Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Kidney ; 2(1)2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27104220

RESUMEN

Sepsis can provoke kidney injury, which increases mortality. Human and animal studies have documented increased renal oxidative injury and mitochondrial damage during sepsis. However, few studies have attempted to dissect specific renal targets and/or types of oxidative injury using the cecal ligation and puncture (CLP) murine model of sepsis. The purpose of this short communication is to examine the extent of lipid peroxidation within renal mitochondria using CLP and blue native gel electrophoresis which separates intact mitochondrial respiratory complexes. Our results show that CLP induced increased 4-hydroxy-nonenal protein adduction (marker of lipid peroxidation) in renal homogenates and mitochondrial fractions. Blue native gel electrophoresis revealed that respiratory complex III was selectively targeted within mitochondrial fractions. This supports our prior report showing renal complex III inactivation following CLP. Future studies will identify specific renal proteins within complex III that are modified during sepsis to provide mechanistic insight on how mitochondrial respiration is inhibited during sepsis.

2.
Clin Pharmacol Ther ; 96(6): 662-4, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25148376

RESUMEN

Sepsis-induced acute kidney injury (SAKI) is a frequent complication of infant sepsis that approximately doubles the mortality rate. The poor prognosis of these patients is a result of care that is mainly supportive, nontargeted, and usually begun only after symptoms of the systemic inflammatory response syndrome are observed. Preclinical studies from relevant rodent models of SAKI suggest that mitochondria-targeted antioxidants may be a new mode of therapy that could promote recovery.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Antioxidantes/uso terapéutico , Mitocondrias/efectos de los fármacos , Compuestos Organofosforados/uso terapéutico , Piperidinas/uso terapéutico , Sepsis/complicaciones , Animales , Modelos Animales de Enfermedad , Humanos , Lactante , Ratones , Mitocondrias/metabolismo , Ratas
3.
Nitric Oxide ; 5(5): 432-41, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11587558

RESUMEN

We recently reported that nitrotyrosine and acetaminophen (APAP)-cysteine protein adducts colocalize in the hepatic centrilobular cells following a toxic dose of APAP to mice. Whereas APAP-adducts are formed by reaction of the metabolite N-acetyl-p-benzoquinone imine with cysteine, nitrotyrosine residues are formed by reaction of tyrosine with peroxynitrite. Peroxynitrite is formed from nitric oxide (NO) and superoxide. This manuscript examines APAP (300 mg/kg) hepatotoxicity in mice lacking inducible nitric oxide synthase activity (NOS2 null or knockout mice; C57BL/6-Nos2(tm1Lau)) and in the wildtype mice. In a time course the ALT levels in the exposed NOS2 null mice were approximately 50% of the wildtype mice; however, histological examination of liver sections indicated similar levels of centrilobular hepatic necrosis in both wild-type and NOS2 null mice. Serum nitrate plus nitrite levels (NO synthesis) were identical in saline-treated NOS2 null and wild-type mice (53 +/- 2 microM). APAP increased NO synthesis in wild-type mice only. The increases paralleled the increases in ALT levels with peak levels of serum nitrate plus nitrite at 6 h (168 +/- 27 microM). In wild-type mice hepatic tyrosine nitration was greatly increased relative to saline treated controls. Tyrosine nitration increased in NOS2 null mice also, but the increase was much less. APAP increased hepatic malonaldehyde levels (lipid peroxidation) in NOS2 null mice only. The results suggest the presence of multiple pathways to APAP-mediated hepatic necrosis, one via nitrotyrosine, as in the wild-type mice, and another that is not dependent upon inducible nitric oxide synthase activity, but which may involve increased superoxide.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Hígado/efectos de los fármacos , Óxido Nítrico Sintasa/deficiencia , Tirosina/análogos & derivados , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Eliminación de Gen , Técnicas para Inmunoenzimas , Peroxidación de Lípido , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nitratos/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Nitritos/metabolismo , Tirosina/metabolismo
4.
Toxicol Sci ; 63(1): 143-8, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11509754

RESUMEN

Previous evidence suggests that both oxygen radicals and nitric oxide (NO) are important mediators of injury during renal ischemia-reperfusion (I-R) injury. However, the generation of reactive nitrogen species (RNS) has not been evaluated in this model at early time points. The purpose of these studies was to examine the development of oxidant stress and the formation of RNS during I-R injury. Male Sprague-Dawley rats were anesthetized and subjected to 40 min of bilateral renal ischemia followed by 0, 3, or 6 h of reperfusion. Control animals received a sham operation. Plasma urea nitrogen and creatinine levels were monitored as markers of renal injury. Glutathione (GSH) oxidation and 4-hydroxynonenal (4-HNE)-protein adducts were used as markers of oxidant stress. 3-Nitrotyrosine (3-NT) was used as a biomarker of RNS formation. Significant increases in plasma creatinine concentrations and urea nitrogen levels were found following both 3 and 6 h of reperfusion. Increases in GSH oxidation, 4-HNE-protein adduct levels, and 3-NT levels were observed following 40 min of ischemia with no reperfusion. Since these results suggested RNS generation during the 40 min of ischemia, a time course of RNS generation following 0, 5, 10, 20, and 40 min of ischemia was evaluated. Significant increases in 3-NT generation was detected as early as 10 min of ischemia and rose to values nearly 10-fold higher than Control at 40 min of ischemia. No additional increase was observed following reperfusion. The data clearly demonstrate that oxidative stress and RNS generation occur in the kidney during ischemia.


Asunto(s)
Riñón/irrigación sanguínea , Nitratos/metabolismo , Estrés Oxidativo , Daño por Reperfusión/metabolismo , Tirosina/análogos & derivados , Aldehídos/metabolismo , Animales , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Radicales Libres/metabolismo , Glutatión/metabolismo , Riñón/patología , Masculino , Oxidación-Reducción , Proteínas/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/sangre , Daño por Reperfusión/patología , Factores de Tiempo , Tirosina/metabolismo
5.
Am J Physiol Renal Physiol ; 280(3): F474-9, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11181409

RESUMEN

ANG II exerts a biphasic effect on Na+ transport in the kidney through its effects on Na+-K+-ATPase activity. Beginning at 10(-13) M, ANG II increased Na+-K+-ATPase in freshly isolated rat proximal tubules to a maximum stimulation at 10(-11) M of 1.43 +/- 0.08-fold above control. Stimulation decreased progressively at concentrations >10(-10) M to a value of 0.96 +/- 0.1-fold at 10(-7) M. In the presence of additional L-arginine, the substrate for NO synthesis, the stimulatory effect of ANG II (10(-11) M) was lost. Conversely, N-monomethyl-L-arginine (L-NMMA), the nitric oxide (NO) synthase inhibitor, unmasked the stimulatory effect of ANG II at 10(-7) M (1.40 +/- 0.1-fold). 1H-[1,2,4]oxadiazole-[4,3-a]quinoxalin-1-one, the soluble guanylyl cyclase inhibitor, like L-NMMA, unmasked the stimulatory effect of ANG II at 10(-7) M (1.30 +/- 0.1-fold). The intracellular cGMP concentration was increased 1.58 +/- 0.28-fold at 10(-7) M ANG II. The ANG II AT(1) receptor antagonist SK&F 108566 blocked the stimulatory effect of ANG II at 10(-11) M. These data suggest that the NO/cGMP signaling pathway serves as a negative component in the regulation of Na+-K+-ATPase activity by ANG II.


Asunto(s)
Angiotensina II/fisiología , GMP Cíclico/fisiología , Túbulos Renales Proximales/enzimología , Óxido Nítrico/fisiología , Transducción de Señal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Angiotensina II/farmacología , Antagonistas de Receptores de Angiotensina , Animales , Arginina/farmacología , Calcio/metabolismo , GMP Cíclico/biosíntesis , Inhibidores Enzimáticos/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Membranas Intracelulares/metabolismo , Túbulos Renales Proximales/metabolismo , Masculino , Oxadiazoles/farmacología , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1 , Receptor de Angiotensina Tipo 2 , Solubilidad , omega-N-Metilarginina/farmacología
6.
J Pharmacol Exp Ther ; 295(1): 417-22, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10992009

RESUMEN

Reactive oxygen species are suggested to participate in ischemia-reperfusion (I-R) injury. However, induction of inducible nitric oxide synthase (iNOS) and production of high levels of nitric oxide (NO) also contribute to this injury. NO can combine with superoxide to form the potent oxidant peroxynitrite (ONOO(-)). NO and ONOO(-) were investigated in a rat model of renal I-R injury using the selective iNOS inhibitor L-N(6)-(1-iminoethyl)lysine (L-NIL). Sprague-Dawley rats were subjected to 40 min of bilateral renal ischemia followed by 6 h of reperfusion with or without L-NIL administration. Control animals received a sham surgery and had plasma creatinine values of 0.4 +/- 0.1 mg/dl. I-R surgery significantly increased plasma creatinine levels to 1.9 +/- 0.3 mg/dl (P <.05) and caused renal cortical necrosis. L-NIL administration (3 mg/kg) in animals subjected to I-R significantly decreased plasma creatinine levels to 1.2 +/- 0.10 mg/dl (P <.05 compared with I-R) and reduced tubular damage. ONOO(-) formation was evaluated by detecting 3-nitrotyrosine-protein adducts, a stable biomarker of ONOO(-) formation. Immunohistochemistry and HPLC revealed that the kidneys from I-R animals had increased levels of 3-nitrotyrosine-protein adducts compared with control animals. L-NIL-treated rats (3 mg/kg) subjected to I-R showed decreased levels of 3-nitrotyrosine-protein adducts. These results support the hypothesis that iNOS-generated NO mediates damage in I-R injury possibly through ONOO(-) formation.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Isquemia/metabolismo , Riñón/irrigación sanguínea , Lisina/análogos & derivados , Nitratos/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Daño por Reperfusión/metabolismo , Animales , Lisina/farmacología , Masculino , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo II , Ratas , Ratas Sprague-Dawley
7.
J Pharmacol Exp Ther ; 293(3): 968-72, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10869399

RESUMEN

The role of inducible nitric-oxide synthase (iNOS) in lipopolysaccharide (LPS)-induced hepatic oxidant stress was evaluated using the iNOS inhibitor L-iminoethyl-lysine (L-NIL). Male rats were divided into three groups. One group received LPS (Salmonella minnesota) 2 mg/kg i.v. A second group received LPS plus L-NIL (3 mg/kg i.p.) at the time of LPS administration followed by a second dose 3 h later. A third group received saline i.v. At 6 h, blood and liver tissue were collected. Serum nitrate/nitrite (metabolic products of nitric oxide) levels were increased from 5.4 +/- 1.5 nmol/ml in the saline group to 360 +/- 48 nmol/ml in the LPS group (n = 5). Values for the LPS + L-NIL group were significantly reduced to 35 +/- 7 nmol/ml. Tissue malondialdehyde levels were increased from 0.20 +/- 0.02 nmol/mg (n = 4) in the saline group to 0.41 +/- 0.03 nmol/mg (n = 4) in the LPS group. L-NIL significantly reduced the values in the LPS group to 0.29 +/- 0.02 nmol/mg (n = 4). 4-Hydroxynonenal-protein adducts levels were increased 3.6-fold by LPS treatment as compared with saline. L-NIL significantly reversed the levels to 1.6-fold (n = 4). Intracellular GSH levels were decreased from 8.49 +/- 0.64 nmol/mg (n = 4) in the saline group to 5.63 +/- 0.51 nmol/mg in the LPS group (n = 7). L-NIL significantly increased the levels in the LPS group to 7.04 +/- 0.46 nmol/mg (n = 7). These data indicate that LPS-induced nitric oxide generation can result in oxidant stress in the liver, and that inhibitors of iNOS may offer some protection in LPS-induced hepatic toxicity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Lipopolisacáridos/toxicidad , Hígado/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Estrés Oxidativo , Animales , Western Blotting , Glutatión/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Nitratos/metabolismo , Óxido Nítrico Sintasa de Tipo II , Ratas , Ratas Sprague-Dawley
8.
Biochem Pharmacol ; 59(2): 203-9, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10810455

RESUMEN

Lipopolysaccharide (LPS)-induced renal oxidant injury and the role of nitric oxide (NO) were evaluated using the inducible nitric oxide synthase (iNOS) inhibitor L-iminoethyl-lysine (L-NIL). One group of male rats received LPS (Salmonella minnesota; 2 mg/kg, i.v.). A second group received LPS plus L-NIL (3 mg/kg, i.p.). A third group received saline i.v. At 6 hr, iNOS protein was induced in the kidney cortex, and plasma nitrate/nitrite levels were increased from 4 +/- 2 nmol/mL in the Saline group to 431 +/- 23 nmol/mL in the LPS group. The value for the LPS + L-NIL group was reduced significantly to 42 +/- 9 nmol/mL. LPS increased blood urea nitrogen levels from 13 +/- 1 to 47 +/- 3 mg/dL. LPS + L-NIL reduced these levels significantly to 29 +/- 2 mg/dL. Plasma creatinine levels were unchanged in all groups. Tissue lipid peroxidation products in the kidney were increased from 0.16 +/- 0.01 nmol/mg in the Saline group to 0.30 +/- 0.03 nmol/mg in the LPS group. LPS + L-NIL reduced the values significantly to 0.22 +/- 0.02 nmol/mg. Intracellular glutathione levels were decreased in the kidneys from 1.32 +/- 0.1 nmol/mg in the Saline group to 0.66 +/- 0.08 nmol/mg in the LPS group. LPS + L-NIL increased the levels significantly to 0.99 +/- 0.13 nmol/mg. LPS increased the 3-nitrotyrosine-protein adducts in renal tubules as detected by immunohistochemistry, indicating the generation of peroxynitrite. L-NIL decreased adduct formation. These data indicated that LPS-induced NO generation resulted in peroxynitrite formation and oxidant stress in the kidney and that inhibitors of iNOS may offer protection against LPS-induced renal toxicity.


Asunto(s)
Riñón/efectos de los fármacos , Lipopolisacáridos/farmacología , Óxido Nítrico/fisiología , Estrés Oxidativo/fisiología , Animales , Nitrógeno de la Urea Sanguínea , Western Blotting , Creatina/sangre , Inhibidores Enzimáticos/farmacología , Glutatión/metabolismo , Inmunohistoquímica , Riñón/fisiología , Peroxidación de Lípido/efectos de los fármacos , Lisina/análogos & derivados , Lisina/farmacología , Masculino , Nitratos/sangre , Nitratos/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Nitritos/sangre , Ratas , Ratas Sprague-Dawley , Tirosina/análogos & derivados , Tirosina/análisis , Tirosina/metabolismo
9.
Biochem Pharmacol ; 60(1): 137-43, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10807955

RESUMEN

Puromycin aminonucleoside (PAN) administration in rats produces an experimental model of nephrotic syndrome characterized by glomerular epithelial cell injury and proteinuria. The purpose of this study was to examine the role of nitric oxide (NO) in this model of minimal change glomerular disease. Aminoguanidine (AG) was used to inhibit inducible nitric oxide synthase (iNOS). Sprague-Dawley rats were divided into Control (N = 9), PAN (N = 14), AG (N = 2), and PAN + AG (N = 12) treatment groups. Control animals received saline (i.v. ), PAN animals received PAN (75 mg/kg, i.v.), and PAN + AG animals received PAN plus AG (50 mg/kg, i.p., twice daily). AG animals received a saline injection (i.v.) on day 0 in the place of PAN and then AG on the same schedule as the PAN + AG group. Animals were kept in metabolic cages, and urinary protein excretion and nitrite (NO(2)(-)) excretion were measured daily. PAN administration increased urinary NO(2)(-) excretion by day 2, and levels remained elevated through day 7. AG prevented this PAN-induced increase in urinary NO(2)(-) excretion. Plasma nitrate (NO(3)(-)) and NO(2)(-) (NOx) concentrations were also increased in the PAN and PAN + AG groups. iNOS protein expression was not detected in either the glomeruli or the cortex at day 7. Proteinuria developed in PAN animals on day 4 and increased steadily through day 7. PAN + AG animals showed a pattern similar to that of the PAN group. These results indicated that in contrast to models of proliferative glomerulonephritis, NO formation during PAN-induced nephrotic syndrome is increased but does not participate in the development of glomerular injury as measured by proteinuria.


Asunto(s)
Síndrome Nefrótico/enzimología , Óxido Nítrico Sintasa/fisiología , Animales , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Masculino , Síndrome Nefrótico/inducido químicamente , Síndrome Nefrótico/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Puromicina Aminonucleósido , Ratas , Ratas Sprague-Dawley
10.
Biol Reprod ; 62(1): 8-15, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10611061

RESUMEN

In chickens, oviposition is correlated with increased plasma levels of the neurohypophysial hormone vasotocin, and vasotocin stimulates contraction of uterine strips in vitro. A gene encoding a vasotocin receptor subtype that we have designated the VT1 receptor was cloned from the domestic chicken. The open reading frame encodes a 370-amino acid polypeptide that displays seven segments of hydrophobic amino acids, typical of guanine nucleotide-protein-coupled receptors. Other structural features of the VT1 receptor include two potential N-linked glycosylation sites in the extracellular N-terminal region, a conserved aspartic acid in transmembrane domain 2 that is found in nearly all guanine nucleotide-protein-coupled receptors, and two potential protein kinase C phosphorylation sites in the third intracellular loop and C-terminal tail. Expressed VT1 receptors in COS7 cells bind neurohypophysial hormones with the following rank order of potency: vasotocin congruent with vasopressin > oxytocin congruent with mesotocin > isotocin. In addition, the expressed VT1 receptor mediates vasotocin-induced phosphatidylinositol turnover and Ca(2+) mobilization. In the chicken, expression of VT1 receptor gene transcripts is limited to the shell gland (uterus) and the brain. Thus, the VT1 receptor that we have cloned may mediate contractions of the shell gland during oviposition and activate reproductive behaviors known to be stimulated by vasotocin in lower vertebrates.


Asunto(s)
Encéfalo/metabolismo , Pollos/genética , Clonación Molecular , Expresión Génica , Oviposición , Receptores de Vasopresinas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Femenino , Datos de Secuencia Molecular , Receptores de Vasopresinas/química , Transfección , Útero/metabolismo
11.
Hepatology ; 30(1): 186-95, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10385655

RESUMEN

Hepatotoxic doses of acetaminophen to mice produce not only acetaminophen-protein adducts in the centrilobular cells of the liver, but nitrotyrosine-protein adducts in the same cells, the site of the necrosis. Nitration of tyrosine occurs with peroxynitrite, a species formed by reaction of nitric oxide (NO.) with superoxide (O2. -). Because NO. and O2.- may be produced by activated Kupffer cells and/or infiltrated macrophages, we pretreated mice with the macrophage inactivators/depeleters gadolinium chloride (7 mg/kg, intravenously [iv]) or dextran sulfate (10 mg/kg, iv) 24 hours before administration of acetaminophen (300 mg/kg). Mice treated with acetaminophen plus gadolinium chloride, or acetaminophen plus dextran sulfate, had significantly less evidence of hepatotoxicity as evidenced by lower serum alanine transaminase (ALT) levels (28 +/- 1 IU/L and 770 +/- 240 IU/L, respectively) at 8 hours compared with acetaminophen (6,380 +/- 408 IU/L). Analysis of hepatic homogenates for acetaminophen-protein adducts at 2 hours, a time of maximal covalent binding and before hepatocyte lysis, indicated that these pretreatments did not decrease covalent binding. Western blot analysis for the macrophage marker protein F4/80 in homogenates revealed not only the expected decrease by the macrophage inactivators/depleters, but also an apparent increase in acetaminophen-only-treated mice. At 8 hours nitrotyrosine-protein adducts were present in the acetaminophen-only-treated mice, but not in the acetaminophen plus gadolinium chloride-treated mice, or acetaminophen plus dextran sulfate-treated mice. High levels of heme-protein adducts, a measure of oxidative stress, were detected in livers of the 8 hour acetaminophen-only-treated mice. These data suggest that acetaminophen hepatotoxicity is mediated by an initial metabolic activation and covalent binding, and subsequent activation of macrophages to form O2.-, NO., and peroxynitrite. Nitration of tyrosine correlates with toxicity.


Asunto(s)
Acetaminofén/toxicidad , Sulfato de Dextran/farmacología , Gadolinio/farmacología , Macrófagos del Hígado/fisiología , Hígado/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Acetaminofén/antagonistas & inhibidores , Acetaminofén/farmacocinética , Alanina Transaminasa/sangre , Animales , Antiinflamatorios/farmacología , Aspartato Aminotransferasas/sangre , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/patología , Hígado/patología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos , Necrosis , Nitratos/metabolismo , Oxidantes/metabolismo , Estrés Oxidativo/efectos de los fármacos , Superóxidos/metabolismo
12.
Life Sci ; 63(4): PL75-80, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9698043

RESUMEN

We examined the signal transduction cascade of angiotensin II in isolated rat proximal tubules. Angiotensin II induced a rapid (15 sec) concentration-dependent rise in intracellular free Ca2+ (EC50 = 1.7 nM). The rise in Ca2+ was blocked by the angiotensin II receptor AT1 specific antagonist SK&F 108566. This indicates that the rise in Ca2+ is fully mediated by AT1 receptors. To characterize further the antagonism by SK&F 108566, the Schild analysis was performed (pA2 = 10.9 +/- 0.14 and slope = 0.94 +/- 0.11; n=3). It indicated that SK&F 108566 is a high affinity competitive antagonist at AT1 receptors in the proximal tubule. Angiotensin II signaling also induced a rapid (5 min) rise in cGMP formation. This response was blocked by SK&F 108566, by inhibition of nitric oxide synthase, or by inhibition of soluble guanylyl cyclase. This indicates that the formation of cGMP elicited by angiotensin II is mediated by AT1 receptors and activation of the NO-cGMP pathway. Since cGMP can inhibit Na, K-ATPase activity, activation of the NO-cGMP pathway may act as a negative feedback component of angiotensin II signaling in renal proximal tubules.


Asunto(s)
Angiotensina II/metabolismo , GMP Cíclico/metabolismo , Túbulos Renales Proximales/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Tiofenos , Acrilatos/farmacología , Animales , Antihipertensivos/farmacología , Calcio/metabolismo , GMP Cíclico/biosíntesis , Imidazoles/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
13.
Chem Res Toxicol ; 11(6): 604-7, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9625727

RESUMEN

Treatment of mice with a toxic dose of acetaminophen (300 mg/kg, ip) significantly increased hepatotoxicity at 4 h, as evidenced by histological necrosis in the centrilobular areas of the liver, and increased serum levels of alanine aminotransferase (ALT) (from 8 +/- 1 IU/L in saline-treated mice to 3226 +/- 892 IU/L in the acetaminophen-treated mice). Serum levels of nitrate plus nitrite (a marker of nitric oxide synthesis) were also increased from 62 +/- 8 microM in saline-treated mice to 110 +/- 14 microM in acetaminophen-treated mice (P < 0.05). Regression analysis of serum ALT levels to serum nitrate plus nitrite levels in individual mice revealed a positive, linear relationship between serum ALT levels and serum nitrate plus nitrite levels with a correlation coefficient of 0.9 (P < 0.05). The y intercept value (nitrate plus nitrite level) was 63 +/- 15 microM. Immunohistochemical analysis of liver sections from acetaminophen-intoxicated mice using an anti-3-nitrotyrosine antibody indicated tyrosine nitration in the proteins of the centrilobular cells. Tyrosine nitration has been shown to occur by peroxynitrite, a reactive intermediate formed by an extremely rapid reaction of nitric oxide and superoxide and a species which also has hydroxyl radical-like activity. Analysis of liver sections using an anti-acetaminophen antiserum indicated the centrilobular cells also contained acetaminophen-protein adducts, a reaction of the metabolite N-acetyl-p-benzoquinone imine with cysteine residues on proteins. These data are consistent with acetaminophen metabolic activation leading to increased synthesis of nitric oxide and superoxide and to peroxynitrite as an important intermediate in the toxicity.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Hígado/efectos de los fármacos , Proteínas/metabolismo , Tirosina/análogos & derivados , Acetaminofén/farmacocinética , Alanina Transaminasa/sangre , Animales , Biotransformación , Hígado/metabolismo , Masculino , Ratones , Nitratos/metabolismo , Óxido Nítrico/biosíntesis , Tirosina/metabolismo
14.
J Toxicol Environ Health ; 51(5): 415-35, 1997 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-9233377

RESUMEN

Lipopolysaccharide is a component of the gram-negative bacterial cell wall that is responsible for 25,000-50,000 deaths in the United States each year. The sequelae of gram-negative infection and septicemia leading to death include fever, hypotension with inadequate tissue perfusion, and disseminated intravascular coagulation. It is clear that different cell types respond differently to lipopolysaccharide. Furthermore, various autacoids and cytokines are released that can affect cellular function even in cell types that do not recognize lipopolysaccharide. Despite advances made in the etiology of septic shock and organ failure, therapy is still for the most part supportive and largely ineffective. The aim of this review is to summarize the current understanding of the role of lipopolysaccharide in the development of septicemia by examining signal transduction and therapeutic approaches.


Asunto(s)
Proteínas de Fase Aguda , Escherichia coli , Lipopolisacáridos/toxicidad , Glicoproteínas de Membrana , Salmonella typhimurium , Animales , Bacteriemia/inducido químicamente , Bacteriemia/metabolismo , Bacteriemia/patología , Proteínas Portadoras/metabolismo , Endotoxemia/inducido químicamente , Endotoxemia/metabolismo , Endotoxemia/patología , Humanos , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/química , Transducción de Señal/fisiología
15.
Arch Biochem Biophys ; 338(2): 129-35, 1997 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9028863

RESUMEN

In previous studies, we found that lipid A, the biologically active component of lipopolysaccharide, triggers a rapid release of intracellular calcium, the activation of nitric oxide synthase (NOS), and nitric oxide (NO) production in rat proximal tubules. This pathway leads ultimately to cell death [as measured by the release of lactate dehydrogenase (LDH)], initiated by early generation of NO. In the present studies we found that lipid A produces a time- and concentration-dependent increase in lipid peroxidation [malondialdehyde (MDA) formation] prior to cell death. Furthermore, preventing lipid peroxidation protected against cell death. Lipid A (50 micro;g/ml) produced significant MDA formation in 30 min. The addition of two antioxidants 5 min prior to lipid A completely inhibited MDA formation and LDH release at 90 min. Preincubation with 5 mm GSH also significantly reduced MDA formation. The involvement of NOS activation in lipid A-induced lipid peroxidation was established when an NOS inhibitor and an inhibitor of intracellular calcium release completely blocked MDA formation. In addition, superoxide generation was significantly increased in the presence of lipid A, and the involvement of superoxide was established when superoxide dismutase protected against oxidant injury. The iron chelators deferoxamine (also a scavenger of peroxynitrite) and diethylenetriaminepentaacetic acid prevented lipid A-induced lipid peroxidation and cell death, indicating a role for iron and peroxynitrite. The addition of an NO scavenger, 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-3-oxide-1-oxyl, prior to lipid A also completely protected tubule cells from lipid peroxidation and subsequent cell death. These results indicate that lipid A-stimulated NO generation in the rat proximal tubule initiates oxidant injury.


Asunto(s)
Enfermedades Renales/inducido químicamente , Túbulos Renales Proximales/fisiopatología , Lípido A/toxicidad , Óxido Nítrico/biosíntesis , Animales , Antioxidantes/farmacología , Calcio/metabolismo , Quelantes/farmacología , Inducción Enzimática , Hierro , Túbulos Renales Proximales/efectos de los fármacos , Peróxidos Lipídicos/metabolismo , Masculino , Óxido Nítrico Sintasa/biosíntesis , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Superóxidos/metabolismo
16.
Nitric Oxide ; 1(5): 432-8, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9441914

RESUMEN

Lipid A, the biologically active component of lipopolysaccharide, initiates a specific cytotoxic signaling cascade in the renal proximal tubule that involves a rapid release of intracellular calcium, the activation of nitric oxide synthase (NOS) and NO production. Superoxide (O2-) generation is also a component of this cascade and both NO and O2- are required for the development of oxidant stress and cytotoxicity. Here we examined whether NOS activity was responsible for O2- generation. In renal proximal tubules isolated from the rat, the NOS inhibitor N(G)-monomethyl-L-arginine (L-NMMA) but not D-NMMA blocked lipid A (50 microg/ml)-stimulated O2- generation as measured by the reduction of cytochrome c during a 30-min incubation period. When L-arginine (2 mM) was added to the tubule suspensions, O2- generation was significantly inhibited, while NO2- (a marker of NO generation) was significantly increased. The addition of L-arginine also reduced lipid A-stimulated malondialdehyde formation at 30 min (a marker of lipid peroxidation) and lactate dehydrogenase release at 90 min (a marker of cell death). Thus, lipid A-stimulated the generation of both NO and O2- via NOS activation. Furthermore, increasing L-arginine availability shifted NOS activity toward NO generation and reduced oxidant injury. These results offer an explanation of why scavengers of NO or oxygen radicals ameliorate endotoxin-induced acute renal failure in vivo.


Asunto(s)
Túbulos Renales Proximales/metabolismo , Lípido A/farmacología , Óxido Nítrico Sintasa/metabolismo , Superóxidos/metabolismo , omega-N-Metilarginina/farmacología , Análisis de Varianza , Animales , Arginina/farmacología , Técnicas In Vitro , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , L-Lactato Deshidrogenasa/análisis , Peroxidación de Lípido/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Ratas , Ratas Sprague-Dawley , Estereoisomerismo
17.
J Pharmacol Exp Ther ; 279(1): 91-6, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8858980

RESUMEN

Lipid A, the biologically active component of lipopolysaccharide, stimulated nitric oxide (NO) production by isolated rat proximal tubules (as measured by NO2- release) in a time-dependent manner. At a concentration of 50 micrograms/ml, lipid A stimulated NO2- generation and guanosine 3',5'-cyclic phosphate (cGMP) production within 5 min. Both of these effects were blocked by NG-methyl-L-arginine (L-NMMA), an inhibitor of NO synthase or by 8-(N,N'-diethylamino)-octyl-3,4,5-trimethoxybenzoate (TMB-8), an inhibitor of intracellular Ca++ release. Because an increase in NO production may be cytotoxic, we examined the cytotoxic potential of lipid A. At 90 min, lipid A (50 micrograms/ml) produced significant lactate dehydrogenase release (42 +/- 5%) compared to control (25 +/- 5%; P < .05). Both L-NMMA (1 mM) and TMB-8 (100 microM) completely protected against lipid A-induced cytotoxicity. TMB-8 but not L-NMMA inhibited the rise intracellular Ca++ concentration ([Ca++]i) in isolated proximal tubules elicited by lipid A. L-NMMA but not TMB-8 inhibited proximal tubule soluble NO synthase activity. Thus, in the proximal tubule, lipid A stimulates a rise in [Ca++]i that in turn activates constitutive NO synthase. Furthermore, these events lead ultimately to NO-dependent cytotoxicity. Therefore, these findings suggest the potential for lipopolysaccharide to have a direct impact on proximal tubule physiology and renal function in vivo and support the potential therapeutic benefits of NO synthase inhibitors in the treatment of endotoxemia.


Asunto(s)
Túbulos Renales Proximales/metabolismo , Lípido A/toxicidad , Óxido Nítrico/fisiología , Animales , Calcio/metabolismo , GMP Cíclico/biosíntesis , Ácido Gálico/análogos & derivados , Ácido Gálico/farmacología , L-Lactato Deshidrogenasa/metabolismo , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Sprague-Dawley , omega-N-Metilarginina/farmacología
18.
J Pharmacol Exp Ther ; 276(2): 555-60, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8632321

RESUMEN

It is clear that lipopolysaccharides (LPS) are responsible for the multiorgan failure often associated with endotoxemia. However, little is known of the direct effects of LPS on kidney cells. We examined the effects of lipid A, the biologically active component of LPS, on rat proximal tubule Ca++ homeostasis. Lipid A produced a rapid, transient, concentration-dependent rise in intracellular Ca++ concentration, [Ca++]i, as monitored by fura-2. At 50 micrograms/ml [Ca++]i rose to 138 +/- 12 nM (n = 4) above basal [Ca++]i levels. The response to lipid A was not significantly inhibited by chelating extracellular Ca++ with EGTA (5 mM). However, the rise in [Ca++]i was significantly inhibited by 8-(N,N-dimethylamino)octyl-3,4,5-trimethoxybenzoate hydrochloride) and thapsigargin (17 +/- 7 nM and 13 +/- 9 nM rise, respectively; P < .05). These data indicate that the rise in [Ca++]i induced by lipid A is due to release of intracellular stores, and not extracellular influx. We also examined the role of inositol 1,4,5-trisphosphate in the lipid A response. Lipid A caused a time-dependent increase in inositol 1,4,5-trisphosphate that paralleled the rise in [Ca++]i, suggesting the release in [Ca++]i is through an inositol 1,4,5-trisphosphate-mediated release of intracellular stores. The ability of lipid A to alter Ca++ homeostasis suggests a potential for LPS to directly alter proximal tubule physiology and renal function in vivo.


Asunto(s)
Calcio/metabolismo , Homeostasis/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Lípido A/farmacología , Animales , Ácido Egtácico/farmacología , Ácido Gálico/análogos & derivados , Ácido Gálico/farmacología , Técnicas In Vitro , Túbulos Renales Proximales/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
19.
Biochem Pharmacol ; 49(1): 115-8, 1995 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-7530953

RESUMEN

Renal proximal tubules isolated from the rat possess nitric oxide synthase (NOS) activity that is calcium/calmodulin dependent and stereoselectively inhibited by NG-monomethyl-arginine (NMMA). In the absence of added Ca2+ and calmodulin, activity was reduced 84 +/- 13% compared with the activity in the presence of 2 mM Ca2+ and 25 micrograms/mL calmodulin. Inhibition by EGTA (10 mM) was 95 +/- 5% and by calmidazolium (R24571, 250 microM) was 99 +/- 1%. Inhibition by L-NMMA (100 microM) was 78 +/- 13% and by D-NMMA (100 microM) was 7 +/- 7%. The majority of NOS activity was found in the soluble fraction. NOS activity in isolated proximal tubules was also examined 6 hr after a single i.v. injection of lipopolysaccharide. Activity was increased significantly (P < 0.05) in the soluble fraction by 2-fold [from 0.320 +/- 0.052 to 0.648 +/- 0.046 (nmol/mg protein/30 min)] and in the particulate fraction by 3-fold [from 0.081 +/- 0.030 to 0.256 +/- 0.034 (nmol/mg protein/30 min)]. All activities were inhibited by EGTA. These data demonstrate that proximal tubules express a calcium/calmodulin-dependent NOS activity that is increased in vivo by lipopolysaccharide.


Asunto(s)
Aminoácido Oxidorreductasas/efectos de los fármacos , Aminoácido Oxidorreductasas/metabolismo , Túbulos Renales Proximales/enzimología , Lipopolisacáridos/farmacología , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Arginina/farmacología , Citrulina/análisis , Citrulina/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Cinética , Masculino , NADP/metabolismo , Óxido Nítrico Sintasa , Ratas , Ratas Sprague-Dawley , omega-N-Metilarginina
20.
J Pharmacol Exp Ther ; 266(1): 47-51, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8331573

RESUMEN

Acute renal failure is a frequent and serious complication of endotoxemia. However, the biochemical events that might ultimately lead to renal cell injury have not been examined previously. The renal tubular epithelial cell line LLC-PK1 was used to evaluate the role of intracellular-free calcium ([Ca++]i) in lipid A-induced cytotoxicity. Lipid A, considered to be the active component of bacterial endotoxin, produced a time- and concentration-dependent cytotoxicity as assessed by loss of trypan blue exclusion (38 +/- 3% cell deaths in 120 min with 50 micrograms/ml, n = 4). Chelation of [Ca++]i by quin 2 and inhibition of Ca++ release by 8-(N,N-dimethylamino)octyl-3,4,5-trimethoxybenzoate hydrochloride each protected against lipid A (50 micrograms/ml) cytotoxicity whereas removal of extracellular Ca++ was without effect. Fura-2 fluorescence was used to demonstrate that lipid A induces a rise in [Ca++]i in a concentration-dependent manner. Extracellular Ca++ was not required for the rise in [Ca++]i. Lipid A (50 micrograms/ml) produced a similar rise in [Ca++]i in the presence of 1 mM Ca++ (144 +/- 3 nM above basal) or in the absence of added Ca++ (155 +/- 28 nM, n = 4). The peak rise in [Ca++]i occurred rapidly (within 30 sec), clearly preceding cell death. The observations that lipid A induces a rise in [Ca++]i that precedes cell death, and that quin 2 and 8-(N,N-dimethylamino)octyl-3,4,5-trimethoxybenzoate hydrochloride protect against cell death suggest release of intracellular Ca++ mediates the cytotoxicity of lipid A in this renal tubular epithelial cell line.


Asunto(s)
Calcio/fisiología , Túbulos Renales/efectos de los fármacos , Lípido A/toxicidad , Aminoquinolinas/farmacología , Animales , Tampones (Química) , Calcio/metabolismo , Células Cultivadas/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Colorantes Fluorescentes/farmacología , Homeostasis/efectos de los fármacos , Líquido Intracelular/metabolismo , Túbulos Renales/citología , Túbulos Renales/metabolismo , Cinética , Modelos Biológicos , Porcinos , Azul de Tripano/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA