Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Toxicol Appl Pharmacol ; 275(3): 189-97, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24486436

ABSTRACT

Previously we reported a gene expression signature in rat liver for detecting a specific type of oxidative stress (OS) related to reactive metabolites (RM). High doses of the drugs disulfiram, ethinyl estradiol and nimesulide were used with another dozen paradigm OS/RM compounds, and three other drugs flutamide, phenacetin and sulindac were identified by this signature. In a second study, antiepileptic drugs were compared for covalent binding and their effects on OS/RM; felbamate, carbamazepine, and phenobarbital produced robust OS/RM gene expression. In the present study, liver RNA samples from drug-treated rats from more recent experiments were examined for statistical fit to the OS/RM signature. Of all 97 drugs examined, in addition to the nine drugs noted above, 19 more were identified as OS/RM-producing compounds-chlorpromazine, clozapine, cyproterone acetate, dantrolene, dipyridamole, glibenclamide, isoniazid, ketoconazole, methapyrilene, naltrexone, nifedipine, sulfamethoxazole, tamoxifen, coumarin, ritonavir, amitriptyline, valproic acid, enalapril, and chloramphenicol. Importantly, all of the OS/RM drugs listed above have been linked to idiosyncratic hepatotoxicity, excepting chloramphenicol, which does not have a package label for hepatotoxicity, but does have a black box warning for idiosyncratic bone marrow suppression. Most of these drugs are not acutely toxic in the rat. The OS/RM signature should be useful to avoid idiosyncratic hepatotoxicity of drug candidates.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Gene Expression Profiling , Liver/drug effects , Oxidative Stress/drug effects , Oxidative Stress/genetics , Toxicogenetics/methods , Animals , Databases, Genetic , Gene Expression Regulation/drug effects , Genetic Markers , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley
3.
Drug Metab Lett ; 2(1): 11-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-19356064

ABSTRACT

Membrane transporters are critical for the uptake as well as elimination of chemicals and by-products of metabolism from the liver and kidneys. Since these proteins are important determinants of chemical disposition, changes in their expression in different disease states can modulate drug pharmacokinetics. The present study investigated alterations in the renal and hepatic expression of organic anion and cation transporters (Oats/Octs), multidrug resistance-associated proteins (Mrps), breast cancer resistance protein (Bcrp), P-glycoprotein (Pgp), and hepatic Na(+)-taurocholate cotransporting polypeptide (Ntcp) in type 2 diabetic rats. For this purpose, type 2 diabetes was induced by feeding male Sprague-Dawley rats a high fat diet followed by a single dose of streptozotocin (45 mg/kg, i.p., in 0.01 M citrate buffer pH 4.3) on day 14. Controls received normal diet and vehicle. Kidney and liver samples were collected on day 24 for generation of crude plasma membrane fractions and Western blot analysis of Oat, Oct, Mrp, Bcrp, Pgp, and Ntcp proteins. With regards to renal uptake transporters, type 2 diabetes increased levels of Oat2 (2.3-fold) and decreased levels of Oct2 to 50% of control kidneys. Conversely, efflux transporters Mrp2, Mrp4, and Bcrp were increased 5.4-fold, 2-fold, and 1.6-fold, respectively in type 2 diabetic kidneys with no change in levels of Mrp1, Mrp5, or Pgp. Studies of hepatic transporters in type 2 diabetic rats reveal that the protein level of Mrp5 was reduced to 4% of control livers with no change in levels of Bcrp, Mrp1, Mrp2, Mrp4, Ntcp, or Pgp. The changes reported in this study may have implications in type 2 diabetic patients.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation , Membrane Transport Proteins/genetics , Animals , Blotting, Western , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Kidney/metabolism , Kidney/physiopathology , Liver/metabolism , Liver/physiopathology , Male , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , Rats , Rats, Sprague-Dawley , Streptozocin
4.
Toxicology ; 232(3): 200-15, 2007 Apr 11.
Article in English | MEDLINE | ID: mdl-17298859

ABSTRACT

Liver injury initiated by non-lethal doses of CCl(4) and thioacetamide (TA) progresses to hepatic failure and death of type 2 diabetic (DB) rats due to failed advance of liver cells from G(0)/G(1) to S-phase and inhibited tissue repair. Objective of the present study was to investigate cellular signaling mechanisms of failed cell division in DB rats upon hepatotoxicant challenge. In CCl(4)-treated non-diabetic (non-DB) rats, increased IL-6 levels, sustained activation of extracellular regulated kinases 1/2 (ERK1/2) MAPK, and sustained phosphorylation of retinoblastoma protein (p-pRB) via cyclin D1/cyclin-dependent kinase (cdk) 4 and cyclin D1/cdk6 complexes stimulated G(0)/G(1) to S-phase transition of liver cells. In contrast to the non-DB rats, CCl(4) administration led to lower plasma IL-6, decreased ERK1/2 activation, lower cyclin D1, and cdk 4/6 expression resulting in decreased p-pRB and inhibition of liver cell division in the DB rats. Furthermore, higher TGFbeta1 expression and p21 activation may also contribute to decreased p-pRB in DB rats compared to non-DB rats. Similarly, after TA administration to DB rats, down-regulation of cyclin D1 and p-pRB leads to markedly decreased advance of liver cells from G(0)/G(1) to S-phase and tissue repair compared to the non-DB rats. Hepatic ATP levels did not differ between the DB and non-DB rats obviating its role in failed tissue repair in the DB rats. In conclusion, decreased p-pRB may contribute to blocked advance of cells from G(0)/G(1) to S-phase and failed cell division in DB rats exposed to CCl(4) or TA, leading to progression of liver injury and hepatic failure.


Subject(s)
Carbon Tetrachloride Poisoning/pathology , Chemical and Drug Induced Liver Injury , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Liver Diseases/pathology , Thioacetamide/toxicity , Adenosine Triphosphate/metabolism , Animals , Carbon Tetrachloride Poisoning/enzymology , Carbon Tetrachloride Poisoning/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cyclin D1/biosynthesis , Cyclin-Dependent Kinases/biosynthesis , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Immunoblotting , Interleukin-6/blood , Liver Diseases/enzymology , Liver Diseases/metabolism , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Retinoblastoma Protein/metabolism , Transforming Growth Factor beta1/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Toxicol Appl Pharmacol ; 216(3): 416-25, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16926038

ABSTRACT

Heme oxygenase-1 (HO-1) is one of several enzymes induced by hepatotoxicants, and is thought to have an important protective role against cellular stress during liver inflammation and injury. The objective of the present study was to evaluate the role of HO-1 in estradiol-induced liver injury. A single dose of ethinyl estradiol (500 mg/kg, po) resulted in mild liver injury. Repeated administration of ethinyl estradiol (500 mg/kg/day for 4 days, po) resulted in no detectable liver injury or dysfunction. Using RT-PCR analysis, we demonstrate that HO-1 gene expression in whole liver tissue is elevated (>20-fold) after the single dose of ethinyl estradiol. The number and intensity of HO-1 immunoreactive macrophages were increased after the single dose of ethinyl estradiol. HO-1 expression was undetectable in hepatic parenchymal cells from rats receiving Methocel control or a single dose of ethinyl estradiol, however cytosolic HO-1 immunoreactivity in these cells after repeated dosing of ethinyl estradiol was pronounced. The increases in HO-1 mRNA and HO-1 immunoreactivity following administration of a single dose of ethinyl estradiol suggested that this enzyme might be responsible for the observed protection of the liver during repeated dosing. To investigate the effect of HO-1 expression on ethinyl estradiol-induced hepatotoxicity, rats were pretreated with hemin (50 micromol/kg, ip, a substrate and inducer of HO-1), with tin protoporphyrin IX (60 micromol/kg, ip, an HO-1 inhibitor), or with gadolinium chloride (10 mg/kg, iv, an inhibitor/toxin of Kupffer cells) 24 h before ethinyl estradiol treatment. Pretreatment with modulators of HO-1 expression and activity had generally minimal effects on ethinyl estradiol-induced liver injury. These data suggest that HO-1 plays a limited role in antioxidant defense against ethinyl estradiol-induced oxidative stress and hepatotoxicity, and suggests that other coordinately induced enzymes are responsible for protection observed with repeated administration of high doses of this compound.


Subject(s)
Antioxidants/metabolism , Estrogens/pharmacology , Ethinyl Estradiol/pharmacology , Heme Oxygenase-1/biosynthesis , Liver/enzymology , Animals , Biomarkers , Enzyme Induction/drug effects , Female , Gadolinium/pharmacology , Gene Expression/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Hemin/pharmacology , Immunohistochemistry , Liver/drug effects , Macrophages/drug effects , Metalloporphyrins/pharmacology , Protoporphyrins/pharmacology , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Response Elements , Reverse Transcriptase Polymerase Chain Reaction
6.
Hepatology ; 44(2): 379-88, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16871587

ABSTRACT

Acute liver failure induced by hepatotoxic drugs results from rapid progression of injury. Substantial research has shown that timely liver regeneration can prevent progression of injury leading to a favorable prognosis. However, the mechanism by which compensatory regeneration prevents progression of injury is not known. We have recently reported that calpain released from necrotic hepatocytes mediates progression of liver injury even after the hepatotoxic drug is cleared from the body. By examining expression of calpastatin (CAST), an endogenous inhibitor of calpain in three liver cell division models known to be resistant to hepatotoxicity, we tested the hypothesis that increased CAST in the dividing hepatocytes affords resistance against progression of injury. Liver regeneration that follows CCl(4)-induced liver injury, 70% partial hepatectomy, and postnatal liver development were used. In all three models, CAST was upregulated in the dividing/newly divided hepatocytes and declined to normal levels with the cessation of cell proliferation. To test whether CAST overexpression confers resistance against hepatotoxicity, CAST was overexpressed in the livers of normal SW mice using adenovirus before challenging them with acetaminophen (APAP) overdose. These mice exhibited markedly attenuated progression of liver injury and 57% survival. Whereas APAP-bioactivating enzymes and covalent binding of the APAP-derived reactive metabolites remained unaffected, degradation of calpain specific target substrates such as fodrin was significantly reduced in these mice. In conclusion, CAST overexpression could be used as a therapeutic strategy to prevent progression of liver injury where liver regeneration is severely hampered.


Subject(s)
Calcium-Binding Proteins/metabolism , Liver Failure, Acute/metabolism , Liver Regeneration , Liver/metabolism , Acetaminophen/toxicity , Animals , Animals, Newborn , Calcium-Binding Proteins/genetics , Calpain/antagonists & inhibitors , Carbon Tetrachloride/toxicity , Cell Division , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2E1/metabolism , Disease Models, Animal , Disease Progression , Hepatocytes/metabolism , Hepatocytes/pathology , Immunohistochemistry , Liver/growth & development , Liver Failure, Acute/chemically induced , Liver Failure, Acute/pathology , Male , Mice , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Up-Regulation
7.
Toxicol Appl Pharmacol ; 215(2): 146-57, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16546232

ABSTRACT

Previously we have shown that 90% of streptozotocin (STZ)-induced type-1 diabetic (DB) mice survive from acute renal failure (ARF) and death induced by a normally LD(90) dose (75 mg/kg, i.p.) of the nephrotoxicant S-1,2-dichlorovinyl-l-cysteine (DCVC). This remarkable protection is due to a combination of slower progression of DCVC-initiated renal injury and increased compensatory nephrogenic tissue repair in the DB kidneys. BRDU immunohistochemistry revealed that the DB condition led to 4-fold higher number of proximal tubular cells (PTC) entering S-phase of cell cycle. In the present study, we tested the hypothesis that DB-induced augmentation of PTC into S-phase is accompanied by overexpression of the calpain-inhibitor calpastatin, which endogenously prevents the progression of DCVC-initiated renal injury mediated by the calpain escaping out of damaged PTCs. Immunohistochemical detection of renal calpain and its activity in the urine, over a time course after treatment with the LD(90) dose of DCVC, indicated progressive increase in leakage of calpain into the extracellular spaces of the injured PTCs of the non-diabetic (NDB) kidneys as compared to the DB kidneys. Calpastatin expression was minimally detected in the NDB kidneys, using immunohistochemistry, over the time course. On the other hand, consistently higher number of tubules in the DB kidney showed calpastatin expression over the time course. The lower leakage of calpain in the DB kidneys was commensurate with constitutively higher expression of calpastatin in the S-phase-laden PTCs of these mice. To test the protective role of newly divided/dividing PTCs, DB mice were given the anti-mitotic agent colchicine (CLC) (2 mg/kg and 1.5 mg/kg, i.p., on days 8 and 10 after STZ injection) prior to challenge with a LD(90) dose of DCVC, which led to 100% mortality by 48 h. Mortality was due to rapid progression of DCVC-initiated renal injury, suggesting that newly divided/dividing cells are instrumental in mitigating the progression of DCVC-initiated renal injury in DB. The anti-mitotic effect of CLC in DB kidney is associated with lower expression of calpastatin and higher leakage of calpain in the injured tubules. These findings suggest that constitutively higher cell division in the DB kidney is associated with overexpression of calpastatin, which reduces the progression of DCVC-initiated renal injury mediated by calpain on the one hand and accelerates nephrogenic tissue repair on the other, thereby restoring renal structure and function.


Subject(s)
Acetylcysteine/analogs & derivatives , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Calcium-Binding Proteins/metabolism , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/metabolism , Diabetes Mellitus, Experimental/metabolism , Acetylcysteine/toxicity , Acute Kidney Injury/pathology , Animals , Antimitotic Agents/pharmacology , Calpain/urine , Colchicine/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Drug Therapy, Combination , Kidney Function Tests , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Longevity/drug effects , Male , Mice , Regeneration/drug effects
8.
Toxicol Appl Pharmacol ; 211(2): 133-47, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16125744

ABSTRACT

Streptozotocin (STZ)-induced diabetic (DB) rats are protected from nephrotoxicity of gentamicin, cisplatin and mercuric chloride, although the mechanisms remain unclear. Ninety percent of DB mice receiving a LD90 dose (75 mg/kg, ip) of S-1,2-dichlorovinyl-l-cysteine (DCVC) survived in contrast to only 10% of the nondiabetic (NDB) mice surviving the same dose. We tested the hypothesis that the mechanism of protection is upregulated tissue repair. In the NDB mice, DCVC produced steep temporal increases in blood urea nitrogen (BUN) and plasma creatinine, which were associated with proximal tubular cell (PTC) necrosis, acute renal failure (ARF), and death within 48 h. In contrast, in the DB mice, BUN and creatinine increased less steeply, declining after 36 h to completely resolve by 96 h. HPLC analysis of plasma and urine revealed that DB did not alter the toxicokinetics of DCVC. Furthermore, activity of renal cysteine conjugate beta-lyase, the enzyme that bio-activates DCVC, was unaltered in DB mice, undermining the possibility of lower bioactivation of DCVC leading to lower injury. [3H]-thymidine pulse labeling and PCNA analysis indicated an early onset and sustained nephrogenic tissue repair in DCVC-treated DB mice. BRDU immunohistochemistry revealed a fourfold increase in the number of cells in S-phase in the DB kidneys even without exposure to DCVC. Blocking the entry of cells into S-phase by antimitotic intervention using colchicine abolished stimulated nephrogenic tissue repair and nephro-protection. These findings suggest that pre-placement of S-phase cells in the kidney due to diabetes is critical in mitigating the progression of DCVC-initiated renal injury by upregulation of tissue repair, leading to survival of the DB mice by avoiding acute renal failure.


Subject(s)
Cysteine/analogs & derivatives , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Animals , Area Under Curve , Blood Urea Nitrogen , Bromodeoxyuridine/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Colchicine/pharmacology , Creatinine/blood , Cysteine/blood , Cysteine/pharmacokinetics , Cysteine/toxicity , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/mortality , Dose-Response Relationship, Drug , Half-Life , Immunohistochemistry , Injections, Intraperitoneal , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Lyases/metabolism , Male , Mice , Proliferating Cell Nuclear Antigen/analysis , Regeneration/drug effects , S Phase/drug effects , Streptozocin , Thymidine/metabolism , Tritium
9.
J Pharmacol Exp Ther ; 316(2): 507-19, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16207833

ABSTRACT

Type 2 diabetic (DB) mice exposed to CCl(4) (LD(50) = 1.25 ml/kg), acetaminophen (LD(80) = 600 mg/kg; APAP), and bromobenzene (LD(80) = 0.5 ml/kg) i.p. yielded 30, 20, and 20% mortality, respectively, indicating hepatotoxic resistance. Male Swiss-Webster mice were made diabetic by feeding high fat and administrating streptozotocin (120 mg/kg i.p.) on day 60. On day 71, time-course studies after APAP (600 mg/kg) treatment revealed identical initial liver injury in non-DB and DB mice, which progressed only in non-DB mice, resulting in 80% mortality. The hypothesis that decreased APAP bioactivation, altered toxicokinetics, and/or increased tissue repair are the underlying mechanisms was investigated. High-performance liquid chromatography analysis revealed no difference in plasma and urinary APAP or detoxification of APAP via glucuronidation between DB and non-DB mice. Hepatic CYP2E1 protein and activity, glutathione, and [(14)C]APAP covalent binding did not differ between DB and non-DB mice, suggesting that lower bioactivation-based injury is not the mechanism of decreased hepatotoxicity in DB mice. Diabetes increased cells in S phase by 8-fold in normally quiescent liver of these mice. Immunohistochemistry revealed overexpression of calpastatin in the newly dividing/divided cells, explaining inhibition of hydrolytic enzyme calpain in perinecrotic areas and lower progression of APAP-initiated injury in the DB mice. Antimitotic intervention of diabetes-associated cell division with colchicine before APAP administration resulted in 70% mortality in APAP-treated colchicine-intervened DB mice. These studies suggest that advancement of cells in the cell division cycle and higher tissue repair protect DB mice by preventing progression of APAP-initiated liver injury that normally leads to mortality.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/enzymology , Liver/drug effects , Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Animals , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2E1/biosynthesis , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Enzyme Induction , Glutathione/metabolism , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred Strains
10.
Toxicol Appl Pharmacol ; 211(3): 221-32, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16153671

ABSTRACT

Previously, we reported high hepatotoxic sensitivity of type 2 diabetic (DB) rats to three dissimilar hepatotoxicants. Additional work revealed that a normally nonlethal dose of CCl4 was lethal in DB rats due to inhibited compensatory tissue repair. The present study was conducted to investigate the importance of compensatory tissue repair in determining the final outcome of hepatotoxicity in diabetes, using another structurally and mechanistically dissimilar hepatotoxicant, thioacetamide (TA), to initiate liver injury. A normally nonlethal dose of TA (300 mg/kg, ip), caused 100% mortality in DB rats. Time course studies (0 to 96 h) showed that in the non-DB rats, liver injury initiated by TA as assessed by plasma alanine or aspartate aminotransferase and hepatic necrosis progressed up to 48 h and regressed to normal at 96 h resulting in 100% survival. In the DB rats, liver injury rapidly progressed resulting in progressively deteriorating liver due to rapidly expanding injury, hepatic failure, and 100% mortality between 24 and 48 h post-TA treatment. Covalent binding of 14C-TA-derived radiolabel to liver tissue did not differ from that observed in the non-DB rats, indicating similar bioactivation-based initiation of hepatotoxicity. S-phase DNA synthesis measured by [3H]-thymidine incorporation, and advancement of cells through the cell division cycle measured by PCNA immunohistochemistry, were substantially inhibited in the DB rats compared to the non-DB rats challenged with TA. Thus, inhibited cell division and compromised tissue repair in the DB rats resulted in progressive expansion of liver injury culminating in mortality. In conclusion, it appears that similar to type 1 diabetes, type 2 diabetes also increases sensitivity to dissimilar hepatotoxicants due to inhibited compensatory tissue repair, suggesting that sensitivity to hepatotoxicity in diabetes occurs in the absence as well as presence of insulin.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Thioacetamide/toxicity , Animals , Chemical and Drug Induced Liver Injury/pathology , Disease Susceptibility , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley
11.
J Pharmacol Exp Ther ; 308(2): 694-704, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14610242

ABSTRACT

There is a need for well characterized and economical type 2 diabetic model that mimics the human disease. We have developed a type 2 diabetes rat model that closely resembles the diabetic patients and takes only 24 days to develop robust diabetes. Nonlethal doses of allyl alcohol (35 mg/kg i.p.), CCl(4) (2 ml/kg i.p.), or thioacetamide (300 mg/kg i.p.) yielded 80 to 100% mortality in diabetic rats. The objective of the present study was to investigate two hypotheses: higher CCl(4) bioactivation and/or inhibited compensatory tissue repair were the underlying mechanisms for increased CCl(4) hepatotoxicity in diabetic rats. Diabetes was induced by feeding high fat diet followed by a single dose of streptozotocin on day 14 (45 mg/kg i.p.) and was confirmed on day 24 by hyperglycemia, normoinsulinemia, and oral glucose intolerance. Time course studies (0-96 h) of CCl(4) (2 ml/kg i.p.) indicated that although initial liver injury was the same in nondiabetic and diabetic rats, it progressed only in the latter, culminating in hepatic failure, and death. Hepatomicrosomal CYP2E1 protein and activity, lipid peroxidation, glutathione, and (14)CCl(4) covalent binding to liver tissue were the same in both groups, suggesting that higher bioactivation-based injury is not the mechanism. Inhibited tissue repair resulted in progression of injury and death in diabetic rats, whereas in the nondiabetic rats robust tissue repair resulted in regression of injury and survival after CCl(4) administration. These studies show high sensitivity of type 2 diabetes to model hepatotoxicants and suggest that CCl(4) hepatotoxicity is potentiated due to inhibited tissue repair.


Subject(s)
Carbon Tetrachloride Poisoning , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/pathology , Diabetes Mellitus, Type 2/pathology , Animals , Chemical and Drug Induced Liver Injury/mortality , Cytochrome P-450 CYP2E1/metabolism , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/mortality , Disease Models, Animal , Glutathione/metabolism , Lipid Peroxidation/physiology , Proliferating Cell Nuclear Antigen/analysis , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...