Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Toxicol Mech Methods ; 30(9): 656-671, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32746684

RESUMO

Pyroxasulfone induced a low incidence of urinary bladder tumors in male rats in a 2-year bioassay at 1000 and 2000 ppm, with occasional urinary calculi. No increased incidence of tumors of any tissue occurred in female rats or in mice of either gender. We performed three short-term studies to evaluate early development of pyroxasulfone-induced urinary crystals and urothelial cytotoxicity with consequent regenerative proliferation. First, male rats were treated with dietary 50, 1000 or 2000 ppm pyroxasulfone for 1, 3 or 7 days. The urothelium was examined by light and scanning electron microscopy (LM, SEM) and bromodeoxyuridine labeling index (BrdU LI). In two other studies, male rats were treated with dietary 20 000 ppm pyroxasulfone for 1 week. Urine collected at various times of day was examined by SEM and energy dispersive spectroscopy (EDS) or by LM, SEM, EDS, and infrared spectroscopy (IFS). Urinary crystals were present at various time points. EDS and IFS showed some contained calcium; others contained organic matter. Cytotoxicity was detected by SEM as cellular swelling, craters, and necrosis and by LM as cellular hypertrophy. Increased cell proliferation was detected by LM (hyperplasia), SEM (piling up of round cells), and by increased BrdU LI. There was no evidence of increased apoptosis. These findings support a mode of action for pyroxasulfone-associated bladder tumors in male rats involving formation of urinary crystals leading to urothelial cytotoxicity and regenerative proliferation. This is a high dose phenomenon, therefore, pyroxasulfone is not likely to be carcinogenic to humans at exposure levels that do not cause crystals with subsequent calculi formation in the urinary tract.


Assuntos
Proliferação de Células/efeitos dos fármacos , Herbicidas/toxicidade , Isoxazóis/toxicidade , Sulfonas/toxicidade , Neoplasias da Bexiga Urinária/induzido quimicamente , Cálculos Urinários/induzido quimicamente , Urotélio/efeitos dos fármacos , Animais , Testes de Carcinogenicidade , Cristalização , Relação Dose-Resposta a Droga , Hiperplasia , Masculino , Necrose , Ratos Sprague-Dawley , Medição de Risco , Fatores de Tempo , Neoplasias da Bexiga Urinária/patologia , Cálculos Urinários/urina , Urotélio/ultraestrutura
2.
Toxicol Mech Methods ; 29(7): 488-498, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31050326

RESUMO

The increased incidence of testicular disorders in young men and the possible influence of environmental chemicals, such as dibutyl phthalate (DBP) and acrylamide (AA), requires experimental models for identifying modes of action. Most published reproductive toxicologic studies use RNA samples from the total testis to evaluate testicular gene expression; however, analyses of isolated cell types could provide a more specific tool. Among testicular germ cells, spermatogonia are critical since they represent the onset of spermatogenesis. This study aimed, (1) to establish a technique for spermatogonia isolation; (2) to apply this isolation technique to verify possible gene expression alterations (Pou5f1, Kitlg, Mki-67, Bak1 and Spry4) in prepubertal post-natal day, (PND24) and pubertal (PND45) testes after in utero and postnatal exposure to DBP or AA. The technique was efficient for isolation of a majority of spermatogonia. In utero DBP exposure led to reduced litter body weight at birth, reduced anogenital distance of male pups on PND4, and increased frequency of male nipple retention on PND14 compared to controls. DBP-exposed relative testes weights were reduced only at PND24 compared to control but they did not differ at PND45. DBP-exposed animals showed reduced expression levels of Pou5f1 and Mki67 on PND24, and reduced expression of Pou5f1 and Spry4 on PND45. AA exposure reduced expression of Pou5f1, Mki67, and Spry4 at PND45 although not significantly. Our results suggest that DBP acts by reducing cell proliferation and impairing differentiation in prepubertal and pubertal testes.


Assuntos
Acrilamida/toxicidade , Dibutilftalato/toxicidade , Poluentes Ambientais/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Espermatogônias/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Peso Corporal , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Masculino , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos Sprague-Dawley , Espermatogônias/patologia , Testículo/embriologia , Testículo/crescimento & desenvolvimento , Testículo/patologia
3.
Toxicol Pathol ; 46(3): 266-272, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29504493

RESUMO

Chronic progressive nephropathy (CPN) occurs commonly in rats, more frequently and severely in males than females. High-grade CPN is characterized by increased layers of the renal papilla lining, designated as urothelial hyperplasia in the International Harmonization of Nomenclature and Diagnostic Criteria classification. However, urothelium lining the pelvis is not equivalent to the epithelium lining the papilla. To evaluate whether the epithelium lining the renal papilla is actually urothelial in nature and whether CPN-associated multicellularity represents proliferation, kidney tissues from aged rats with CPN, from rats with multicellularity of the renal papilla epithelium of either low-grade or marked severity, and from young rats with normal kidneys were analyzed and compared. Immunohistochemical staining for uroplakins (urothelial specific proteins) was negative in the papilla epithelium in all rats with multicellularity or not, indicating these cells are not urothelial. Mitotic figures were rarely observed in this epithelium, even with multicellularity. Immunohistochemical staining for Ki-67 was negative. Papilla lining cells and true urothelium differed by scanning electron microscopy. Based on these findings, we recommend that the epithelium lining the papilla not be classified as urothelial, and the CPN-associated lesion be designated as vesicular alteration of renal papilla instead of hyperplasia and distinguished in diagnostic systems from kidney pelvis urothelial hyperplasia.


Assuntos
Epitélio/anatomia & histologia , Medula Renal/citologia , Insuficiência Renal Crônica/patologia , Animais , Masculino , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Urotélio/citologia
4.
Nutr Cancer ; 68(1): 105-12, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26699517

RESUMO

A reported linkage between processed (nitrite-treated) meat products and the incidence of colon cancer could be due to sodium nitrite (NaNO2) itself or to N-nitroso compounds produced from the nitrite. Exposure to nitrite occurs due to residual nitrite in processed meat and to salivary nitrite arising by reduction of nitrate in vegetables and drinking water. Here we tested whether NaNO2 could induce colonic aberrant crypts (ABC) or ABC foci (ACF), which are putative precursors of colon cancer. We fed NaNO2 in drinking water for 20-25 wk to groups of 8-20 adult female mice. After sacrifice, ABC and ACF were counted in 2-cm distal colonic segments. In Experiment 1, no significant differences in ABC/ACF induction were seen between groups of 13-14 A/J mice fed 0, 0.5, or 1.0 g NaNO2/l drinking water. NaNO2 also did not affect fasting blood glucose levels. In Experiment 2, we fed 0, 1.0, 1.25, or 1.5 g NaNO2/l water to groups of 15 CF-1 mice. Five of the mice fed 1.5 g NaNO2/l showed ABC, whereas all other groups showed only 0-2 ABC/group, including 0 ABC for the group fed 1.25 g NaNO2/l. Overall statistical analysis indicated a dose-response p trends of 0.04. Pairwise comparison of ABC between groups fed 1.25 and 1.5 g NaNO2/l showed p 0.02 for both ABC and ACF, but a similar comparison between the untreated and 1.5 g/l groups showed no significant effects. In Experiment 3, hot dogs (18% of diet), which were fed to CF-1 mice previously treated with azoxymethane, inhibited ABC and ACF induction, but this effect was not significant (P = 0.10-0.12). In conclusion, these results support the view that NaNO2 may be a risk factor for colon carcinogenesis.


Assuntos
Focos de Criptas Aberrantes/induzido quimicamente , Neoplasias Colorretais/induzido quimicamente , Nitrito de Sódio/toxicidade , Animais , Azoximetano/toxicidade , Feminino , Hemina/toxicidade , Camundongos
5.
Toxicol Pathol ; 42(5): 855-62, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23690446

RESUMO

Inorganic arsenic (arsenite and arsenate) at high exposures is a known human carcinogen, inducing tumors of the urinary bladder, skin, and lungs. In two experiments, we examined the urothelial proliferative effects of treatment with 173 ppm sodium arsenite (100 ppm arsenic) in the drinking water for 6 and 24 hr, and 3, 7, and 14 days in female F344 rats and 43.3 ppm sodium arsenite (25 ppm arsenic) in female C57BL/6 wild-type and arsenic (+3 oxidation state) methyltransferase knockout (As3mt KO) mice that are unable to methylate arsenicals. In the rat and both mouse genotypes, scanning electron microscopy showed cytotoxic urothelial changes as early as 6 hr after the start of arsenic exposure. The severity of As(III)-induced cytotoxic urothelial changes increased over time in the rat and in the As3mt KO mouse. Light microscopy showed an increase in urothelial hyperplasia in the rat. No significant increases in bromodeoxyuridine-labeling index were observed. The data support the hypothesis that the sequence of events in the mode of action for urothelial effects of orally administered inorganic arsenic in the rat and mouse involves superficial cytotoxicity with consequent regenerative increased cell proliferation similar to the findings associated with the administration of dimethylarsinic acid (DMA(V)) in rats.


Assuntos
Arsenitos/administração & dosagem , Arsenitos/toxicidade , Urotélio/efeitos dos fármacos , Administração Oral , Animais , Peso Corporal , Ácido Cacodílico/administração & dosagem , Ácido Cacodílico/toxicidade , Carcinógenos/administração & dosagem , Carcinógenos/toxicidade , Proliferação de Células/efeitos dos fármacos , Feminino , Hiperplasia/induzido quimicamente , Hiperplasia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Ratos , Ratos Endogâmicos F344 , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/patologia , Urotélio/patologia
6.
Toxicol Pathol ; 41(5): 709-21, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23125116

RESUMO

It is unclear whether the process of spontaneous and chemically induced hemangiosarcoma and hemangioma formation in mice involves the transformation of differentiated endothelial cells (ECs) or recruitment of multipotential bone marrow-derived hematopoietic stem cells or endothelial progenitor cells (EPCs), which show some degree of endothelial differentiation. In the present study, immunohistochemical staining for hematopoietic stem cell markers (CD45 and CD34), EC markers (vascular endothelial growth factor receptor 2 [VEGFR2], CD31, and factor VIII-related antigen), and a myeloid lineage marker (CD14) was employed to better define the origin of hemangiosarcomas and hemangiomas in mice. Staining was negative for CD45, factor VIII-related antigen, and CD14 and positive for CD34, VEGFR2, and CD31, indicating that mouse hemangiosarcomas and hemangiomas are composed of cells derived from EPCs expressing CD34, VEGFR2, and CD31 but not factor VIII-related antigen. The lack of CD45 expression suggests that mouse vascular tumors may arise from EPCs that are at a stage later than hematopoietic stem cells. Since factor VIII-related antigen expression is known to occur later than CD31 expression in EPCs, our observations may indicate that these tumor cells are arrested at a stage prior to complete differentiation.  In addition, myeloid lineage cells do not appear to contribute to hemangiosarcoma and hemangioma formation in mice.


Assuntos
Antígenos CD/análise , Células Endoteliais/metabolismo , Hemangioma/metabolismo , Hemangiossarcoma/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células Mieloides/metabolismo , Animais , Antígenos CD/química , Biomarcadores/análise , Biomarcadores/química , Células Endoteliais/química , Células Endoteliais/imunologia , Feminino , Hemangioma/induzido quimicamente , Hemangioma/imunologia , Hemangiossarcoma/induzido quimicamente , Hemangiossarcoma/imunologia , Células-Tronco Hematopoéticas/química , Células-Tronco Hematopoéticas/imunologia , Imuno-Histoquímica , Masculino , Camundongos , Mutagênicos/toxicidade , Células Mieloides/química , Células Mieloides/imunologia
7.
Toxicol Pathol ; 39(7): 1032-45, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21937740

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) agonists and PPARγ/α dual agonists are used in the treatment of type 2 diabetes mellitus and hyperlipidemias. In carcinogenicity studies, some of these agonists induced hemangiomas/hemangiosarcomas in mice, but not in rats. We hypothesized that increased endothelial cell (EC) proliferation may be involved in the mechanism of PPAR agonist-induced vascular tumors in mice. We previously showed that the sarcomagenic PPARγ agonist troglitazone (TG) increased EC proliferation in brown and white adipose tissue and liver in mice at sarcomagenic doses (400 and 800 mg/kg) after four weeks of treatment. In vitro, TG had a mitogenic effect on mouse microvascular mouse ECs by increasing cell proliferation and survival. The current studies showed that treatment of mouse ECs in vitro induced alterations in proliferation pathway gene expression, especially the expression of insulin-like growth factor-1, but had no effect on mouse oxidative stress pathways. In vivo, treatment with vitamin E did not inhibit TG-induced EC proliferation in liver and adipose tissue. In addition, no hypoxic effect was detected in adipose tissue of TG-treated mice; however, TG had a minor effect on hepatocellular hypoxia. These results provide additional evidence supporting a direct mitogenic effect in the mode of action of TG-induced hemangiosarcomas in mice.


Assuntos
Processos de Crescimento Celular/efeitos dos fármacos , Cromanos/farmacologia , Células Endoteliais/efeitos dos fármacos , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Tecido Adiposo/química , Animais , Antineoplásicos/farmacologia , Peso Corporal/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Ingestão de Líquidos/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fígado Gorduroso , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Hemangiossarcoma , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Histocitoquímica , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fígado/química , Fígado/efeitos dos fármacos , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Reprodutibilidade dos Testes , Troglitazona , Vitamina E/farmacologia
8.
Toxicol Lett ; 336: 32-38, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33176187

RESUMO

Tobacco smoking is a major risk factor for human cancers including urinary bladder carcinoma. In a previous study, nicotine enhanced rat urinary bladder carcinogenesis in a two-stage carcinogenesis model. Nicotine also induced cytotoxicity in the bladder urothelium in a short-term study. In the present study, male rats were treated with nicotine (40 ppm) in drinking water co-administered with the NADPH oxidase inhibitor, apocynin (0, 250 or 750 mg/kg) in diet for 4 weeks. The apocynin treatment induced no clinical toxic effects. Reduction of reactive oxygen species (ROS) by apocynin was confirmed by immunohistochemistry of 8-OHdG in the bladder urothelium. Incidences of simple hyperplasia, cell proliferation and apoptosis were reduced by apocynin treatment in the bladder urothelium. However, despite reduction of cell proliferation (labeling index), apocynin did not affect the incidence of simple hyperplasia, apoptosis, or ROS generation in the kidney pelvis urothelium, in addition to 8-OHdG positivity induced by nicotine being lower. In vitro, apocynin (500 µM) reduced ROS generation, but induced cell proliferation in bladder cancer cell lines (T24 and UMUC3 cells). These data suggest that oxidative stress may play a role in the cell proliferation of the bladder urothelium induced by nicotine.


Assuntos
Acetofenonas/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , NADPH Oxidases/antagonistas & inibidores , Nicotina , Neoplasias da Bexiga Urinária/prevenção & controle , Bexiga Urinária/efeitos dos fármacos , Urotélio/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Hiperplasia , Masculino , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos Endogâmicos F344 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Bexiga Urinária/enzimologia , Bexiga Urinária/ultraestrutura , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/enzimologia , Neoplasias da Bexiga Urinária/ultraestrutura , Urotélio/enzimologia , Urotélio/ultraestrutura
9.
Toxicol Appl Pharmacol ; 244(2): 99-105, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20045014

RESUMO

Based on epidemiological data, chronic exposure to high levels of inorganic arsenic in drinking water is carcinogenic to humans, inducing skin, urinary bladder and lung tumors. In vivo, inorganic arsenic is metabolized to organic methylated arsenicals including the highly toxic dimethylarsinous acid (DMA(III)) and monomethylarsonous acid (MMA(III)). Short-term treatment of rats with 100 microg/g trivalent arsenic (As(III)) as sodium arsenite in the diet or in drinking water induced cytotoxicity and necrosis of the urothelial superficial layer, with increased cell proliferation and hyperplasia. The objectives of this study were to determine if these arsenic-induced urothelial effects are dose responsive, the dose of arsenic at which urothelial effects are not detected, and the urinary concentrations of the arsenical metabolites. We treated female F344 rats for 5 weeks with sodium arsenite at dietary doses of 0, 1, 10, 25, 50, and 100 ppm. Cytotoxicity, cell proliferation and hyperplasia of urothelial superficial cells were increased in a dose-responsive manner, with maximum effects found at 50 ppm As(III). There were no effects at 1 ppm As(III). The main urinary arsenical in As(III)-treated rats was the organic arsenical dimethylarsinic acid (DMA(V)). The thio-metabolites dimethylmonothioarsinic acid (DMMTA(V)) and monomethylmonothioarsinic acid (MMMTA(V)) were also found in the urine of As(III)-treated rats. The LC(50) concentrations of DMMTA(V) for rat and human urothelial cells in vitro were similar to trivalent oxygen-containing arsenicals. These data suggest that dietary As(III)-induced urothelial cytotoxicity and proliferation are dose responsive, and the urothelial effects have a threshold corresponding to the urinary excretion of measurable reactive metabolites.


Assuntos
Arsenitos/administração & dosagem , Arsenitos/urina , Dieta , Compostos de Sódio/administração & dosagem , Compostos de Sódio/urina , Bexiga Urinária/efeitos dos fármacos , Urotélio/efeitos dos fármacos , Animais , Intoxicação por Arsênico/metabolismo , Intoxicação por Arsênico/urina , Arsenicais/metabolismo , Arsenitos/metabolismo , Linhagem Celular , Linhagem Celular Transformada , Relação Dose-Resposta a Droga , Feminino , Humanos , Metilação , Ratos , Ratos Endogâmicos F344 , Compostos de Sódio/metabolismo , Compostos de Sulfidrila/metabolismo , Compostos de Sulfidrila/urina , Bexiga Urinária/metabolismo , Urotélio/metabolismo
10.
Toxicol Appl Pharmacol ; 246(1-2): 1-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20423714

RESUMO

Arsenic (+3 oxidation state) methyltransferase (As3mt) catalyzes reactions which convert inorganic arsenic to methylated metabolites. This study determined whether the As3mt null genotype in the mouse modifies cytotoxic and proliferative effects seen in urinary bladders of wild type mice after exposure to inorganic arsenic. Female wild type C57BL/6 mice and As3mt KO mice were divided into 3 groups each (n=8) with free access to a diet containing 0, 100 or 150 ppm of arsenic as arsenite (As(III)). During the first week of As(III) exposure, As3mt KO mice exhibited severe and lethal systemic toxicity. At termination, urinary bladders of both As3mt KO and wild type mice showed hyperplasia by light microscopy. As expected, arsenic-containing granules were found in the superficial urothelial layer of wild type mice. In As3mt KO mice these granules were present in all layers of the bladder epithelium and were more abundant and larger than in wild type mice. Scanning electron microscopy of the bladder urothelium of As3mt KO mice treated with 100 ppm As(III) showed extensive superficial necrosis and hyperplastic changes. In As3mt KO mice, livers showed severe acute inflammatory changes and spleen size and lymphoid areas were decreased compared with wild type mice. Thus, diminished arsenic methylation in As3mt KO mice exacerbates systemic toxicity and the effects of As(III) on the bladder epithelium, showing that altered kinetic and dynamic behavior of arsenic can affect its toxicity.


Assuntos
Arsenitos/toxicidade , Metiltransferases/metabolismo , Bexiga Urinária/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Hiperplasia , Masculino , Metiltransferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Microscopia Eletrônica de Varredura , Bexiga Urinária/patologia , Bexiga Urinária/ultraestrutura
11.
Toxicology ; 429: 152325, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31678612

RESUMO

Tobacco smoking is a major risk factor for human cancers including urinary bladder carcinoma. In a previous study, nicotine enhanced rat urinary bladder carcinogenesis using a rat urinary bladder two-stage carcinogenesis model. In the present study, nicotine metabolites (cotinine, trans-3'-hydroxy cotinine and N'-nitrosonornicotine) were evaluated in a cell proliferation assay using urinary bladder urothelial cell lines. Cotinine (0.1 to 1 mM) induced the highest cell proliferation compared to the others, including nicotine, in three bladder cancer cell lines (RT4, T24 and UMUC3 cells). By Western blot, cotinine induced phosphorylation of Stat3 and expression of cyclin D1 in UMUC3 cells. The cell proliferation induced by cotinine was blocked by inhibitors of nicotinic receptors (10 nM SR16584 or 10 µM methyllycaconitine citrate) and Stat3 (100 nM stattic). In an in vivo study, cotinine (13, 40 and 120 ppm) in drinking water also induced cell proliferation and simple hyperplasia in urinary bladder and renal pelvis urothelium of rats, but to a lesser degree compared to nicotine (40 ppm). Cytotoxicity detected by scanning electron microscopy and apoptosis in the bladder urothelium were induced by nicotine but not cotinine. These data suggest that cotinine is able to induce urothelial cell proliferation both in vitro and in vivo, and high urinary concentrations may enhance urothelial carcinogenesis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cotinina/toxicidade , Nicotina/toxicidade , Urotélio/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Carcinogênese/induzido quimicamente , Linhagem Celular Tumoral , Cotinina/análogos & derivados , Humanos , Masculino , Nicotina/metabolismo , Nitrosaminas/toxicidade , Ratos , Ratos Endogâmicos F344 , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/patologia , Urotélio/citologia
12.
Toxicol Appl Pharmacol ; 237(1): 83-90, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19285096

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) agonists and PPARgamma/alpha dual agonists have been or are being developed for clinical use in the treatment of type 2 diabetes mellitus and hyperlipidemias. A common tumor finding in rodent carcinogenicity studies for these agonists is hemangioma/hemangiosarcoma in mice but not in rats. We hypothesized that increased endothelial cell proliferation may be involved in the mechanism of PPAR agonist-induced vascular tumors in mice, and we investigated the effects on endothelial cells utilizing troglitazone, the first clinically used PPARgamma agonist, in vivo and in vitro. Troglitazone (400 and 800 mg/kg/day) induced hemangiosarcomas in mice in a 2-year bioassay. We showed that troglitazone increased endothelial cell proliferation in brown and white adipose tissue and liver in mice at sarcomagenic doses after 4 weeks of treatment. Troglitazone was cytotoxic both to human dermal microvascular endothelial cells (HMEC1) and mouse mammary fat pad microvascular endothelial cells (MFP MVEC) at high concentrations. However, MFP MVEC were more resistant to the cytotoxic effects of troglitazone based on the much lower LC(50) in HMEC1 (17.4 muM) compared to MFP MVEC (92.2 muM). Troglitazone increased the proliferation and survival of MFP MVEC but not HMEC1 in growth factor reduced conditions. Our data demonstrate that troglitazone may induce hemangiosarcomas in mice, at least in part, through enhancement of survival and proliferation of microvascular endothelial cells. Such an effect does not occur with human cells, suggesting that human may react differently to exposure to PPAR agonists compared with mice.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cromanos/toxicidade , Células Endoteliais/efeitos dos fármacos , Hemangiossarcoma/induzido quimicamente , PPAR gama/metabolismo , Tiazolidinedionas/toxicidade , Tecido Adiposo/efeitos dos fármacos , Animais , Testes de Carcinogenicidade , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Feminino , Humanos , Dose Letal Mediana , Estudos Longitudinais , Camundongos , Microvasos/citologia , Microvasos/efeitos dos fármacos , PPAR gama/agonistas , PPAR gama/genética , RNA Mensageiro/análise , Distribuição Aleatória , Especificidade da Espécie , Estatísticas não Paramétricas , Troglitazona
13.
Toxicol Lett ; 187(2): 124-9, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19429254

RESUMO

Arsenite (As(III)), an inorganic arsenical, is a known human carcinogen, inducing tumors of the skin, urinary bladder and lung. It is known to be metabolized to organic methylated arsenicals in vivo. As(III) has been reported to have the ability to up-regulate the epidermal growth factor receptor (EGFR)-associated pathway in epithelial cells, including human urothelial cells in vitro. EGFR is a cell-surface receptor belonging to the ErbB family of receptor tyrosine kinases, and the EGFR-associated signaling pathway has been reported to play an important role in carcinogenesis and cancer progression, including in bladder cancer. In this study, we investigated the growth effects of As(III) and an organic trivalent arsenical, dimethylarsinous acid (DMA(III)), and the effects of co-exposure of gefitinib, an EGFR inhibitor, with As(III) to a rat urothelial cell line (MYP3). We also investigated the effects of co-administration of dietary As(III) and gefitinib in vivo. In vitro, concentrations of 1.0microM As(III) or 0.5microM DMA(III) induced cytotoxicity. However, lower concentrations of As(III) treatment had a slight mitogenic growth effect whereas lower concentrations of DMA(III) did not. Gefitinib blocked As(III)-induced cell growth in vitro. In vivo, a high dose of gefitinib alone induced slight urothelial cytotoxicity, and did not reduce cytotoxicity and regenerative cell proliferation when co-administered with As(III). The majority of arsenic metabolites present in the urine of As(III)-treated rats were organic arsenicals, mainly dimethylarsinic acid (DMA(V)). As(III) was also present, and its concentration was higher than the concentration required to produce cytotoxicity in vitro. These data suggest that an EGFR inhibitor has the ability to block As(III)-induced cell proliferation in vitro but not in vivo in a short-term study.


Assuntos
Arsenitos/toxicidade , Ácido Cacodílico/análogos & derivados , Receptores ErbB/antagonistas & inibidores , Quinazolinas/farmacologia , Bexiga Urinária/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Ácido Cacodílico/toxicidade , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Gefitinibe , Histocitoquímica , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Bexiga Urinária/enzimologia
14.
Food Chem Toxicol ; 133: 110793, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31473338

RESUMO

The toxicity of dietary E 171, a food grade titanium dioxide was evaluated. A recent study reported rats receiving E 171 in water developed inflammation and aberrant crypt foci (ACF) in the gastrointestinal tract. Here, rats received food containing E 171 (7 or 100 days). The 100-day study included feeding E 171 after dimethylhydrazine (DMH) or vehicle only pretreatment. Food consumption was similar between treatment groups with maximum total cumulative E 171 exposure being 2617 mg/kg in 7 days and 29,400 mg/kg in 100 days. No differences were observed due to E 171 in the percentage of dendritic, CD4+ T or Treg cells within Peyer's patches or the periphery, or in cytokine production in plasma, sections of jejunum, and colon in 7- or 100-day E 171 alone fed rats. Differences were observed for IL-17A in colon (400 ppm E 171 + DMH) and IL-12p70 in plasma (40 ppm E 171 + DMH). E 171 had no effect on histopathologic evaluations of small and large intestines, liver, spleen, lungs, or testes, and no effects on ACF, goblet cell numbers, or colonic gland length. Dietary E 171 administration (7- or 100-day), even at high doses, produced no effect on the immune parameters or tissue morphology.


Assuntos
Aditivos Alimentares/toxicidade , Mucosa Intestinal/efeitos dos fármacos , Titânio/toxicidade , 1,2-Dimetilidrazina/farmacologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Carcinógenos/farmacologia , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Aditivos Alimentares/química , Masculino , Tamanho da Partícula , Nódulos Linfáticos Agregados/efeitos dos fármacos , Ratos Wistar , Linfócitos T Reguladores/efeitos dos fármacos , Titânio/química
15.
Toxicol Pathol ; 36(7): 999-1005, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19126794

RESUMO

Based on epidemiological data, chronic exposure to high levels of inorganic arsenic in the drinking water is carcinogenic to the urinary bladder of humans. Recently, models have been developed involving transplacental administration of inorganic arsenic and subsequent administration of another substance that produces a low incidence of urogenital neoplasms. Administration of arsenite or arsenate in the diet or drinking water to five-to eight-week-old mice or rats rapidly induces urothelial cytotoxicity and regenerative hyperplasia. In mice administered arsenite, we observed eosinophilic intracytoplasmic granules present in the urothelial cells. These granules were not present in urothelial cells of untreated mice or in treated or untreated rats. By transmission electron microscopy, the granules were located within the mitochondrial matrix, that is, mitochondrial inclusions. Arsenic, primarily as arsenite, was present in partially purified mitochondria containing these granules. Cells containing the granules were not usually associated with degenerative changes. Lack of these granules in rats suggests that they are not necessary for inorganic arsenic-induced urothelial cytotoxicity or hyperplasia. These granules have also been observed with exposures to other metals in other tissues and other species, suggesting that they represent a protective mechanism against metal-induced toxicity.


Assuntos
Arsênio/toxicidade , Carcinógenos/toxicidade , Grânulos Citoplasmáticos/ultraestrutura , Mitocôndrias/ultraestrutura , Urotélio/efeitos dos fármacos , Animais , Arsênio/administração & dosagem , Arsenicais/análise , Testes de Carcinogenicidade , Carcinógenos/administração & dosagem , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/metabolismo , Feminino , Hiperplasia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Animais , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Urotélio/patologia , Urotélio/ultraestrutura
16.
Cancer Res ; 66(15): 7793-800, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16885383

RESUMO

Exposure to estrogens is associated with an increased risk of breast cancer. Our laboratory has shown that the ACI rat is uniquely susceptible to 17beta-estradiol (E2)-induced mammary cancer. We previously mapped two loci, Emca1 and Emca2 (estrogen-induced mammary cancer), that act independently to determine susceptibility to E2-induced mammary cancer in crosses between the susceptible ACI rat strain and the genetically related, but resistant, Copenhagen (COP) rat strain. In this study, we evaluate susceptibility to E2-induced mammary cancer in a cross between the ACI strain and the unrelated Brown Norway (BN) rat strain. Whereas nearly 100% of the ACI rats developed mammary cancer when treated continuously with E2, BN rats did not develop palpable mammary cancer during the 196-day course of E2 treatment. Susceptibility to E2-induced mammary cancer segregated as a dominant or incompletely dominant trait in a cross between BN females and ACI males. In a population of 251 female (BN x ACI)F(2) rats, we observed evidence for a total of five genetic determinants of susceptibility. Two loci, Emca4 and Emca5, were identified when mammary cancer status at sacrifice was evaluated as the phenotype, and three additional loci, Emca6, Emca7, and Emca8, were identified when mammary cancer number was evaluated as the phenotype. A total of three genetic interactions were identified. These data indicate that susceptibility to E2-induced mammary cancer in the BN x ACI cross behaves as a complex trait controlled by at least five loci and multiple gene-gene interactions.


Assuntos
Cocarcinogênese , Estradiol/farmacologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Animais , Mapeamento Cromossômico , Feminino , Predisposição Genética para Doença , Neoplasias Hipofisárias/genética , Ratos , Ratos Endogâmicos ACI , Ratos Endogâmicos BN
17.
Toxicol Lett ; 285: 94-103, 2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29305328

RESUMO

Polyhexamethylene biguanide (PHMB), an amphiphilic polymeric biocide, increased liver tumor incidence in male and female rats at 1000 and 1500 mg/L in drinking water, but not at 500 mg/L in previous studies. In another study, PHMB administered in diet at 4000 mg/kg was negative for hepatocellular tumors. The present studies evaluated bioavailability and distribution of PHMB administered in drinking water and diet and possible modes of action (MOA). PHMB in drinking water was unpalatable during the first 3 days, resulting in markedly decreased food consumption and decreased body weight. Ki-67 labeling index was increased in hepatocytes and endothelial cells dose responsively with PHMB administered in drinking water but not diet. Vitamin E had no effect on this. There was no cytotoxicity by histopathology or serum enzymes, and no increase in cytokines TNFα, IL-1α or NF-κB. Focal iron deposition in sinusoidal lining cells was detected. Microarray analyses were non-contributory. No effect on CAR or PPARα activation was detected. 14C-PHMB administered at 500, 1000, or 1500 mg/L in the drinking water or 4000 mg/kg in the diet was nearly completely absorbed and excreted in urine, with some fecal excretion. The hypothesized MOA for liver tumors induced by PHMB in drinking water is: 1) severe dehydration and starvation because of unpalatability, followed by ingestion with rapid absorption and urinary excretion; 2) increased hepatocyte proliferation; and 3) induction of hepatocellular foci and tumors. The PHMB-induced rat hepatocellular tumors are unlikely to pose a human cancer risk. However, the actual MOA has not been determined.


Assuntos
Biguanidas/toxicidade , Desinfetantes/toxicidade , Fígado/efeitos dos fármacos , Administração Oral , Animais , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Testes de Função Hepática , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar , Testes de Toxicidade
18.
Toxicology ; 398-399: 31-40, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29501575

RESUMO

Tobacco smoking is a major risk factor for human cancers including urinary bladder carcinoma. Cigarette smoke inhalation in mice and orally administered nicotine in rats and mice increased urothelial cell proliferation. Nicotine, a major component of smoke, induced cell proliferation in multiple cell types in vitro. In the present study, the enhancing effects of nicotine on F344 rat bladder carcinogenesis induced by N-butyl-N-(4-hydroxybutyl)nitrosamine (BBN) were examined. Nicotine administered in drinking water for 32 weeks following 4 weeks of BBN treatment significantly increased the incidence and number of urothelial carcinomas dose-dependently. Ki67 and pSTAT3 labeling indices and expression of nicotinic acetylcholine receptor alpha 7 (nAChRα7) in non-tumor bladder urothelial lesions were significantly increased by nicotine, but the TUNEL assay for apoptosis showed no increase. In a 4 week study, inhibitors of nicotinic acetylcholine receptor decreased nicotine-induced urothelial simple hyperplasia and Ki67 labeling index in the bladder and kidney pelvis at a single cytotoxic dose of nicotine (40 ppm). Urothelial cytotoxicity with regenerative proliferation was observed by light and scanning electron microscopy. In vitro, nicotine was not cytotoxic to rat or human immortalized urothelial cells (do not express nicotine receptors) below millimolar concentrations, nor in human RT4, T24 or UMUC3 urothelial carcinoma cells (express nicotine receptors). However, nicotine slightly, but statistically significantly, increased cell proliferation at micromolar concentrations in human urothelial carcinoma cells. These data suggest that nicotine enhances urinary bladder carcinogenesis by inducing cytotoxicity with regenerative proliferation. The possible role of direct mitogenesis, involving nAChR and STAT3 signaling and of nicotine receptors requires further investigation at non-cytotoxic doses of nicotine.


Assuntos
Nicotina/toxicidade , Neoplasias da Bexiga Urinária/induzido quimicamente , Bexiga Urinária/efeitos dos fármacos , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Carcinogênese/induzido quimicamente , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Nicotina/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/patologia
19.
Toxicol Rep ; 4: 32-38, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28959622

RESUMO

The in vitro arsenite (AsIII) cytotoxicity dose-response (DR) of human keratinocytes (HEK001) was examined at greater statistical resolution than ever previously reported using the MTT assay to determine cell viability. Fifty-four 96-well plates were treated with AsIII concentrations of 0.25, 0.5, 1, 2, 3, 4, 5, 7, 10, 15, 20, 25, or 30 µM. Because of unexpected variation in viability response patterns, a two-stage DR analysis was used in which data on plate-specific viability (%), estimated as 100% times the ratio of measured viability in exposed to unexposed cells, were fit initially to a generalized lognormal response function positing that HEK001 cells studied consisted of: a proportion P of relatively highly sensitive (HS) cells, a proportion Po of relatively resistant cells, and a remaining (1-P-Po) fraction of typical-sensitivity (TS) cells exhibiting the intermediate level of AsIII sensitivity characteristic of most cells in each assay. The estimated fractions P and Po were used to adjust data from all 54 plates (and from the 28 plates yielding the best fits) to reflect the condition that P = Po = 0 to provide detailed DR analysis specifically for TS cells. Four DR models fit to the combined adjusted data were each very predictive (R2 > 0.97) overall but were inconsistent with at least one of the data set examined (p < 10-5). Adjusted mean responses at ≤3 µM were approximately equal (p > 0.30) and exceeded 100% significance (p ≤ 10-6). A low-dose hormetic model provided the best fit to the combined adjusted data for TS cells (R2 = 0.995). Marked variability in estimates of P (the proportion of apparent HS cells) was unexpected, not readily explained, and warrants further study using additional cell lines and assay methods, and in vivo.

20.
Genetics ; 169(4): 2189-97, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15687265

RESUMO

Estrogens stimulate proliferation and enhance survival of the prolactin (PRL)-producing lactotroph of the anterior pituitary gland and induce development of PRL-producing pituitary tumors in certain inbred rat strains but not others. The goal of this study was to elucidate the genetic bases of estrogen-induced pituitary tumorigenesis in reciprocal intercrosses between the genetically related ACI and Copenhagen (COP) rat strains. Following 12 weeks of treatment with the synthetic estrogen diethylstilbestrol (DES), pituitary mass, an accurate surrogate marker of absolute lactotroph number, was increased 10.6-fold in ACI rats and 4.5-fold in COP rats. Composite interval mapping analyses of the phenotypically defined F(2) progeny from the reciprocal crosses identified six quantitative trait loci (QTL) that determine the pituitary growth response to DES. These loci reside on chromosome 6 [Estrogen-induced pituitary tumor (Ept)1], chromosome 3 (Ept2 and Ept6), chromosome 10 (Ept9), and chromosome 1 (Ept10 and Ept13). Together, these six Ept loci and one additional suggestive locus on chromosome 4 account for an estimated 40% of the phenotypic variance exhibited by the combined F(2) population, while 34% of the phenotypic variance was estimated to result from environmental factors. These data indicate that DES-induced pituitary mass behaves as a quantitative trait and provide information that will facilitate identification of genes that determine the tumorigenic response of the pituitary gland to estrogens.


Assuntos
Estrogênios/farmacologia , Hipófise/efeitos dos fármacos , Neoplasias Hipofisárias/induzido quimicamente , Neoplasias Hipofisárias/genética , Animais , Biomarcadores , Mapeamento Cromossômico , Cruzamentos Genéticos , Epistasia Genética , Estrogênios/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Marcadores Genéticos , Genótipo , Masculino , Modelos Genéticos , Modelos Estatísticos , Tamanho do Órgão , Fenótipo , Hipófise/patologia , Prolactina/metabolismo , Locos de Características Quantitativas , Ratos , Sensibilidade e Especificidade , Fatores Sexuais , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA