Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Int J Clin Pharmacol Ther ; 56(11): 501-506, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30148451

RESUMEN

With the increasing popularity of E-cigarettes, chronic exposure to nicotine (NIC) is emerging as a novel risk factor for the kidney. NIC increases oxidative stress in the kidneys, which impairs the viability and function of renal tubular and endothelial cells, alters renal hemodynamics, and compromises overall kidney function. Moreover, long-term NIC exposure increases the risk of development and progression of chronic kidney diseases and may escalate the impact of coexisting morbidities such as obesity-associated renal disease, hypertension, renal transplant status, or the toxicity of various anticancer agents. In this review, we summarize experimental findings describing increased renal risk of chronic NIC exposure and explore therapeutic interventions to alleviate adverse effects of NIC.
.


Asunto(s)
Enfermedades Renales/inducido químicamente , Nicotina/efectos adversos , Agonistas Nicotínicos/efectos adversos , Animales , Sistemas Electrónicos de Liberación de Nicotina , Humanos , Enfermedades Renales/epidemiología , Enfermedades Renales/prevención & control , Riesgo , Fumar/efectos adversos
2.
Int J Clin Pharmacol Ther ; 56(10): 467-475, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29974857

RESUMEN

Baclofen is a centrally-acting γ-amino butyric acid agonist used mainly in the symptomatic management of spasticity originating from the spinal cord. It is absorbed completely from the gastrointestinal tract, metabolized by the liver to a minor degree, and excreted unchanged by the kidneys. Baclofen is moderately lipophilic and can cross the blood-brain barrier easily. At the usual dosage, it acts mainly at the spinal level without central nervous system (CNS) side effects. During renal failure, however, the elimination of the drug will decrease with a prolonged half-life, resulting in a larger area-under-the-curve exposure and disproportionate CNS toxicity. Clinically, these patients with renal failure may present with a variety of toxic symptoms manifesting at therapeutic/sub-therapeutic doses of baclofen. In cases of unexplained mental status changes in a patient receiving baclofen therapy, a careful assessment of renal function and a high suspicion of baclofen-induced encephalopathy will be key to the diagnosis.
.


Asunto(s)
Baclofeno/efectos adversos , Enfermedades del Sistema Nervioso Central/inducido químicamente , Relajantes Musculares Centrales/efectos adversos , Insuficiencia Renal/complicaciones , Baclofeno/administración & dosificación , Humanos , Relajantes Musculares Centrales/administración & dosificación
3.
Apoptosis ; 22(2): 220-228, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27770269

RESUMEN

Chronic nicotine exposure (via smoking, E-cigarettes) increases oxidative stress in the kidney that sensitizes it to additional injury in experimental models and in the renal patient. The pro-apoptotic p66shc protein-via serine36 phosphorylation that facilitates its mitochondrial translocation and therein cytochrome c binding-generates oxidative stress that leads to injury of renal proximal tubule cells during chronic nicotine exposure. Coenzyme Q10-a clinically safe antioxidant-has been used against nicotine/smoke extract-associated oxidative stress in various non-renal cells. This study explored the anti-oxidant/anti-apoptotic effect of Coenzyme Q10 on nicotine-induced oxidative stress and its impact on p66shc in cultured rat renal proximal tubule cells (NRK52E). We studied the anti-oxidant effect of 10 µM Coenzyme Q10 using various mutants of the p66shc gene and also determined the induction of selected anti-oxidant entities (antioxidant response element, promoter of the manganese superoxide dismutase gene) in reporter luciferase assay during oxidative stress induced by 200 µM nicotine. Our studies revealed that Coenzyme Q10 strongly inhibits nicotine-mediated production of reactive oxygen species and consequent apoptosis that requires serine36 phosphorylation but not mitochondrial translocation/cytochrome c binding of p66shc. While both nicotine and Coenzyme Q10 stimulates the p66shc promoter, only nicotine exposure results in mitochondrial translocation of p66shc. In contrast, the Coenzyme Q10-stimulated and non-mitochondrial p66shc activates the anti-oxidant manganese superoxide dismutase promoter via the antioxidant response elements and hence, rescues cells from nicotine-induced oxidative stress and consequent apoptosis.


Asunto(s)
Apoptosis/genética , Túbulos Renales Proximales/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Ubiquinona/análogos & derivados , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Citocromos c/metabolismo , Sistemas Electrónicos de Liberación de Nicotina , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nicotina/toxicidad , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Ratas , Especies Reactivas de Oxígeno , Fumar/efectos adversos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Ubiquinona/genética
4.
Pediatr Res ; 82(5): 822-828, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28665927

RESUMEN

BackgroundMany adolescents are exposed to nicotine via smoking, e-cigarette use, or second-hand smoke. Nicotine-induced renal oxidative stress and its long-term consequences may be higher in adolescents than in adults because of intrinsic factors in the adolescent kidney.MethodsAdolescent and adult male C57Bl/6J mice were subjected to 2 or 200 µg/ml nicotine, which closely emulates passive or active smoking, respectively, for 4 weeks. Extent of nicotine exposure (cotinine content), oxidative stress (HNE), renal function (creatinine), tubular injury (KIM-1), and pretreatment renal levels of select pro-oxidant (p66shc) and antioxidant (Nrf2/MnSOD) genes were determined. Impact of p66shc overexpression or Nrf2/MnSOD knockdown on low-/high-dose nicotine-induced oxidative stress was determined in cultured renal proximal tubule cells.ResultsDespite similar plasma/renal cotinine levels, renal HNE and KIM-1 contents were higher in adolescents compared with those in adults, whereas renal function was unaltered after passive or active smoking-equivalent nicotine exposure. Pretreatment levels of p66shc were higher, whereas Nrf2/MnSOD levels were lower in the adolescent kidney. In agreement with this, overexpression of p66shc or knockdown of Nrf2/MnSOD augmented nicotine-induced ROS production in renal proximal tubule cells.ConclusionChronic nicotine exposure incites higher oxidative stress in the adolescent than in adult kidney because of a pre-existent pro-oxidant milieu.


Asunto(s)
Enfermedades Renales/etiología , Túbulos Renales Proximales/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/farmacología , Estrés Oxidativo/efectos de los fármacos , Fumar/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Factores de Edad , Aldehídos/metabolismo , Animales , Células Cultivadas , Cotinina/metabolismo , Cotinina/toxicidad , Creatinina/metabolismo , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Nicotina/metabolismo , Agonistas Nicotínicos/metabolismo , Factores de Riesgo , Fumar/metabolismo , Fumar/patología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Superóxido Dismutasa/metabolismo , Factores de Tiempo
5.
Pediatr Res ; 79(2): 243-50, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26492285

RESUMEN

BACKGROUND: Statins elicit antioxidant effects independently of their lipid-lowering properties. Heme oxygenase-1 (HO-1) induction may be a part of these pleiotropic effects, which are insufficiently described in the kidney. We hypothesize that simvastatin (SIM) transcriptionally activates HO-1 that protects renal proximal tubule cells from lipotoxic injury. METHODS: Impact of SIM on 100 µmol/l oleic acid (OA)-mediated reactive oxygen species (ROS) production and consequent oxidative stress (4-hydroxynonenal (HNE) content) as well as cell injury/apoptosis (lactate dehydrogenase (LDH) release, caspase-3 activation) were determined in cultured renal proximal tubule (NRK52E) cells. Effect of SIM on the HO-1 promoter and its enhancer elements (antioxidant response element (ARE), CCAAT, AP1, and cAMP response element (CRE)) was also determined in reporter luciferase assays. Dominant-negative (dnMEK, M1CREB) and pharmacologic (H89) approaches were used to inhibit activation of extracellular signal regulated kinase (ERK), CREB, and protein kinase A (PKA), respectively. RESULTS: SIM dose-dependently activated the HO-1 promoter that was essential for protection against OA-dependent ROS production/oxidative stress and LDH release/caspase-3 activation. We found that the HO-1 promoter was induced through ERK and PKA-dependent activation of the CRE by SIM. CONCLUSION: SIM may protect the kidney from adverse effects of circulating fatty acids by upregulating the antioxidant HO-1, aside from its well-described lipid-lowering effects.


Asunto(s)
Antioxidantes/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hemo Oxigenasa (Desciclizante)/biosíntesis , Túbulos Renales Proximales/efectos de los fármacos , Ácido Oléico/toxicidad , Estrés Oxidativo/efectos de los fármacos , Simvastatina/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoprotección , Relación Dosis-Respuesta a Droga , Inducción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Regiones Promotoras Genéticas , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transfección
6.
Nicotine Tob Res ; 18(7): 1628-34, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26896163

RESUMEN

INTRODUCTION: Life expectancy of an obese smoker is 13 years less than a normal weight smoker, which could be linked to the increased renal risk imposed by smoking. Both smoking-through nicotine (NIC)-and obesity-by free fatty acid overload-provoke oxidative stress in the kidney, which ultimately results in development of chronic kidney injury. Their combined renal risk, however, is virtually unknown. We tested the hypothesis that chronic NIC exposure worsens renal oxidative stress in mice on high-fat diet (HFD) by altering the balance between expression of pro-oxidant and antioxidant genes. METHODS: Nine-week-old male C57Bl/6J mice consumed normal diet (ND) or HFD and received either NIC (200 µg/ml) or vehicle (2% saccharine) in their drinking water. Body weight, plasma clinical parameters, renal lipid deposition, markers of renal oxidative stress and injury, as well as renal expression of the pro-oxidant p66shc and the antioxidant MnSOD were determined after 12 weeks. RESULTS: NIC significantly augmented levels of circulating free fatty acid, as well as lipid deposition, oxidative stress and sublethal injury in the kidneys of mice on HFD. In addition, NIC exposure suppressed HFD-mediated induction of MnSOD while increased expression of p66shc in the kidney. CONCLUSIONS: Tobacco smoking or the increasingly popular E-cigarettes-via NIC exposure-could worsen obesity-associated lipotoxicity in the kidney. Hence, our findings could help to develop strategies that mitigate adverse effects of NIC on the obese kidney. IMPLICATIONS: Life expectancy of an obese smoker is 13 years less than a normal weight smoker, which could be linked to the increased renal risk imposed by smoking. NIC-the main component of tobacco smoke, E-cigarettes and replacement therapies-links smoking to renal injury via oxidative stress, which could superimpose renal oxidative stress caused by obesity. Our results substantiate this scenario using a mouse model of diet induced obesity and NIC exposure and imply the augmented long-term renal risk in obese smokers. Also, our study may help to develop strategies that mitigate adverse effects of NIC on the obese kidney.


Asunto(s)
Dieta Alta en Grasa , Riñón/metabolismo , Nicotina/farmacología , Obesidad , Estrés Oxidativo/efectos de los fármacos , Fumar/efectos adversos , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
7.
Cytokine ; 66(1): 7-16, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24548419

RESUMEN

STAT3 has been implicated in mitochondrial function; however, the physiological relevance of this action is not established. Here we studied the importance of STAT3 to the cellular response to stimuli, TNFα and serum deprivation, which increase mitochondrial reactive oxygen species (ROS) formation. Experiments were performed using wild type (WT) and STAT3 knockout (KO) mouse embryonic fibroblasts (MEF). Both WT and STAT3 KO MEF expressed similar levels of tumor necrosis factor receptor 1 (TNFR1) and exhibited comparable IκBα degradation with TNFα. However, in the absence of STAT3 nuclear accumulation of NFκB p65 with TNFα was attenuated and induction of the survival protein c-FLIPL was eliminated. Nonetheless, WT MEF were more sensitive to TNFα-induced death which was attributed to necrosis. Deletion of STAT3 decreased ROS formation induced by TNFα and serum deprivation. STAT3 deletion was associated with lower levels of complex I and rates of respiration. Relative to WT cells, mitochondria of STAT3 KO cells released significantly more cytochrome c in response to oxidative stress and had greater caspase 3 cleavage due to serum deprivation. Our findings are consistent with STAT3 being important for mitochondrial function and cell viability by ensuring mitochondrial integrity and the expression of pro-survival genes.


Asunto(s)
Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Mitocondrias/metabolismo , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/metabolismo , Animales , Caspasa 3/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Citoprotección/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Humanos , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Modelos Biológicos , FN-kappa B/metabolismo , Unión Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
8.
Am J Nephrol ; 38(3): 226-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23988748

RESUMEN

BACKGROUND/AIMS: Adult and childhood obesity is an independent risk factor in development of chronic kidney disease (CKD) and its progression to end-stage kidney disease. Pathologic consequences of obesity include non-esterified fatty acid-induced oxidative stress and consequent injury. Since the serine36-phosphorylated p66shc is a newly recognized mediator of oxidative stress and kidney injury, we studied its role in oleic acid (OA)-induced production of reactive oxygen species (ROS), mitochondrial depolarization and injury in cultured renal proximal tubule cells. METHODS: Renal proximal tubule cells were used and treated with OA: ROS production, mitochondrial depolarization as well as injury were determined. Transcriptional effects of OA on the p66shc gene were determined in a reporter luciferase assay. The role of p66shc in adverse effects of OA was determined using knockdown, p66shc serine36 phosphorylation and cytochrome c binding-deficient cells. RESULTS: We found that OA increased ROS production via the mitochondria - and to a less extent via the NADPH oxidase - resulting in ROS-dependent mitochondrial depolarization and consequent injury. Interestingly, OA also stimulated the promoter of p66shc. Hence, knockdown of p66shc, impairment its Ser36 phosphorylation (mutation of Ser36 residue to alanine) or cytochrome c binding (W134F mutation) significantly attenuated OA-dependent lipotoxicity. CONCLUSION: These results offer a novel mechanism by which obesity may lead to renal tubular injury and consequently development of CKD. Manipulation of this pathway may offer therapeutic means to ameliorate obesity-dependent renal lipotoxicity.


Asunto(s)
Riñón/efectos de los fármacos , Riñón/metabolismo , Ácido Oléico/toxicidad , Proteínas Adaptadoras de la Señalización Shc/fisiología , Animales , Línea Celular , Citocromos c/metabolismo , Relación Dosis-Respuesta a Droga , Túbulos Renales/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Ratones , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Obesidad/metabolismo , Fosforilación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno , Factores de Riesgo , Serina/química , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
9.
Nephrol Dial Transplant ; 28(6): 1417-25, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23328708

RESUMEN

BACKGROUND: Chronic nicotine (Ch-NIC) exposure exacerbates ischemia/reperfusion (I/R)-induced oxidative stress and acute kidney injury (AKI), and mitochondrial production of reactive oxygen species (ROS) in cultured renal proximal tubule cells (RPTCs). Because Ser36-phosphorylated p66shc modulates mitochondrial ROS production and injury of RPTCs, we hypothesized that Ch-NIC exacerbates AKI by increasing stress-induced phosphorylation of p66shc. METHODS: We first tested whether Ch-NIC augments I/R-AKI-induced expression and phosphorylation of p66shc in vivo. We then examined whether knocking down p66shc, or impairing its Ser36 phosphorylation or binding to cytochrome c, alters the effects of Ch-NIC on oxidative stress (H2O2)-induced production of ROS, mitochondrial depolarization and injury in RPTCs in vitro. RESULTS: We found that Ch-NIC increased the expression of p66shc in the control and ischemic kidneys, but only increased its Ser36 phosphorylation after renal I/R. Knocking down p66shc or impairing phosphorylation of its Ser36 residue, via the S36A mutation (but not the phosphomimetic S36D mutation), blunted Ch-NIC + H2O2-dependent ROS production, mitochondrial depolarization and injury in RPTCs. Additionally, Ch-NIC + H2O2-dependent binding of p66shc to mitochondrial cytochrome c was attenuated by S36A mutation of p66shc, and impairing cytochrome c binding (via W134F mutation) abolished ROS production, mitochondrial depolarization and injury, while ectopic overexpression of p66shc (which mimics Ch-NIC treatment) augmented oxidant injury. We determined that Ch-NIC stimulates the p66shc promoter through p53- and epigenetic modification (promoter hypomethylation). CONCLUSIONS: Ch-NIC worsens oxidative stress-dependent acute renal injury by increasing expression and consequent oxidative stress-dependent Ser36 phosphorylation of p66shc. Thus, targeting this pathway may have therapeutic relevance in preventing/ameliorating tobacco-related kidney injury.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Túbulos Renales Proximales/efectos de los fármacos , Nicotina/toxicidad , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/inducido químicamente , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Western Blotting , Células Cultivadas , Citocromos c/metabolismo , Peróxido de Hidrógeno/metabolismo , Inmunoprecipitación , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Agonistas Nicotínicos/toxicidad , Fosforilación Oxidativa , Fosforilación , Regiones Promotoras Genéticas/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Serina/metabolismo , Proteínas Adaptadoras de la Señalización Shc/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Activación Transcripcional
10.
Am J Physiol Renal Physiol ; 302(6): F722-9, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22169004

RESUMEN

Previous data from our group have demonstrated (Arany I, Grifoni S, Clark JS, Csongradi, Maric C, Juncos LA. Am J Physiol Renal Physiol 301: F125-F133, 2011) that chronic nicotine (NIC) exposure exacerbates acute renal ischemic injury (AKI) in mice that could increase the risk for development and progression of chronic kidney disease (CKD). It has been shown that proximal tubules of the kidney can acquire characteristics that may compromise structural recovery and favor development of inflammation and fibrosis following injury. Chronic NIC exposure can amplify this epithelial process although the mechanism is not identified. Recently, the unphosphorylated form of signal transducer and activator of transcription-3 (U-STAT3) has emerged as a noncanonical mediator of inflammation and fibrosis that may be responsible for the effects of chronic NIC. We found that levels of transforming growth factor ß-1 (TGF-ß1), α-smooth muscle actin (α-SMA), fibronectin, monocyte chemotactic protein-1 (MCP-1), and expression of U-STAT3 were increased in the ischemic kidneys of NIC-exposed mice. Chronic NIC exposure also increased TGF-ß1-dependent F-actin reorganization, vimentin, fibronectin, and α-SMA expression as well as promoter activity of α-SMA and MCP-1 without significant loss of epithelial characteristics (E-cadherin) in cultured renal proximal tubule cells. Importantly, transduction of cells with a U-STAT3 mimetic (Y705F-STAT3) augmented stress fiber formation and also amplified NIC+TGF-ß1-induced expression of α-SMA, vimentin, fibronectin, as well as promoter activity of α-SMA and MCP-1. Our results reveal a novel, chronic NIC-exposure-related and U-STAT3-dependent mechanism as mediator of a sustained transcription of genes that are linked to remodeling and inflammation in the kidney during injury. This process may facilitate progression of AKI to CKD. The obtained data may lead to devising therapeutic methods to specifically enhance the protective and/or inhibit adverse effects of STAT3 in the kidney.


Asunto(s)
Enfermedades Renales/inducido químicamente , Nicotina/toxicidad , Factor de Transcripción STAT3/metabolismo , Actinas , Animales , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nicotina/administración & dosificación , Fosforilación , Factor de Transcripción STAT3/genética , Factor de Crecimiento Transformador beta1/metabolismo
11.
Am J Physiol Regul Integr Comp Physiol ; 302(5): R620-6, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22237591

RESUMEN

Placental hypoxia/ischemia has been implicated as a central factor in the development of preeclampsia. One particularly useful animal model to study the impact of placental ischemia is the reduced uterine perfusion pressure (RUPP) model. We have previously demonstrated that RUPP animals exhibit elevated placental oxidative stress, which plays an important role in the development of the associated maternal hypertension. Recently, we have demonstrated that cobalt protoporphyrin (CoPP)-mediated induction of heme oxygenase-1 (HO-1) attenuates RUPP-induced oxidative stress and consequent hypertension. However, signaling pathways that are involved in this process are virtually unknown. Here, we show that placentas from RUPP animals exhibit increased phosphorylation of JNK, STAT1, STAT3, and p52shc with a concomitant increase in caspase-3 activation and depletion of intracellular ATP. Treatment with CoPP decreased RUPP-induced phosphorylation of JNK and STAT1, while it increased phosphorylation of ERK and STAT3, leading to decreased caspase-3 activation and restoration of intracellular ATP content. Our data imply that RUPP induces oxidative stress and the consequent injurious state by increasing phosphorylation of mediators of injury (STAT1, JNK) and, to a lesser extent, survival (STAT3, p52shc) in placentas of pregnant rats. HO-1 induction shifts this balance to a prosurvival phenotype by augmenting phosphorylation of the prosurvival ERK and STAT3, while suppressing phosphorylation of JNK and STAT1. This attenuates the resulting injury, as indicated by caspase-3 activation and ATP depletion. These results demonstrate a novel therapeutic activity of HO-1 induction in placental cell survival during ischemia and support the HO-1 pathway as a promising therapeutic target for the management of preeclampsia.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemo-Oxigenasa 1/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Placenta/metabolismo , Preñez/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Útero/irrigación sanguínea , Adenosina Trifosfato/metabolismo , Animales , Caspasa 3/metabolismo , Supervivencia Celular/fisiología , Femenino , Isquemia/metabolismo , Modelos Animales , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Placenta/irrigación sanguínea , Placenta/citología , Preeclampsia/metabolismo , Embarazo , Protoporfirinas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
12.
J Biol Chem ; 285(52): 40901-10, 2010 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20947506

RESUMEN

Membranous nephropathy is a disease that affects the filtering units of the kidney, the glomeruli, and results in proteinuria accompanied by loss of kidney function. Passive Heymann nephritis is an experimental model that mimics membranous nephropathy in humans, wherein the glomerular epithelial cell (GEC) injury induced by complement C5b-9 leads to proteinuria. We examined the role of cytochrome P450 2B1 (CYP2B1) in this complement-mediated sublytic injury. Overexpression of CYP2B1 in GECs significantly increased the formation of reactive oxygen species, cytotoxicity, and collapse of the actin cytoskeleton following treatment with anti-tubular brush-border antiserum (anti-Fx1A). In contrast, silencing of CYP2B1 markedly attenuated anti-Fx1A-induced reactive oxygen species generation and cytotoxicity with preservation of the actin cytoskeleton. Gelsolin, which maintains an organized actin cytoskeleton, was significantly decreased by complement C5b-9-mediated injury but was preserved in CYP2B1-silenced cells. In rats injected with anti-Fx1A, the cytochrome P450 inhibitor cimetidine blocked an increase in catalytic iron and ROS generation, reduced the formation of malondialdehyde adducts, maintained a normal distribution of nephrin in the glomeruli, and provided significant protection at the onset of proteinuria. Thus, GEC CYP2B1 contributes to complement C5b-9-mediated injury and plays an important role in the pathogenesis of passive Heymann nephritis.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Citocromo P-450 CYP2B1/metabolismo , Glomerulonefritis Membranosa/enzimología , Glomérulos Renales/enzimología , Túbulos Renales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Anticuerpos/farmacología , Cimetidina/farmacología , Complejo de Ataque a Membrana del Sistema Complemento/genética , Citocromo P-450 CYP2B1/antagonistas & inhibidores , Citocromo P-450 CYP2B1/genética , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Silenciador del Gen , Glomerulonefritis Membranosa/genética , Glomerulonefritis Membranosa/patología , Complejo Antigénico de Nefritis de Heymann/metabolismo , Glomérulos Renales/patología , Túbulos Renales/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microvellosidades/metabolismo , Microvellosidades/patología , Ratas
13.
Am J Physiol Renal Physiol ; 301(1): F125-33, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21511693

RESUMEN

Recent epidemiological reports showed that smoking has a negative impact on renal function and elevates the renal risk not only in the renal patient but perhaps also in the healthy population. Studies suggested that nicotine, a major tobacco alkaloid, links smoking to renal dysfunction. While several studies showed that smoking/chronic nicotine exposure exacerbates the progression of chronic renal diseases, its impact on acute kidney injury is virtually unknown. Here, we studied the effects of chronic nicotine exposure on acute renal ischemic injury. We found that chronic nicotine exposure increased the extent of renal injury induced by warm ischemia-reperfusion as evidenced by morphological changes, increase in plasma creatinine level, and kidney injury molecule-1 expression. We also found that chronic nicotine exposure elevated markers of oxidative stress such as nitrotyrosine as well as malondialdehyde. Interestingly, chronic nicotine exposure alone increased oxidative stress and injury in the kidney without morphological alterations. Chronic nicotine treatment not only increased reactive oxygen species (ROS) production and injury but also exacerbated oxidative stress-induced ROS generation through NADPH oxidase and mitochondria in cultured renal proximal tubule cells. The resultant oxidative stress provoked injury through JNK-mediated activation of the activator protein (AP)-1 transcription factor in vitro. This mechanism might exist in vivo as phosphorylation of JNK and its downstream target c-jun, a component of the AP-1 transcription factor, is elevated in the ischemic kidneys exposed to chronic nicotine. Our results imply that smoking may sensitize the kidney to ischemic insults and perhaps facilitates progression of acute kidney injury to chronic kidney injury.


Asunto(s)
Lesión Renal Aguda/patología , Isquemia/patología , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Adenoviridae/genética , Animales , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cotinina/sangre , Cotinina/metabolismo , Riñón/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Luciferasas/metabolismo , MAP Quinasa Quinasa 4/genética , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/patología , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/fisiología
14.
Am J Physiol Renal Physiol ; 298(5): F1214-21, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20053790

RESUMEN

Mitochondrial dysfunction is involved in pathopysiology of ischemia-reperfusion-induced acute kidney injury (AKI). The p66shc adaptor protein is a newly recognized mediator of mitochondrial dysfunction, which might play a role in AKI-induced renal tubular injury. Oxidative stress-mediated Serine36 phosphorylation of p66shc facilitates its transportation to the mitochondria where it oxidizes cytochrome c and generates excessive amount of reactive oxygen species (ROS). The consequence is mitochondrial depolarization and injury. Earlier we determined that p66shc plays an essential role in injury of cultured mouse renal proximal tubule cells during oxidative stress. Here, we studied the role of p66shc in ROS generation and consequent mitochondrial dysfunction during oxidative injury in renal proximal tubule cells. We employed p66shc knockdown renal proximal tubule cells and cells that overexpress wild-type, Serine phosphorylation (S36A), or cytochrome c-binding (W134F) mutants of p66shc. Inhibition of the mitochondrial electron transport chain or the mitochondrial permeability transition revealed that hydrogen peroxide-induced injury is mitochondrial ROS and consequent mitochondrial depolarization dependent. We also found that through Ser36 phosphorylation and mitochondria/cytochrome c binding, p66shc mediates those effects. We propose a similar mechanism in vivo as we demonstrated mitochondrial binding of p66shc as well as its association with cytochrome c in the postischemic kidneys of mice. Thus, manipulating p66shc might offer a new therapeutic modality to ameliorate renal ischemic injury.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Túbulos Renales Proximales/fisiopatología , Mitocondrias/fisiología , Estrés Oxidativo/fisiología , Proteínas Adaptadoras de la Señalización Shc/fisiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Línea Celular , Citocromos c/metabolismo , Peróxido de Hidrógeno/farmacología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Serina/metabolismo
15.
Kidney Int ; 78(2): 182-90, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20407477

RESUMEN

Previously, we demonstrated that cytochrome P450 2B1 (CYP2B1) can generate reactive oxygen species in puromycin aminonucleoside (PAN)-induced nephrotic syndrome, an animal model of minimal-change disease in humans. In this study we found that overexpression of CYP2B1 in rat glomerular epithelial cells in vitro significantly increased PAN-induced reactive oxygen species generation, cytotoxicity, cell death, and collapse of the actin cytoskeleton. All of these pathological changes were markedly attenuated by siRNA-induced CYP2B1 silencing. The cellular CYP2B1 protein content was significantly decreased whereas its mRNA level was markedly increased, suggesting regulation by protein degradation rather than transcriptional inhibition in the PAN-treated glomerular epithelial cells. This degradation of CYP2B1 was accompanied by the induction of heme oxygenase-1, an important indicator of heme-induced oxidative stress. In PAN-treated CYP2B1-silenced glomerular epithelial cells the induction of heme oxygenase-1 and caspase-3 activity were significantly decreased. Further, cleavage of the stress-induced pro-apoptotic endoplasmic reticulum-specific pro-caspase-12 was prevented in the silenced cells. Our results support a pivotal role of CYP2B1 for reactive oxygen species production in the endoplasmic reticulum in PAN-induced cytotoxicity.


Asunto(s)
Citocromo P-450 CYP2B1/genética , Células Epiteliales/efectos de los fármacos , Silenciador del Gen , Glomérulos Renales/citología , Glomérulos Renales/efectos de los fármacos , Puromicina Aminonucleósido/toxicidad , Animales , Células Cultivadas , Citocromo P-450 CYP2B1/biosíntesis , Expresión Génica , Ratas
16.
Anticancer Res ; 29(6): 2295-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19528494

RESUMEN

BACKGROUND: Increased production of reactive oxygen species (ROS) by anticancer drugs has been described in patients with various malignancies, which might attribute to their nephrotoxicity. MATERIALS AND METHODS: The effects of two epigenetic modifiers - trichostatin A (TSA) and 5-aza-deoxycytidine (5AZA) - on ROS production and cell injury alone or in combination with mild oxidative stress were studied in mouse renal proximal tubule cells. RESULTS: Both agents increased mitochondrial ROS production and consequent lactate dehydrogenase (LDH) release either alone or in combination with a low dose of H(2)O(2). The antioxidant N-acetyl-cysteine (NAC) abolished LDH release. It was also found that CREB-mediated transcription, vital for survival of proximal tubule cells, is attenuated by these anticancer agents. CONCLUSION: The ROS-inducing activity of TSAI and 5AZA might explain the in vivo nephrotoxicity of epigenetic modifiers. The mechanisms that are responsible for this injury could involve attenuation of pro-survival signaling and/or activation of death signaling pathway(s) associated with mitochondrial ROS release.


Asunto(s)
Antifúngicos/farmacología , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Ácidos Hidroxámicos/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Azacitidina/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Decitabina , Túbulos Renales Proximales/metabolismo , Ratones
17.
In Vivo ; 23(6): 975-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20023242

RESUMEN

BACKGROUND: Cancer therapies and cancer progression can increase oxidative stress that might account for renal toxicity in cancer patients. Flavin 7 (F7) is a natural polyphenol-containing dietary supplement with potential antioxidant activity. Therefore, it might help to attenuate renal toxicity of chemotherapeutics. MATERIALS AND METHODS: Cultured mouse renal proximal tubule cells were subjected to H(2)O(2)-mediated oxidative stress. Potential antioxidant effects of F7 were assessed by measuring the production of reactive oxygen species (ROS), mitochondrial depolarization and injury (lactate dehydrogenase release as well as trypan blue exclusion) in cells that were pretreated with F7 prior to treatment with H(2)O(2). RESULTS: F7 pretreatment significantly attenuated H(2)O(2)-induced ROS production, mitochondrial depolarization and consequent injury in renal proximal tubule cells. CONCLUSION: F7 supplementation might be beneficial for cancer patients in order to prevent renal toxicity of anticancer drug- or cancer progression-related oxidative stress.


Asunto(s)
Antioxidantes/farmacología , Frutas/química , Túbulos Renales Proximales/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Bencimidazoles/metabolismo , Carbocianinas/metabolismo , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Colorantes Fluorescentes/metabolismo , Peróxido de Hidrógeno/toxicidad , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , L-Lactato Deshidrogenasa/metabolismo , Ratones , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo
18.
In Vivo ; 23(4): 591-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19567395

RESUMEN

7,12-Dimethylbenz[a]anthracene (DMBA) and N-methyl-N-nitrosourea (MNU) are important environmental carcinogens. Their different biological effects were examined in CBA/Ca H-2(K) haplotype inbred mice on the gene expression of c-myc, Ha-ras and p53 through a 24 hour period. Elevated expression of c-myc and Ha-ras genes was found in the spleen, lung, thymus and lymph nodes 6 and 12 hours after DMBA treatment and in the lung and thymus 3 hours after MNU treatment. In the liver, DMBA induced strong onco/suppressor gene expression as early as 6 hours after the treatment, but MNU increased the p53 gene expression 12 hours after the treatment. The gene expression patterns reflected the different mechanism of the direct acting MNU and metabolically activated DMBA. This phenomenon provides evidence as to the usefulness of detection of onco/supressor key gene expression as early molecular epidemiological biomarkers of carcinogenesis and carcinogenic exposure in animal model, useful in human cancer prevention practice as well.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/toxicidad , Carcinógenos/toxicidad , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , 9,10-Dimetil-1,2-benzantraceno/química , Alquilantes/química , Alquilantes/toxicidad , Animales , Carcinógenos/química , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino , Metilnitrosourea/química , Metilnitrosourea/toxicidad , Ratones , Ratones Endogámicos CBA
19.
Kidney Int ; 74(3): 261-2, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18626494

RESUMEN

The paper by Ma and Devarajan suggests that the subtle apoptotic events that occur in the distal nephron after acute kidney injury might have a greater than expected impact on the adjacent proximal tubules and kidney function. Understanding these events might facilitate development of therapeutic means to ameliorate acute kidney injury.


Asunto(s)
Lesión Renal Aguda/patología , Apoptosis/fisiología , Daño por Reperfusión/patología , Lesión Renal Aguda/etiología , Animales , Humanos , Riñón/fisiopatología , Túbulos Renales Proximales/fisiopatología , Nefronas/fisiopatología
20.
In Vivo ; 32(5): 1033-1037, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30150424

RESUMEN

BACKGROUND/AIM: We have previously reported that simvastatin exhibits antioxidant properties via extracellular signal-regulated kinase (ERK)/cAMP-response element binding (CREB) protein-dependent induction of heme oxygenase-1 (HO1) and chronic nicotine exposure inhibits ERK/CREB signaling in renal proximal tubule cells (through p66shc). Herein, whether nicotine dampens simvastatin-dependent HO1 induction was determined. MATERIALS AND METHODS: Renal proximal tubule (NRK52E) cells were pre-treated with 200 µM nicotine for 24 h followed by 10 µM simvastatin. Promoter activity of HO1 and manganese superoxide dismutase (MnSOD) and activation of CREB and ERK (via ELK1) were determined in luciferase reporter assays. CREB and p66shc were modulated via genetic means. RESULTS: Nicotine suppressed simvastatin-dependent activation of HO1 and MnSOD promoters and activity of CREB and ELK1 via p66shc. Overexpression of CREB or knockdown of p66shc restored simvastatin-dependent induction of HO1 and MnSOD in the presence of nicotine. CONCLUSION: Antioxidant efficiency of simvastatin might be significantly lessened in smokers/E-cigarette users.


Asunto(s)
Antioxidantes/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Nicotina/efectos adversos , Simvastatina/farmacología , Animales , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Nicotina/administración & dosificación , Regiones Promotoras Genéticas , Ratas , Fumar/efectos adversos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA