Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90062-4
Joachim Kremer, Zhi-Wei Lai, Charlotte Esser
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a ligand for the arylhydrocarbon receptor (Ah receptor), abundant in the murine thymus. In the thymus immunocompetent T cells develop. Upon exposure of murine fetal thymi in organ cultures to TCDD the distribution of mature and immature thymocytes is skewed towards apparently mature, prospective cytotoxic cells of the CD4−CD8+T cell receptor+ phenotype. The normally abundant CD4+CD8+ cells are decreased. Proliferation of the most immature thymocyte subpopulations is inhibited and maturation of thymocytes appears accelerated by TCDD. Eventually the thymocyte number is significantly decreased. Selective treatment of stroma cells showed them to be the primary target cells of TCDD action. Thymus stroma plays a pivotal role in thymocyte maturation and is indispensable for the selection of thymocytes bearing T cell receptors specific for foreign antigen in the context of self. We tested whether the effects of TCDD on thymocyte differentiation and maturation has further consequences for the selection processes by analysing (a) the repertoire of Vβ genes used as a measure for negative selection and (b) the expression of CD69 and bcl-2 by thymocytes as a parameter for positive selection. Our data indicate that TCDD does not cause gross disturbance of negative selection but provide evidence for more cells auditioning for positive selection by TCDD exposure.
{"title":"Evidence for the promotion of positive selection of thymocytes by Ah receptor agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin","authors":"Joachim Kremer, Zhi-Wei Lai, Charlotte Esser","doi":"10.1016/0926-6917(95)90062-4","DOIUrl":"10.1016/0926-6917(95)90062-4","url":null,"abstract":"<div><p>2,3,7,8-tetrachlorodibenzo-<em>p</em>-dioxin (TCDD) is a ligand for the arylhydrocarbon receptor (Ah receptor), abundant in the murine thymus. In the thymus immunocompetent T cells develop. Upon exposure of murine fetal thymi in organ cultures to TCDD the distribution of mature and immature thymocytes is skewed towards apparently mature, prospective cytotoxic cells of the CD4<sup>−</sup>CD8<sup>+</sup>T cell receptor<sup>+</sup> phenotype. The normally abundant CD4<sup>+</sup>CD8<sup>+</sup> cells are decreased. Proliferation of the most immature thymocyte subpopulations is inhibited and maturation of thymocytes appears accelerated by TCDD. Eventually the thymocyte number is significantly decreased. Selective treatment of stroma cells showed them to be the primary target cells of TCDD action. Thymus stroma plays a pivotal role in thymocyte maturation and is indispensable for the selection of thymocytes bearing T cell receptors specific for foreign antigen in the context of self. We tested whether the effects of TCDD on thymocyte differentiation and maturation has further consequences for the selection processes by analysing (a) the repertoire of V<sub>β</sub> genes used as a measure for negative selection and (b) the expression of CD69 and bcl-2 by thymocytes as a parameter for positive selection. Our data indicate that TCDD does not cause gross disturbance of negative selection but provide evidence for more cells auditioning for positive selection by TCDD exposure.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 413-427"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90062-4","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19722557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
We have studied the hypothesis that 6,7-dihydroxy-1-methyl-1,2,3,4-tetrahydroisoquinoline (salsolinol) is neurotoxic. Salsolinol induced a significant time and dose related inhibition of 3[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide; thiazoyl blue (MTT) reduction, and increased lactate dehydrogenase release (LDH) release from human SH-SY5Y neuroblastoma cells, at concentrations within the range of 1-methyl-4-phenylpyridinium (MPP+) cytotoxicity, in vitro. Cytotoxicity was not inhibited by the addition of antioxidants, monoamine oxidase inhibitors or imipramine. In confluent monolayers, salsolinol stimulated catecholamine uptake with EC50 values of 17 μM and 11 μM, for noradrenaline and dopamine, respectively. Conversely, at concentrations above 100 μM, salsolinol inhibited the uptake of noradrenaline and dopamine, with IC50 values of 411 μM and 379 μM, respectively. The inhibition of catecholamine uptake corresponded to the increased displacement of [3H]nisoxetine from the uptake1 site by salsolinol, as the Ki (353 μM) for displacement was similar to the IC50 (411 and 379 μM) for uptake. Salsolinol stimulated catecholamine uptake does not involve the uptake recognition site, or elevation of cAMP, cGMP, or inhibition of protein kinase C. Salsolinol also inhibited both carbachol (1 mM) and K+ (100 mM, Na+ adjusted) evoked release of noradrenaline from SH-SY5Y cells, with IC50 values of 500 μM and 120 μM, respectively. In conclusion, salsolinol appears to be cytotoxic to SH-SY5Y cells, via a mechanism that does not require uptake1, bioactivation by monoamine oxidase, or membrane based free radical damage. The effects of salsolinol on catecholamine uptake, and the mechanism of toxicity require further investigation.
{"title":"Studies on the neurotoxicity of 6,7-dihydroxy-1-methyl-1,2,3,4-tetrahydroisoquinoline (salsolinol) in SH-SY5Y cells","authors":"J.M. Willets , D.G. Lambert , J. Lunec , H.R. Griffiths","doi":"10.1016/0926-6917(95)90051-9","DOIUrl":"10.1016/0926-6917(95)90051-9","url":null,"abstract":"<div><p>We have studied the hypothesis that 6,7-dihydroxy-1-methyl-1,2,3,4-tetrahydroisoquinoline (salsolinol) is neurotoxic. Salsolinol induced a significant time and dose related inhibition of 3[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide; thiazoyl blue (MTT) reduction, and increased lactate dehydrogenase release (LDH) release from human SH-SY5Y neuroblastoma cells, at concentrations within the range of 1-methyl-4-phenylpyridinium (MPP<sup>+</sup>) cytotoxicity, in vitro. Cytotoxicity was not inhibited by the addition of antioxidants, monoamine oxidase inhibitors or imipramine. In confluent monolayers, salsolinol stimulated catecholamine uptake with EC<sub>50</sub> values of 17 μM and 11 μM, for noradrenaline and dopamine, respectively. Conversely, at concentrations above 100 μM, salsolinol inhibited the uptake of noradrenaline and dopamine, with IC<sub>50</sub> values of 411 μM and 379 μM, respectively. The inhibition of catecholamine uptake corresponded to the increased displacement of [<sup>3</sup>H]nisoxetine from the uptake<sub>1</sub> site by salsolinol, as the <em>K</em><sub><em>i</em></sub> (353 μM) for displacement was similar to the IC<sub>50</sub> (411 and 379 μM) for uptake. Salsolinol stimulated catecholamine uptake does not involve the uptake recognition site, or elevation of cAMP, cGMP, or inhibition of protein kinase C. Salsolinol also inhibited both carbachol (1 mM) and K<sup>+</sup> (100 mM, Na<sup>+</sup> adjusted) evoked release of noradrenaline from SH-SY5Y cells, with IC<sub>50</sub> values of 500 μM and 120 μM, respectively. In conclusion, salsolinol appears to be cytotoxic to SH-SY5Y cells, via a mechanism that does not require uptake<sub>1</sub>, bioactivation by monoamine oxidase, or membrane based free radical damage. The effects of salsolinol on catecholamine uptake, and the mechanism of toxicity require further investigation.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 319-326"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90051-9","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19720880","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In the present study we investigated the influence of acute lead poisoning upon the expression of benzodiazepine receptors. In addition, we examined if administration of PK 11195, an isoquinoline carboxamide derivative, to lead-poisoned rats could modulate the changes in receptor binding properties achieved by lead alone. Lead poisoning was ascertained by determination of urine σ-aminolevulinic acid levels and lead levels in rat livers. Scatchard analysis of saturation curves of [3H]PK 11195 binding to liver membranes of rats treated with lead alone or with both lead and PK 11195 showed an approximately two-fold decrease in receptor density in comparison with control groups. Peripheral benzodiazepine receptor density in the kidneys and adrenals of poisoned rats was not changed by lead intoxication per se or by coadministration of PK 11195. Scatchard analysis of saturation curves of [3H]Ro 15-1788 binding in rat cerebral cortex tissue showed no difference in the receptor density between the various groups. The Kd values of all organs were in the nanomolar range (1–4 nM). We conclude that PK 11195 is not a protective agent of hepatic peripheral benzodiazepine receptors in lead intoxication. Moreover, it causes over-accumulation of lead in hepatocytes in an unknown mechanism of action.
{"title":"Down-regulation of hepatic peripheral-type benzodiazepine receptors caused by acute lead intoxication","authors":"Ora Fonia , Ronit Weizman , Eliyahu Zisman , Ruth Ashkenazi , Moshe Gavish","doi":"10.1016/0926-6917(95)90053-5","DOIUrl":"10.1016/0926-6917(95)90053-5","url":null,"abstract":"<div><p>In the present study we investigated the influence of acute lead poisoning upon the expression of benzodiazepine receptors. In addition, we examined if administration of PK 11195, an isoquinoline carboxamide derivative, to lead-poisoned rats could modulate the changes in receptor binding properties achieved by lead alone. Lead poisoning was ascertained by determination of urine σ-aminolevulinic acid levels and lead levels in rat livers. Scatchard analysis of saturation curves of [<sup>3</sup>H]PK 11195 binding to liver membranes of rats treated with lead alone or with both lead and PK 11195 showed an approximately two-fold decrease in receptor density in comparison with control groups. Peripheral benzodiazepine receptor density in the kidneys and adrenals of poisoned rats was not changed by lead intoxication per se or by coadministration of PK 11195. Scatchard analysis of saturation curves of [<sup>3</sup>H]Ro 15-1788 binding in rat cerebral cortex tissue showed no difference in the receptor density between the various groups. The <em>K</em><sub>d</sub> values of all organs were in the nanomolar range (1–4 nM). We conclude that PK 11195 is not a protective agent of hepatic peripheral benzodiazepine receptors in lead intoxication. Moreover, it causes over-accumulation of lead in hepatocytes in an unknown mechanism of action.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 335-339"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90053-5","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19720882","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90067-5
Jac M.M.J.G. Aarts , Michael S. Denison , Mary A. Cox , Marjolijn A.C. Schalk , Patricia M. Garrison , Kathryn Tullis , Laura H.J. de Haan , Abraham Brouwer
Using recombinant cell lines showing Ah receptor-controlled expression of a luciferase reporter gene, the interaction of di-ortho-substituted polychlorinated biphenyls (PCBs) with Ah receptor agonists was studied. In the recombinant Hepa1c1c7 mouse hepatoma (H1L1.1c7) cells strong antagonistic interaction of 2,2′,5,5′-tetrachlorobiphenyl (PCB52) with luciferase expression induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 3,3′,4,4′-tetrachlorobiphenyl (PCB77) was observed, and similarly, between 2,2′,3,3′,4,4′-hexachlorobiphenyl (PCB128) and PCB77. Accordingly, PCB52 was found to inhibit ethoxyresorufin-O-deethylase (EROD) induction by PCB77 in wild-type Hepa1c1c7 cells. In contrast, the antagonistic effect of PCB52 on TCDD-induced luciferase expression was only minor in recombinant guinea pig GPC16 colon adenocarcinoma (G16L1.1c8) and human HepG2 hepatoma (HG2L1.1c3) cells, and intemediate in recombinant H4IIE rat hepatoma (H4L1.1c4) cells. Gel retardation studies using a 32P-labelled dioxin responsive element (DRE)-containing oligonucleotide, and ligand binding studies using [3H]TCDD, demonstrated that the species-specific antagonistic activity of PCB52 on Ah receptor-controlled luciferase expression is due to inhibition of Ah receptor ligand and DNA binding. We conclude, that Ah-mediated luciferase expression provides a useful tool to study the species specificity of Ah receptor (ant)agonists.
{"title":"Species-specific antagonism of Ah receptor action by 2,2′,5,5′-tetrachloro- and 2,2′,3,3′,4,4′-hexachlorobiphenyl","authors":"Jac M.M.J.G. Aarts , Michael S. Denison , Mary A. Cox , Marjolijn A.C. Schalk , Patricia M. Garrison , Kathryn Tullis , Laura H.J. de Haan , Abraham Brouwer","doi":"10.1016/0926-6917(95)90067-5","DOIUrl":"10.1016/0926-6917(95)90067-5","url":null,"abstract":"<div><p>Using recombinant cell lines showing Ah receptor-controlled expression of a luciferase reporter gene, the interaction of di-<em>ortho</em>-substituted polychlorinated biphenyls (PCBs) with Ah receptor agonists was studied. In the recombinant Hepa1c1c7 mouse hepatoma (H1L1.1c7) cells strong antagonistic interaction of 2,2′,5,5′-tetrachlorobiphenyl (PCB52) with luciferase expression induced by 2,3,7,8-tetrachlorodibenzo-<em>p</em>-dioxin (TCDD) or 3,3′,4,4′-tetrachlorobiphenyl (PCB77) was observed, and similarly, between 2,2′,3,3′,4,4′-hexachlorobiphenyl (PCB128) and PCB77. Accordingly, PCB52 was found to inhibit ethoxyresorufin-<em>O</em>-deethylase (EROD) induction by PCB77 in wild-type Hepa1c1c7 cells. In contrast, the antagonistic effect of PCB52 on TCDD-induced luciferase expression was only minor in recombinant guinea pig GPC16 colon adenocarcinoma (G16L1.1c8) and human HepG2 hepatoma (HG2L1.1c3) cells, and intemediate in recombinant H4IIE rat hepatoma (H4L1.1c4) cells. Gel retardation studies using a <sup>32</sup>P-labelled dioxin responsive element (DRE)-containing oligonucleotide, and ligand binding studies using [<sup>3</sup>H]TCDD, demonstrated that the species-specific antagonistic activity of PCB52 on Ah receptor-controlled luciferase expression is due to inhibition of Ah receptor ligand and DNA binding. We conclude, that Ah-mediated luciferase expression provides a useful tool to study the species specificity of Ah receptor (ant)agonists.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 463-474"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90067-5","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19721855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90059-4
Amaia Irizar , Christopher R. Barnett , Peter R. Flatt , Costas Ioannides
The hepatic expression of xenobiotic-metabolising cytochrome P450 isoforms in the genetically obese Zucker rat, a model of obesity, was compared to that of its lean littermate. Cytochrome P450 (CYP) levels were determined using diagnostic substrates and/or immunologically in Western blot analyses. When compared with the lean Zucker rat, the obese animal exhibited hyperglycaemia, hypercholesterolaemia, marked hyperinsulinaemia and hypertriglyceridaemia but was normoketonaemic. CYP3A and CYP1A2 levels were higher in the obese Zucker rat when compared with the lean littermate but, in contrast, a protein recognised by human CYP2D6 and, to a lesser extent, CYP2C11 levels were lower. Pretreatment with acetone, dexamethasone and clofibrate resulted in enhanced p-nitrophenol hydroxylase (CYP2E), erythromycin N-demethylase (CYP3A) and lauric acid hydroxylase (CYP4A) activities respectively in the liver of the lean Zucker rat but, in contrast, the obese Zucker rat was refractive to such treatment; similarly, hepatic apoprotein levels of the CYP2E and CYP4A subfamilies were increased markedly only in the lean Zucker rat. It is concluded that CYP2E, CYP3A and CYP4A subfamilies are poorly expressed in the obese Zucker rat, and this rat strain may serve as a good model for elucidating the molecular mechanisms of induction of these cytochrome P450 proteins.
{"title":"Defective expression of cytochrome P450 proteins in the liver of the genetically obese Zucker rat","authors":"Amaia Irizar , Christopher R. Barnett , Peter R. Flatt , Costas Ioannides","doi":"10.1016/0926-6917(95)90059-4","DOIUrl":"10.1016/0926-6917(95)90059-4","url":null,"abstract":"<div><p>The hepatic expression of xenobiotic-metabolising cytochrome P450 isoforms in the genetically obese Zucker rat, a model of obesity, was compared to that of its lean littermate. Cytochrome P450 (CYP) levels were determined using diagnostic substrates and/or immunologically in Western blot analyses. When compared with the lean Zucker rat, the obese animal exhibited hyperglycaemia, hypercholesterolaemia, marked hyperinsulinaemia and hypertriglyceridaemia but was normoketonaemic. CYP3A and CYP1A2 levels were higher in the obese Zucker rat when compared with the lean littermate but, in contrast, a protein recognised by human CYP2D6 and, to a lesser extent, CYP2C11 levels were lower. Pretreatment with acetone, dexamethasone and clofibrate resulted in enhanced <em>p</em>-nitrophenol hydroxylase (CYP2E), erythromycin <em>N</em>-demethylase (CYP3A) and lauric acid hydroxylase (CYP4A) activities respectively in the liver of the lean Zucker rat but, in contrast, the obese Zucker rat was refractive to such treatment; similarly, hepatic apoprotein levels of the CYP2E and CYP4A subfamilies were increased markedly only in the lean Zucker rat. It is concluded that CYP2E, CYP3A and CYP4A subfamilies are poorly expressed in the obese Zucker rat, and this rat strain may serve as a good model for elucidating the molecular mechanisms of induction of these cytochrome P450 proteins.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 385-393"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90059-4","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19722554","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90072-1
{"title":"Acknowledgement to reviewers","authors":"","doi":"10.1016/0926-6917(95)90072-1","DOIUrl":"https://doi.org/10.1016/0926-6917(95)90072-1","url":null,"abstract":"","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 495-496"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90072-1","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"137289479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90055-1
Vanaja Paul , Easwaramoorthy Balasubramaniam , Arumugam Radhakrishnan Jayakumar , Mehboob Kazi
The neurobehavioral and hepatic effects following chronic endosulfan administration were studied in adult male and female rats. The neurobehavioral effect was determined by testing spontaneous motor activity, motor coordination and learning and memory processes in rats of either sex, 30 days after treating the animal orally with endosulfan (3.0 and 6.0 mg/kg per day). Mortality occurring during the treatment and body weight gain at the termination of treatment were also recorded. Liver weight and liver and serum concentrations of glutamic oxaloacetic transamine, glutamic pyruvic transaminase and acetylinesterase were measured in order to determine the hepatotoxic effect of endosulfan. Body weight gain, motor coordination and acetylcholinesterase activity were unaltered in either sex. Learning and memory processes were impaired in both groups indistinguishably. Liver weight and liver and serum transaminases concentrations were increased more markedly in female than in male animals. A 30% mortality occurred in female group that received 6 mg/kg of endosulfan. Endosulfan stimulated spontaneous motor activity more markedly in male than in female animals. These findings suggest that a sex-related difference seems to occur in the stimulation of spontaneous motor activity, liver injury and mortality that result from repeated exposure to sublethal doses of endosulfan in rats.
{"title":"A sex-related difference in the neurobehavioral and hepatic effects following chronic endosulfan treatment in rats","authors":"Vanaja Paul , Easwaramoorthy Balasubramaniam , Arumugam Radhakrishnan Jayakumar , Mehboob Kazi","doi":"10.1016/0926-6917(95)90055-1","DOIUrl":"10.1016/0926-6917(95)90055-1","url":null,"abstract":"<div><p>The neurobehavioral and hepatic effects following chronic endosulfan administration were studied in adult male and female rats. The neurobehavioral effect was determined by testing spontaneous motor activity, motor coordination and learning and memory processes in rats of either sex, 30 days after treating the animal orally with endosulfan (3.0 and 6.0 mg/kg per day). Mortality occurring during the treatment and body weight gain at the termination of treatment were also recorded. Liver weight and liver and serum concentrations of glutamic oxaloacetic transamine, glutamic pyruvic transaminase and acetylinesterase were measured in order to determine the hepatotoxic effect of endosulfan. Body weight gain, motor coordination and acetylcholinesterase activity were unaltered in either sex. Learning and memory processes were impaired in both groups indistinguishably. Liver weight and liver and serum transaminases concentrations were increased more markedly in female than in male animals. A 30% mortality occurred in female group that received 6 mg/kg of endosulfan. Endosulfan stimulated spontaneous motor activity more markedly in male than in female animals. These findings suggest that a sex-related difference seems to occur in the stimulation of spontaneous motor activity, liver injury and mortality that result from repeated exposure to sublethal doses of endosulfan in rats.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 355-360"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90055-1","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19720884","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90066-7
Marie Vahter , Gabriela Concha , Barbro Nermell , Robert Nilsson , Fernando Dulout , A.T. Natarajan
The metabolism of inorganic arsenic (As) in native women in four Andean villages in north-western Argentina with elevated levels of As in the drinking water (2.5, 14, 31, and 200 μg/l, respectively) has been investigated. Collected foods contained 9–427 μg As/kg wet weight, with the highest concentrations in soup. Total As concentrations in blood were markedly elevated (median 7.6 μg/l) only in the village with the highest concentration in the drinking water. Group median concentrations of metabolites of inorganic As (inorganic As, methylarsonic acid (MMA) and dimethylarsinic acid (DMA) in the urine varied between 14 and 256 μg/l. Urinary concentrations of total As were only slightly higher (18–258 μg/l), indicating that inorganic As was the main form of As ingested. In contrast to all other populations studied so far, arsenic was excreted in the urine mainly as inorganic As and DMA. There was very little MMA in the urine (overall median 2.2%, range 0.0–11%), which should be compared to 10–20% of the urinary arsenic in all other populations studied. This may indicate the existence of genetic polymorphism in the control of the methyltransferase activity involved in the methylation of As. Furthermore, the percentage of DMA in the urine was significantly higher in the village with 200 μg As/l in the water, indicating an induction of the formation of DMA. Such an effect has not been observed in other studies on human subjects with elevated exposure to arsenic.
{"title":"A unique metabolism of inorganic arsenic in native Andean women","authors":"Marie Vahter , Gabriela Concha , Barbro Nermell , Robert Nilsson , Fernando Dulout , A.T. Natarajan","doi":"10.1016/0926-6917(95)90066-7","DOIUrl":"10.1016/0926-6917(95)90066-7","url":null,"abstract":"<div><p>The metabolism of inorganic arsenic (As) in native women in four Andean villages in north-western Argentina with elevated levels of As in the drinking water (2.5, 14, 31, and 200 μg/l, respectively) has been investigated. Collected foods contained 9–427 μg As/kg wet weight, with the highest concentrations in soup. Total As concentrations in blood were markedly elevated (median 7.6 μg/l) only in the village with the highest concentration in the drinking water. Group median concentrations of metabolites of inorganic As (inorganic As, methylarsonic acid (MMA) and dimethylarsinic acid (DMA) in the urine varied between 14 and 256 μg/l. Urinary concentrations of total As were only slightly higher (18–258 μg/l), indicating that inorganic As was the main form of As ingested. In contrast to all other populations studied so far, arsenic was excreted in the urine mainly as inorganic As and DMA. There was very little MMA in the urine (overall median 2.2%, range 0.0–11%), which should be compared to 10–20% of the urinary arsenic in all other populations studied. This may indicate the existence of genetic polymorphism in the control of the methyltransferase activity involved in the methylation of As. Furthermore, the percentage of DMA in the urine was significantly higher in the village with 200 μg As/l in the water, indicating an induction of the formation of DMA. Such an effect has not been observed in other studies on human subjects with elevated exposure to arsenic.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 455-462"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90066-7","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19721854","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90069-1
Juthika Koley , Sukti Sinha , A.K. Basak , M. Das , S.N. Dube , Pradip K. Majumder , Arvind K. Gupta , Shyamal Dasgupta , Biswanath Koley
The present study demonstrated cardiorespiratory effects of a synthetic phosphorus-containing ichthyotoxic metabolite elaborated by the marine dinoflagellate Ptychodiscus brevis in anaesthetised cats. The metabolite at a dose of 0.25–1.5 mg/kg i.v., resulted in a dose-dependent fall in blood pressure and such vasodepressor effect was associated with bradycardia. There is initial respiratory apnoea ollowed by increased rate and depth of respiration (hyperapnoea) following the administration of the toxin. The hypotensive response was accompanied by a decrease in aortic baroreceptor activity. The ECG showed atrioventricular conduction block, arrhythmia and depression of S-T segment and T wave which indicated coronary insufficiency. Vasodepressive property of the toxin is presumably muscarinic in nature as atropine counteracted the vasodepression.
{"title":"Cardiovascular and respiratory changes following exposure to a synthetic toxin of Ptychodiscus brevis","authors":"Juthika Koley , Sukti Sinha , A.K. Basak , M. Das , S.N. Dube , Pradip K. Majumder , Arvind K. Gupta , Shyamal Dasgupta , Biswanath Koley","doi":"10.1016/0926-6917(95)90069-1","DOIUrl":"10.1016/0926-6917(95)90069-1","url":null,"abstract":"<div><p>The present study demonstrated cardiorespiratory effects of a synthetic phosphorus-containing ichthyotoxic metabolite elaborated by the marine dinoflagellate <em>Ptychodiscus brevis</em> in anaesthetised cats. The metabolite at a dose of 0.25–1.5 mg/kg i.v., resulted in a dose-dependent fall in blood pressure and such vasodepressor effect was associated with bradycardia. There is initial respiratory apnoea ollowed by increased rate and depth of respiration (hyperapnoea) following the administration of the toxin. The hypotensive response was accompanied by a decrease in aortic baroreceptor activity. The ECG showed atrioventricular conduction block, arrhythmia and depression of S-T segment and T wave which indicated coronary insufficiency. Vasodepressive property of the toxin is presumably muscarinic in nature as atropine counteracted the vasodepression.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 483-486"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90069-1","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19721857","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 1995-12-07DOI: 10.1016/0926-6917(95)90049-7
Diadelis Remirez , Jan N.M. Commandeur, Ed Groot, Nico P.E. Vermeulen
The protective effects of lobenzarit, an antioxidative agent and antirheumatic drug, on the cytotoxicity of paracetamol in rat hepatocytes were studied, as well as the inhibitory effects of lobenzarit on cytochrome P-450s and glutathione S-transferases (GSTs) in rat liver. Paracetamol was selected as a model toxin, since it is known to be bioactivated by specific cytochrome P-450s presumably to N-acetyl-p-benzoquinoneimine, a reactive metabolite which upon overdasage of paracetamol causes protein and non-protein thiol depletion, lipid peroxidation and cytotoxicity measurable as LDH leakage. At concentrations of lobenzarit of 0.2 and 0.3 mM, added 30 min before paracetamol, the drug prevented paracetamol-induced leakage of lactate dehydrogenase (LDH) almost completely and lipid peroxidation (LPO) and depletion of glutathione (GSH) substantially and also the formation of the 3-glutathionyl conjugate of paracetamol. However, at a concentration of 0.05 mM lobenzarit did not protect anymore against the paracetamol toxicity. When added to the hepatocytes 1 h and 2 h before paracetamol, 0.05 and 0.2 and 0.3 mM concentrations of lobenzarit did not protect against the cytotoxicity induced by paracetamol either. Lobenzarit did not inhibit cytochromes P-450 1A1/1A2, 2B1/2B2 abd 2E1 which were measured as ethoxyresorufin O-deethylation (EROD) activity in β-naphthoflavone-induced rat liver microsomes, as pentoxyresorufin de-pentylation (PROD) activity in phenobarbital-induced microsomes and as p-nitrophenol hydroxylation (PNPH) activity in pyrazol-induced microsomes. Lobenzarit did not show inhibition of glutathione S-transferase (GST) activity towards 1-chloro-2,4-dinitrobenzene (CDNB) in cytosol from liver of rats treated with phenobarbital, pyrazol and β-naphthoflavone either. It is concluded that the cytoprotective effect of lobenzarit is most likely due to its antioxidant effects and/or to its ability to stimulate GSH reductase.
{"title":"Mechanism of protection of lobenzarit against paracetamol-induced toxicity in rat hepatocytes","authors":"Diadelis Remirez , Jan N.M. Commandeur, Ed Groot, Nico P.E. Vermeulen","doi":"10.1016/0926-6917(95)90049-7","DOIUrl":"10.1016/0926-6917(95)90049-7","url":null,"abstract":"<div><p>The protective effects of lobenzarit, an antioxidative agent and antirheumatic drug, on the cytotoxicity of paracetamol in rat hepatocytes were studied, as well as the inhibitory effects of lobenzarit on cytochrome P-450s and glutathione <em>S</em>-transferases (GSTs) in rat liver. Paracetamol was selected as a model toxin, since it is known to be bioactivated by specific cytochrome P-450s presumably to <em>N</em>-acetyl-<em>p</em>-benzoquinoneimine, a reactive metabolite which upon overdasage of paracetamol causes protein and non-protein thiol depletion, lipid peroxidation and cytotoxicity measurable as LDH leakage. At concentrations of lobenzarit of 0.2 and 0.3 mM, added 30 min before paracetamol, the drug prevented paracetamol-induced leakage of lactate dehydrogenase (LDH) almost completely and lipid peroxidation (LPO) and depletion of glutathione (GSH) substantially and also the formation of the 3-glutathionyl conjugate of paracetamol. However, at a concentration of 0.05 mM lobenzarit did not protect anymore against the paracetamol toxicity. When added to the hepatocytes 1 h and 2 h before paracetamol, 0.05 and 0.2 and 0.3 mM concentrations of lobenzarit did not protect against the cytotoxicity induced by paracetamol either. Lobenzarit did not inhibit cytochromes P-450 1A1/1A2, 2B1/2B2 abd 2E1 which were measured as ethoxyresorufin <em>O</em>-deethylation (EROD) activity in β-naphthoflavone-induced rat liver microsomes, as pentoxyresorufin de-pentylation (PROD) activity in phenobarbital-induced microsomes and as <em>p</em>-nitrophenol hydroxylation (PNPH) activity in pyrazol-induced microsomes. Lobenzarit did not show inhibition of glutathione <em>S</em>-transferase (GST) activity towards 1-chloro-2,4-dinitrobenzene (CDNB) in cytosol from liver of rats treated with phenobarbital, pyrazol and β-naphthoflavone either. It is concluded that the cytoprotective effect of lobenzarit is most likely due to its antioxidant effects and/or to its ability to stimulate GSH reductase.</p></div>","PeriodicalId":100501,"journal":{"name":"European Journal of Pharmacology: Environmental Toxicology and Pharmacology","volume":"293 4","pages":"Pages 301-308"},"PeriodicalIF":0.0,"publicationDate":"1995-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/0926-6917(95)90049-7","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"19720878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}