kidney and in cultured mouse proximal tubule (TKPTS) cells and demonstrated concomitant activation of ERK as well as the cyclic adenosine monophosphate (cAMP)-responsive element binding protein (CREB), during survival in vitro. We now show that CREB is a necessary prosurvival target of ERK. METHODS: Ischemia/reperfusion (I/R) injury was induced in 129Sv mice. Oxidant stress was induced by hydrogen peroxide (H(2)O(2)) in TKPTS cells. Activation of CREB was determined by immunohistochemistry and Western blotting. Inhibition and activation of CREB was achieved by mutant or activated CREB-containing adenoviruses in vitro. The effects of oxidant stress on cell survival, CREB binding, and CREB-mediated transcription was determined by cell counting, gelshift analysis, and luciferase assay, respectively. RESULTS: I/R activates CREB in the surviving distal nephron segments of the kidney. Inhibition of ERK and CREB abrogates survival after 0.5 mmol/L H(2)O(2) treatment, while overexpression of CREB ameliorates necrotic death caused by 1 mmol/L H(2)O(2). Inhibition of ERK also inhibited CREB activation. Binding of phosphorylated CREB to a CREB oligonucleotide was significantly increased after 0.5 mmol/L H(2)O(2) but decreased after 1 mmol/L H(2)O(2). Similarly, CREB-mediated transcription was significantly increased after 0.5 mmol/L H(2)O(2) treatment, while 1 mmol/L H(2)O(2) inhibited it. Interestingly, transcription from the CREB-driven bcl-2 promoter was unchanged after 0.5 mmol/L but decreased after 1 mmol/L H(2)O(2) treatment in agreement with Western blot studies. CONCLUSION: We show that survival during oxidant stress is mediated through CREB and identification of its downstream targets will reveal important survival pathways.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Estrés Oxidativo/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Peróxido de Hidrógeno/farmacología , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transcripción Genética/fisiología
20.
Am J Physiol Renal Physiol ; 286(2): F378-84, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12965891

RESUMEN

The optimal use of cisplatin as a chemotherapeutic drug has been limited by its nephrotoxicity. Murine models have been used to study cisplatin-induced acute renal failure. After cisplatin administration, cells of the S3 segment in the renal proximal tubule are especially sensitive and undergo extensive necrosis in vivo. Similarly, cultured proximal tubule cells undergo apoptosis in vitro after cisplatin exposure. We have shown in vivo that kidney cells enter the cell cycle after cisplatin administration but that cell cycle-inhibitory proteins p21 and 14-3-3sigma are also upregulated. These proteins coordinate the cell cycle, and deletion of either of the genes resulted in increased nephrotoxicity in vivo or increased cell death in vitro after exposure to cisplatin. However, it was not known whether cell cycle inhibition before acute renal failure could protect from cisplatin-induced cell death, especially in cells with functional p21 and 14-3-3sigma genes. Using several cell cycle inhibitors, including a p21 adenovirus, and the drugs roscovitine and olomoucine, we have been able to completely protect a mouse kidney proximal tubule cell culture from cisplatin-induced apoptosis. The protection by p21 was independent of an effect on the cell cycle and was likely caused by selective inhibition of caspase-dependent and -independent cell death pathways in the cells.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Células Epiteliales/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Proteínas 14-3-3 , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Células Epiteliales/citología , Exonucleasas/metabolismo , Exorribonucleasas , Túbulos Renales Proximales/citología , Ratones , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Purinas/farmacología , Roscovitina , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA