Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Kidney Int ; 74(1): 47-51, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18368029

RESUMO

Vascular access dysfunction contributes to patient morbidity during maintenance hemodialysis. In this study we determined if knockout of heme oxygenase-1 predisposed to malfunction of arteriovenous fistulas. After three weeks, all fistulas in wild type mice were patent whereas a third of the fistulas in knockout mice were occluded and these exhibited increased neointimal hyperplasia and venous wall thickening. Heme oxygenase-1 mRNA and protein were robustly induced in the fistulas of the wild type mice. In the knockout mice there was increased PAI-1 and MCP-1 expression, marked induction of MMP-2 and MMP-9, but similar expression of PDGF alpha, IGF-1, TGF-beta1, VEGF, and osteopontin compared to wild type mice. We conclude that heme oxygenase-1 deficiency promotes vasculopathic gene expression, accelerates neointimal hyperplasia and impairs the function of arteriovenous fistulas.


Assuntos
Cateteres de Demora/efeitos adversos , Heme Oxigenase-1/deficiência , Animais , Derivação Arteriovenosa Cirúrgica , Vasos Sanguíneos/lesões , Vasos Sanguíneos/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Heme Oxigenase-1/genética , Heme Oxigenase-1/fisiologia , Camundongos , Camundongos Knockout , Túnica Íntima/patologia
2.
Kidney Int ; 72(9): 1073-80, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17728706

RESUMO

Heme oxygenase-1 may exert cytoprotective effects. In this study we examined the sensitivity of heme oxygenase-1 knockout (HO-1(-/-)) mice to renal ischemia by assessing glomerular filtration rate (GFR) and cytokine expression in the kidney, and inflammatory responses in the systemic circulation and in vital extrarenal organs. Four hours after renal ischemia, the GFR of HO-1(-/-) mice was much lower than that of wild-type mice in the absence of changes in renal blood flow or cardiac output. Eight hours after renal ischemia, there was a marked induction of interleukin-6 (IL-6) mRNA and its downstream signaling effector, phosphorylated signal transducer and activator of transcription 3 (pSTAT3), in the kidney, lung, and heart in HO-1(-/-) mice. Systemic levels of IL-6 were markedly and uniquely increased in HO-1(-/-) mice after ischemia as compared to wild-type mice. The administration of an antibody to IL-6 protected against the renal dysfunction and mortality observed in HO-1(-/-) mice following ischemia. We suggest that the exaggerated production of IL-6, occurring regionally and systemically following localized renal ischemia, in an HO-1-deficient state may underlie the heightened sensitivity observed in this setting.


Assuntos
Taxa de Filtração Glomerular/fisiologia , Heme Oxigenase-1/metabolismo , Isquemia/fisiopatologia , Rim/irrigação sanguínea , Animais , Débito Cardíaco/fisiologia , Citocinas/sangue , Feminino , Heme Oxigenase-1/genética , Interleucina-6/metabolismo , Isquemia/metabolismo , Rim/metabolismo , Rim/fisiopatologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Fator de Transcrição STAT3/metabolismo , Fatores de Tempo
3.
Kidney Int ; 71(7): 619-28, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17311071

RESUMO

Renal ischemia-reperfusion injury (IRI) rapidly induces production of inflammatory mediators including, and in particular, tumor necrosis factor (TNF). Possible sources include resident parenchymal and bone marrow-derived cells as well as recruited leukocytes. Cell suspensions from kidneys subjected to IRI were examined by cell separation followed by in vitro culture and enzyme-linked immunosorbent assay (ELISA), immunoperoxidase and immunofluorescence microscopy, and multicolor flow cytometry to determine the contribution of dendritic cells (DCs) to early production of TNF and other inflammatory mediators. Secretion of TNF, interleukin (IL-6), monocyte chemoattractant protein-1 (MCP-1), and regulated on activation normal T cell expressed and secreted (RANTES) was increased in cell suspensions from IRI compared with control kidneys and was higher in DC-enriched preparations. Immunostaining identified TNF(+ve) cells that coexpressed the DC marker CD11c. Flow cytometry of bone marrow-derived (CD45(+ve)) cell populations at 24 h post-IRI demonstrated that F4/80(+ve)/CD11c(+ve) DCs remained proportionately stable and exhibit higher levels of DC maturation markers, whereas the proportion of F4/80(-ve) DCs, monocytes, neutrophils, and T cells increased. Intracellular staining for TNF confirmed that F4/80(+ve) DCs were the predominant TNF(+ve) cell and expressed higher levels than other TNF(+ve) cells. In vivo depletion of DCs from the kidney substantially attenuated TNF secretion by total and CD45(+ve) cells following IRI. The results uncover a role for resident F4/80(+ve) DCs as the predominant secretors of TNF within 24 h of IRI.


Assuntos
Células Dendríticas/metabolismo , Rim/irrigação sanguínea , Traumatismo por Reperfusão/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Antígenos CD11/biossíntese , Camundongos , Camundongos Endogâmicos C57BL
4.
Kidney Int ; 60(6): 2181-91, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11737592

RESUMO

BACKGROUND: Induction of heme oxygenase-1 (HO-1) protects against diverse insults in the kidney and other tissues. We examined the effect of overexpression of HO-1 on cell growth, expression of p21, and susceptibility to apoptosis. METHODS: LLC-PK1 cells were genetically engineered to exhibit stable overexpression of HO-1. The effects of such overexpression on cell growth, the cell cycle, and the cell cycle-inhibitory protein, p21, were assessed; additionally, the susceptibility of these HO-1 overexpressing cells to apoptosis induced by three different stimuli (TNF-alpha/cycloheximide, staurosporine, or serum deprivation) was evaluated by such methods as the quantitation of caspase-3 activity, phase contrast microscopy, and the TUNEL method. RESULTS: HO-1 overexpressing LLC-PK1 cells demonstrated cellular hypertrophy, decreased hyperplastic growth, and growth arrest in the G0/G1 phase of the cell cycle. HO-1 overexpressing cells were markedly resistant to apoptosis induced by TNFalpha/cycloheximide or staurosporine as assessed by the caspase-3 activity assay. Such overexpression also conferred resistance to apoptosis induced by serum deprivation as evaluated by the TUNEL method; in these studies, inhibition of HO attenuated the resistance to apoptosis. Expression of the cyclin dependent kinase inhibitor, p21CIP1, WAF1, SDI1, as judged by Northern and Western analyses, was significantly increased in HO-1 overexpressing cells, and decreased as HO activity was inhibited. Moreover, this reduction in expression of p21 attendant upon the inhibition of HO activity in HO-1 overexpressing cells paralleled the loss of resistance of these cells to apoptosis when HO activity is inhibited. The pharmacologic inducer of HO-1, hemin, increased expression of p21 in wild-type cells and decreased apoptosis provoked by TNF-alpha/cycloheximide. CONCLUSION: Cellular overexpression of HO-1 up-regulates p21, diminishes proliferative cell growth, and confers marked resistance to apoptosis. We speculate that such up-regulation of p21 contributes to the altered pattern of cell growth and resistance to apoptosis. Our studies uncover the capacity of HO-1 to markedly influence the cell cycle in renal epithelial cells. In light of the profound importance of the cell cycle as a determinant of cell fate, we speculate that the inductive effect of HO-1 on p21 and the attendant inhibitory effect on the cell cycle provide a hitherto unsuspected mechanism underlying the cytoprotective actions of HO-1.


Assuntos
Apoptose/efeitos dos fármacos , Ciclinas/metabolismo , Heme Oxigenase (Desciclizante)/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Heme Oxigenase-1 , Células LLC-PK1/efeitos dos fármacos , Células LLC-PK1/metabolismo , Células LLC-PK1/patologia , Suínos , Regulação para Cima
5.
Kidney Int ; 60(3): 1047-57, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11532099

RESUMO

BACKGROUND: It is widely held that liver disease predisposes toward acute tubular necrosis. The present study examines the effect of acute cholestatic liver disease on the susceptibility to glycerol-induced acute tubular necrosis in the rat. METHODS: Acute cholestatic liver disease was induced by ligation of the common bile duct, while the intramuscular injection of hypertonic glycerol was used to induce acute tubular necrosis. Renal injury was assessed by plasma creatinine concentration and renal histology. An in vitro model of heme protein-induced renal injury (hemoglobin in conjunction with glutathione depletion) was employed to assess the cytoprotective effects of bilirubin. RESULTS: Ligation of the common bile duct markedly reduced acute renal injury that occurs in the glycerol model (7.5 mL/kg body weight), as evidenced by a lower plasma creatinine concentration and less severe renal histologic injury. At a higher dose of glycerol (10 mL/kg body weight), ligation of the common bile duct again reduced renal injury and cumulative mortality that occurs five days after the induction of this model of acute renal failure. These protective effects of ligation of the common bile duct could not be ascribed to less severe muscle injury or red cell damage. Ligation of the common bile duct induced heme oxygenase-1 in the kidney and markedly so in the liver. Inhibition of heme oxygenase significantly attenuated, but did not prevent, the protective effects conferred by ligation of the common bile duct. Bilirubin, in low micromolar concentrations, was cytoprotective against heme protein-induced cell injury in vitro. CONCLUSIONS: Ligation of the common bile duct confers resistance to glycerol-induced acute tubular necrosis in the rat, actions that arise, in part, from the induction of heme oxygenase-1 in the kidney and liver. Bilirubin, in micromolar concentrations, protects against heme protein-induced renal injury. Our studies uncover a novel form of acquired resistance to renal injury, occurring, unexpectedly, in the setting of acute cholestatic liver disease. We speculate that such potentially cytoprotective alterations may safeguard the kidney against irreversible functional and structural injury in the hepatorenal syndrome.


Assuntos
Injúria Renal Aguda/prevenção & controle , Colestase Intra-Hepática/fisiopatologia , Síndrome Hepatorrenal/fisiopatologia , Injúria Renal Aguda/etiologia , Animais , Colestase Intra-Hepática/sangue , Creatinina/sangue , Modelos Animais de Doenças , Glicerol , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase-1 , Necrose Tubular Aguda/complicações , Masculino , Ratos , Ratos Sprague-Dawley
6.
Am J Pathol ; 158(3): 893-903, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238038

RESUMO

Chronic nephropathy is a recognized complication of sickle cell disease. Using a transgenic sickle mouse, we examined whether oxidative stress occurs in the sickle kidney, the origins and functional significance of such oxidant stress, and the expression of the oxidant-inducible, potentially protective gene, heme oxygenase-1 (HO-1); we also examined the expression of HO-1 in the kidney and in circulating endothelial cells in sickle patients. We demonstrate that this transgenic sickle mouse exhibits renal enlargement, medullary congestion, and a reduced plasma creatinine concentration. Oxidative stress is present in the kidney as indicated by increased amounts of lipid peroxidation; heme content is markedly increased in the kidney. Exacerbation of oxidative stress by inhibiting glutathione synthesis with buthionine-sulfoximine dramatically increased red blood cell sickling in the sickle kidney: in buthionine-sulfoximine-treated sickle mice, red blood cell sickling extended from the medulla into the cortical capillaries and glomeruli. HO activity is increased in the sickle mouse kidney, and is due to induction of HO-1. In the human sickle kidney, HO-1 is induced in renal tubules, interstitial cells, and in the vasculature. Expression of HO-1 is increased in circulating endothelial cells in patients with sickle cell disease. These results provide the novel demonstration that oxidative stress occurs in the sickle kidney, and that acute exacerbation of oxidative stress in the sickle mouse precipitates acute vaso-occlusive disease. Additionally, the oxidant-inducible, heme-degrading enzyme, HO-1, is induced regionally in the murine and human sickle kidney, and systemically, in circulating endothelial cells in sickle patients.


Assuntos
Anemia Falciforme/enzimologia , Heme Oxigenase (Desciclizante)/biossíntese , Rim/enzimologia , Estresse Oxidativo , Anemia Falciforme/sangue , Anemia Falciforme/patologia , Animais , Butionina Sulfoximina/farmacologia , Creatinina/sangue , Endotélio Vascular/enzimologia , Inibidores Enzimáticos/farmacologia , Eritrócitos/patologia , Glutationa/biossíntese , Heme/metabolismo , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Humanos , Rim/irrigação sanguínea , Rim/patologia , Peroxidação de Lipídeos , Proteínas de Membrana , Camundongos , Camundongos Transgênicos , Ativação Transcricional
7.
Kidney Int ; 59(1): 106-17, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11135063

RESUMO

BACKGROUND: Heme oxygenase (HO) is the rate-limiting enzyme in the degradation of heme; its inducible isozyme, HO-1, protects against acute heme protein-induced nephrotoxicity and other forms of acute tissue injury. This study examines the induction of HO-1 in the kidney chronically inflamed by heme proteins and the functional significance of such an induction of HO-1. METHODS: Studies were undertaken in a patient with chronic tubulointerstitial disease in the setting of paroxysmal nocturnal hemoglobinuria (PNH), in a rat model of chronic tubulointerstitial nephropathy caused by repetitive exposure to heme proteins, and in genetically engineered mice deficient in HO-1 (HO-1 -/-) in which hemoglobin was repetitively administered. RESULTS: The kidney in PNH evinces robust induction of HO-1 in renal tubules in the setting of chronic inflammation. The heme protein-enriched urine from this patient, but not urine from a healthy control subject, induced expression of HO-1 in renal tubular epithelial cells (LLC-PK1 cells). A similar induction of HO-1 and related findings are recapitulated in a rat model of chronic inflammation induced by repetitive exposure to heme proteins. Additionally, in the rat, the administration of heme proteins induces monocyte chemoattractant protein (MCP-1). The functional significance of HO-1 so induced was uncovered in the HO-1 knockout mouse: Repeated administration of hemoglobin to HO-1 +/+ and HO-1 -/- mice led to intense interstitial cellular inflammation in HO-1 -/- mice accompanied by striking up-regulation of MCP-1 and activation of one of its stimulators, nuclear factor-kappaB (NF-kappaB). These findings were not observed in similarly treated HO-1 +/+ mice or in vehicle-treated HO-1 -/- and HO-1 +/+ mice. CONCLUSION: We conclude that up-regulation of HO-1 occurs in the kidney in humans and rats repetitively exposed to heme proteins. Such up-regulation represents an anti-inflammatory response since the genetic deficiency of HO-1 markedly increases activation of NF-kappaB, MCP-1 expression, and tubulointerstitial cellular inflammation.


Assuntos
Heme Oxigenase (Desciclizante)/fisiologia , Heme/fisiologia , Nefrite/etiologia , Adulto , Animais , Doença Crônica , Esquema de Medicação , Indução Enzimática , Heme/farmacologia , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Hemoglobinúria Paroxística/enzimologia , Humanos , Rim/enzimologia , Proteínas de Membrana , Camundongos , Camundongos Knockout/genética , Ratos , Regulação para Cima
8.
Am J Physiol Regul Integr Comp Physiol ; 279(6): R1949-55, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11080057

RESUMO

We investigated a transgenic mouse model of sickle cell disease, homozygous for deletion of mouse beta-globin and containing transgenes for human beta(S) and beta(S-antilles) globins linked to the transgene for human alpha-globin. In these mice, basal cGMP production in aortic rings is increased, whereas relaxation to an endothelium-dependent vasodilator, A-23187, is impaired. In contrast, aortic expression of endothelial nitric oxide synthase (NOS) is unaltered in sickle mice, whereas expression of inducible NOS is not detected in either group; plasma nitrate/nitrite concentrations and NOS activity are similar in both groups. Increased cGMP may reflect the stimulatory effect of peroxides (an activator of guanylate cyclase), because lipid peroxidation is increased in aortae and in plasma in sickle mice. Despite increased vascular cGMP levels in sickle mice, conscious systolic blood pressure is comparable to that of aged-matched controls; sickle mice, however, evince a greater rise in systolic blood pressure in response to nitro-L-arginine methyl ester, an inhibitor of NOS. Systemic concentrations of the vasoconstrictive oxidative product 8-isoprostane are increased in sickle mice. We conclude that vascular responses are altered in this transgenic sickle mouse and are accompanied by increased lipid peroxidation and production of cGMP; we suggest that oxidant-inducible vasoconstrictor systems such as isoprostanes may oppose nitric oxide-dependent and nitric oxide-independent mechanisms of vasodilatation in this transgenic sickle mouse. Destabilization of the vasoactive balance in the sickle vasculature by clinically relevant states may predispose to vasoocclusive disease.


Assuntos
Anemia Falciforme/fisiopatologia , Globinas/genética , Hemoglobina Falciforme/genética , Músculo Liso Vascular/fisiopatologia , Anemia Falciforme/genética , Animais , Aorta/fisiologia , Aorta/fisiopatologia , Autoanticorpos/genética , Pressão Sanguínea , Calcimicina/farmacologia , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/fisiologia , Endotélio Vascular/fisiopatologia , Globinas/deficiência , Hemoglobina Falciforme/imunologia , Humanos , Técnicas In Vitro , Peroxidação de Lipídeos , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Nitratos/sangue , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Nitritos/sangue , Papaverina/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
9.
Am J Physiol Renal Physiol ; 279(4): F728-35, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10997923

RESUMO

We examined whether nitric oxide-generating agents influence expression of heme oxygenase-1 (HO-1) in renal proximal tubular epithelial cells, LLC-PK(1) cells, and the mechanisms underlying any such effects. In sublytic amounts, the nitric oxide donor sodium nitroprusside induced HO-1 mRNA and protein and HO activity in a dose-dependent and time-dependent fashion; this induction was specific for nitric oxide since the nitric oxide scavenger carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide significantly reduced such induction. The induction of HO activity by sodium nitroprusside, or by another nitric oxide donor, spermine NONOate, was markedly reduced by the iron chelator deferoxamine. Two different thiol-containing agents, N-acetylcysteine and dithiothreitol, blunted such induction of HO by nitric oxide. Downstream products of nitric oxide, such as peroxynitrite or cGMP, were not involved in inducing HO. In higher concentrations (millimolar amounts), sodium nitroprusside induced appreciable cytotoxicity as assessed by lactate dehydrogenase (LDH) release and lipid peroxidation, and both of these effects were markedly reduced by deferoxamine. Inhibition of HO did not affect the cytotoxic effects (measured by LDH release) of sodium nitroprusside. We thus provide the novel description of the induction of HO-1 in renal proximal tubular epithelial cells exposed to nitric oxide donors and provide the first demonstration in kidney-derived cells for the involvement of a redox-based mechanism in such expression. We also demonstrate that, in LLC-PK(1) cells exposed to nitric oxide donors, chelatable iron is involved in eliciting the HO-1 response observed at lower concentrations of these donors, and in mediating the cytotoxic effects of these donors when present in higher concentrations.


Assuntos
Heme Oxigenase (Desciclizante)/metabolismo , Túbulos Renais Proximais/enzimologia , Óxido Nítrico/fisiologia , Animais , Quelantes/farmacologia , Desferroxamina/farmacologia , Relação Dose-Resposta a Droga , Indução Enzimática/fisiologia , Células Epiteliais/enzimologia , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Túbulos Renais Proximais/citologia , L-Lactato Desidrogenase/antagonistas & inibidores , L-Lactato Desidrogenase/metabolismo , Células LLC-PK1 , Peróxidos Lipídicos/antagonistas & inibidores , Peróxidos Lipídicos/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio , Nitroprussiato/farmacologia , Estresse Oxidativo/fisiologia , RNA Mensageiro/metabolismo , Espermina/análogos & derivados , Espermina/farmacologia , Suínos
10.
Kidney Int ; 58(1): 144-52, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10886559

RESUMO

BACKGROUND: Angiotensin II is strongly incriminated in progressive renal injury. There is recent evidence that angiotensin II induces oxidative stress in vitro. We examined the capacity of angiotensin II to induce oxidative stress in vivo and the functional significance of such stress. The capacity of angiotensin II to induce the oxidant-sensitive gene heme oxygenase (HO) in vivo and in vitro was also examined. METHODS: Angiotensin II was administered via mini-osmotic pumps to rats maintained on standard diets. Indices of oxidative stress, including thiobarbituric acid reactive substance, carbonyl protein content, and HO activity, were determined. Indices of oxidative stress and functional markers were also determined in the DOCA salt model. The effect of angiotensin II was studied in rats maintained on antioxidant-deficient diets so as to examine the functional significance of oxidative stress induced by angiotensin II. We also explored the inductive effect of angiotensin II on HO in vivo and whether such actions occur in vitro. RESULTS: Angiotensin II administered in vivo increased kidney content of thiobarbituric acid reactive substances protein carbonyl content, and HO activity. These indices were not present in the kidney of rats treated with DOCA salt for three weeks. Such oxidative stress was functionally significant, since the administration of angiotensin II to rats maintained on a prooxidant diet demonstrated increased proteinuria and decreased creatinine clearance. The stimulatory effect on HO activity was due to induction of HO-1 mRNA, with HO-2 mRNA remaining unchanged. Expression of HO-1 was localized to the renal proximal tubules in vivo. We also demonstrate that angiotensin II at concentrations of 10-8 and 10-7 mol/L induces expression of HO-1 mRNA in LLC-PK1 cells. CONCLUSIONS: Angiotensin II induces oxidative stress in vivo, which contributes to renal injury. This study also demonstrates that angiotensin II induces renal HO activity caused by up-regulation of HO-1 in renal proximal tubules. Finally, angiotensin II directly induces HO-1 in renal proximal tubular epithelial cells in vitro.


Assuntos
Angiotensina II/farmacologia , Heme Oxigenase (Desciclizante)/metabolismo , Túbulos Renais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Vasoconstritores/farmacologia , Animais , Antioxidantes/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Northern Blotting , Desoxicorticosterona , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Hipertensão Renal/metabolismo , Técnicas In Vitro , Falência Renal Crônica/metabolismo , Túbulos Renais/irrigação sanguínea , Túbulos Renais/citologia , Células LLC-PK1/efeitos dos fármacos , Células LLC-PK1/enzimologia , Masculino , Nefrectomia , Proteinúria/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Circulação Renal/fisiologia , Suínos , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
11.
Kidney Int ; 57(6): 2423-33, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10844611

RESUMO

BACKGROUND: Renal diseases are conventionally classified into acute and chronic disorders. We questioned whether acute, reversible, renal insults may be induced to incite a chronic scarring process, employing as an acute insult the glycerol model of heme protein-induced renal injury. METHODS: Rats were subjected to weekly injections of hypertonic glycerol for up to six months. Renal function was serially determined, and the effect of such insults on renal histology and renal expression of collagen and fibrogenic cytokines was assessed. RESULTS: After the first injection of glycerol, which, expectedly, induced a prompt fall in the glomerular filtration rate (GFR), subsequent injections encountered a remarkable renal resistance in that the fall in GFR was markedly blunted. This resistance to acute decline in renal function in rats subjected to repetitive injections of glycerol was accompanied by less necrosis and apoptosis of renal tubular epithelial cells after such injections. The attenuation in the fall in GFR in response to repetitive exposure to glycerol-induced heme protein injury was maintained for up to six months. A progressive decline in GFR appeared after three months and was accompanied by histologic tubulointerstitial injury, the latter assessed at six months. These kidneys demonstrated up-regulation of collagen I, III, and IV in conjunction with increased expression of the oxidant-inducible, chemotactic cytokine, monocyte chemoattractant protein-1 (MCP-1), and the oxidant-inducible, fibrogenic cytokine, transforming growth factor-beta1 (TGF-beta1). The exposure of the kidney to a single injection of hypertonic glycerol increased the expression of both cytokines some three to five days following this exposure, while the exposure of NRK 49F cells in culture to an iron-dependent model of oxidative stress also increased expression of TGF-beta1 and collagen mRNAs. CONCLUSIONS: We conclude that this nephrotoxic insult, repetitively administered, encounters a resistance in the kidney such that the expected fall in GFR does not occur. However, with time, such resistance is accompanied by a decrease in GFR, the latter associated with chronic tubulointerstitial disease. Thus, a long-term cost is exacted, either along with, or as a consequence of, such resistance. We suggest that chronic up-regulation of such oxidant-inducible genes such as TGF-beta1 and MCP-1 contributes to tubulointerstitial disease, and iron-mediated oxidative stress may directly induce TGF-beta1.


Assuntos
Hemeproteínas/farmacologia , Nefropatias/induzido quimicamente , Rim/efeitos dos fármacos , Doença Aguda , Animais , Quimiocina CCL2/metabolismo , Doença Crônica , Colágeno/metabolismo , Creatinina/metabolismo , Glicerol/administração & dosagem , Glicerol/farmacologia , Injeções Intramusculares , Rim/metabolismo , Rim/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Estresse Oxidativo/fisiologia , Ratos , Fator de Crescimento Transformador beta/metabolismo
12.
Am J Pathol ; 156(5): 1527-35, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10793064

RESUMO

Heme oxygenase (HO) is the rate limiting enzyme in the degradation of heme, and its isozyme, HO-1, may protect against tissue injury. One posited mechanism is the degradation of heme released from destabilized heme proteins. We demonstrate that HO-1 is a critical protectant against acute heme protein-induced toxicity in vivo. In the glycerol model of heme protein toxicity-one characterized by myolysis, hemolysis, and kidney damage-HO-1 is rapidly induced in the kidney of HO-1 +/+ mice as the latter sustain mild, reversible renal insufficiency without mortality. In stark contrast, after this insult, HO-1 -/- mice exhibit fulminant, irreversible renal failure and 100% mortality; HO-1 -/- mice do not express HO-1, and evince an eightfold increment in kidney heme content as compared to HO-1 +/+ mice. We also demonstrate directly the critical dependency on HO-1 in protecting against a specific heme protein, namely, hemoglobin: doses of hemoglobin which exert no nephrotoxicity or mortality in HO-1 +/+ mice, however, precipitate rapidly developing, acute renal failure and marked mortality in HO-1 -/- mice. We conclude that the induction of HO-1 is an indispensable response in protecting against acute heme protein toxicity in vivo.


Assuntos
Injúria Renal Aguda/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Hemeproteínas/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/mortalidade , Animais , Creatinina/sangue , Feminino , Glicerol/administração & dosagem , Glicerol/efeitos adversos , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Hemeproteínas/farmacologia , Hemoglobinas/farmacologia , Rim/efeitos dos fármacos , Rim/enzimologia , Rim/patologia , Testes de Função Renal , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Análise de Sobrevida , Taxa de Sobrevida
13.
Am J Physiol ; 277(1): F58-65, 1999 07.
Artigo em Inglês | MEDLINE | ID: mdl-10409298

RESUMO

In the glycerol model of renal injury we describe an acute rise in systemic arterial pressure which is attended by a reduced vasodilatory response to acetylcholine in vivo; vasodilatory responses to verapamil, however, were not impaired. Neither arginine nor sodium nitroprusside diminished this rise in blood pressure; N(omega)-nitro-L-arginine methyl ester (L-NAME) elevated basal mean arterial pressure and markedly blunted the rise in mean arterial pressure following the administration of glycerol. Aortic rings from the glycerol-treated rat demonstrate an impaired vasodilatory response to acetylcholine, an effect not repaired by arginine; the vasodilatory responses to nitric oxide donors, sodium nitroprusside and SIN-1, were also impaired; 8-bromo-cGMP, at higher doses, evinced a vasodilatory response comparable to that observed in the control rings. This pattern of responses was not a nonspecific effect of aortic injury, since aortic rings treated with mercuric chloride, a potent oxidant, displayed an impaired vasodilatory response to acetylcholine but not to sodium nitroprusside. We conclude that in the glycerol model of heme protein-induced tissue injury, there is an acute elevation in mean arterial pressure attended by impaired endothelium-dependent vasodilatation in vitro and in vivo. We suggest that the acute scavenging of nitric oxide by heme proteins depletes the blood vessel wall of its endogenous vasodilator and permeation of heme proteins into the blood vessel wall may contribute to such sustained effects as observed in vitro.


Assuntos
Hemeproteínas , Hipertensão/fisiopatologia , Rim/fisiopatologia , Acetilcolina/farmacologia , Doença Aguda , Animais , Pressão Sanguínea/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Modelos Animais de Doenças , Glicerol/farmacologia , Rim/efeitos dos fármacos , Masculino , Nitroprussiato/farmacologia , Ratos , Ratos Sprague-Dawley , Vasodilatadores/farmacologia
14.
Kidney Int ; 53(2): 367-81, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9461096

RESUMO

Growth and injury represent recurrent and related themes in the study of progressive renal disease. We have previously demonstrated that a prooxidant diet, one deficient in antioxidants, selenium and vitamin E, induces renal enlargement, proteinuria, mild tubulointerstitial disease and diminished glomerular filtration rate (GFR). Our present study represents continued examination of these processes. We demonstrate that these diets increase thymidine incorporation into DNA and net DNA content in renal tissue, and induce expression of the mRNA for the proto-oncogene, c-myc, and the histone, H2b. We localize increased DNA synthesis as occurring mainly in the distal renal tubular epithelium. These deficient kidneys also exhibit interstitial expansion that parallels the pattern of DNA synthesis in that both processes are more prominent in the medulla than in the cortex. mRNAs for collagens I, III and IV in conjunction with transforming growth factor-beta1 (TGF-beta1) are up-regulated in the kidney in rats maintained on the deficient diet. In complementary in vitro studies, the exposure of rat kidney fibroblasts, NRK 49F cells, to noncytolytic doses of hydrogen peroxide, induces collagen III, collagen IV and TGF-beta1 mRNA. Induction of these genes is also observed in mesangial cells so exposed to noncytolytic doses of hydrogen peroxide. A final aspect of our study was the examination of renal generation of hydrogen peroxide and the profile of the hydrogen peroxide-degrading enzymes. Deficient kidneys exhibit increased mitochondrial generation of hydrogen peroxide independent of oxygen consumption but in conjunction with suppression of glutathione peroxidase mRNA and activity. Lipid peroxidation was increased twofold in the cortex and medulla of the deficient kidneys. Surprisingly, catalase activity, measured in the cortex and medulla, and whole kidney catalase mRNA were also reduced in rats maintained on the antioxidant deficient diet, effects that may further compromise the clearance of hydrogen peroxide. These changes in catalase represent an adverse response to this dietary deficiency, and may be relevant to decreased catalase activity described in chronic renal insufficiency. Thus, a chronic prooxidant state, with features that mimic those of clinical uremia, increases DNA synthesis of renal tubular epithelium, induces mRNA expression for collagens I, III and IV in conjunction with the mRNA for the fibrogenic cytokine, TGF-beta1. Oxidants also induce collagen III, collagen IV and TGF-beta1 mRNA in vitro.


Assuntos
Colágeno/genética , Expressão Gênica/fisiologia , Rim/metabolismo , Fator de Crescimento Transformador beta/genética , Animais , Antioxidantes/metabolismo , Northern Blotting , Catalase/metabolismo , DNA/análise , DNA/biossíntese , Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Rim/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Masculino , Mitocôndrias/enzimologia , Oxidantes/farmacologia , Oxirredução , Proto-Oncogenes/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Selênio/deficiência , Deficiência de Vitamina E/metabolismo
15.
Kidney Int ; 53(1): 100-11, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9453005

RESUMO

We examined two potential intracellular targets in the glycerol model of acute renal failure, namely, the mitochondrion and the nucleus. Within three hours, alterations in mitochondrial function are already apparent. With either glutamate/malate or succinate/rotenone, state 3 and uncoupled respirations were decreased at three hours, and at 24 hours, such decrements were quite pronounced; in the presence of glutamate/malate, state 2 respiration was also depressed at 24 hours, while with succinate/rotenone state 2 was increased. Marked ultrastructural changes were observed in mitochondria studied at three hours, including the novel finding of degenerate mitochondria in autophagic vacuoles. Since the heme content in mitochondria was increased some tenfold within three hours, mitochondrial function was studied after exposure to concentrations of heme that reproduced such contents of heme: mitochondria initially displayed increased respiration, and subsequently, a persistent decline in oxygen consumption until oxygen consumption was virtually undetectable. With higher concentrations of heme, the early increase in oxygen consumption was blunted and the progressive decline in oxygen consumption was hastened. The antioxidant iron chelator, deferoxamine, prevented the early rise in oxygen consumption but did not prevent or delay the subsequent decline. We also assessed nuclear damage as a potential lesion in the glycerol model. DNA laddering was not observed at any time point. At 3 and 24 hours there was DNA injury by the TUNEL technique in the distal nephron but not in the proximal nephron. The 8-hydroxydeoxyguanosine/deoxyguanosine content was increased in the glycerol kidneys at 24 hours but not at three hours. At neither time point was evidence of apoptosis observed by light or electron microscopy. In studies undertaken in cell culture models, heme, at concentrations of 10 microM, failed to evince any such changes in LLC-PK1 cells, a cell line from the proximal tubule, or in MDCK cells, a cell line derived from the distal tubule. At concentrations of 50 microM, heme induced approximately 20% positivity in MDCK cells but none in LLC-PK1 cells by the TUNEL technique. We conclude that mitochondria and nuclei are prominent targets for injury in the glycerol model of acute renal failure. The presence of TUNEL-positive cells in the distal nephron but not at proximal sites in vivo underscores the increasing appreciation of the distinct responses of these nephron sites to nephrotoxic insults.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Glicerol/toxicidade , Hemeproteínas/toxicidade , Mitocôndrias/efeitos dos fármacos , Animais , Hidroxitolueno Butilado/farmacologia , Núcleo Celular/efeitos dos fármacos , Dano ao DNA , Rim/ultraestrutura , Mitocôndrias/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Ratos
16.
J Clin Invest ; 98(9): 2139-45, 1996 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8903334

RESUMO

Considerable attention is directed to a surprising biologic phenomenon wherein tissues exposed to one insult acquire resistance to another. We identify a novel example of acquired resistance to acute renal failure and a mechanism that contributes to such resistance. Nephrotoxic serum, administered to rats 24 h before the induction of glycerol-induced acute renal failure, reduces functional and structural injury that occurs in this model. Since heme oxygenase, the rate-limiting enzyme in heme degradation, protects against heme protein-induced renal injury, we questioned whether induction of heme oxygenase underlies the protection afforded by nephrotoxic serum. Kidney heme oxygenase (HO-1) mRNA was induced 6 h after nephrotoxic serum and renal tubules were identified as the site of expression of heme oxygenase protein. Induction of heme oxygenase was accompanied by increased renal content of ferritin but not by induction of other antioxidant enzymes. Inhibition of heme oxygenase prevented the protection afforded by nephrotoxic serum. Nephrotoxic serum did not protect against ischemic acute renal failure, a model in which heme oxygenase is not induced. Thus, nephrotoxic serum protects against glycerol-induced acute renal failure by inducing heme oxygenase in tubules. This study provides the first demonstration of resistance to tubular injury acquired from glomerular inflammation, uncovers a mechanism for such resistance, and exposes the dialogue that occurs between glomeruli and tubules.


Assuntos
Glomerulonefrite/complicações , Heme Oxigenase (Desciclizante)/biossíntese , Nefropatias/etiologia , Injúria Renal Aguda/fisiopatologia , Animais , Indução Enzimática , Ferritinas/metabolismo , Regulação Enzimológica da Expressão Gênica , Glomerulonefrite/fisiopatologia , Glicerol/toxicidade , Heme/urina , Heme Oxigenase (Desciclizante)/genética , Hemoglobinúria/metabolismo , Imunidade Inata , Nefropatias/enzimologia , Túbulos Renais/metabolismo , Masculino , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/fisiopatologia , Ovinos
17.
Am J Physiol ; 271(4 Pt 2): F814-23, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8898011

RESUMO

In protein-uric states, renal tubular epithelial cells are exposed to diverse macromolecules, including low-density lipoproteins (LDL), normally excluded from the urinary space. Oxidized LDL (LDLox) is incriminated in atherogenesis and glomerulosclerosis. Since urine is prooxidant, we considered whether LDLox injuries renal tubular epithelial cells (LLC-PK1). We demonstrate that the cytotoxicity of LDLox on LLC-PK1 cells resembles its toxicity to human umbilical vein endothelial cells (HUVEC) in that oxidized but not native LDL is injurious. Pretreatment of LLC-PK1 cells and HUVEC with antioxidants markedly reduced the cytotoxicity of LDLox. Pretreatment of LDL with antioxidants, prior to oxidation of LDL, vitiated its cytotoxicity. That LDLox is prooxidant was supported by expression of heme oxygenase, a redox-sensitive enzyme. LDLox induced heme oxygenase mRNA and enzyme activity. Pretreatment of LDL with antioxidants prior to oxidation attenuated heme oxygenase mRNA induction in LLC-PK1 and HUVEC. An iron chelator prevented cytotoxicity and heme oxygenase expression induced by LDLox. Based on these effects of LDLox, we draw an analogy between tubulointerstitial disease and atherogenesis and speculate that LDLox contributes to tubulointerstitial disease in proteinuric states.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Animais , Antioxidantes/farmacologia , Bleomicina , Sobrevivência Celular/efeitos dos fármacos , Endotélio Vascular/citologia , Indução Enzimática/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/metabolismo , Humanos , Ferro/metabolismo , Túbulos Renais/citologia , Células LLC-PK1 , Lipoproteínas LDL/efeitos dos fármacos , Suínos
18.
Kidney Int ; 50(3): 1032-43, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8872981

RESUMO

The role of oxidative stress in mercuric chloride (HgCl2)-induced nephrotoxicity is uncertain and controversial. We demonstrate that I.L.C-PK1 cells, exposed to HgCl2, generate massive amounts of hydrogen peroxide, the latter completely quenched by the hydrogen peroxide scavenger, pyruvate. HgCl2 exerts a dose-dependent cytotoxicity which is attenuated by pyruvate and catalase. Cellular generation of hydrogen peroxide arises, at least in part, from mitochondria since mitochondrial rates of generation of hydrogen peroxide increase in response to HgCl2; HgCl2 also provokes a shift in absorbance spectra in rhodamine 123 loaded-mitochondria and stimulates mitochondrial state 4 respiration. HgCl2, applied for one hour, impairs cellular vitality as demonstrated by the MTT assay, an assay dependent in part on mitochondrial function. HgCl2 impairs function in other organelles such as lysosomes that maintain a transmembrane proton gradient; these latter effects are partially attenuated by pyruvate. We complement these in vitro findings with in vivo evidence demonstrating that HgCl2 stimulates renal generation of hydrogen peroxide. The functional significance of such generation of hydrogen peroxide was evaluated in rats deficient in selenium and vitamin E, a nutrient deficiency that impairs the scavenging of hydrogen peroxide and promotes the toxicity of this oxidant. In these rats serum creatinine values were significantly higher on sequential days following the administration of HgCl2. To probe the renal response to oxidative stress induced by HgCl2, we examined hydrogen peroxide-scavenging enzymes and redox-sensitive genes. Catalase activity was unaltered whereas glutathione peroxidase activity was decreased, effects that may contribute to the net renal generation of hydrogen peroxide. The redox sensitive enzyme, heme oxygenase, was markedly up-regulated in the kidney in response to HgCl2. HgCl2 also induced members of the bcl family, bcl2 and bclx, genes that protect against apoptosis and oxidant injury. In another model of oxidant-induced renal injury, the glycerol model, bcl2 mRNA was not induced at 6 and 24 hours after the administration of glycerol. In summary, we demonstrate that HgCl2 potently stimulates renal generation of hydrogen peroxide in vitro and in vivo and such generation of peroxide contributes to renal dysfunction in vitro and in vivo. We also demonstrate that in response to HgCl2, redox sensitive genes are expressed including heme oxygenase and members of the bcl family.


Assuntos
Nefropatias/metabolismo , Mercúrio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Northern Blotting , Citotoxinas/farmacologia , Expressão Gênica/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/genética , Peróxido de Hidrogênio/metabolismo , Rim/citologia , Rim/efeitos dos fármacos , Rim/enzimologia , Nefropatias/induzido quimicamente , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Proteína bcl-X
19.
Kidney Int ; 48(4): 1298-307, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8569092

RESUMO

Cellular content of heme is regulated by heme oxygenase, the rate limiting enzyme in the degradation of heme. Induction of heme oxygenase is a protective response in an in vivo model of heme protein mediated renal injury, the glycerol model of acute renal failure. In addition to heme, heme oxygenase is induced by diverse forms of oxidative stress, the functional significance of which is currently unknown. We examined whether heme oxygenase is induced, and the functional significance of such induction, in two in vivo models of oxidant-induced toxic nephropathy, namely, cisplatin and gentamicin nephropathies; nephrotoxicity in these models is not dependent on the delivery of a burden of heme proteins to the kidney as occurs in the glycerol model. We demonstrate induction of heme oxygenase mRNA and protein in the kidney as early as 6 and 12 hours after a single dose of cisplatin (6 mg/kg i.v.). Pretreatment with tin protoporphyrin, a competitive inhibitor of heme oxygenase, led to higher serum creatinine values on days 3 through 5 and lower inulin clearances on day 5; tin protoporphyrin also exacerbated renal injury in this model. Renal hemodynamics studied at day 2 after cisplatin demonstrate reduced renal blood flow rates, increased renal vascular resistance and increased fractional excretion of sodium in rats treated with tin protoporphyrin. Tin protoporphyrin alone had no significant effect on serum creatinine and renal hemodynamics in rats with intact, disease-free kidneys. We confirmed that tin protoporphyrin prevented the increase in heme oxygenase activity induced by cisplatin. Induction of heme oxygenase by cisplatin was associated with increased kidney heme content and ferritin content.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Cisplatino/toxicidade , Heme Oxigenase (Desciclizante)/biossíntese , Rim/efeitos dos fármacos , Rim/metabolismo , Animais , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ferritinas/metabolismo , Gentamicinas/toxicidade , Heme/metabolismo , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Heme Oxigenase (Desciclizante)/genética , Rim/lesões , Masculino , Metaloporfirinas/farmacologia , Protoporfirinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Compostos de Sulfidrila/metabolismo , Fatores de Tempo
20.
Lab Invest ; 72(4): 474-83, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7723286

RESUMO

BACKGROUND: Prior administration of endotoxin has conferred resistance to tissue damage in a number of models of organ injury. The mechanisms by which this resistance is conferred are enigmatic. Recognizing that enhanced tissue oxidative stress may be a feature of endotoxin-associated injury and is present in many models of tissue injury, we questioned whether the beneficial effect conferred by endotoxin is dependent on the up-regulation of antioxidant defenses. EXPERIMENTAL DESIGN: We employed the glycerol model of acute renal failure (Gly-ARF), a model in which oxidant damage occurs in the kidney and other organs as a result of rhabdomyolysis and hemolysis. Rats were pretreated with endotoxin 24 hours before, or at the time of, induction of Gly-ARF. Renal functional studies and assessment of renal antioxidant status were performed. The effect of prior treatment with endotoxin was also examined in models of methemoglobin-induced and ischemic ARF. RESULTS: Renal function was improved in rats pretreated with endotoxin but worsened in rats subjected to Gly-ARF and endotoxin simultaneously. Endotoxin induced heme oxygenase activity and ferritin content in the kidney but did not induce other antioxidant systems such as catalase and glutathione peroxidase. Treatment with a competitive inhibitor of heme oxygenase blocked endotoxin-induced protection on days 2 and 3, while markedly attenuating the protective effect on day 1. Pretreatment with endotoxin reduced renal injury induced by methemoglobin, but not ischemia. CONCLUSIONS: The resistance to injury conferred by endotoxin in Gly-ARF involves induction of an antioxidant response, consisting of increased heme oxygenase and ferritin synthesis. This coupled response allows degradation of heme as well as chelation of iron, thus decreasing oxidant-mediated tissue injury. This novel mechanism of endotoxin-induced resistance may be applicable not only to Gly-ARF but also to other models of tissue injury in which enhanced oxidative stress is implicated.


Assuntos
Injúria Renal Aguda/fisiopatologia , Ferritinas/fisiologia , Heme Oxigenase (Desciclizante)/fisiologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Injúria Renal Aguda/induzido quimicamente , Animais , Suscetibilidade a Doenças , Endotoxinas/farmacologia , Glicerol , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Hemeproteínas/metabolismo , Hemólise/efeitos dos fármacos , Isquemia/fisiopatologia , Rim/metabolismo , Masculino , Metemoglobina , Músculos/efeitos dos fármacos , Músculos/fisiopatologia , Oxirredutases/metabolismo , Ratos , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA