Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 336: 32-38, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33176187

ABSTRACT

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.


Subject(s)
Acetophenones/pharmacology , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , NADPH Oxidases/antagonists & inhibitors , Nicotine , Urinary Bladder Neoplasms/prevention & control , Urinary Bladder/drug effects , Urothelium/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Disease Models, Animal , Humans , Hyperplasia , Male , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Signal Transduction , Urinary Bladder/enzymology , Urinary Bladder/ultrastructure , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/ultrastructure , Urothelium/enzymology , Urothelium/ultrastructure
2.
Toxicol Mech Methods ; 30(9): 656-671, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32746684

ABSTRACT

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.


Subject(s)
Cell Proliferation/drug effects , Herbicides/toxicity , Isoxazoles/toxicity , Sulfones/toxicity , Urinary Bladder Neoplasms/chemically induced , Urinary Calculi/chemically induced , Urothelium/drug effects , Animals , Carcinogenicity Tests , Crystallization , Dose-Response Relationship, Drug , Hyperplasia , Male , Necrosis , Rats, Sprague-Dawley , Risk Assessment , Time Factors , Urinary Bladder Neoplasms/pathology , Urinary Calculi/urine , Urothelium/ultrastructure
3.
Toxicology ; 429: 152325, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31678612

ABSTRACT

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.


Subject(s)
Cell Proliferation/drug effects , Cotinine/toxicity , Nicotine/toxicity , Urothelium/drug effects , Animals , Apoptosis/drug effects , Carcinogenesis/chemically induced , Cell Line, Tumor , Cotinine/analogs & derivatives , Humans , Male , Nicotine/metabolism , Nitrosamines/toxicity , Rats , Rats, Inbred F344 , Urinary Bladder/drug effects , Urinary Bladder/pathology , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/pathology , Urothelium/cytology
4.
Food Chem Toxicol ; 133: 110793, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31473338

ABSTRACT

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.


Subject(s)
Food Additives/toxicity , Intestinal Mucosa/drug effects , Titanium/toxicity , 1,2-Dimethylhydrazine/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , Carcinogenesis/drug effects , Carcinogens/pharmacology , Cytokines/metabolism , Dendritic Cells/drug effects , Food Additives/chemistry , Male , Particle Size , Peyer's Patches/drug effects , Rats, Wistar , T-Lymphocytes, Regulatory/drug effects , Titanium/chemistry
5.
Toxicol Mech Methods ; 29(7): 488-498, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31050326

ABSTRACT

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.


Subject(s)
Acrylamide/toxicity , Dibutyl Phthalate/toxicity , Environmental Pollutants/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Spermatogonia/drug effects , Testis/drug effects , Animals , Body Weight , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Gene Expression/drug effects , Male , Organ Size/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/pathology , Rats, Sprague-Dawley , Spermatogonia/pathology , Testis/embryology , Testis/growth & development , Testis/pathology
6.
Toxicology ; 398-399: 31-40, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29501575

ABSTRACT

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.


Subject(s)
Nicotine/toxicity , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder/drug effects , Administration, Oral , Animals , Body Weight/drug effects , Carcinogenesis/chemically induced , Cell Line , Cell Proliferation/drug effects , Humans , Male , Nicotine/administration & dosage , Random Allocation , Rats , Rats, Inbred F344 , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
7.
Toxicol Pathol ; 46(3): 266-272, 2018 04.
Article in English | MEDLINE | ID: mdl-29504493

ABSTRACT

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.


Subject(s)
Epithelium/anatomy & histology , Kidney Medulla/cytology , Renal Insufficiency, Chronic/pathology , Animals , Male , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Urothelium/cytology
8.
Toxicol Lett ; 285: 94-103, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29305328

ABSTRACT

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.


Subject(s)
Biguanides/toxicity , Disinfectants/toxicity , Liver/drug effects , Administration, Oral , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Hepatocytes/drug effects , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Liver Function Tests , Male , Oxidative Stress/drug effects , Rats, Wistar , Toxicity Tests
9.
Toxicol Rep ; 4: 32-38, 2017.
Article in English | MEDLINE | ID: mdl-28959622

ABSTRACT

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.

10.
Nutr Cancer ; 68(1): 105-12, 2016.
Article in English | MEDLINE | ID: mdl-26699517

ABSTRACT

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.


Subject(s)
Aberrant Crypt Foci/chemically induced , Colorectal Neoplasms/chemically induced , Sodium Nitrite/toxicity , Animals , Azoxymethane/toxicity , Female , Hemin/toxicity , Mice
11.
Toxicol Rep ; 2: 833-837, 2015.
Article in English | MEDLINE | ID: mdl-28962419

ABSTRACT

Chronic exposure to high levels of inorganic arsenic (iAs) has been associated with cancerous and non-cancerous health effects, including cardiovascular effects. However, the mechanism for a presumed toxic effect of arsenic on vascular tissue is not clear. Our working hypothesis is that inorganic trivalent arsenic and its methylated metabolites react with cysteine-containing cellular proteins and alter their function leading to adverse events such as cytotoxicity or proliferation. In this study, human microvascular endothelial cells (HMEC1) and mouse microvascular endothelial cells (MFP-MVEC) were exposed to arsenite (iAsIII), monomethylarsonous acid (MMAIII), or dimethylarsinous acid (DMAIII) for 72 h to evaluate cytotoxicity, and for 24, 48 or 72 h to evaluate cell proliferation. Both cell lines showed similar LC50 values, from 0.1 to 2.4 µM, for all three trivalent arsenicals. The endothelial cells treated with1 nM to 1 µM concentrations of the three trivalent arsenicals did not show increased cell proliferation at 24, 48 or 72 h or increased rate of proliferation at 72 h of exposure. Overall, cytotoxicity of trivalent arsenicals to microvascular endothelial cells is similar to their cytotoxicity to epithelial cells, and that these compounds are not mitogenic.

12.
Toxicol Pathol ; 42(5): 855-62, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23690446

ABSTRACT

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.


Subject(s)
Arsenites/administration & dosage , Arsenites/toxicity , Urothelium/drug effects , Administration, Oral , Animals , Body Weight , Cacodylic Acid/administration & dosage , Cacodylic Acid/toxicity , Carcinogens/administration & dosage , Carcinogens/toxicity , Cell Proliferation/drug effects , Female , Hyperplasia/chemically induced , Hyperplasia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Rats , Rats, Inbred F344 , Urinary Bladder/drug effects , Urinary Bladder/pathology , Urothelium/pathology
13.
Toxicology ; 315: 49-54, 2014 Jan 06.
Article in English | MEDLINE | ID: mdl-24269753

ABSTRACT

Tobacco smoking is a major risk factor for multiple human cancers including urinary bladder carcinoma. Tobacco smoke is a complex mixture containing chemicals that are known carcinogens in humans and/or animals. Aromatic amines a major class of DNA-reactive carcinogens in cigarette smoke, are not present at sufficiently high levels to fully explain the incidence of bladder cancer in cigarette smokers. Other agents in tobacco smoke could be excreted in urine and enhance the carcinogenic process by increasing urothelial cell proliferation. Nicotine is one such major component, as it has been shown to induce cell proliferation in multiple cell types in vitro. However, in vivo evidence specifically for the urothelium is lacking. We previously showed that cigarette smoke induces increased urothelial cell proliferation in mice. In the present study, urothelial proliferative and cytotoxic effects were examined after nicotine treatment in mice and rats. Nicotine hydrogen tartrate was administered in drinking water to rats (52 ppm nicotine) and mice (514 ppm nicotine) for 4 weeks and urothelial changes were evaluated. Histopathologically, 7/10 rats and 4/10 mice showed simple hyperplasia following nicotine treatment compared to none in the controls. Rats had an increased mean BrdU labeling index compared to controls, although it was not statistically significantly elevated in either species. Scanning electron microscopic visualization of the urothelium did not reveal significant cytotoxicity. These findings suggest that oral nicotine administration induced urothelial hyperplasia (increased cell proliferation), possibly due to a mitogenic effect of nicotine and/or its metabolites.


Subject(s)
Cell Proliferation/drug effects , Nicotine/toxicity , Nicotinic Agonists/toxicity , Urothelium/drug effects , Administration, Oral , Animals , Female , Hyperplasia/chemically induced , Hyperplasia/pathology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Rats , Rats, Wistar , Urothelium/pathology
14.
Toxicol Sci ; 137(1): 36-46, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24097667

ABSTRACT

Inorganic arsenic (iAs) is a known human carcinogen at high exposures, increasing the incidences of urinary bladder, skin, and lung cancers. In most mammalian species, ingested iAs is excreted mainly through urine primarily as dimethylarsinic acid (DMA(V)). In wild-type (WT) mice, iAs, DMA(V), and dimethylarsinous acid (DMA(III)) exposures induce formation of intramitochondrial urothelial inclusions. Arsenite (iAs(III)) also induced intranuclear inclusions in arsenic (+3 oxidation state) methyltransferase knockout (As3mt KO) mice. The arsenic-induced formation of inclusions in the mouse urothelium was dose and time dependent. The inclusions do not occur in iAs-treated rats and do not appear to be related to arsenic-induced urothelial cytotoxicity. Similar inclusions in exfoliated urothelial cells from humans exposed to iAs have been incorrectly identified as micronuclei. We have characterized the urothelial inclusions using transmission electron microscopy (TEM), DNA-specific 4',6-diamidino-2-phenylindole (DAPI), and non-DNA-specific Giemsa staining and determined the arsenical content. The mouse inclusions stained with Giemsa but not with the DAPI stain. Analysis of urothelial mitochondrial- and nuclear-enriched fractions isolated from WT (C57BL/6) and As3mt KO mice exposed to arsenate (iAs(V)) for 4 weeks showed higher levels of iAs(V) in the treated groups. iAs(III) was the major arsenical present in the enriched nuclear fraction from iAs(V)-treated As3mt KO mice. In conclusion, the urothelial cell inclusions induced by arsenicals appear to serve as a detoxifying sequestration mechanism similar to other metals, and they do not represent micronuclei.


Subject(s)
Cacodylic Acid/analogs & derivatives , Carcinogens/toxicity , Cell Nucleus/drug effects , Inclusion Bodies/drug effects , Mitochondria/drug effects , Urinary Bladder/drug effects , Urothelium/drug effects , Animals , Cacodylic Acid/toxicity , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Dose-Response Relationship, Drug , Female , Inclusion Bodies/metabolism , Inclusion Bodies/ultrastructure , Methyltransferases/deficiency , Methyltransferases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Time Factors , Urinary Bladder/metabolism , Urinary Bladder/ultrastructure , Urothelium/metabolism , Urothelium/ultrastructure
15.
Toxicology ; 314(2-3): 238-46, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24172598

ABSTRACT

Diuron is carcinogenic to the rat urinary bladder at high dietary levels. The proposed mode of action (MOA) for diuron is urothelial cytotoxicity and necrosis followed by regenerative urothelial hyperplasia. Diuron-induced urothelial cytotoxicity is not due to urinary solids. Diuron is extensively metabolized, and in rats, N-(3,4-dichlorophenyl)urea (DCPU) and 4,5-dichloro-2-hydroxyphenyl urea (2-OH-DCPU) were the predominant urinary metabolites; lesser metabolites included N-(3,4-dichlorophenyl)-3-methylurea (DCPMU) and trace levels of 3,4-dichloroaniline (DCA). In humans, DCPMU and DCPU have been found in the urine after a case of product abuse. To aid in elucidating the MOA of diuron and to evaluate the metabolites that are responsible for the diuron toxicity in the bladder epithelium, we investigated the urinary concentrations of metabolites in male Wistar rats treated with 2500ppm of diuron, the urothelial cytotoxicity in vitro of the metabolites and their gene expression profiles. DCPU was found in rat urine at concentrations substantially greater than the in vitro IC50 and induced more gene expression alterations than the other metabolites tested. 2-OH-DCPU was present in urine at a concentration approximately half of the in vitro IC50, whereas DCPMU and DCA were present in urine at concentrations well below the IC50. For the diuron-induced MOA for the rat bladder, we suggest that DCPU is the primary metabolite responsible for the urothelial cytotoxicity with some contribution also by 2-OH-DCPU. This study supports a MOA for diuron-induced bladder effects in rats consisting of metabolism to DCPU (and 2-OH-DCPU to a lesser extent), concentration and excretion in urine, urothelial cytotoxicity, and regenerative proliferation.


Subject(s)
Diuron/metabolism , Diuron/toxicity , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Animals , Cell Line , Herbicides/metabolism , Herbicides/toxicity , Humans , Male , Rats , Rats, Wistar , Urothelium/cytology , Urothelium/drug effects , Urothelium/metabolism
16.
Toxicology ; 305: 130-5, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23376817

ABSTRACT

Chronic exposure to inorganic arsenic (iAs) is carcinogenic to the human urinary bladder. It produces urothelial cytotoxicity and proliferation in rats and mice. DMA(V), a major methylated urinary metabolite of iAs, is a rat bladder carcinogen, but without effects on the mouse urothelium. DMA(III) was shown to be the likely urinary metabolite of DMA(V) inducing urothelial changes and is also postulated to be one of the active metabolites of iAs. To evaluate potential DMA(III)-induced urothelial effects, it was administered to As3mt knockout mice which cannot methylate arsenicals. Female C57BL/6 wild type and As3mt knockout mice (10/group) were administered DMA(III), 77.3ppm in water for four weeks. Urothelial effects were evaluated by light and scanning electron microscopy (EM) and immunohistochemical detection of bromodeoxyuridine (BrdU) incorporation. EM findings were rated 1-5, with higher rating indicating greater extent of cytotoxicity visualized. DMA(III) significantly increased the BrdU labeling index, a ratio of BrdU labeled cells to non-labeled cells, in the treated knockout group compared to control and wild type treated groups. DMA(III) induced simple hyperplasia in more knockout mice (4/10) compared to wild type mice (2/10). All treated knockout mice had more and larger intracytoplasmic granules compared to the treated wild type mice. Changes in EM classification were not significant. In conclusion, DMA(III) induces urothelial toxicity and regenerative hyperplasia in mice and most likely plays a role in inorganic arsenic-induced urothelial changes. However, DMA(V) does not induce hyperplasia in mice, suggesting that urinary concentrations of DMA(III) do not reach cytotoxic levels in DMA(V)-treated mice.


Subject(s)
Cacodylic Acid/analogs & derivatives , Diet , Methyltransferases/metabolism , Urinary Bladder/drug effects , Urothelium/drug effects , Animals , Antimetabolites , Bromodeoxyuridine , Cacodylic Acid/toxicity , Cell Death/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Female , Genotype , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Oxidation-Reduction , Urinary Bladder/pathology , Urothelium/pathology
17.
Toxicol Pathol ; 41(5): 709-21, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23125116

ABSTRACT

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.


Subject(s)
Antigens, CD/analysis , Endothelial Cells/metabolism , Hemangioma/metabolism , Hemangiosarcoma/metabolism , Hematopoietic Stem Cells/metabolism , Myeloid Cells/metabolism , Animals , Antigens, CD/chemistry , Biomarkers/analysis , Biomarkers/chemistry , Endothelial Cells/chemistry , Endothelial Cells/immunology , Female , Hemangioma/chemically induced , Hemangioma/immunology , Hemangiosarcoma/chemically induced , Hemangiosarcoma/immunology , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/immunology , Immunohistochemistry , Male , Mice , Mutagens/toxicity , Myeloid Cells/chemistry , Myeloid Cells/immunology
18.
Toxicol Sci ; 130(2): 281-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22923491

ABSTRACT

Diuron, a substituted urea herbicide, is carcinogenic to the rat urinary bladder at high dietary levels (2500 ppm). To further elucidate the mode of action, this study aimed to determine the time course and sequence of bladder cytotoxic and proliferative changes induced by diuron treatment of male Wistar rats. Rats were randomized into two groups (control and 2500 ppm diuron) and treated for 28 days. Ten rats from each group were terminated on each of study days 1, 3, 7, or 28. Scanning electron micro scopy (SEM) showed urothelial cell swelling beginning on day 1, and by day 28, showed extensive necrosis, exfoliation and piling up of cells suggestive of hyperplasia. No difference in the bromo deoxyuridine labeling index was detected. In a second experiment, rats were randomized into control and diuron-treated groups and treated for 7 days or 8 weeks. After 7 days, transmission electron microscopy showed cell degenerative changes and distention of the cytoplasm, organelles, and nuclei characteristic of cytolysis. This resulted in protrusion of the superficial cells into the lumen, corresponding to the cell swelling observed previously by SEM. After 8 weeks, bladders in the diuron-treated group showed an increased incidence of simple hyperplasia by light microscopy (6/10, p < 0.05) compared with controls (0/10) and a significantly different SEM classification. In summary, our results support the hypothesis that urothelial cytotoxicity followed by regenerative cell proliferation are the sequential key events that occur with high-dose diuron exposure in rats.


Subject(s)
Diuron/toxicity , Epithelial Cells/drug effects , Herbicides/toxicity , Urinary Bladder/drug effects , Urothelium/drug effects , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Drinking/drug effects , Eating/drug effects , Epithelial Cells/ultrastructure , Hyperplasia , Kidney/drug effects , Kidney/pathology , Male , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Necrosis , Rats , Rats, Wistar , Regeneration/drug effects , Time Factors , Urinary Bladder/ultrastructure , Urothelium/ultrastructure
19.
Toxicology ; 299(2-3): 155-9, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22664484

ABSTRACT

Inorganic arsenic is a known human carcinogen, inducing tumors of the skin, urinary bladder and lung. It is metabolized to organic methylated arsenicals. 2,3-Dimercaptopropane-1-sulfonic acid (DMPS), a chelating agent, is capable of reducing pentavalent arsenicals to the trivalent state and binding to the trivalent species, and it has been used in the treatment of heavy metal poisoning in humans. Therefore, we investigated the ability of DMPS to inhibit the cytotoxicity and regenerative urothelial cell proliferation induced by arsenate administration in vivo. Female rats were treated for 4 weeks with 100 ppm As(V). DMPS (2800 ppm) co-administered in the diet significantly reduced the As(V)-induced cytotoxicity of superficial cells detected by scanning electron microscopy (SEM), and the incidence of simple hyperplasia observed by light microscopy and the bromodeoxyuridine (BrdU) labeling index. It also reduced the total concentration of arsenicals in the urine and the methylation of arsenic. There were no differences in oxidative stress as assessed by immunohistochemical staining for 8-hydroxy-2'-deoxyguanosine (8OHdG) of the bladder urothelium. No differences were detected in urine sediments between groups. These data suggest that DMPS has the ability to inhibit both arsenate-induced acute toxicity and regenerative proliferation of the rat bladder epithelium, most likely by decreasing exposure of the urothelium to trivalent arsenicals excreted in the urine. These data provide additional evidence that the effects of arsenate exposure in vivo do not appear to be related to oxidative effects on dG in DNA.


Subject(s)
Arsenates/toxicity , Chelating Agents/pharmacology , Unithiol/pharmacology , Urinary Bladder/drug effects , Urothelium/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Arsenates/urine , Cell Line , Cell Survival/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Female , Immunohistochemistry , Microscopy, Electron, Scanning , Random Allocation , Rats , Rats, Inbred F344 , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urothelium/metabolism , Urothelium/pathology
20.
Toxicol Sci ; 128(1): 1-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22499580

ABSTRACT

Essential oils from mint plants, including peppermint and pennyroyal oils, are used at low levels as flavoring agents in various foods and beverages. Pulegone is a component of these oils. In a 2-year bioassay, oral administration of pulegone slightly increased the urothelial tumor incidence in female rats. We hypothesized that its mode of action (MOA) involved urothelial cytotoxicity and increased cell proliferation, ultimately leading to tumors. Pulegone was administered by gavage at 0, 75, or 150 mg/kg body weight to female rats for 4 and 6 weeks. Fresh void urine and 18-h urine were collected for crystal and metabolite analyses. Urinary bladders were evaluated by light microscopy and scanning electron microscopy (SEM) and bromodeoxyuridine (BrdU) labeling index. Pulegone and its metabolites, piperitenone, piperitone, menthofuran, and menthone, were tested for cytotoxicity in rat (MYP3) and human (1T1) urothelial cells by the 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. No abnormal urinary crystals were observed by light microscopy. Urine samples (18-h) showed the presence of pulegone, piperitone, piperitenone, and menthofuran in both treated groups. By SEM, bladders from treated rats showed superficial necrosis and exfoliation. There was a significant increase in the BrdU labeling index in the high-dose group. In vitro studies indicated that pulegone and its metabolites, especially piperitenone, are excreted and concentrated in the urine at cytotoxic levels when pulegone is administered at high doses to female rats. The present study supports the hypothesis that cytotoxicity followed by regenerative cell proliferation is the MOA for pulegone-induced urothelial tumors in female rats.


Subject(s)
Monoterpenes/pharmacology , Urinary Bladder/drug effects , Animals , Body Weight/drug effects , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Cells, Cultured , Cyclohexane Monoterpenes , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Microscopy, Electron, Scanning , Rats , Rats, Inbred F344 , Urinary Bladder/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...