Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Pharm Biol ; 59(1): 986-997, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34347571

ABSTRACT

CONTEXT: The roots of Tagetes lucida Cav. (Asteraceae) have antioxidant and antimicrobial properties. OBJECTIVE: This study aimed to examine the hepatoprotective effects of T. lucida roots ethanol extract (TLRE) using carbon tetrachloride (CCl4)-induced hepatotoxicity in rats. MATERIALS AND METHODS: The active ingredients of TLRE were identified by high-performance liquid chromatography, infra-red spectrum, and mass spectrometric procedures. Ninety rats were distributed into four main groups: positive, therapeutic, protective, and negative group. The therapeutic group was implemented using CCl4 (a single dose of 2 mL/kg) before TLRE or silymarin administration. Meanwhile, the protective group was implemented by administering CCl4 (a single dose of 2 mL/kg) after force-feeding TLRE or silymarin. Each therapeutic and protective group was divided into three subgroups: force-fed with saline, TLRE (500 mg/kg), and silymarin (25 mg/kg). The positive group was split into two subgroups that were force-fed TLRE and silymarin. Positive, therapeutic, and protective groups were compared to the negative group (untreated rats). CCl4, TLRE, and silymarin were orally administrated using a gastric tube. RESULTS: In the therapeutic and protective groups, TLRE significantly reduced liver enzymes, i.e., aspartate aminotransferase (12.47 and 6.29%), alanine aminotransferase (30.48 and 11.39%), alkaline phosphatase (17.28 and 15.90%), and cytochrome P450-2E1 (39.04 and 48.24%), and tumour necrosis factor-α (53.72 and 53.72%) in comparison with CCl4-induced hepatotoxicity controls. CONCLUSIONS: TLRE has a potent hepatoprotective effect with a good safety margin. After a repeated study on another type of small experimental animal, their offspring, and an experiment with a large animal, this study may lead to clinical trials.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/pathology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Tagetes/chemistry , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Carbon Tetrachloride/pharmacokinetics , Cytochrome P-450 CYP2E1/metabolism , Lipids/blood , Liver/pathology , Male , Models, Animal , Rats , Rats, Wistar , Silymarin/pharmacology , Tumor Necrosis Factor-alpha/metabolism
2.
Cell Death Dis ; 12(1): 76, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436540

ABSTRACT

The biological clock is an endogenous biological timing system, which controls metabolic functions in almost all organs. Nutrient metabolism, substrate processing, and detoxification are circadian controlled in livers. However, how the clock genes respond to toxins and influence toxicity keeps unclear. We identified the clock gene Per1 was specifically elevated in mice exposed to toxins such as carbon tetrachloride (CCl4). Mice lacking Per1 slowed down the metabolic rate of toxins including CCl4, capsaicin, and acetaminophen, exhibiting relatively more residues in the plasma. Liver injury and fibrosis induced by acute and chronic CCl4 exposure were markedly alleviated in Per1-deficient mice. These processes involved the binding of PER1 protein and hepatocyte nuclear factor-1alpha (HNF-1α), which enhances the recruitment of HNF-1α to cytochrome P450 2E1 (Cyp2e1) promoter and increases Cyp2e1 expression, thereby promoting metabolism for toxins in the livers. These results indicate that PER1 mediates the metabolism of toxins and appropriate suppression of Per1 response is a potential therapeutic target for toxin-induced hepatotoxicity.


Subject(s)
Cytochrome P-450 CYP2E1/metabolism , Period Circadian Proteins/metabolism , Animals , Carbon Tetrachloride/pharmacokinetics , Cytochrome P-450 CYP2E1/genetics , Gene Expression , Hep G2 Cells , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Period Circadian Proteins/genetics , Transfection
3.
Food Chem Toxicol ; 145: 111591, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32739454

ABSTRACT

The present study investigated whether a single pretreatment with clofibric acid suppresses liver injury in rats after CCl4 intoxication. Rats received a single pretreatment with clofibric acid (100 mg/kg, i.p.) 1 h prior to a CCl4 (1 mL/kg, p.o.) challenge, and were euthanized 24 h after the CCl4 administration. A single pretreatment with clofibric acid effectively suppressed increases in the serum aminotransferase activities and the severity of necrosis following the CCl4 challenge, whereas the pretreatment did not protect against CCl4-induced fatty liver. The clofibric acid pretreatment did not affect blood concentrations of CCl4 in the early stage after CCl4 dosing, or the level of the CCl4 reaching the liver 1 h after the CCl4 challenge. Moreover, the clofibric acid pretreatment did not affect the intensity of the covalent binding of the [14C]CCl4 metabolite to microsomal proteins and lipids. The clofibric acid pretreatment did not alter microsomal cytochrome P450 2E1 activity. Based on these results, we conclude that protection against CCl4-induced hepatocellular necrosis by a clofibric acid pretreatment does not require its repeated administration, and that a single and brief pre-exposure to clofibric acid prior to CCl4 dosing markedly suppresses necrosis without affecting the development and progression of steatosis.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Clofibric Acid/therapeutic use , Necrosis/prevention & control , Protective Agents/therapeutic use , Animals , Carbon Tetrachloride/metabolism , Carbon Tetrachloride/pharmacokinetics , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2E1/metabolism , Fatty Liver/chemically induced , Fatty Liver/pathology , Liver/pathology , Male , Microsomes, Liver , Necrosis/chemically induced , Necrosis/pathology , Rats, Wistar
4.
Radiology ; 287(2): 581-589, 2018 05.
Article in English | MEDLINE | ID: mdl-29156148

ABSTRACT

Purpose To evaluate the biodistribution, metabolism, and pharmacokinetics of a new type I collagen-targeted magnetic resonance (MR) probe, CM-101, and to assess its ability to help quantify liver fibrosis in animal models. Materials and Methods Biodistribution, pharmacokinetics, and stability of CM-101 in rats were measured with mass spectrometry. Bile duct-ligated (BDL) and sham-treated rats were imaged 19 days after the procedure by using a 1.5-T clinical MR imaging unit. Mice were treated with carbon tetrachloride (CCl4) or with vehicle two times a week for 10 weeks and were imaged with a 7.0-T preclinical MR imaging unit at baseline and 1 week after the last CCl4 treatment. Animals were imaged before and after injection of 10 µmol/kg CM-101. Change in contrast-to-noise ratio (ΔCNR) between liver and muscle tissue after CM-101 injection was used to quantify liver fibrosis. Liver tissue was analyzed for Sirius Red staining and hydroxyproline content. The institutional subcommittee for research animal care approved all in vivo procedures. Results CM-101 demonstrated rapid blood clearance (half-life = 6.8 minutes ± 2.4) and predominately renal elimination in rats. Biodistribution showed low tissue gadolinium levels at 24 hours (<3.9% injected dose [ID]/g ± 0.6) and 10-fold lower levels at 14 days (<0.33% ID/g ± 12) after CM-101 injection with negligible accumulation in bone (0.07% ID/g ± 0.02 and 0.010% ID/g ± 0.004 at 1 and 14 days, respectively). ΔCNR was significantly (P < .001) higher in BDL rats (13.6 ± 3.2) than in sham-treated rats (5.7 ± 4.2) and in the CCl4-treated mice (18.3 ± 6.5) compared with baseline values (5.2 ± 1.0). Conclusion CM-101 demonstrated fast blood clearance and whole-body elimination, negligible accumulation of gadolinium in bone or tissue, and robust detection of fibrosis in rat BDL and mouse CCl4 models of liver fibrosis. © RSNA, 2017 Online supplemental material is available for this article.


Subject(s)
Fibrosis/pathology , Gadolinium/pharmacokinetics , Liver Cirrhosis/diagnostic imaging , Liver/pathology , Magnetic Resonance Imaging , Polysaccharides, Bacterial/pharmacokinetics , Animals , Carbon Tetrachloride/pharmacokinetics , Disease Models, Animal , Fibrosis/diagnostic imaging , Half-Life , Liver/diagnostic imaging , Mass Spectrometry , Mice , Rats , Tissue Distribution
5.
Int J Toxicol ; 31(6): 551-63, 2012.
Article in English | MEDLINE | ID: mdl-23197488

ABSTRACT

Liver disease is a major health issue characterized by several pathological changes, with steatosis (fatty liver) representing a common initial step in its pathogenesis. Steatosis is of critical importance because prevention of fatty liver can obviate downstream pathologies of liver disease (eg, fibrosis). Recent studies have shown a strong correlation between chemical exposure and steatosis. The work described here identifies chemicals on the US Environmental Protection Agency's Integrated Risk Information System (IRIS) that induce steatosis and investigates putative mechanisms by which these chemicals may contribute to this pathological condition. Mitochondrial impairment, insulin resistance, impaired hepatic lipid secretion, and enhanced cytokine production were identified as potential mechanisms that could contribute to steatosis. Taken together, this work is significant because it identifies multiple mechanisms by which environmental chemicals may cause fatty liver and expands our knowledge of the possible role of environmental chemical exposure in the induction and progression of liver disease.


Subject(s)
Environmental Pollutants/toxicity , Fatty Liver/chemically induced , Mitochondria, Liver/drug effects , Xenobiotics/toxicity , Animals , Carbon Tetrachloride/pharmacokinetics , Carbon Tetrachloride/toxicity , Cytokines/metabolism , Databases, Factual , Dogs , Dose-Response Relationship, Drug , Environmental Pollutants/pharmacokinetics , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hydrocarbons, Chlorinated/toxicity , Insulin Resistance , Lipid Metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/prevention & control , Male , Mice , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Rats , Risk Assessment , Vinyl Chloride/pharmacokinetics , Vinyl Chloride/toxicity , Xenobiotics/pharmacokinetics
6.
J. physiol. biochem ; 68(1): 29-35, mar. 2012.
Article in English | IBECS | ID: ibc-122375

ABSTRACT

No disponible


Liver fibrosis is a major health problem that can lead to the development of liver cirrhosis and hepatocellular carcinoma. On the other hand, several antioxidants have been shown to possess protective effect against liver fibrosis. Therefore, in the present work, the effectiveness of curcumin, alpha-lipoic acid, and N-acetylcysteine in protecting against carbon tetrachloride (CCl4)-induced liver fibrosis as well as the mechanism(s) implicated in this protective effect was studied. The antioxidants used in this study resulted in hepatoprotective effect as evident by substantial (..) (AU)


Subject(s)
Animals , Rats , Curcumin/pharmacokinetics , Liver Cirrhosis/drug therapy , Carbon Tetrachloride/pharmacokinetics , Protective Agents/pharmacokinetics , Disease Models, Animal , Thioctic Acid/pharmacokinetics , Acetylcysteine/pharmacokinetics , Antioxidant Response Elements
7.
J. physiol. biochem ; 67(4): 519-530, dic. 2011.
Article in English | IBECS | ID: ibc-122389

ABSTRACT

No disponible


Acetyl-L-carnitine (ALCAR) has been shown to prevent experimental selenite cataractogenesis, a manifestation of oxidative stress, but little is known about its potential in other settings of oxidative stress. The present study was based on the hypothesis that ALCAR prevents carbon tetrachloride (CCl4)-induced oxidative stress in vital tissues. Male albino Wistar rats were divided into three groups, each of six rats. Group I (control) rats received only vehicle (1 ml/kg b.w.) for 4 days; Group II (CCl4-exposed, untreated) rats received CCl4 (2 ml/kg b.w.) on the second and third days and vehicle on the first and fourth days; Group III (CCl4-exposed, ALCAR-treated) rats received ALCAR (200 mg/kg b.w.) for 4 days and CCl4 on the second and third days. All administrations were made intraperitoneally. After the experimental period, significantly (P < 0.05) elevated mean serum levels of aspartate transaminase, alanine transaminase, alkaline phosphatase, and lactate dehydrogenase were observed in Group II rats when compared to Group I and Group III rats. The mean levels of vitamin C, vitamin E, and reduced glutathione and the mean activities of superoxide dismutase, catalase, and glutathione peroxidase were significantly (P < 0.05) lower in samples of hemolysate and of liver, kidney, and brain tissues of Group II rats than those in Group I and Group III rats. The mean level of lipid peroxidation was significantly (P < 0.05) higher in Group II rats than that in Group I and Group III rats. Moreover, the CCl4-induced upregulation of inducible nitric oxide synthase expression was prevented by ALCAR in the liver and brain tissues. These results suggest that ALCAR is able to prevent the CCl4-induced oxidative stress (AU)


Subject(s)
Animals , Rats , Oxidative Stress , Acetylcarnitine/pharmacokinetics , Protective Agents/pharmacokinetics , Disease Models, Animal , Carbon Tetrachloride/pharmacokinetics , Case-Control Studies
8.
J. physiol. biochem ; 67(4): 569-576, dic. 2011. ilus
Article in English | IBECS | ID: ibc-122394

ABSTRACT

No disponible


Coumarins are a vast group of natural compounds and some of them possess antioxidant activities. The comparison of the antioxidant activity of some coumarins with various chemical molecular structure has not been investigated in previous studies. Therefore, this study was aimed to investigate the hepatoprotective effect against carbon tetrachloride (CCl4) -induced hepatic injury by coumarin (1,2-benzopyrone) and coumarin derivatives, esculetin (6,7-dihydroxycoumarin), scoparone (6,7-dimethoxycoumarin), and 4-methylumbelliferone (7-hyroxy-4-methyl) in male Sprague–Dawley rats. Product of lipid peroxidation, malondialdehyde (MDA), activities of antioxidant enzymes, superoxide dismutase (SOD), catalase (CAT) were evaluated for oxidative stress in hepatic injury. Gamma glutamyl transpeptidase (GGT), lactate dehydrogenase (LDH) were detected in plasma as a biomarker of hepatic injury. Significantly elevated levels of MDA and lowered levels of SOD and CAT activities were observed in liver of rats exposed to CCl4, when compared to control values. Similarly, administration of CCl4 increased LDH and GGT levels in serum. Pre-treatment of rats with esculetin (35 mg kg−1, orally) and scoparone (35 mg kg−1, orally) significantly prevented CCl4-induced decrease in MDA levels and increase in SOD and CAT, whereas 4-methylumbelliferone (35 mg kg−1) and coumarin (30 mg kg−1) had no effect against CCl4-induced rise in serum enzymes. Esculetin and scoparone also showed protective properties as was evidenced in reduced LDH and GGT levels in serum. The results of this study indicate that the chemical structures of coumarins play an important role in the prevention of oxidative stress (AU)


Subject(s)
Animals , Rats , Coumarins/pharmacokinetics , Chemical and Drug Induced Liver Injury, Chronic/physiopathology , Oxidative Stress , Protective Agents/pharmacokinetics , Disease Models, Animal , Carbon Tetrachloride/pharmacokinetics
9.
Toxicol Sci ; 115(1): 253-66, 2010 May.
Article in English | MEDLINE | ID: mdl-20106946

ABSTRACT

Biologically based dose-response (BBDR) modeling of environmental pollutants can be utilized to inform the mode of action (MOA) by which compounds elicit adverse health effects. Chemicals that produce tumors are typically labeled as either genotoxic or nongenotoxic. Though both the genotoxic and the nongenotoxic MOA may be operative as a function of dose, it is important to note that the label informs but does not define a MOA. One commonly proposed MOA for nongenotoxic carcinogens is characterized by the key events cytotoxicity and regenerative proliferation. The increased division rate associated with such proliferation can cause an increase in the probability of mutations, which may result in tumor formation. We included these steps in a generalized computational pharmacodynamic (PD) model incorporating cytotoxicity as a MOA for three carcinogens (chloroform, CHCl(3); carbon tetrachloride, CCL(4); and N,N-dimethylformamide, DMF). For each compound, the BBDR model is composed of a chemical-specific physiologically based pharmacokinetic model linked to a PD model of cytotoxicity and cellular proliferation. The rate of proliferation is then linked to a clonal growth model to predict tumor incidences. Comparisons of the BBDR simulations and parameterizations across chemicals suggested that significant variation among the models for the three chemicals arises in a few parameters expected to be chemical specific (such as metabolism and cellular injury rate constants). Optimization of model parameters to tumor data for CCL(4) and DMF resulted in similar estimates for all parameters related to cytotoxicity and tumor incidences. However, optimization of the CHCl(3) data resulted in a higher estimate for one parameter (BD) related to death of initiated cells. This implies that additional steps beyond cytotoxicity leading to induced cellular proliferation can be quantitatively different among chemicals that share cytotoxicity as a hypothesized carcinogenic MOA.


Subject(s)
Carbon Tetrachloride/toxicity , Carcinogens/toxicity , Chemical and Drug Induced Liver Injury, Chronic/pathology , Chloroform/toxicity , Dimethylformamide/toxicity , Liver Neoplasms/pathology , Animals , Carbon Tetrachloride/pharmacokinetics , Carcinogens/pharmacokinetics , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury, Chronic/etiology , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Chloroform/pharmacokinetics , Computational Biology , Computers , Dimethylformamide/pharmacokinetics , Female , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Male , Mice , Models, Biological , Regeneration/drug effects , Risk Assessment
10.
J. physiol. biochem ; 65(3): 225-233, sept. 2009.
Article in English | IBECS | ID: ibc-122867

ABSTRACT

No disponible


This study aimed to investigate whether treatments with vitamin E, L-carnitine and melatonin can protect against CCl4 and diabetes-induced hepatic oxidative stress. Hepatic oxidative stress was performed in rats through 50% v/v carbon tetrachloride (CCl4) (1 ml/kg/3days, i.p.), and through diabetes mellitus induced by streptozotocin (STZ) (40 mg/kg, i.p.). Vitamin E (100 mg/kg/day, i.p), L-carnitine (300 mg/kg/day, i.p.) and melatonin (10 mg/kg/day, i.p.) were injected for a period of 6 weeks. Thereafter, changes in serum glucose level, liver function tests, hepatic malondialdehyde (MDA) content, hepatic reduced glutathione (GSH) content, hepatic superoxide (..) (AU)


Subject(s)
Animals , Rats , Vitamin E/pharmacokinetics , Melatonin/pharmacokinetics , Carnitine/pharmacokinetics , Oxidative Stress , Diabetes Mellitus/physiopathology , Carbon Tetrachloride/pharmacokinetics , Disease Models, Animal , Protective Agents/pharmacokinetics , Antioxidants/pharmacokinetics
11.
Eksp Klin Farmakol ; 71(6): 42-4, 2008.
Article in Russian | MEDLINE | ID: mdl-19140516

ABSTRACT

It is established in experiments on noninbred rats that 2,4,6-triphenyl-4H-selenopyrane (peroral administration in a dose of 0.8 mg/kg during 3 days) induces cytochrome P450, thus increasing the toxicity and immunotoxicity of carbon tetrachloride (metabolized via "lethal synthesis"), and reduces the analogous effects of carbophos, the biotransformation of which proceeds via the formation of low-toxicity and nontoxic metabolites.


Subject(s)
Antibody Formation/drug effects , Carbon Tetrachloride/toxicity , Cytochrome P-450 Enzyme System/biosynthesis , Immunity, Cellular/drug effects , Malathion/toxicity , Organoselenium Compounds/pharmacology , Animals , Biotransformation , Carbon Tetrachloride/pharmacokinetics , Female , Lethal Dose 50 , Malathion/pharmacokinetics , Male , Rats
12.
J Toxicol Environ Health A ; 70(18): 1527-41, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17710613

ABSTRACT

Physiologically based pharmacokinetic (PBPK) models are increasingly available for environmental chemicals and applied in risk assessments. Volatile organic compounds (VOCs) are important pollutants in air, soil, and water. CYP2E1 metabolically activates many VOCs in animals and humans. Despite its presence in extrahepatic tissues, the metabolism by CYP2E1 is often described as restricted to the liver in PBPK models, unless target tissue dose metrics in extrahepatic tissues are needed for the model application, including risk assessment. The impact of accounting for extrahepatic metabolism by CYP2E1 on the estimation of metabolic parameters and the prediction of dose metrics was evaluated for three lipophilic VOCs: vinyl chloride, trichloroethylene, and carbon tetrachloride. Metabolic parameters estimated from fitting gas uptake data with and without extrahepatic metabolism were similar. The impact of extrahepatic metabolism on PBPK predictions was evaluated using inhalation exposure scenarios relevant for animal toxicity studies and human risk assessment. Although small, the relative role of extrahepatic metabolism and the differences in the predicted dose metrics were greater at low exposure concentrations. The impact was species dependent and influenced by Km for CYP2E1. The current study indicates that inhalation modeling for several representative VOCs that are CYP2E1 substrates is not affected by the inclusion of extrahepatic metabolism, implying that liver-only metabolism may be a reasonable simplification for PBPK modeling of lipophilic VOCs. The PBPK predictions using this assumption can be applied confidently for risk assessment, but this conclusion should not necessarily be applied to VOCs that are metabolized by other enzymes.


Subject(s)
Carbon Tetrachloride/pharmacokinetics , Cytochrome P-450 CYP2E1/metabolism , Models, Biological , Trichloroethylene/pharmacokinetics , Vinyl Chloride/pharmacokinetics , Adipose Tissue/metabolism , Adult , Animals , Brain/metabolism , Humans , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Microsomes/enzymology , Rats , Rats, Sprague-Dawley , Skin/metabolism , Volatilization
13.
Toxicol Appl Pharmacol ; 223(1): 56-65, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17610925

ABSTRACT

Injury to liver, resulting in loss of its normal physiological/biochemical functions, may adversely affect a secondary organ. We examined the response of the liver and kidney to chemical substances that require metabolic activation for their toxicities in mice with a preceding liver injury. Carbon tetrachloride treatment 24 h prior to a challenging dose of carbon tetrachloride or acetaminophen decreased the resulting hepatotoxicity both in male and female mice as determined by histopathological examination and increases in serum enzyme activities. In contrast, the renal toxicity of the challenging toxicants was elevated markedly in male, but not in female mice. Partial hepatectomy also induced similar changes in the hepatotoxicity and nephrotoxicity of a challenging toxicant, suggesting that the contrasting response of male liver and kidney was associated with the reduction of the hepatic metabolizing capacity. Carbon tetrachloride pretreatment or partial hepatectomy decreased the hepatic xenobiotic-metabolizing enzyme activities in both sexes but elevated the renal p-nitrophenol hydroxylase, p-nitroanisole O-demethylase and aminopyrine N-demethylase activities significantly only in male mice. Increases in Cyp2e1 and Cyp2b expression were also evident in male kidney. Castration of males or testosterone administration to females diminished the sex-related differences in the renal response to an acute liver injury. The results indicate that reduction of the hepatic metabolizing capacity induced by liver injury may render secondary target organs susceptible to chemical substances activated in these organs. This effect may be sex-specific. It is also suggested that an integrated approach should be taken for proper assessment of chemical hazards.


Subject(s)
Acetaminophen/pharmacokinetics , Carbon Tetrachloride/pharmacokinetics , Chemical and Drug Induced Liver Injury/metabolism , Kidney/enzymology , Liver/enzymology , Testosterone/physiology , Acetaminophen/toxicity , Aminopyrine N-Demethylase/metabolism , Animals , Biotransformation , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/blood , Cytochrome P-450 CYP2E1/metabolism , Female , Hepatectomy , Hepatocytes/drug effects , Hepatocytes/pathology , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred ICR , Necrosis , Orchiectomy , Organ Specificity/drug effects , Oxidoreductases, O-Demethylating/metabolism , Sex Factors , Testosterone/pharmacology
14.
Regul Toxicol Pharmacol ; 48(1): 93-101, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17367907

ABSTRACT

The derivation of reference concentrations (RfCs) for systemically acting volatile organic chemicals (VOCs) uses a default factor of 10 to account for the interindividual variability in pharmacokinetics (PK) and pharmacodynamics (PD). The magnitude of the PK component of the interindividual variability factor (IVF; also referred to as human kinetic adjustment factor (HKAF)) has previously been estimated using Monte Carlo approaches and physiologically based pharmacokinetic (PBPK) models. Since the RfC derivation considers continuous lifetime human exposure to VOCs in the environment, algorithms to compute steady-state internal dose (SS-ID), such as steady-state arterial blood concentration (Ca) and the steady-state rate of amount metabolized (RAM), can be used to derive IVF-PKs. In this context, probability-bounds (P-bounds) approach is potentially useful for computing an interval of probability distribution of SS-ID from knowledge of population distribution of input parameters. The objective of this study was therefore to compute IVF-PK using the P-bounds approach along with an algorithm for SS-ID in an adult population exposed to VOCs. The existing steady-state algorithms, derived from PBPK models, were rewritten such that SS-ID could be related, without any interdependence, to the following input parameters: alveolar ventilation (Qp), hepatic blood flow (Ql), intrinsic clearance (CL(int)) and blood:air partition coefficient (Pb). The IVF-PK was calculated from the P-bounds of SS-ID corresponding to the 50th and 95th percentiles. Following either specification of probability distribution-free bounds (characterized by minimal, maximal, and mean values) or distribution-defined values (mean, standard deviation and shape of probability distribution where: Qp=lognormal, Ql=lognormal, CL(int)=lognormal and Pb=normal) in RAMAS Risk Calc software version 3.0 (Applied Biomathematics, Setauket, NY), the P-bound estimates of SS-ID for benzene, carbon tetrachloride, chloroform and methyl chloroform were obtained for low level exposures (1ppm). Using probability distribution-defined inputs, the IVF-PK for benzene, carbon tetrachloride, chloroform and methyl chloroform were, respectively, 1.18, 1.28, 1.24, and 1.18 (based on P-bounds for Ca), and 1.31, 1.58, 1.30, and 1.24 (based on P-bounds for RAM). A validation of the P-bounds computation was performed by comparing the results with those obtained using Monte Carlo simulation of the steady-state algorithms. In data-poor situations, when the statistical distributions for all input parameters were not known or available, the P-bounds approach allowed the estimation of IVF-PK. The use of P-bounds method along with steady-state algorithms, as done in this study for the first time, is a practical and scientifically sound way of computing IVF-PKs for systemically acting VOCs.


Subject(s)
Inhalation , Models, Biological , Organic Chemicals/pharmacokinetics , Probability , Air Pollutants/pharmacokinetics , Algorithms , Benzene/pharmacokinetics , Carbon Tetrachloride/pharmacokinetics , Chloroform/pharmacokinetics , Humans , Monte Carlo Method , Pharmacokinetics , Probability Theory , Trichloroethanes/pharmacokinetics , Volatilization
15.
Ars pharm ; 48(4): 351-360, 2007. ilus, tab
Article in Es | IBECS | ID: ibc-64398

ABSTRACT

Se examinó el extracto acuoso y etanólico (100 mg/kg) de Luffa acutangula Linn (frutos) para determinar la actividadantihepatotóxica en ratas druckrey mediante la hepatotoxicidad inducida por tetracloruro de carbono (CCl4)y paracetamol (PCM). Se demostró que el extracto posee un efecto hepatoprotector signifi cativo, ya que reduce losniveles séricos de transaminasas (SGPT y SGOT), fosfatasa alcalina (ALP) y bilirrubina. La signifi cativa actividadhepatoprotectora de Luffa acutangula es comparable a la de la silimarina, agente hepatoprotector estándar, lo quejustifi ca su uso en afecciones del hígado


The ethanolic and aqueous extract (100mg/kg) of Luffa acutangula Linn(fruits) was examined for antihepatotoxic activityin druckrey rats by inducing hepatotoxicity with Carbon tetrachloride(CCl4) and Paracetamol(PCM). The extracthas shown to posses signifi cant hepatoprotective effect by lowering the serum level of transaminases (SGPT & SGOT),Alkaline phosphatase (ALP) and bilirubin. The signifi cant hepatoprotective activity of Luffa acutangula is comparableto that standard hepatoprotective agent silymarin, which justify its use in liver affection


Subject(s)
Rats , Male , Female , Animals , Plant Extracts/chemistry , Luffa acutangula/analysis , Luffa acutangula/pharmacology , Luffa acutangula/therapeutic use , Carbon Tetrachloride/chemistry , Carbon Tetrachloride/chemical synthesis , Acetaminophen/chemistry , Acetaminophen/pharmacology , Silymarin/pharmacology , Phytotherapy/classification , Phytotherapy , Luffa acutangula/supply & distribution , Carbon Tetrachloride/analysis , Carbon Tetrachloride/pharmacology , Carbon Tetrachloride/pharmacokinetics , Silymarin/pharmacokinetics
16.
Toxicol Appl Pharmacol ; 191(3): 211-26, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-13678654

ABSTRACT

Liver injury is known to progress even after the hepatotoxicant is long gone and the mechanisms of progressive injury are not understood. We tested the hypothesis that hydrolytic enzymes such as calpain, released from dying hepatocytes, destroy the surrounding cells causing progression of injury. Calpain inhibitor, N-CBZ-VAL-PHE-methyl ester (CBZ), administered 1 h after a toxic but nonlethal dose of CCl(4) (2 ml/kg, ip) to male Sprague Dawley rats substantially mitigated the progression of liver injury (6 to 48 h) and also led to 75% protection against CCl(4)-induced lethality following a lethal dose (LD75) of CCl(4) (3 ml/kg). Calpain leakage in plasma and in the perinecrotic areas increased until 48 h and decreased from 72 h onward paralleling progression and regression of liver injury, respectively, after CCl(4) treatment. Mitigation of progressive injury was accompanied by substantially low calpain in perinecrotic areas and in plasma after CBZ treatment. Normal hepatocytes incubated with the plasma collected from CCl(4)-treated rats (collected at 12 h when most of the CCl(4) is eliminated) resulted in extensive cell death prevented by CBZ. Cell-impermeable calpain inhibitor E64 also protected against progression of CCl(4)-induced liver injury, thereby confirming the role of released calpain in progression of liver injury. Following CCl(4) treatment, calpain-specific breakdown of alpha-fodrin increased, while it was negligible in rats receiving CBZ after CCl(4). Hepatocyte cell death in incubations containing calpain was completely prevented by CBZ. Eighty percent of Swiss Webster mice receiving a lethal dose (LD80) of acetaminophen (600 mg/kg, ip) survived if CBZ was administered 1 h after acetaminophen, suggesting that calpain-mediated progression of liver injury is neither species nor chemical specific. These findings suggest the role of calpain in progression of liver injury.


Subject(s)
Calpain/metabolism , Hepatocytes/enzymology , Liver Diseases/enzymology , Acetaminophen/metabolism , Acetaminophen/pharmacokinetics , Acetaminophen/toxicity , Animals , Blotting, Western , Calpain/antagonists & inhibitors , Calpain/blood , Carbon Tetrachloride/metabolism , Carbon Tetrachloride/pharmacokinetics , Carbon Tetrachloride/toxicity , Carrier Proteins/metabolism , Chemical and Drug Induced Liver Injury , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP2E1 Inhibitors , Dipeptides/pharmacology , Disease Progression , Immunohistochemistry , Liver Diseases/blood , Liver Diseases/pathology , Male , Mice , Microfilament Proteins/metabolism , Necrosis , Random Allocation , Rats , Rats, Sprague-Dawley
17.
Biol Pharm Bull ; 25(11): 1494-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12419969

ABSTRACT

Animal models prepared by treatment with toxic compounds such as a carbon tetrachloride have been used to examine drug disposition in hepatic diseases. However, it is possible that these compounds accumulate and cause damage to other organs as they are administered systemically. In this study, we used the liver surface application technique to deliver a toxic compound to the liver to prepare an appropriate animal model in which only the liver is significantly damaged. To restrict the absorption area in the liver, a cylindrical diffusion cell was attached to the liver surface of male Wistar rats. Twenty-four hours after direct addition of carbon tetrachloride to the diffusion cell, plasma levels of glutamic-oxaloacetic transaminase (GOT) and glutamic-pyruvic transaminase (GPT), and hepatic malondialdehyde (MDA) concentration were increased, while there were no changes in plasma creatinine or renal MDA level. On the other hand, not only GOT, GPT and hepatic MDA, but also creatinine and renal MDA levels were markedly increased by p.o. and i.p. administration of carbon tetrachloride, suggesting renal damage. These results indicated that the animal models of liver damage prepared by utilizing drug delivery techniques to accumulate toxic compounds in the liver would enable us to investigate the precise effects of hepatic disorder on drug disposition.


Subject(s)
Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Disease Models, Animal , Drug Delivery Systems/methods , Liver/drug effects , Animals , Carbon Tetrachloride/pharmacokinetics , Liver/metabolism , Liver/pathology , Male , Rats , Rats, Wistar
18.
Risk Anal ; 22(3): 623-31, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12088237

ABSTRACT

Because ethical considerations often preclude directly determining the human health effects of treatments or interventions by experimentation, such effects are estimated by extrapolating reactions predicted from animal experiments. Under such conditions, it must be demonstrated that the reliability of the extrapolated predictions is not excessively affected by inherent data limitations and other components of model specification. This is especially true of high-level models composed of ad hoc algebraic equations whose parameters do not correspond to specific physical properties or processes. Models based on independent experimental data restricting the numerical space of parameters that do represent actual physical properties can be represented at a more detailed level. Sensitivities of the computed trajectories to parameter variations permit more detailed attribution of uncertainties in the predictions to these low-level properties. S-systems, in which parameters are estimated empirically, and physiological models, whose parameters can be estimated accurately from independent data, are used to illustrate the applicability of trajectory sensitivity analysis to lower-level models.


Subject(s)
Models, Biological , Animals , Carbon Tetrachloride/pharmacokinetics , Ethylene Glycols/pharmacokinetics , Humans , Metabolism , Models, Statistical , Pentanols/pharmacokinetics , Physiology , Reproducibility of Results , Risk Assessment , Sensitivity and Specificity
19.
J Pharmacol Toxicol Methods ; 48(1): 41-4, 2002.
Article in English | MEDLINE | ID: mdl-12750040

ABSTRACT

INTRODUCTION: The purpose of this study was to optimize carbon tetrachloride-induced hepatotoxicity in the rat with respect to dose, route of injection, and time course. METHODS: Male Wistar albino rats, 4 to 6 weeks old and weighing 130-180 g were used. Hepatotoxicity was evaluated by measuring the activity of serum enzymes (alkaline phosphatase [ALP], alanine aminotransferase [ALT], and aspartate aminotransferase [AST]) as well as serum total bilirubin level. RESULTS: Intraperitoneal injection of carbon tetrachloride (CCl(4)) increased the activity of ALP (from 64.9 to 137.3 U/l), ALT (from 106.6 to 693.1 U/l), and AST (from 113.8 to 693.9 U/l). Plasma bilirubin level increased (from 0.119 to 0.42 mg/dl). In contrast, subcutaneous injection of CCl(4) had no effect on these variables. The optimum intraperitoneal dose of CCl(4) was found to be 2 ml/kg body weight (dissolved in an equal volume of olive oil), and this increased the level of bilirubin and the activity of the three enzymes significantly, without causing death of the animals. Hepatotoxicity was observed within 2 h of intraperitoneal injection of CCl(4) and reached a peak after 24 h. Bilirubin level and serum enzyme activities declined gradually to normal levels by 3 days after CCl(4) injection. CONCLUSION: It is possible to reliably evoke reversible hepatotoxicity in rats by intraperitoneal injection of 2 ml/kg CCl(4).


Subject(s)
Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/pharmacokinetics , Chemical and Drug Induced Liver Injury/pathology , Alanine Transaminase/blood , Alkaline Phosphatase/metabolism , Animals , Aspartate Aminotransferases/blood , Bilirubin/metabolism , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Injections, Subcutaneous , Liver Function Tests , Male , Models, Biological , Rats , Rats, Wistar
20.
Z Naturforsch C J Biosci ; 56(7-8): 649-59, 2001.
Article in English | MEDLINE | ID: mdl-11531102

ABSTRACT

CCl4-induced liver damage was modeled in monolayer cultures of rat primary hepatocytes with a focus on involvement of covalent binding of CCl4 metabolites to cell components and/or peroxidative damage as the cause of injury. (1) Covalent binding of 14C-labeled metabolites was detected in hepatocytes immediately after exposure to CCl4. (2) Low oxygen partial pressure increased the reductive metabolism of CCl4 and thus covalent binding. (3) [14C]-CCl4 was bound to lipids and to proteins throughout subcellular fractions. Binding occurred preferentially to triacylglycerols and phospholipids, with phosphatidylcholine containing the highest amount of label. (4) The lipid peroxidation potency of CCl4 revealed subtle differences compared to other peroxidative substances, viz., ADP-Fe3+ and cumol hydroperoxide, respectively. (5) CCl4, but not the other peroxidative substances, decreased the rate of triacylglycerol secretion as very low density lipoproteins. (6) The anti-oxidant vitamin E (alpha-tocopherol) blocked lipid peroxidation, but not covalent binding, and secretion of lipoproteins remained inhibited. (7) The radical scavenger piperonyl butoxide prevented CCl4-induced lipid peroxidation as well as covalent binding of CCl4 metabolites to cell components, and also restored lipoprotein metabolism. The results confirm that covalent binding of the CCl3* radical to cell components initiates the inhibition of lipoprotein secretion and thus steatosis, whereas reaction with oxygen, to form CCl3-OO*, initiates lipid peroxidation. The two processes are independent of each other, and the extent to which either process occurs depends on partial oxygen pressure. The former process may result in adduct formation and, ultimately, cancer initiation, whereas the latter results in loss of calcium homeostasis and, ultimately, apoptosis and cell death.


Subject(s)
Carbon Tetrachloride/pharmacokinetics , Carbon Tetrachloride/toxicity , Hepatocytes/metabolism , Liver/pathology , Animals , Biotransformation , Carbon Radioisotopes , Hepatocytes/drug effects , Kinetics , Lipid Metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Microsomes, Liver/metabolism , Phospholipids/metabolism , Protein Biosynthesis , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...