Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 5(1): e8955, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-20126461

RESUMEN

TWEAK is a member of the TNF superfamily of cytokines that contribute to kidney tubulointerstitial injury. It has previously been reported that TWEAK induces transient nuclear translocation of RelA and expression of RelA-dependent cytokines in renal tubular cells. Additionally, TWEAK induced long-lasting NFkappaB activation suggestive of engagement of the non-canonical NFkappaB pathway. We now explore TWEAK-induced activation of NFkappaB2 and RelB, as well as expression of CCL21, a T-cell chemotactic factor, in cultured murine tubular epithelial cells and in healthy kidneys in vivo. In cultured tubular cells, TWEAK and TNFalpha activated different DNA-binding NFkappaB complexes. TWEAK-induced sustained NFkappaB activation was associated with NFkappaB2 p100 processing to p52 via proteasome and nuclear translocation and DNA-binding of p52 and RelB. TWEAK, but not TNFalpha used as control), induced a delayed increase in CCL21a mRNA (3.5+/-1.22-fold over control) and CCL21 protein (2.5+/-0.8-fold over control), which was prevented by inhibition of the proteasome, or siRNA targeting of NIK or RelB, but not by RelA inhibition with parthenolide. A second NFkappaB2-dependent chemokine, CCL19, was upregulates by TWEAK, but not by TNFalpha. However, both cytokines promoted chemokine RANTES expression (3-fold mRNA at 24 h). In vivo, TWEAK induced nuclear NFkappaB2 and RelB translocation and CCL21a mRNA (1.5+/-0.3-fold over control) and CCL21 protein (1.6+/-0.5-fold over control) expression in normal kidney. Increased tubular nuclear RelB and tubular CCL21 expression in acute kidney injury were decreased by neutralization (2+/-0.9 vs 1.3+/-0.6-fold over healthy control) or deficiency of TWEAK (2+/-0.9 vs 0.8+/-0.6-fold over healthy control). Moreover, anti-TWEAK treatment prevented the recruitment of T cells to the kidney in this model (4.1+/-1.4 vs 1.8+/-1-fold over healthy control). Our results thus identify TWEAK as a regulator of non-canonical NFkappaB activation and CCL21 expression in tubular cells thus promoting lymphocyte recruitment to the kidney during acute injury.


Asunto(s)
Quimiocina CCL21/fisiología , Túbulos Renales/fisiología , FN-kappa B/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Células Cultivadas , Citocina TWEAK , Ensayo de Inmunoadsorción Enzimática , Ratones , Microscopía Confocal
2.
J Cell Mol Med ; 13(9B): 3329-42, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19426154

RESUMEN

The tumour necrosis factor (TNF) family member TWEAK activates the Fn14 receptor and has pro-apoptotic, proliferative and pro-inflammatory actions that depend on the cell type and the microenvironment. We explored the proliferative actions of TWEAK on cultured tubular cells and in vivo on renal tubules. Additionally, we studied the role of TWEAK in compensatory proliferation following unilateral nephrectomy and in an inflammatory model of acute kidney injury (AKI) induced by a folic acid overdose. TWEAK increased the proliferation, cell number and cyclin D1 expression of cultured tubular cells, in vitro. Exposure to serum increased TWEAK and Fn14 expression and the proliferative response to TWEAK. TWEAK activated the mitogen-activated protein kinases ERK and p38, the phosphatidyl-inositol 3-kinase (PI3K)/Akt pathway and NF-kappaB. TWEAK-induced proliferation was prevented by inhibitors of these protein kinases and by the NF-kappaB inhibitor parthenolide. TWEAK-induced tubular cell proliferation as assessed by PCNA and cyclin D1 expression in the kidneys of adult healthy mice in vivo. By contrast, TWEAK knock-out mice displayed lower tubular cell proliferation in the remnant kidney following unilateral nephrectomy, a non-inflammatory model. This is consistent with TWEAK-induced proliferation on cultured tubular cells in the absence of inflammatory cytokines. Consistent with our previously published data, in the presence of inflammatory cytokines TWEAK promoted apoptosis, not proliferation, of cultured tubular cells. In this regard, TWEAK knock-out mice with AKI displayed less tubular apoptosis and proliferation, as well as improved renal function. In conclusion, TWEAK actions in tubular cells are context dependent. In a non-inflammatory milieu TWEAK induces proliferation of tubular epithelium. This may be relevant for compensatory renal hyperplasia following nephrectomy.


Asunto(s)
Epitelio/metabolismo , Hiperplasia/metabolismo , Riñón/metabolismo , Riñón/patología , Factores de Necrosis Tumoral/metabolismo , Animales , Apoptosis , Proliferación Celular , Ciclina D1/biosíntesis , Citocina TWEAK , Ácido Fólico/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrectomía/métodos , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor de TWEAK
3.
J Am Soc Nephrol ; 19(4): 695-703, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18235096

RESUMEN

TNF-like weak inducer of apoptosis (TWEAK) is a member of the TNF superfamily of cytokines. In addition to binding and activating the fibroblast growth factor-inducible 14 receptor, TWEAK may regulate apoptosis, proliferation, and inflammation; however, the role of this system in kidney injury is unknown. In vitro, it was found that TWEAK induced the sustained activation of NF-kappaB in a murine tubular epithelial cell line (MCT). NF-kappaB activation was associated with degradation of IkappaB-alpha; translocation of RelA to the nucleus; and increased mRNA and protein expression of monocyte chemoattractant protein-1, RANTES, and IL-6. Similarly, in vivo, the systemic administration of TWEAK induced renal NF-kappaB activation, chemokine and IL-6 expression, and interstitial inflammation in mice. Parthenolide, which prevents IkappaB-alpha degradation, inhibited TWEAK-induced NF-kappaB activation and prevented the aforementioned changes in vitro and in vivo. After folic acid-induced acute kidney injury, fibroblast growth factor-inducible 14 expression increased in mouse tubular epithelium. Neutralization of TWEAK decreased the expression of chemokines in tubular cells and reduced interstitial inflammation. In conclusion, TWEAK has NF-kappaB-dependent proinflammatory effects on tubular epithelial cells in vitro and in vivo. Moreover, blockade of TWEAK reduces tubular chemokine expression and macrophage infiltration, suggesting that TWEAK modulates acute kidney injury by regulating the inflammatory response.


Asunto(s)
Nefritis Intersticial/inmunología , Factores de Necrosis Tumoral/fisiología , Animales , Células Cultivadas , Citocina TWEAK , Humanos , Ratones , Factores de Necrosis Tumoral/farmacología
4.
Front Biosci ; 13: 580-9, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17981571

RESUMEN

TWEAK is a recently identified cytokine of the TNF superfamiliy. Through activation of the Fn14 receptor, TWEAK regulates cell proliferation, cell death and inflammation. Recent studies show increased TWEAK and Fn14 expression in tubular cells during acute kidney injury as well as elevated urinary TWEAK levels in patients with active lupus nephritis. Furthermore, glomerular mesangial cells and renal tubular epithelial cells express the Fn14 receptor under the regulation of proinflammatory cytokines. TWEAK weakly increases cell death and promotes secretion of inflammatory mediators in non-stimulated mesangial cells. In addition, in a proinflammatory milieu, TWEAK induces apoptosis of mesangial and tubular cells. The available data suggest that TWEAK is a new player in kidney injury both at the glomerular and tubulointerstitial levels and might be a target for therapeutic intervention.


Asunto(s)
Riñón/lesiones , Riñón/patología , Factores de Necrosis Tumoral/fisiología , Enfermedad Aguda , Animales , Apoptosis , Muerte Celular , Proliferación Celular , Citocina TWEAK , Humanos , Inflamación , Riñón/metabolismo , Glomérulos Renales/metabolismo , Ratones , Modelos Biológicos , Estructura Terciaria de Proteína , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo
5.
Curr Med Chem ; 13(22): 2695-702, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17017920

RESUMEN

Breakdown of glucose under physiological conditions gives rise to glucose degradation products (GDPs). GDPs are also formed during heat sterilization of glucose-containing peritoneal dialysis fluids (PD-fluids). In PD-fluids GDPs have been shown in many different in vitro assays to be responsible for adverse effects such as growth inhibition, and impaired leukocyte function and impaired wound healing of peritoneal mesothelial cells. They have been linked to changes in the peritoneal membrane as well as to the decline in residual renal function of PD-patients. In diabetes one of the GDPs, 3-deoxyglucosone (3-DG), has been proposed as responsible for side-effects rather the glucose itself. 3,4-dideoxyglucosone-3-ene (3,4-DGE) was recently identified as the most bio-reactive GDP in PD-fluids. It exists in equilibrium with a pool of precursors, consisting of 3-DG but also of other hitherto unidentified GDPs. In PD-fluids the concentration of GDPs in this pool is 10-20 times as high as that of 3,4-DGE. In vitro 3,4-DGE induces caspase-dependent apoptosis of neutrophils and peripheral blood mononuclear cells. Such induction may explain immunosuppressive properties of 3,4-DGE and contribute to an impaired peritoneal antibacterial defense. 3,4-DGE also induces renal cell apoptosis. This may explain the better preservation of residual renal function in PD patients not exposed to GDPs. The concentration of 3-DG increases with worsening glycemic control and has been implicated in the genesis of diabetic microangiopathy. As 3,4-DGE is much more bio-reactive than 3-DG and as it may be easily recruited from the pool, it seems probable that 3,4-DGE is the molecule involved in the diabetic lesions rather than 3-DG itself. Thus, 3,4-DGE might contribute to diabetic nephropathy and to the impaired antibacterial defenses in diabetics. Unraveling of the pool dynamics of the GDPs and the molecular mechanisms of GDP-mediated cell injury may provide new therapeutic insights in PD and diabetes.


Asunto(s)
Desoxiglucosa/análogos & derivados , Diabetes Mellitus/terapia , Diálisis Peritoneal , Apoptosis/efectos de los fármacos , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/prevención & control , Desoxiglucosa/efectos adversos , Desoxiglucosa/química , Desoxiglucosa/farmacología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/microbiología , Diabetes Mellitus/patología , Glucosa/química , Glucosa/metabolismo , Humanos
6.
Nephrol Dial Transplant ; 21 Suppl 2: ii12-5, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16825252

RESUMEN

Peritoneal dialysis (PD) and diabetes mellitus share the high glucose concentration in the cell microenvironment. This has led to the suggestion that they may also share pathogenic pathways of cell and tissue injury. Hypotheses have been formulated on the pathogenesis of peritoneal injury in the course of PD that take into account knowledge of the mechanisms of tissue injury in diabetes patients. More recently, research on the pathways of PD complications has uncovered potentially novel mediators of diabetes complications. Accelerated leucocyte apoptosis has been identified as a cause of impaired peritoneal antibacterial defence in PD, which may lead to new therapeutic interventions. In this regard, interference with leucocyte apoptosis by the use of caspase inhibitors may accelerate the clearance of bacteria such as Staphylococcus aureus, which cause significant morbidity in both PD and diabetes patients. Evidence suggests that glucose degradation products in PD solutions accelerate leucocyte apoptosis. In particular, 3,4-di-deoxyglucosone-3-ene (3,4-DGE) accounted for most, if not all, the cytotoxicity of PD fluids against neutrophils and lymphocytes. Interestingly, 3,4-DGE also induces apoptosis in cells, such as renal epithelium, from organs that are targets of diabetes complications. This raises the possibility that apoptosis induction by glucose metabolites that are the key participants in PD complications may underlie the pathogenesis of some features of diabetic tissue injury.


Asunto(s)
Complicaciones de la Diabetes , Soluciones para Diálisis/toxicidad , Diálisis Peritoneal , Peritoneo/patología , Animales , Apoptosis , Glucosa/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Productos Finales de Glicación Avanzada/toxicidad , Humanos , Peritonitis/etiología
7.
Antioxid Redox Signal ; 7(9-10): 1285-93, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16115034

RESUMEN

Arterial hypertension is a leading contributor to the progression of chronic renal disease. Short-term studies had addressed the role of oxidative stress in hypertensive nephropathy. We have now studied oxidative stress and caspase activation in a long-term model of hypertensive renal injury. Nontreated spontaneously hypertensive rats with uninephrectomy displayed severe arterial hypertension over a 36-week follow-up. Uncontrolled high blood pressure in the context of modest renal mass reduction resulted in significant histological renal injury. Blood pressure control by the angiotensin-converting enzyme (ACE) inhibitor, quinapril, or the AT1 receptor antagonist, losartan, decreased the degree of renal injury. Hypertensive renal injury was associated with evidence of activation of the apoptotic pathway (increased activation of caspase-3) and local renal (increased staining for 4-hydroxy-2-nonenal) and systemic [increased serum levels of 8-iso-prostaglandin F2alpha (8-iso-PGF2alpha)] lipid oxidation when compared with normotensive control rats. In addition, severe hypertension decreased the renal antioxidant defenses, as exemplified by decreased expression of Cu/Zn superoxide dismutase. Treatment with quinapril or losartan decreased caspase-3 activation, 4-hydroxy-2-nonenal staining, and 8-iso-PGF2alpha levels and increased Cu/Zn superoxide dismutase expression. These results suggest that hypertension-associated oxidative stress and its consequences may be decreased by either ACE inhibition or AT1 receptor antagonist, emphasizing the role of angiotensin II in hypertensive renal damage.


Asunto(s)
Presión Sanguínea , Regulación de la Expresión Génica , Hipertensión/fisiopatología , Hipertensión/terapia , Riñón/lesiones , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Antioxidantes/farmacología , Apoptosis , Caspasa 3 , Caspasas/metabolismo , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Activación Enzimática , Humanos , Hipertensión Renal/patología , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Peroxidación de Lípido , Losartán/farmacología , Masculino , Estrés Oxidativo , Quinapril , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Superóxido Dismutasa/metabolismo , Tetrahidroisoquinolinas/farmacología , Factores de Tiempo
8.
Diabetes ; 54(8): 2424-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16046310

RESUMEN

Diabetes complications are caused by hyperglycemia. Hyperglycemia results in increased concentrations of glucose degradation products. The study of peritoneal dialysis solution biocompatibility has highlighted the adverse effects of glucose degradation products. Recently, 3,4-dideoxyglucosone-3-ene (3,4-DGE) has been identified as the most toxic glucose degradation product in peritoneal dialysis fluids. Its role in renal pathophysiology has not been addressed. 3,4-DGE induces apoptosis in murine renal tubular epithelial cells in a dose- and time-dependent manner. Peak apoptosis is observed after 72 h of culture. The lethal concentration range is 25-50 micromol/l. 3,4-DGE results in Bax oligomerization, release of cytochrome c from mitochondria, activation of caspases-9 and -3, and Bid proteolysis. Apoptosis induced by 3,4-DGE is caspase dependent and could be prevented by the broad-spectrum caspase inhibitor zVAD-fmk (Z-Val-Ala-DL-Asp-fluoromethylketone) and by specific inhibitors of caspases-2, -8, and -9. However, caspase inhibition did not prevent eventual cell death. In contrast, antagonism of Bax by a Ku-70-derived peptide or antisense oligonucleotides prevented both apoptosis and cell death. In conclusion, 3,4-DGE promotes apoptosis of cultured renal parenchymal cells by a Bax- and caspase-dependent mechanism. A role for 3,4-DGE in diabetes complications in the kidney and in the modulation of residual renal function in peritoneal dialysis should be further explored.


Asunto(s)
Apoptosis/efectos de los fármacos , Túbulos Renales Proximales/citología , Pironas/farmacología , Animales , Caspasa 3 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular , Citocromos c/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Células Epiteliales/citología , Ratones , Mitocondrias/metabolismo , Oligonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factores de Tiempo , Proteína X Asociada a bcl-2
9.
Antivir Ther ; 10(1): 185-90, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15751777

RESUMEN

Cidofovir is an antiviral drug with activity against a wide array of DNA viruses including poxvirus. The therapeutic use of cidofovir is marred by a dose-limiting side effect, nephrotoxicity, leading to proximal tubular cell injury and acute renal failure. Treatment with cidofovir requires the routine use of prophylactic measures. A correct knowledge of the cellular and molecular mechanisms of cidofovir toxicity may lead to the development of alternative prophylactic strategies. We recently cared for a patient with irreversible acute renal failure due to cidofovir. Renal biopsy showed tubular cell apoptosis. Cidofovir induced apoptosis in primary cultures of human proximal tubular cells in a temporal (peak apoptosis at 7 days) and concentration (10-40 microg/ml) pattern consistent with that of clinical toxicity. Apoptosis was identified by the presence of hypodiploid cells, by the exposure of annexin V binding sites and by morphological features and was associated with the appearance of active caspase-3 fragments. Cell death was specific as it was also present in a human proximal tubular epithelial cell line (HK-2), but not in a human kidney fibroblast cell line, and was prevented by probenecid. An inhibitor of caspase-3 (DEVD) prevented cidofovir apoptosis. The survival factors present in serum, insulin-like growth factor-1 and hepatocyte growth factor, were also protective. The present data suggest that apoptosis induction is a mechanism contributing to cidofovir nephrotoxicity. The prophylactic administration of factors with survival activity for tubular epithelium should be further explored in cidofovir renal injury.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antivirales/administración & dosificación , Citosina/análogos & derivados , Citosina/efectos adversos , Túbulos Renales Proximales/efectos de los fármacos , Organofosfonatos/efectos adversos , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Adulto , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Cidofovir , Retinitis por Citomegalovirus/complicaciones , Retinitis por Citomegalovirus/tratamiento farmacológico , Infecciones por VIH/complicaciones , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino
10.
Kidney Int ; 67(2): 592-601, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15673306

RESUMEN

BACKGROUND: Paracetamol overdose causes acute renal failure and chronic exposure to paracetamol has been linked to chronic renal failure. Recently, apoptosis induction has been identified as a possible mechanism of paracetamol nephrotoxicity. METHODS: Murine proximal tubular epithelial MCT cells were cultured in the presence of paracetamol. The effects of Bcl-xL overexpression, Bax antisense oligodeoxynucleotides, and different caspase inhibitors on cell death were studied. RESULTS: While paracetamol did not change the mRNA expression of the antiapoptotic gene bcl-xL, it decreased Bcl-xL protein levels. The decrease in Bcl-xL was prevented by lactacystin, but not by caspase inhibitors. Addition to the culture media of the survival factors present in fetal calf serum (FCS) increased Bcl-xL expression and decreased paracetamol-induced apoptosis. Overexpression of a human bcl-xL transgene decreased apoptosis induced by paracetamol by 60% at 24 hours and increased long-term cell survival. The constitutive expression of the viral caspase inhibitor CrmA decreased the rate of apoptosis by 60% at 24 hours and the broad-specific caspase inhibitor zVAD-fmk prevented paracetamol-induced features of apoptosis. However, caspase inhibitors did not prevent eventual cell death. Bax did not translocate to mitochondria and Bax antisense oligodeoxynucleotides were not protective. CONCLUSION: Our results suggest that induction of apoptosis may underlie the nephrotoxic potential of paracetamol and identify Bcl-xL as a player in toxic tubular cell injury.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Apoptosis/efectos de los fármacos , Túbulos Renales/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Animales , Caspasas/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Humanos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteína X Asociada a bcl-2 , Proteína bcl-X
11.
J Am Soc Nephrol ; 15(2): 380-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14747384

RESUMEN

Paracetamol (also known as acetaminophen) causes acute and chronic renal failure. While the mechanisms leading to hepatic injury have been extensively studied, the molecular mechanisms of paracetamol-induced nephrotoxicity are poorly defined. Paracetamol induced cell death with features of apoptosis in murine proximal tubular epithelial cells. While paracetamol increased the expression of the death receptor Fas on the cell surface, the Fas pathway was not involved in the paracetamol-induced apoptosis of tubular cells. The mitochondrial pathway was not activated during paracetamol-induced apoptosis; there was no dissipation of mitochondrial potential or release of apoptogenic factors such as cytochrome c or Smac/DIABLO. However, paracetamol-induced apoptosis is a caspase-dependent process that involves activation of caspase-9 and caspase-3 in the absence of cytosolic cytochrome c or Smac/DIABLO. The authors also detected induction of endoplasmic reticulum (ER) stress, characterized by GADD153 upregulation and translocation to the nucleus, as well as caspase-12 cleavage. Interestingly, after treatment of murine tubular cells with paracetamol and calpain inhibitors, the caspase-12 cleavage product was still detectable, and calpain inhibitors were unable to protect tubular cells from paracetamol-induced apoptosis. The results suggest that induction of apoptosis may underlie the nephrotoxic potential of paracetamol and identify ER stress as a therapeutic target in nephrotoxicity.


Asunto(s)
Acetaminofén/efectos adversos , Analgésicos no Narcóticos/efectos adversos , Retículo Endoplásmico/efectos de los fármacos , Túbulos Renales , Animales , Apoptosis/efectos de los fármacos , Caspasa 12 , Caspasas/farmacología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Túbulos Renales/citología , Túbulos Renales/patología , Ratones , Tunicamicina/farmacología , Receptor fas/efectos de los fármacos
12.
J Am Soc Nephrol ; 14(12): 3072-80, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14638906

RESUMEN

Tubular cell apoptosis contributes to the pathogenesis of renal injury. However, the intracellular pathways that are active in tubular epithelium are poorly understood. The lethal pathways activated by cyclosporin A (CsA), a nephrotoxin that induces caspase-dependent apoptosis in tubular epithelium, were explored. Fas expression, caspase activation, and mitochondrial injury were assessed by Western blot, flow cytometry, and microscopy in cultured murine tubular epithelial cells exposed to CsA. The influence of FasL antagonists, Bax antisense oligodeoxynucleotides, and caspase inhibitors on cell survival was explored. Tubular cells constitutively express FasL. CsA increased the expression of Fas. However, Fas had no role in CsA-induced apoptosis, as CsA did not sensitize to FasL-induced apoptosis, caspase-8 activity was not increased, and neither blocking anti-FasL antibodies nor caspase-8 inhibition prevented CsA-induced apoptosis. Apoptosis induced by CsA is associated with the translocation of Bax to the mitochondria and Bax antisense oligodeoxynucleotides protected from CsA-induced apoptosis. CsA promoted a caspase-independent release of cytochrome c and Smac/Diablo from mitochondria. CsA also led to a caspase-dependent loss of mitochondrial membrane potential. Caspase-2, caspase-3, and caspase-9 were activated, and specific caspase inhibitor prevented apoptosis and increased long-term survival. Evidence for endoplasmic reticulum stress, such as induction of GADD153, was also uncovered. However, endoplasmic reticulum-specific caspase-12 was not activated. CsA induces changes in several apoptotic pathways. However, the main lethal apoptotic pathway in CsA-exposed tubular epithelial cells involves mitochondrial injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclosporina/farmacología , Túbulos Renales/citología , Animales , Inhibidores de Caspasas , Caspasas/fisiología , Células Cultivadas , Ratones , Mitocondrias/metabolismo , Regulación hacia Arriba , Receptor fas/fisiología
13.
Biochem Pharmacol ; 66(8): 1589-94, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14555238

RESUMEN

Recent research has shown that apoptosis and its regulatory mechanisms contribute to cell number regulation in acute renal failure. Acute tubular necrosis is the most frequent form of parenchymal acute renal failure. The main causes are ischemia-reperfusion, sepsis and nephrotoxic drugs. Exogenous factors such as nephrotoxic drugs and bacterial products, and endogenous factors such as lethal cytokines promote tubular cell apoptosis. Such diverse stimuli engage intracellular death pathways that in some cases are stimulus-specific. We now review the role of apoptosis in acute renal failure, the potential molecular targets of therapeutic intervention, the therapeutic weapons to modulate the activity of these targets and the few examples of therapeutic intervention on apoptosis.


Asunto(s)
Apoptosis/fisiología , Túbulos Renales/lesiones , Enfermedad Aguda , Animales , Humanos , Túbulos Renales/patología
14.
Kidney Int Suppl ; (86): S21-6, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12969123

RESUMEN

BACKGROUND: Activation of the renin angiotensin system has been described in pathologic conditions, including kidney damage. Angiotensin II (Ang II) acts through two receptors, AT1 and AT2. Most of the known actions of Ang II, including vasoconstriction and fibrosis, are due to AT1 activation. Recent data suggest that AT2 participates in the regulation of cell growth and renal inflammatory infiltration. Therefore, we investigated the renal expression of AT2 receptors in several models of renal injury. METHODS: Investigations were done in the following experimental models of kidney damage: systemic infusion of Ang II (inflammation), folic acid nephropathy (tubular cell death), and protein overload proteinuria. AT2 expression was determined by immunohistochemistry (protein) and reverse transcription-polymerase chain reaction (RT-PCR) (gene). RESULTS: In control animals, low levels of renal expression of AT2 were found. Ang II infusion resulted in an up-regulation of AT2 in tubular cells and de novo AT2 expression in glomeruli and vessels, associated with the presence of inflammatory cells. Acute tubular injury induced by folic acid was characterized by AT2 overexpression and apoptosis in tubular cells. Protein overload caused heavy proteinuria and tubular AT2 up-regulation. CONCLUSION: AT2 is re-expressed in pathologic conditions of kidney damage, such as inflammation, apoptosis, and proteinuria, suggesting a potential role of this receptor during renal injury.


Asunto(s)
Enfermedades Renales/metabolismo , Riñón/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Angiotensina II , Animales , Ácido Fólico , Inmunohistoquímica , Enfermedades Renales/inducido químicamente , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteinuria/metabolismo , Ratas , Ratas Wistar , Regulación hacia Arriba
15.
Kidney Int Suppl ; (86): S52-6, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12969128

RESUMEN

BACKGROUND: Apoptosis contributes to tubular cell loss in the course of renal injury. However, the mechanisms regulating tubular cell apoptosis are not well understood. Smac/Diablo is a mitochondrial protein that is released to the cytosol during apoptosis, where it blocks the antiapoptotic activity of inhibitor of apoptosis proteins (IAPs). METHODS: We have studied the regulation of Smac/Diablo mRNA and protein expression in murine toxic acute tubular necrosis, and in cultured tubular epithelial cells exposed to the lethal cytokine tumor necrosis factor (TNF). RESULTS: Folic acid-induced acute renal failure was associated with tubular cell apoptosis. Smac/Diablo mRNA and protein levels increased by 50% at 24 hours. TNF, a cytokine whose renal expression increases in folic acid nephropathy, induced apoptosis in cultured tubular epithelial cells in a time-dependent manner. In addition, TNF increased the mRNA and protein expression of Smac/Diablo. CONCLUSION: These findings support the concept that regulation of Smac/Diablo mRNA and protein expression is a mechanism by which lethal stimuli amplify their lethal potential in renal cells.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteínas Portadoras/metabolismo , Túbulos Renales/metabolismo , Proteínas Mitocondriales/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Células Cultivadas , Células Epiteliales/metabolismo , Ácido Fólico , Túbulos Renales/citología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Túbulos Renales/fisiología , Ratones , Ratones Endogámicos BALB C , Proteínas Mitocondriales/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
16.
Kidney Int ; 64(1): 181-91, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12787408

RESUMEN

BACKGROUND: Hyperplasia is attributed to enhanced tubular cell proliferation with unbalanced cell death. The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors induce apoptosis in a variety of cell lines, including proximal tubular cells. However, the mechanisms by which statins induce apoptosis in tubular cells have not been fully addressed. METHODS: Apoptosis induced by simvastatin was measured in murine tubular cells with and without overexpressing Bcl-xL. Expression of genes implicated in cell death was studied by Northern and Western blot. RESULTS: The treatment of proliferating murine tubular cells (MCT) with simvastatin induced apoptosis in a time- and dose-dependent manner (0.1 to 1 micromol/L). Apoptosis was correlated with Bcl-xL mRNA and protein down-regulation. By contrast, the treatment with simvastatin did not modify the expression of the proapoptotic protein Bax. Simvastatin treatment was associated with cytochrome C release from the mitochondria to the cytosol. We also observed the presence of active caspase 9 and 3 during apoptosis induced by simvastatin. These effects were reversed by mevalonate, farnesylpyrophosphate (FPP), and geranylgeranylpyrophosphate (GGPP), suggesting the involvement of protein prenylation. Simvastatin appears to alter the balance between cell-life and death-promoting genes, as reflected by the decreased Bcl-xL/Bax ratio. Supporting this hypothesis, overexpression of Bcl-xL reduced the amount of apoptosis induced by simvastatin by 80% when compared with control vector-expressing cells. The overexpression of Bcl-xL also prevented the activation of caspase 9 and 3. CONCLUSION: Our results indicate that down-regulation of Bcl-xL expression mediates apoptosis induced by statins in tubular cells. These results may be relevant to the treatment of disorders characterized by altered tubular proliferation.


Asunto(s)
Apoptosis/fisiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Simvastatina/farmacología , Proteínas de Fase Aguda/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Activación Enzimática , Túbulos Renales/citología , Ratones , Prenilación de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/metabolismo , Proteína bcl-X
17.
J Am Soc Nephrol ; 11(7): 1266-1277, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10864583

RESUMEN

Fas ligand (FasL) is a cell membrane cytokine that can promote apoptosis through activation of Fas receptors. Fas receptor activation induces glomerular cell apoptosis in vivo and participates in tubular cell death during acute renal failure. However, there is little information on the expression of FasL in the kidney. This study reports that FasL mRNA and protein are present in normal mouse and rat kidney. In situ hybridization and immunohistochemistry showed that proximal tubular epithelium is the main site of FasL expression in the normal kidney. In addition, increased total kidney FasL mRNA and de novo FasL protein expression by glomerular cells were observed in two different models of glomerular injury : rat immune-complex proliferative glumerulonephritis and murine lupus nephritis. Both full-length and soluble FasL were increased in the kidneys of the mice with nephritis. Cultured murine proximal tubular epithelial MCT cells and primary cultures of murine tubular epithelial cells expressed FasL mRNA and protein. Tubular epithelium-derived FasL induced apoptosis in Fassensitive lymphoid cell lines but not in Fas-resistant lymphoid cell lines. By contrast, MCT cells grown in the presence of the survival factors of serum were resistant to FasL, and only became partially sensitive to apoptosis induced by high concentrations (100 ng/ml) of FasL upon serum deprivation. However, MCT cells stimulated with inflammatory mediators (tumor necrosis factor-alpha, interferon-gamma, and lipopolysaccharide) increased cell surface Fas expression and were sensitized to apoptosis induced by FasL (FasL 55 +/- 5% versus control 8.3 +/- 4.1% apoptotic cells at 24 h, P < 0.05). Cytokine-primed primary cultures of tubular epithelial cells also acquired sensitivity to FasL-induced apoptosis. These results suggest that FasL expression by intrinsic renal cells may play a role in cell homeostasis in the normal kidney and during renal injury.


Asunto(s)
Apoptosis/fisiología , Glomerulonefritis/metabolismo , Túbulos Renales/metabolismo , Glicoproteínas de Membrana/fisiología , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Epitelio/metabolismo , Proteína Ligando Fas , Femenino , Humanos , Glomérulos Renales/metabolismo , Túbulos Renales/citología , Túbulos Renales Proximales/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Valores de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...