Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Toxicol Sci ; 145(1): 108-17, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25666834

RESUMO

Although the importance of mitochondrial dysfunction in acute kidney injury (AKI) has been documented, noninvasive early biomarkers of mitochondrial damage are needed. We examined urinary ATP synthase subunit ß (ATPSß) as a biomarker of renal mitochondrial dysfunction during AKI. Mice underwent sham surgery or varying degrees (5, 10, or 15 min ischemia) of ischemia/reperfusion (I/R)-induced AKI. Serum creatinine, BUN, and neutrophil gelatinase-associated lipocalin were elevated only in the 15 min I/R group at 24 h. Immunoblot analysis of urinary ATPSß revealed two bands (full length ∼52 kDa and cleaved ∼25 kDa), both confirmed as ATPSß by LC-MS/MS, that increased at 24 h in 10- and 15-min I/R groups. These changes were associated with mitochondrial dysfunction evidenced by reduced renal cortical expression of mitochondrial proteins, ATPSß and COX1, proximal tubular oxygen consumption, and ATP. Furthermore, in the 15-min I/R group, urinary ATPSß was elevated until 72 h before returning to baseline 144 h after reperfusion with recovery of renal function. Evaluation of urinary ATPSß in a nonalcoholic steatohepatitis model of liver injury only revealed cleaved ATPSß, suggesting specificity of full-length ATPSß for renal injury. Immunoblot analyses of patient urine samples collected 36 h after cardiac surgery revealed increased urinary ATPSß levels in patients with postcardiac surgery-induced AKI. LC-MS/MS urinalysis in human subjects with AKI confirmed increased ATPSß. These translational studies provide evidence that ATPSß may be a novel and sensitive urinary biomarker of renal mitochondrial dysfunction and could serve as valuable tool for the testing of potential therapies for AKI and chemical-induced nephrotoxicity.


Assuntos
Injúria Renal Aguda/enzimologia , Biomarcadores/urina , Mitocôndrias/fisiologia , ATPases Mitocondriais Próton-Translocadoras/urina , Injúria Renal Aguda/fisiopatologia , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL
2.
J Pharmacol Exp Ther ; 351(3): 663-73, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25292181

RESUMO

Skeletal muscle atrophy remains a clinical problem in numerous pathologic conditions. ß2-Adrenergic receptor agonists, such as formoterol, can induce mitochondrial biogenesis (MB) to prevent such atrophy. Additionally, atomoxetine, an FDA-approved norepinephrine reuptake inhibitor, was positive in a cellular assay for MB. We used a mouse model of dexamethasone-induced skeletal muscle atrophy to investigate the potential role of atomoxetine and formoterol to prevent muscle mass loss. Mice were administered dexamethasone once daily in the presence or absence of formoterol (0.3 mg/kg), atomoxetine (0.1 mg/kg), or sterile saline. Animals were euthanized at 8, 16, and 24 hours or 8 days later. Gastrocnemius muscle weights, changes in mRNA and protein expression of peroxisome proliferator-activated receptor-γ coactivator-1 α (PGC-1α) isoforms, ATP synthase ß, cytochrome c oxidase subunit I, NADH dehydrogenase (ubiquinone) 1 ß subcomplex, 8, ND1, insulin-like growth factor 1 (IGF-1), myostatin, muscle Ring-finger protein-1 (muscle atrophy), phosphorylated forkhead box protein O 3a (p-FoxO3a), Akt, mammalian target of rapamycin (mTOR), and ribosomal protein S6 (rp-S6; muscle hypertrophy) in naive and muscle-atrophied mice were measured. Atomoxetine increased p-mTOR 24 hours after treatment in naïve mice, but did not change any other biomarkers. Formoterol robustly activated the PGC-1α-4-IGF1-Akt-mTOR-rp-S6 pathway and increased p-FoxO3a as early as 8 hours and repressed myostatin at 16 hours. In contrast to what was observed with acute treatment, chronic treatment (7 days) with atomoxetine increased p-Akt and p-FoxO3a, and sustained PGC-1α expression and skeletal muscle mass in dexamethasone-treated mice, in a manner comparable to formoterol. In conclusion, chronic treatment with a low dose of atomoxetine prevented dexamethasone-induced skeletal muscle wasting and supports a potential role in preventing muscle atrophy.


Assuntos
Dexametasona/toxicidade , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/prevenção & controle , Propilaminas/uso terapêutico , Animais , Cloridrato de Atomoxetina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Propilaminas/farmacologia
3.
Hepatology ; 60(5): 1741-52, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24723460

RESUMO

UNLABELLED: Liver eosinophilia has been associated with incidences of drug-induced liver injury (DILI) for more than 50 years, although its role in this disease has remained largely unknown. In this regard, it was recently shown that eosinophils played a pathogenic role in a mouse model of halothane-induced liver injury (HILI). However, the signaling events that drove hepatic expression of eosinophil-associated chemokines, eotaxins, eosinophil infiltration, and subsequent HILI were unclear. We now provide evidence implicating hepatic epithelial-derived cytokine thymic stromal lymphopoietin (TSLP) and type 2 immunity, in particular, interleukin-4 (IL-4) production, in mediating hepatic eosinophilia and injury during HILI. TSLP was constitutively expressed by mouse hepatocytes and increased during HILI. Moreover, the severity of HILI was reduced in mice deficient in either the TSLP receptor (TSLPR) or IL-4 and was accompanied by decreases in serum levels of eotaxins and hepatic eosinophilia. Similarly, concanavalin A-induced liver injury, where type 2 cytokines and eosinophils play a significant role in its pathogenesis, was also reduced in TSLPR-deficient mice. Studies in vitro revealed that mouse and human hepatocytes produce TSLP and eotaxins in response to treatment with combinations of IL-4 and proinflammatory cytokines IL-1ß and tumor necrosis factor alpha. CONCLUSION: This report provides the first evidence implicating roles for hepatic TSLP signaling, type 2 immunity, and eosinophilia in mediating liver injury caused by a drug.


Assuntos
Anestésicos Inalatórios/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Citocinas/metabolismo , Halotano/efeitos adversos , Interleucina-4/metabolismo , Animais , Concanavalina A , Feminino , Hepatite Animal/metabolismo , Hepatócitos/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Linfopoietina do Estroma do Timo
4.
PLoS One ; 8(9): e73655, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24040012

RESUMO

OBJECTIVE: To determine whether delayed administration of a single dose of suramin, a drug that has been used extensively in humans to treat trypanosomiasis, attenuates renal injury in a leptin receptor deficient C57BLKS/J db/db type 2 diabetic nephropathy (T2DN) mouse model. RESEARCH DESIGN AND METHODS: Groups of female non-diabetic (control) db/m and diabetic db/db mice of 8 and 16 weeks of age, respectively, were treated with suramin (10 mg/kg) or saline i.v. All animals were euthanized one week later. Measurements in mice 1 week following treatment included the following: body weight; blood glucose; urinary protein excretion; pathological lesions in glomeruli and proximal tubules; changes in protein expression of pro-inflammatory transcription factor nuclear factor κB (NF-κB) and intracellular adhesion molecule-1 (ICAM-1), profibrotic transforming growth factor-ß1 (TGF-ß1), phospho-SMAD-3 and alpha-smooth muscle actin (α-SMA); and immunohistochemical analysis of leukocyte infiltration and collagen 1A2 (COL1A2) deposition. RESULTS: Immunoblot analysis revealed increased NF-κB, ICAM-1, TGF-ß1, phospho-SMAD-3, and α-SMA proteins in both 9 and 17 week db/db mice as compared to db/m control mice. Immunohistochemical analysis revealed moderate leukocyte infiltration and collagen 1A2 (COL1A2) deposition in 9 week db/db mice that was increased in the 17 week db/db mice. Importantly, suramin significantly decreased expression of all these markers in 9 week db/db mice and partially decreased in 17 week db/db mice without altering body weight, blood glucose or urinary protein excretion. There was no difference in creatinine clearance between 9 week db/m and db/db mice ± suramin. Importantly, in the 17 week db/db mice suramin intervention reversed the impaired creatinine clearance and overt histological damage. CONCLUSIONS: Delayed administration of a single dose of suramin in a model of T2DN attenuated inflammation and fibrosis as well as improved renal function, supporting the use of suramin in T2DN.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Nefropatias Diabéticas/tratamento farmacológico , Suramina/farmacologia , Actinas/metabolismo , Animais , Colágeno Tipo I/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/genética , Modelos Animais de Doenças , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Molécula 1 de Adesão Intercelular/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/química , NF-kappa B/metabolismo , Receptores para Leptina/deficiência , Receptores para Leptina/genética , Proteína Smad3 , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Tripanossomicidas/farmacologia
5.
J Pharmacol Exp Ther ; 343(1): 34-43, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22736507

RESUMO

Progression of hyperglycemia-induced renal injury is a contributing factor for diabetic nephropathy (DN)-induced end-stage renal disease (ESRD), and development of novel therapeutic strategies that act early to prevent progression of DN and ESRD are important. We examined the efficacy and mechanism(s) of suramin on hyperglycemia-induced renal injury before development of overt histological damage. Two groups of male Sprague-Dawley rats received streptozotocin (STZ) and one group received saline. Three weeks later, one STZ group received suramin (10 mg/kg). All animals were euthanized 1 week later (4 weeks). Although there was a decrease in creatinine clearance between control and STZ ± suramin rats, there was no difference in creatinine clearance between STZ rats ± suramin intervention. Liquid chromatography-tandem mass spectroscopy-based analysis revealed increases in urinary proteins that are early indicators of DN (e.g., cystatin C, clusterin, cathepsin B, retinol binding protein 4, and peroxiredoxin-1) in the STZ group, which were blocked by suramin. Endothelial intracellular adhesion molecule-1 (ICAM-1) activation, leukocyte infiltration, and inflammation; transforming growth factor-ß1 (TGF-ß1) signaling; TGF-ß1/SMAD-3-activated fibrogenic markers fibronectin-1, α-smooth muscle actin, and collagen 1A2; activation of proinflammatory and profibrotic transcription factors nuclear factor-κB (NF-κB) and signal transducer and activator of transcription factor-3 (STAT-3), respectively, were all increased in STZ rats and suramin blocked these changes. In conclusion, delayed administration of suramin attenuated 1) urinary markers of DN, 2) inflammation by blocking NF-κB activation and ICAM-1-mediated leukocyte infiltration, and 3) fibrosis by blocking STAT-3 and TGF-ß1/SMAD-3 signaling. These results support the potential use of suramin in DN.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/urina , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/urina , Suramina/uso terapêutico , Animais , Biomarcadores/urina , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/etiologia , Molécula 1 de Adesão Intercelular/urina , Falência Renal Crônica/tratamento farmacológico , Falência Renal Crônica/etiologia , Falência Renal Crônica/urina , Masculino , NF-kappa B/urina , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/urina , Resultado do Tratamento
6.
J Pharmacol Exp Ther ; 341(1): 126-36, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22228809

RESUMO

Acute kidney injury (AKI) is a common and potentially life-threatening complication after ischemia/reperfusion and exposure to nephrotoxic agents. In this study, we examined the efficacy and mechanism(s) of suramin in promoting recovery from glycerol-induced AKI, a model of rhabdomyolysis-induced AKI. After intramuscular glycerol injection (10 ml of 50% glycerol per kilogram) into male Sprague-Dawley rats, serum creatinine maximally increased at 24 to 72 h and then decreased at 120 h. Creatinine clearance (CrCl) decreased 75% at 24 to 72 h and increased at 120 h. Suramin (1 mg/kg i.v.) administered 24 h after glycerol accelerated recovery of renal function as demonstrated by increased CrCl, decreased renal kidney injury molecule-1, and improved histopathology 72 h after glycerol injection. Suramin treatment decreased interleukin-1ß (IL-1ß) mRNA, transforming growth factor-ß(1) (TGF-ß(1)), phospho-p65 of nuclear factor-κB (NF-κB), and cleaved caspase-3 at 48 h compared with glycerol alone. Suramin treatment also decreased glycerol-induced activation of intracellular adhesion molecule-1 (ICAM-1) and leukocyte infiltration at 72 h. Urinary/renal neutrophil gelatinase-associated lipocalin 2 (NGAL) levels, hemeoxygenase-1 expression, and renal cell proliferation were increased by suramin compared with glycerol alone at 72 h. Mechanistically, suramin decreases early glycerol-induced proinflammatory (IL-1ß and NF-κB) and growth inhibitory (TGF-ß(1)) mediators, resulting in the prevention of late downstream inflammatory effects (ICAM-1 and leukocyte infiltration) and increasing compensatory nephrogenic repair. These results support the hypothesis that delayed administration of suramin is effective in abrogating apoptosis, attenuating inflammation, and enhancing nephrogenic repair after glycerol-induced AKI.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Glicerol/toxicidade , Recuperação de Função Fisiológica/efeitos dos fármacos , Suramina/uso terapêutico , Injúria Renal Aguda/fisiopatologia , Animais , Mediadores da Inflamação/fisiologia , Mediadores da Inflamação/uso terapêutico , Masculino , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia
7.
Chem Res Toxicol ; 25(1): 83-93, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22107450

RESUMO

In a recent study, we reported that interleukin (IL)-4 had a protective role against acetaminophen (APAP)-induced liver injury (AILI), although the mechanism of protection was unclear. Here, we carried out more detailed investigations and have shown that one way IL-4 may control the severity of AILI is by regulating glutathione (GSH) synthesis. In the present studies, the protective role of IL-4 in AILI was established definitively by showing that C57BL/6J mice made deficient in IL-4 genetically (IL-4(-/-)) or by depletion with an antibody, were more susceptible to AILI than mice not depleted of IL-4. The increased susceptibility of IL-4(-/-) mice was not due to elevated levels of hepatic APAP-protein adducts but was associated with a prolonged reduction in hepatic GSH that was attributed to decreased gene expression of γ-glutamylcysteine ligase (γ-GCL). Moreover, administration of recombinant IL-4 to IL-4(-/-) mice postacetaminophen treatment diminished the severity of liver injury and increased γ-GCL and GSH levels. We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. Overall, these results show for the first time that IL-4 has a role in regulating the synthesis of GSH in the liver under conditions of cellular stress. This mechanism appears to be responsible at least in part for the protective role of IL-4 against AILI in mice and may have a similar role not only in AILI in humans but also in pathologies of the liver caused by other drugs and etiologies.


Assuntos
Acetaminofen/toxicidade , Analgésicos não Narcóticos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Glutationa/metabolismo , Interleucina-4/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocromo P-450 CYP2E1/metabolismo , Glutamato-Cisteína Ligase/metabolismo , Interleucina-4/deficiência , Interleucina-4/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/metabolismo
8.
Biochem Biophys Res Commun ; 374(1): 6-10, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18586006

RESUMO

Recent studies in mice suggest that stress-activated c-Jun N-terminal protein kinase 2 (JNK2) plays a pathologic role in acetaminophen (APAP)-induced liver injury (AILI), a major cause of acute liver failure (ALF). In contrast, we present evidence that JNK2 can have a protective role against AILI. When male C57BL/6J wild type (WT) and JNK2(-/-) mice were treated with 300mg APAP/kg, 90% of JNK2(-/-) mice died of ALF compared to 20% of WT mice within 48h. The high susceptibility of JNK2(-/-) mice to AILI appears to be due in part to deficiencies in hepatocyte proliferation and repair. Therefore, our findings are consistent with JNK2 signaling playing a protective role in AILI and further suggest that the use of JNK inhibitors as a potential treatment for AILI, as has been recommended by other investigators, should be reconsidered.


Assuntos
Acetaminofen/toxicidade , Analgésicos não Narcóticos/toxicidade , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/enzimologia , Fígado/efeitos dos fármacos , Proteína Quinase 9 Ativada por Mitógeno/fisiologia , Animais , Ciclina D , Ciclinas/metabolismo , Fígado/enzimologia , Fígado/patologia , Falência Hepática Aguda/genética , Regeneração Hepática/genética , Masculino , Camundongos , Camundongos Mutantes , Proteína Quinase 9 Ativada por Mitógeno/genética
9.
Am J Physiol Renal Physiol ; 293(4): F994-F1006, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17581926

RESUMO

Previous studies (Vaidya VS, Shankar K, Lock EA, Bucci TJ, Mehendale HM. Toxicol Sci 74: 215-227, 2003; Korrapati MC, Lock EA, Mehendale HM. Am J Physiol Renal Physiol 289: F175-F185, 2005; Korrapati MC, Chilakapati J, Lock EA, Latendresse JR, Warbritton A, Mehendale HM. Am J Physiol Renal Physiol 291: F439-F455, 2006) demonstrated that renal repair stimulated by a low dose of S-(1,2-dichlorovinyl)l-cysteine (DCVC; 15 mg/kg i.p.) 72 h before administration of a normally lethal dose (75 mg/kg i.p.) protects mice from acute renal failure (ARF) and death (autoprotection). The present study identified the proteins indicative of DCVC-induced ARF and autoprotection in male Swiss Webster mice. Renal dysfunction and injury were assessed by plasma creatinine and histopathology, respectively. Whole-kidney homogenates were run on two-dimensional gel electrophoresis gels, and the expression of 18 common proteins was maximally changed (> or =10-fold) in all the treatment groups and they were conclusively identified by liquid chromatography tandem mass spectrometry. These proteins were mildly downregulated after low dose alone and in autoprotected mice in contrast to severe downregulation with high dose alone. Glucose-regulated protein 75 and proteasome alpha-subunit type 1 were further investigated by immunohistochemistry for their localization in the kidneys of all the groups. These proteins were substantially higher in the proximal convoluted tubular epithelial cells in the low-dose and autoprotected groups compared with high-dose alone group. Proteins involved in energetics were downregulated in all the three groups of mice, leading to a compromise in cellular energy. However, energy is recovered completely in low-dose and autoprotected mice. This study provides the first report on proteomics of DCVC-induced ARF and autoprotection in mice and reflects the application of proteomics in mechanistic studies as well as biomarker development in a variety of toxicological paradigms.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Cisteína/análogos & derivados , Rim/metabolismo , Proteômica , Injúria Renal Aguda/mortalidade , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/metabolismo , Coenzima A Ligases/metabolismo , Creatinina/sangue , Cisteína/efeitos adversos , Cisteína/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Proteínas de Transporte de Ácido Graxo/metabolismo , Sequestradores de Radicais Livres/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/metabolismo , Hemopexina/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos
10.
Toxicol Appl Pharmacol ; 219(1): 72-84, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234228

RESUMO

Thioacetamide (TA) undergoes saturation toxicokinetics in ad libitum (AL) fed rats. Diet restriction (DR) protects rats from lethal dose of TA despite increased bioactivation-mediated liver injury via CYP2E1 induction. While a low dose (50 mg TA/kg) produces 6-fold higher initial injury, a 12-fold higher dose produces delayed and mere 2.5-fold higher injury. The primary objective was to determine if this less-than-expected increase in injury is due to saturation toxicokinetics. Rats on AL and DR for 21 days received either 50 or 600 mg TA/kg i.p. T(1/2) and AUCs for TA and TA-S-oxide were consistent with saturable kinetics. Covalent binding of (14)C-TA-derived-radiolabel to liver macromolecules after low dose was 2-fold higher in DR than AL rats. However, following lethal dose, no differences were found between AL and DR. This lack of dose-dependent response appears to be due to saturation of bioactivation at the higher dose. The second objective was to investigate the effect of phenobarbital pretreatment (PB) on TA-initiated injury following a sub-lethal dose (500 mg/kg). PB induced CYP2B1/2 approximately 350-fold, but did not increase covalent binding of (14)C-TA, TA-induced liver injury and mortality, suggesting that CYP2B1/2 has no major role in TA bioactivation. The third objective was to investigate the role of CYP2E1 using cyp2e1 knockout mice (KO). Injury was assessed over time (0-48 h) in wild type (WT) and KO mice after LD(100) dose (500 mg/kg) in WT. While WT mice exhibited robust injury which progressed to death, KO mice exhibited neither initiation nor progression of injury. These findings confirm that CYP2E1 is responsible for TA bioactivation.


Assuntos
Restrição Calórica , Carcinógenos/farmacocinética , Citocromo P-450 CYP2E1/metabolismo , Fenobarbital/farmacologia , Tioacetamida/farmacocinética , Alanina Transaminase/sangue , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Aspartato Aminotransferases/sangue , Biotransformação/efeitos dos fármacos , Carcinógenos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocromo P-450 CYP2B1/metabolismo , Citocromo P-450 CYP2E1/genética , DNA Complementar/biossíntese , DNA Complementar/genética , Meia-Vida , Cinética , Masculino , Camundongos , Camundongos Knockout , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Ratos , Ratos Sprague-Dawley , Esteroide Hidroxilases/metabolismo , Sulfóxidos/metabolismo , Tioacetamida/toxicidade
11.
Toxicology ; 230(2-3): 105-16, 2007 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-17187915

RESUMO

Thioacetamide (TA) is bioactivated by CYP2E1 to TA sulfoxide (TASO), and to the highly reactive sulfdioxide (TASO(2)), which initiates hepatic necrosis by covalent binding. Previously, we have established that TA exhibits saturation toxicokinetics over a 12-fold dose range, which explains the lack of dose-response for bioactivation-based liver injury. In vivo and in vitro studies indicated that the second step (TASO-->TASO(2)) of TA bioactivation is less efficient than the first one (TA-->TASO). The objective of the present study was to specifically test the saturation of the second step of TA bioactivation by directly administering TASO, which obviates the contribution from first step, i.e. TA-->TASO. Male SD rats were injected with low (50mg/kg, ip), medium (100mg/kg) and high (LD(70), 200mg/kg) doses of TASO. Bioactivation-mediated liver injury that occurs in the initial time points (6 and 12h), estimated by plasma ALT, AST and liver histopathology over a time course, was not dose-proportional. Escalation of liver injury thereafter was dose dependent: low dose injury subsided; medium dose injury escalated upto 36h before declining; high dose injury escalated from 24h leading to 70% mortality. TASO was quantified in plasma by HPLC at various time points after administration of the three doses. With increasing dose (i.e., from 50 to 200mg/kg), area under the curve (AUC) and C(max) increased more than dose proportionately, indicating that TASO bioactivation exhibits saturable kinetics. Toxicokinetics and initiation of liver injury of TASO are similar to that of TA, although TASO-initiated injury occurs at lower doses. These findings indicate that bioactivation of TASO to its reactive metabolite is saturable in the rat as suggested by previous studies with TA.


Assuntos
Fígado/efeitos dos fármacos , Tioacetamida/análogos & derivados , Alanina Transaminase/sangue , Animais , Área Sob a Curva , Aspartato Aminotransferases/sangue , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Replicação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Histocitoquímica , Fígado/patologia , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Ratos , Ratos Sprague-Dawley , Tioacetamida/sangue , Tioacetamida/metabolismo , Tioacetamida/farmacocinética , Tioacetamida/toxicidade , Tioacetamida/urina , Timidina/metabolismo
12.
Am J Physiol Renal Physiol ; 291(2): F439-55, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16495211

RESUMO

Previous studies have shown that renal injury initiated by a lethal dose of S-1,2-dichlorovinyl-l-cysteine (DCVC) progresses due to inhibition of cell division and hence renal repair, leading to acute renal failure (ARF) and death in mice. Renal injury initiated by low to moderate doses of DCVC is repaired by timely and adequate stimulation of renal cell division, tubular repair, restoration of renal structure and function leading to survival of mice. Recent studies have established that mice primed with a low dose of DCVC (15 mg/kg i.p.) 72 h before administration of a normally lethal dose (75 mg/kg i.p.) are protected from ARF and death (nephro-autoprotection). We showed that renal cell division and tissue repair stimulated by the low dose are sustained even after the lethal dose administration resulting in survival from ARF and death. If renal cell division induced by the low dose is indeed the critical mechanism of this autoprotection, then its ablation by the antimitotic agent colchicine (1.5 mg CLC/kg i.p.) should abolish autoprotection. The present interventional experiments were designed to test the hypothesis that DCVC autoprotection is due to stimulated cell division and tissue repair by the priming low dose. CLC intervention at 42 and 66 h after the priming dose resulted in marked progressive elevation of plasma blood urea nitrogen and creatinine resulting in ARF and death of mice. Light microscopic examination of hematoxylin and eosin-stained kidney sections revealed progression of renal necrosis concordant with progressively failing renal function. With CLC intervention, S-phase stimulation (as assessed by BrdU pulse labeling), G(1)-to-S phase clearance, and cell division were diminished essentially abolishing the promitogenic effect of the priming low dose of DCVC. Phospho-retinoblastoma protein (P-pRB), a crucial protein for S-phase stimulation, and other cellular signaling mechanisms regulating P-pRB were investigated. We report that decreased P-pRB via activation of protein phosphatase-1 by CLC is the critical mechanism of this inhibited S-phase stimulation and ablation of autoprotection with CLC intervention. These findings lend additional support to the notion that stimulated cell division and renal tissue repair by the priming dose of DCVC are the critical mechanisms that allow sustained compensatory tissue repair and survival of mice in nephro-autoprotection.


Assuntos
Acetilcisteína/análogos & derivados , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/fisiopatologia , Divisão Celular/fisiologia , Acetilcisteína/administração & dosagem , Acetilcisteína/toxicidade , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Animais , Antimitóticos/farmacologia , Nitrogênio da Ureia Sanguínea , Divisão Celular/efeitos dos fármacos , Colchicina/farmacologia , Creatinina/urina , Quinases Ciclina-Dependentes/análise , Quinases Ciclina-Dependentes/fisiologia , DNA/biossíntese , Relação Dose-Resposta a Droga , Fase G1/efeitos dos fármacos , Fase G1/fisiologia , Rim/química , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Masculino , Camundongos , Fosfoproteínas Fosfatases/análise , Fosfoproteínas Fosfatases/fisiologia , Proteína Fosfatase 1 , Fase S/efeitos dos fármacos , Fase S/fisiologia , Transdução de Sinais/fisiologia , Fatores de Tempo
13.
Drug Metab Dispos ; 33(12): 1877-85, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16183780

RESUMO

Thioacetamide (TA), a potent centrilobular hepatotoxicant, undergoes a two-step bioactivation mediated by microsomal CYP2E1 to TA sulfoxide (TASO), and further to TA-S,S-dioxide (TASO2), a reactive metabolite that initiates cellular necrosis. Our earlier studies showed that bioactivation-mediated liver injury of TA is not dose-proportional. The objective of this study was to examine whether increasing doses of TA lead to enzyme saturation, thereby resulting in lack of dose-response for injury: bioactivation of TA --> TASO --> TASO2 may follow zero-order kinetics. A 12-fold dose range of TA (50, 300, and 600 mg/kg i.p.) was injected into male Sprague-Dawley rats. TA and TASO were quantified in plasma, liver, and urine by high-performance liquid chromatography. With increasing doses, the apparent elimination half-lives of TA and TASO increased linearly, indicating that TA bioactivation exhibits saturation kinetics. Increasing TA dose resulted in greater-than-proportional increases in plasma TA and TASO levels. The TASO/TA ratio was inversely proportional to the dose of TA. Covalent binding of 14C-TA-derived radiolabel to liver macromolecules showed a less-than-dose-proportionate increase with a 12-fold higher dose. Less than dose-proportional covalent binding was confirmed in liver microsomal incubations with 14C-TA. Three-fold higher excretion of TASO was seen in urine at the highest dose (600 mg/kg) compared with the lowest dose (50 mg TA/kg). Incubation of TA with rat liver microsomes and purified baculovirus-expressed rat and human CYP2E1 Supersomes, over a concentration range of 0.01 to 10 mM, revealed saturation of TA conversion to TASO at and above 0.05 mM TA concentration, comparable to in vivo plasma and liver levels achieved upon administration of higher doses. Calculated K(m) values for TA (0.1 mM) and TASO (0.6 mM) suggest that the second step of TA bioactivation is 6-fold less efficient. Collectively, the findings indicate saturation of CYP2E1 at the first (TA to TASO) and second (TASO to TASO2) steps of TA bioactivation.


Assuntos
Fígado/efeitos dos fármacos , Tioacetamida/farmacocinética , Tioacetamida/toxicidade , Animais , Proteínas Sanguíneas/metabolismo , Citocromo P-450 CYP2E1/fisiologia , Rim/metabolismo , Masculino , Microssomos Hepáticos/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Sulfóxidos/metabolismo
14.
Am J Physiol Renal Physiol ; 289(1): F175-85, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15741605

RESUMO

Sustained activation of ERK 1/2 by a low dose (15 mg/kg ip) of S-1,2-dichlorovinyl-l-cysteine (DCVC) 72 h before administration of a lethal dose of DCVC (75 mg/kg ip) enhances renal cell division and protects mice against acute renal failure (ARF) and death (autoprotection). The objective of this study was to determine correlation among extent of S-phase DNA synthesis, activation of transcription factors, expression of G(1)/S cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors downstream of ERK 1/2 following DCVC-induced ARF in autoprotection. Administration of the lethal dose alone caused a general downregulation or an unsustainable increase, in transcriptional and posttranscriptional events thereby preventing G(1)-S transition of renal cell cycle. Phosphorylation of IkappaBalpha was inhibited resulting in limited nuclear translocation of NF-kappaB. However, cyclin D1 expression was high probably due to transcriptional cooperation of AP-1. Cyclin D1/cyclin-dependent kinase 4 (cdk4)-cdk6 system-mediated phosphorylation of retinoblastoma protein was downregulated due to overexpression of p16 at 24 h after exposure to the lethal dose alone. Inhibition of S-phase stimulation was confirmed by proliferating cell nuclear antigen assay (PCNA). This inhibitory response was prevented if the lethal dose was administered 72 h after the low priming dose of DCVC due to promitogenic effect of the low dose. NF-kappaB-DNA binding is not limited if mice were pretreated with the priming dose. Cyclin D1/cdk4-cdk6 expression stimulated by the priming dose of DCVC was unaltered even after the lethal dose in the autoprotected group, explaining higher phosphorylated-pRB and S-phase stimulation found in this group. These results were corroborated with PCNA immunohistochemistry. These findings suggest that the priming dose relieves the block on compensatory tissue repair by upregulation of promitogenic mechanisms, normally blocked by the high dose when administered without the prior priming dose.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/fisiopatologia , Cisteína/análogos & derivados , Cisteína/toxicidade , Fase G1/efeitos dos fármacos , Fase S/efeitos dos fármacos , Animais , Replicação do DNA , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Fase G1/fisiologia , Rim/fisiologia , Masculino , Camundongos , Fase S/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA