RESUMO
A single germ line gene mutation at a tumor susceptibility locus in a rodent model of hereditary human renal cancer caused a 70-fold increase in susceptibility to chemical carcinogenesis. A carcinogen that targeted both renal epithelial and mesenchymal cells caused an increase in tumors of epithelial origin in susceptible animals; the number of carcinogen-induced mesenchymal tumors was unaffected by the presence of the mutation at the susceptibility locus. Thus, this mutation defines a genetic locus for susceptibility to carcinogen-induced tumors and modulation of carcinogen susceptibility by this locus exhibits cell-type specificity.
Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Animais , Carcinoma de Células Renais/patologia , Dimetilnitrosamina , Modelos Animais de Doenças , Córtex Renal/patologia , Neoplasias Renais/patologia , Ratos , Ratos MutantesRESUMO
We tested whether a concentration of unleaded gasoline (UG) vapor that was selectively hepatocarcinogenic in female mice in a chronic bioassay is antiestrogenic and whether liver tumor promotion by UG is secondary to antiestrogenicity. Twelve-day-old female C57BL/6 x C3H F1 mice (hereafter called B6C3F1) received i.p. injections of N-nitrosodiethylamine (5 mg/kg) or vehicle. Beginning at 5-7 weeks of age, mice were exposed to 0, 292, or 2056 ppm of PS-6 blend UG vapor for 6 h/day, 5 days/week for 16 weeks, 1 ppm ethinyl estradiol (EE2) in the diet, or 2056 ppm UG vapor and 1 ppm EE2 in the diet. Treatment with 2026 ppm UG but not 292 ppm UG increased relative liver weight, the number of macroscopic hepatic neoplasms, and the size and volume fraction of altered hepatic foci in N-nitrosodiethylamine-initiated mice. Treatment with 2056 ppm UG reduced relative uterus, ovary, and pituitary weights but did not change serum 17 beta-estradiol levels, uterine peroxidase activity, or uterine cytosolic estrogen receptor levels. EE2 treatment reduced the number and size of altered hepatic foci in N-nitrosodiethylamine-initiated mice, caused weight loss, anestrus, vaginal keratinization, decreased uterine peroxidase activity, and decreased uterine cytosolic estrogen receptor levels. UG/EE2 co-treatment attenuated the weight loss, anestrus, and vaginal keratinization caused by EE2 treatment alone but dramatically increased the number of macroscopic hepatic neoplasms and the size and volume fraction of altered hepatic foci as compared to UG treatment alone. Thus, in this two-stage model of carcinogenesis (a) 2056 ppm UG had antiestrogenic effects, particularly with respect to pharmacological actions of EE2; (b) 2056 ppm UG but not 292 ppm UG acted as a liver tumor promoter; (c) EE2 inhibited liver tumor promotion; and (d) EE2 strongly potentiated liver tumor promotion by UG. These data demonstrate significant individual and interactive effects of UG vapor and estrogens in liver tumor promotion in female mice.
Assuntos
Estrogênios/toxicidade , Gasolina/toxicidade , Neoplasias Hepáticas Experimentais/induzido quimicamente , Animais , Peso Corporal/efeitos dos fármacos , Dietilnitrosamina , Interações Medicamentosas , Estradiol/sangue , Antagonistas de Estrogênios/farmacologia , Feminino , Genitália Feminina/efeitos dos fármacos , Genitália Feminina/patologia , Genitália Masculina/efeitos dos fármacos , Genitália Masculina/patologia , Fígado/citologia , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hipófise/patologia , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/patologia , GravidezRESUMO
Recently 4 genes (plasminogen activator inhibitor 2, interleukin 1 beta, clone 1, and clone 141) that are transcriptionally or posttranscriptionally responsive to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) have been cloned from a human skin keratinocyte cell line. We determined whether these genes were expressed in the livers of Sprague-Dawley rats following exposure to TCDD and whether there was a relationship between expression and hepatic cell proliferation. TCDD was administered by using a dose loading/maintenance regimen to achieve rapid quasi-steady-state TCDD liver concentrations of 0.03, 30, or 150 ng/g of liver. Gene expression was determined by Northern analysis using polyadenylated mRNA isolated from liver tissue of male and female animals exposed to TCDD for 1 or 14 days and hybridized with the human complementary DNA clone corresponding to one of the four human TCDD-responsive genes. Under low-stringency hybridization conditions, only the expression of clone 1 could be detected. A dose- and time-dependent expression of this gene was observed in the liver of both male and female rats. Expression of clone 1 was not detected in rats subjected to either a two-thirds partial hepatectomy or exposure to a single administration of the hepatic tumor promoters Wy-14643, carbon tetrachloride, or phenobarbital at doses that induce hepatic cell proliferation. Liver:body weight ratios were elevated in rats exposed to the middle and high TCDD doses. Histopathological observation and analysis of serum enzyme levels indicated no evidence of TCDD-induced liver necrosis. Cell proliferation was evaluated immunohistochemically after 7-day 5-bromo-2'-deoxyuridine administration. No increase in total hepatic labeling index was observed for any of the TCDD-exposed treatment groups compared to controls at week 1 or week 2. An increase in the periportal hepatocyte proliferation labeling pattern was observed in TCDD-treated animals. While these results demonstrate that a human TCDD-responsive gene is expressed in the liver of TCDD-treated male and female Sprague-Dawley rats, the expression of this gene is not linked to hepatic cell proliferation or the sex-specific tumor-promoting activity of TCDD.
Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/fisiologia , Dibenzodioxinas Policloradas/toxicidade , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Fígado/anatomia & histologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Dibenzodioxinas Policloradas/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Fatores Sexuais , Fenômenos Fisiológicos da Pele , TransfecçãoRESUMO
The effect of feeding hypolipidemic peroxisome proliferators on the induction of altered hepatic foci (AHF) in Fischer rats was studied in order to determine whether such agents can induce or promote the development of AHF. In the first study, rats were fed ciprofibrate (10 mg/kg/day) for 1 yr. AHF, neoplastic nodules, and hepatocellular carcinomas were induced. The presence of putative gamma-glutamyltranspeptidase (GGT) activity was numerically the most common marker, although it was absent in larger foci and nodules. A deficiency in canalicular ATPase and glucose-6-phosphatase provided the best markers for the larger foci and nodules. In the second study, rats were subjected to partial hepatectomy, and half of the animals were then intubated with diethylnitrosamine (10 mg/kg). One wk later, rats were fed Wy-14,643 at concentrations of 0, 0.05, and 0.1% in the diet for 6 mo. At 6 mo, the number and volume of foci were increased by the feeding of Wy-14,643 after partial hepatectomy alone and were greatly increased when Wy-14,643 was fed after partial hepatectomy/diethylnitrosamine administration. Canalicular adenosine triphosphatase and glucose-6-phosphatase deficiencies were the most common markers of AHF, and AHF of these phenotypes occupied practically all of the focal volume. The larger AHF did not express GGT, and those foci exhibiting GGT were much less common and occupied very little volume. The absence of the GGT protein itself, as opposed to an inhibition of GGT activity, was verified by immunohistochemical staining using an antibody to GGT. These studies show that hypolipidemic peroxisome proliferators can stimulate an increase in AHF following a single dose of diethylnitrosamine and a mitotic stimulus, and they thus can act as promoters in two-stage liver carcinogenesis. GGT is a poor marker for identifying AHF induced by peroxisome proliferators during the early, premalignant phase of hepatocarcinogenesis.
Assuntos
Clofibrato/análogos & derivados , Ácido Clofíbrico/análogos & derivados , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Pirimidinas/farmacologia , Adenosina Trifosfatases/metabolismo , Animais , Ácido Clofíbrico/farmacologia , Dietilnitrosamina , Esquema de Medicação , Ácidos Fíbricos , Glucose-6-Fosfatase/metabolismo , Hepatectomia , Técnicas Imunológicas , Fígado/patologia , Ratos , Fatores de Tempo , gama-Glutamiltransferase/metabolismoRESUMO
Prolonged exposure to certain alkylating chemicals induces glial and meningeal tumours in rats, probably resulting from DNA damage to dividing neural cells. The present work evaluated DNA synthesis in the brains of untreated, young adult male F344 rats in order to define a BrdUrd infusion protocol to more adequately assess proliferation in slowly dividing neural cell populations. BrdUrd (2.5 to 160 mg/ml) was administered for 6 days via subcutaneous osmotic pumps. Clinical toxicity was not observed at any dose. The labelling index (LI; % of cells per brain area that incorporated BrdUrd) and unit length labelling index (ULLI; % of cells per meningeal length that incorporated BrdUrd) were calculated for selected regions by counting labelled neural cells in defined areas of the right hemisphere in coronal brain sections. Intensely stained cells were numerous in the cerebral subependymal layer (LI = 35.8%); scattered in cerebral white matter tracts (e.g. corpus callosum and internal capsule; LI = 6.2%) as well as cerebral (ULLI = 4.2%) and cerebellar (ULLI = 3.6%) meninges; and rare in the hippocampus (LI > 0.1%). Mildy stained cells were dispersed in the pons (LI = 2.1%), deep cerebral (LI = 1.8%) and cerebellar (LI = 1.0%) grey matter, and thalamus (LI = 0.3%). Phenotypically, BrdUrd-positive cells in neuropil were glial cell precursors and their progeny, while those associated with meninges were usually located in the superficial subarachnoid space and appeared to be fibrocytes. Using BrdUrd infusion, LI for glial precursors at these sites ranged from two- to 10-fold higher than those reported previously after a brief parenteral pulse dose. These data indicate that continuous BrdUrd infusion for 6 days by subcutaneous osmotic pump is an efficient means of labelling neural cells throughout the brain.
Assuntos
Encéfalo/metabolismo , Bromodesoxiuridina/administração & dosagem , DNA/biossíntese , Animais , Astrócitos/química , Encéfalo/citologia , Química Encefálica , Bromodesoxiuridina/análise , Corpo Caloso/química , Imuno-Histoquímica , Bombas de Infusão Implantáveis , Injeções Subcutâneas , Masculino , Ratos , Ratos Endogâmicos F344 , Fatores de TempoRESUMO
Events secondary to induced cell proliferation may play a role in the carcinogenic process. These studies investigated the expression of genes associated with growth control in response to two types of cell proliferation stimuli in the livers of male F344 rats. Regenerative hepatocyte proliferation after partial hepatectomy or a single dose of carbon tetrachloride, and mitogenic liver hyperplasia induced by a single dose of phenobarbital or WY-14,643 were assessed by thymidine incorporation and quantitative autoradiography. The expression of myc, fos, and Ha-ras was evaluated by Northern blot analysis of liver derived poly(A)+ mRNA from these same animals. After each treatment, the level of hepatocyte proliferation (labelling index 4-32%) was observed to peak between 24 and 48 h and return to control values by 8 days. In every case, a peak in myc expression was seen between 0.5 and 18 h depending on the proliferative stimulus treatment. A large peak in fos expression was seen at 0.5-2 h but only with the cytotoxic and regenerative proliferative treatments partial hepatectomy or carbon tetrachloride. A broad peak in Ha-ras expression was observed 12 to 36 h after each treatment. These data demonstrate transient expression of these genes following the synchronous induction of hepatocyte proliferation. The increased expression of fos upon treatment with cytotoxicants, but not mitogens, suggests different modes of growth regulation that may be important in understanding the induction of cell proliferation by these two types of agents.
Assuntos
Expressão Gênica/fisiologia , Genes fos/genética , Genes myc/genética , Genes ras/genética , Fígado/patologia , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Peso Corporal/efeitos dos fármacos , Carcinógenos/farmacologia , Divisão Celular/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Tamanho do Órgão/efeitos dos fármacos , Fenobarbital/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Endogâmicos F344RESUMO
To better understand the mode of action of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced alterations in hepatic cell proliferation and the potential link to tumour-promoting activity, we investigated the effects of TCDD on the expression of certain key genes involved in liver cell growth and the effects of TCDD on induced hepatocyte cell proliferation. Gene expression analysis was conducted, by Northern blot hybridization, using RNA isolated from female Sprague-Dawley rat livers collected at various times during a 14-day dosing period with TCDD known to produce alterations in cell proliferation. No major changes were observed in the expression of the transforming growth factors TGF-alpha and TGF-beta or in oncogenes ras, src and myc. However, the expression of the transcription factors C/EBP, HNF-1 alpha and HNF-4 decreased after 14 days of TCDD treatment. To investigate how TCDD affects hepatic growth, cell proliferation analysis was conducted in rats stimulated to undergo hepatocyte proliferation following either 2/3 partial hepatectomy or lead nitrate treatment. Cell proliferation was quantified by means of immunocytochemical detection of Proliferating Cell Nuclear Antigen (PCNA). Fourteen days of pretreatment with TCDD caused an overall inhibition of hepatocytes in the growth fraction (G1, S, G2 and M) from 61 +/- 3% in the control-partial hepatectomy group to 41 +/- 3% in the TCDD-partial hepatectomy group. A periportal pattern of cell proliferation was observed in the TCDD-partial hepatectomy group as compared to the panlobular pattern of cell proliferation in the control-partial hepatectomy group. TCDD pretreatment also produced an inhibition of cell proliferation induced by the liver mitogen lead nitrate. TCDD-induced inhibition of hepatocyte proliferation could play a role in TCDD tumour promotion and hepatocarcinogenesis through the creation of a environment whereby preneoplastic cells continue to expand while normal hepatocyte proliferation is inhibited.
Assuntos
Inibidores do Crescimento/farmacologia , Fígado/efeitos dos fármacos , Dibenzodioxinas Policloradas/farmacologia , Animais , Northern Blotting , Ciclo Celular/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Substâncias de Crescimento/genética , Hepatectomia , Imuno-Histoquímica , Chumbo/farmacologia , Fígado/citologia , Fígado/cirurgia , Mitógenos/farmacologia , Nitratos/farmacologia , Antígeno Nuclear de Célula em Proliferação/análise , Ratos , Ratos Sprague-DawleyRESUMO
Quantitative knowledge of gene expression can provide valuable information for understanding the action of chemicals that alter cell proliferation and cancer. Accurate quantification of mRNA levels requires the normalization of the gene of interest to a gene with transcriptional levels that do not vary through the cell cycle or with a particular treatment. Changes in expression were examined in proliferating or non-proliferating rat liver for three constitutively expressed 'housekeeping' genes commonly used to normalize mRNA levels from Northern blots. In addition, a direct method of quantifying poly(A)+ mRNA by hybridization with a radiolabelled polythymidylate--poly(T)--probe was compared with traditional methods. Hepatocyte cytolethality and a subsequent wave of hepatocyte proliferation were induced in male Fischer-344 rats by treatment with a single gavage dose of carbon tetrachloride. Induced cell proliferation peaked at 48 h after treatment. Expression of the housekeeping genes actin, glyceraldehyde-3-phosphate-dehydrogenase (GAPDH) and albumin, as well as the proto-oncogene H-ras, was determined by Northern blot analysis at times from 0.5 h to 4 days after treatment. Time-dependent changes were observed in the expression of these genes relative to the levels observed in the untreated control animals. Actin expression peaked at 3.4-fold over control and GAPDH expression was increased by 1.9-fold over control. Albumin mRNA levels varied the least, 1.4-fold over control, indicating that this gene is more appropriate than actin or GAPDH for normalization of proto-oncogene expression under experimental conditions that induce cell proliferation in rat liver. The direct quantification of poly(A)+ mRNA using a poly(T) probe was not influenced by the induction of cell proliferation. This method may be useful when the expression of housekeeping genes is affected by treatment.
Assuntos
Northern Blotting/normas , Divisão Celular/genética , Expressão Gênica , Variação Genética , RNA Mensageiro/análise , Animais , Sondas de DNA , Regeneração Hepática/genética , Masculino , Poli T , Ratos , Ratos Endogâmicos F344 , Transcrição GênicaRESUMO
In previous studies, unleaded gasoline (UG) vapor was found to be a liver tumor promoter and hepatocarcinogen in female mice, but UG was not a hepatocarcinogen in male mice. However, UG vapor had similar transient mitogenic effects in nonlesioned liver of both male and female mice under the conditions of the cancer bioassay. We used an initiation-promotion protocol to determine whether UG vapor acts as a liver tumor promoter in male mice and to examine proliferative effects that may be critical to tumor development. Twelve-day-old male B6C3F1 mice were injected with N-nitrosodiethylamine (DEN; 5 mg/kg, intraperitoneally) or vehicle. Starting at 5-7 weeks of age, mice were exposed by inhalation 6 hr/day, 5 days/week for 16 weeks to 0 or 2046 ppm of PS-6 blend UG. UG treatment caused a significant 2.3-fold increase in the number of macroscopic hepatic masses in DEN-initiated mice, whereas no macroscopic masses were observed in non-initiated mice. Altered hepatic foci (AHF), which were predominantly basophilic in phenotype, were found almost exclusively in DEN-initiated mice. UG treatment significantly increased both the mean volume (threefold) and the volume fraction (twofold) of the AHF without increasing the number of AHF per unit area. UG also induced hepatic pentoxyresorufin-O-dealkylase (PROD) activity, a marker of CYP2B, by more than 12-fold over control with or without DEN cotreatment. To study hepatocyte proliferative effects of UG, we treated mice with 5-bromo-2'-deoxyuridine (BrdU) via osmotic pump for 3 days before necropsy and measured hepatocyte BrdU labeling index (LI) in AHF and nonlesioned liver.(ABSTRACT TRUNCATED AT 250 WORDS)
Assuntos
Gasolina/efeitos adversos , Neoplasias Hepáticas Experimentais/induzido quimicamente , Lesões Pré-Cancerosas/induzido quimicamente , Animais , Divisão Celular , Citocromo P-450 CYP2B1 , Sistema Enzimático do Citocromo P-450/metabolismo , Modelos Animais de Doenças , Exposição Ambiental , Feminino , Fígado/enzimologia , Fígado/patologia , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Oxirredutases/metabolismo , Lesões Pré-Cancerosas/patologiaRESUMO
Proliferating cell nuclear antigen (PCNA), an endogenous nuclear protein, has recently been used to identify replicating cells. PCNA was compared to tritiated thymidine ([3H]-TdR), a reliable and accurate exogenous labeling agent, to ascertain if PCNA gives comparable results for quantitative cell proliferation. Male F344 rats were treated with a single dose of 500 mg/kg 4-acetylaminofluorene (4-AAF), a known liver mitogen. Rats (n = 5) were euthanized and necropsied at 6, 12, 18, 24, 36, 48, 96, or 192 hr after treatment. Two hours before necropsy, rats were pulsed-dosed with [3H]-TdR (2 mCi/kg body weight). Livers were sectioned, autoradiography performed, and labeling indexes (LI), a measurement of the percentage of S-phase hepatocytes, determined. One and a half years after the completion of this study, the archival paraffin blocks of the liver tissue were sectioned and stained for PCNA by an immunohistochemical procedure. Immunocytochemical staining patterns of proliferating cell nuclear antigen antigen expression permitted the recognition of G1, S, G2, M, and quiescent cells. PCNA LI, generated by scoring only cells exhibiting S-phase staining patterns, was compared to the pulse [3H]-TdR LI for each animal. Similar periportal staining patterns of S-phase nuclei were detected by both markers. The [3H]-TdR LI and the PCNA LI exhibited a peak at 24 hr of approximately the same magnitude. However, while the [3H]-TdR LI had returned to near baseline at the 48-hr time point, the PCNA LI remained elevated until the 96-hr time point. This sustained elevation of the PCNA index cannot be explained at this time.(ABSTRACT TRUNCATED AT 250 WORDS)
Assuntos
Divisão Celular/fisiologia , Fígado/citologia , Fígado/metabolismo , Proteínas Nucleares/metabolismo , Timidina/metabolismo , 2-Acetilaminofluoreno/toxicidade , Animais , Biomarcadores , Ciclo Celular/fisiologia , Imuno-Histoquímica , Fígado/efeitos dos fármacos , Masculino , Antígeno Nuclear de Célula em Proliferação , Ratos , Ratos Endogâmicos F344 , TrítioRESUMO
Two different markers for quantitating cell proliferation were evaluated in livers of control and chemically treated mice and rats. Proliferating cell nuclear antigen (PCNA), an endogenous cell replication marker, and bromodeoxyuridine (BrdU), an exogenously administered DNA precursor label, were detected in formalin-fixed, paraffin-embedded tissues using immunohistochemical techniques. The percentage of cells in S phase (labeling indexes, LI) evaluated as PCNA- or BrdU-positive hepatocellular nuclei was compared in recut tissue sections from animals given BrdU by a single IP injection 2 hr before killing the animals. Ten-week-old male B6C3F1 mice and F344 rats were exposed to known mitogenic hepatocarcinogens, Wy-14,643 (WY) in the diet at 0.1% for 2 days or 1,4-dichlorobenzene (DCB) in corn oil by gavage for 2 days (600 mg/kg/day in mice; 300 mg/kg/day in rats). In mice, PCNA and BrdU hepatocyte LI were similar in control, WY-treated, and DCB-treated animals. In rats, PCNA and BrdU gave similar LI in controls and Wy-treated animals. Although PCNA LI was statistically lower than BrdU LI in DCB-treated rats, both PCNA and BrdU LI for DCB-treated rats was increased over LI in control rats. Different patterns of PCNA immunohistochemical staining, interpreted to represent different subpopulations of cells at various phases of the cell cycle, were quantitated using PCNA immunohistochemistry. The proliferating index (PI), defined as the percentage of cells in the cell cycle (G1 + S + G2 + M), was more sensitive than the LI (S phase only) in detecting a chemically induced cell proliferative response.(ABSTRACT TRUNCATED AT 250 WORDS)
Assuntos
Divisão Celular/fisiologia , Fígado/citologia , Fígado/metabolismo , Proteínas Nucleares/metabolismo , Animais , Biomarcadores , Bromodesoxiuridina/metabolismo , Ciclo Celular/fisiologia , Clorobenzenos/toxicidade , DNA/metabolismo , Imuno-Histoquímica , Fígado/efeitos dos fármacos , Masculino , Camundongos , Precursores de Ácido Nucleico/metabolismo , Antígeno Nuclear de Célula em Proliferação , Pirimidinas/toxicidade , Ratos , Ratos Endogâmicos F344RESUMO
Chemicals may induce cell proliferation directly as mitogens or indirectly via cell death with subsequent proliferation to replace lost cells. Chemically induced proliferation has been demonstrated to play a role in the carcinogenic process. A wide range of procedures and techniques are currently being used to define the quantitative relationship between the extent and duration of chemically induced cell proliferation and carcinogenic potential in different species and target organs. However, a limited database and nonstandard protocols and procedures for measuring cell proliferation have made it difficult to compare results between laboratories. Comparison of frequencies of S phase between control and treated animals is the most commonly used end point in cell proliferation studies and may be regarded as an indirect indication of a proliferative response. This response can be ascertained as labeling indexes (LI; percentage of cells in S phase) after the administration of the DNA precursor labels (tritiated thymidine; 3H-TdR; bromodeoxyuridine, BrdU) or through immunostaining of the endogenous cell replication marker, proliferating cell nuclear antigen (PCNA). Both approaches are applicable to tissue sections. An important issue in the design of experimental studies for measuring LI is determining how and when to investigate proliferative responses in relation to the chemical treatment regimen. Variables to consider when designing cell proliferation studies include the animal's age, chemical dose and method of treatment, choice and dose of label, time and length that the label is administered, and methods of quantitation.(ABSTRACT TRUNCATED AT 250 WORDS)
Assuntos
Carcinógenos/toxicidade , Divisão Celular/efeitos dos fármacos , Neoplasias/etiologia , Animais , Divisão Celular/fisiologia , DNA/biossíntese , Humanos , Neoplasias/patologia , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula em Proliferação , Projetos de Pesquisa , RoedoresRESUMO
Chronic exposure to methyl tertiary butyl ether (MTBE) altered the rodent tumor incidence of endocrine-sensitive tissues and decreased the incidence of estrogen-dependent uterine cystic hyperplasia in mice. To test the hypothesis that changes in the incidence of tumors in female B6C3F1 mice after MTBE exposure are secondary to endocrine alterations, we exposed female mice to the carcinogenic dose of MTBE vapor (8000 ppm) for 3 or 21 days or 4 or 8 months under conditions similar to a previous 2-year bioassay. MTBE exposure significantly decreased body weight gain and ovary and pituitary weight at 4 and 8 months and uterine weight at all time points. After 8 months of exposure, MTBE significantly increased the length of the estrous cycle by increasing the mean number of days in both the estrus and the nonestrus stages. Histological evaluation of H&E-stained tissues showed a decrease in the number of uterine glands after subchronic MTBE exposure. DNA synthesis, as measured by the incorporation of 5-bromo-2'-deoxyuridine (BrdU), was decreased in uterine glandular and luminal epithelial cells after MTBE exposure for 3 or 21 days or 4 or 8 months. MTBE exposure decreased the number of epithelial layers in the cervix and vagina at all time points. DNA synthesis was decreased in cervical and vaginal epithelium after 21 days of MTBE. Decreased zona reticularis of adrenal glands was found after 4 and 8 months of MTBE exposure without changes in BrdU incorporation. MTBE did not competitively bind to estrogen receptor. MTBE exposure did not alter serum estrogen levels or alter the location or intensity of estrogen receptor immunoreactivity in the uterus, cervix, and vagina. These data indicate that while MTBE exposure causes multiple endocrine-related tissue and cellular responses, these effects are not mediated through the estrogen receptor.
Assuntos
Carcinógenos/toxicidade , Glândulas Endócrinas/efeitos dos fármacos , Éteres Metílicos/toxicidade , Receptores de Estrogênio/metabolismo , Administração por Inalação , Animais , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Colo do Útero/efeitos dos fármacos , Colo do Útero/patologia , Replicação do DNA/efeitos dos fármacos , Glândulas Endócrinas/metabolismo , Glândulas Endócrinas/patologia , Estradiol/sangue , Estro/efeitos dos fármacos , Feminino , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Vagina/efeitos dos fármacos , Vagina/patologiaRESUMO
The rates of cell proliferation and cell loss in conjunction with the differentiation status of a tissue are among the many factors contributing to carcinogenesis. Nongenotoxic (non-DNA reactive) chemicals may affect this balance by increasing proliferation through direct mitogenesis or through a regenerative response following loss of cells through cytotoxic (oncotic) or apoptotic necrosis. In a recent NTP study in Fischer rats and B6C3F(1) mice, the mycotoxin fumonisin B(1) caused renal carcinomas in male rats and liver cancer in female mice. In an earlier study in male BD-IX rats, fumonisin B(1) caused hepatic toxicity and hepatocellular carcinomas. An early effect of fumonisin B(1) exposure in these target organs is apoptosis. However, there is also some evidence of oncotic necrosis following fumonisin B(1) administration, especially in the liver. Induction of apoptosis may be a consequence of ceramide synthase inhibition and disruption of sphingolipid metabolism by fumonisin B(1). Fumonisin B(1) is not genotoxic in bacterial mutagenesis screens or in the rat liver unscheduled DNA-synthesis assay. Fumonisin B(1) may be the first example of an apparently nongenotoxic (non-DNA reactive) agent producing tumors through a mode of action involving apoptotic necrosis, atrophy, and consequent regeneration.
Assuntos
Apoptose , Ácidos Carboxílicos/metabolismo , Ácidos Carboxílicos/farmacologia , Ácidos Carboxílicos/farmacocinética , Ácidos Carboxílicos/toxicidade , DNA/metabolismo , Neoplasias Esofágicas/complicações , Fumonisinas , Neoplasias Renais/induzido quimicamente , Rim/efeitos dos fármacos , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Esfingolipídeos/metabolismo , África/epidemiologia , Animais , China/epidemiologia , Tomada de Decisões , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/epidemiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Micotoxinas/classificação , Micotoxinas/toxicidade , Ratos , Ratos Endogâmicos , Fatores de RiscoRESUMO
Preliminary results from the National Toxicology Program (NTP) bioassays of furan given by gavage indicate the induction of hepatocellular carcinomas in male F-344 rats and in both sexes of B6C3F1 mice, and cholangiocarcinomas in both sexes of rats. To assess the genotoxicity of furan, chemically induced unscheduled DNA synthesis was evaluated in the in vivo hepatocyte DNA repair assay. Furan did not induce unscheduled DNA synthesis in hepatocytes isolated after single gavage treatment of male F-344 rats (5, 30, and 100 mg/kg) or male B6C3F1 mice (10, 50, 100, and 200 mg/kg). Furan induced cytotoxicity and enhanced cell proliferation were evaluated in livers of rats and mice as events that also might give rise to mutations and/or drive tumor formation. The labeling index (LI, percentage of hepatocyte nuclei in S-phase) was measured histoautoradiographically following a single gavage administration of furan (30 mg/kg, male rats; 50 mg/kg, male mice) followed by an injection of 3H-thymidine 2 hr prior to sacrifice. Hepatocellular necrosis and a sharp increase in LI (23.9 for mice and 17.8 for rats vs. less than 0.5 for controls) was observed 48 hr after treatment with furan, indicative of restorative cell proliferation secondary to cytotoxicity. Hepatocyte proliferation was evaluated also at the highest NTP bioassay dose (15 mg/kg/day for mice and 8 mg/kg/day for rats, 5 days/week) by labeling with 3H-thymidine administered via a 6 day osmotic pump implanted subcutaneously. Necrosis and inflammation were observed along the subcapsular visceral surface of the left or caudate liver lobes, likely due to diffusion of furan directly through the stomach to the liver. After 6 weeks of furan administration, male and female rats, but not mice, exhibited bile duct hyperplasia as well as metaplasia in the areas of fibrosis along the subcapsular visceral surface of the left or caudate liver lobes. The fold increase in hepatocyte LI in treated animals relative to the combined controls measured at weeks 1, 3, and 6 ranged from 39 to 5 for male mice, 18 to 51 for male rats, and 12 to 19 for female rats. Taken together, these data suggest that mechanisms other than direct DNA-reactivity might explain the profile of oncogene mutations observed in the mouse liver tumors, including selective promotion of different subpopulations of preneoplastic cells and/or mutational events secondary to sustained cell proliferation or inflammation. The extensive amount of furan-induced cell proliferation subsequent to cytotoxicity likely had a significant impact on tumor development, and such data should be considered in risk evaluations for this compound.
Assuntos
Furanos/toxicidade , Fígado/efeitos dos fármacos , Mutagênicos/toxicidade , Animais , Divisão Celular , Células Cultivadas , Reparo do DNA , Enzimas/sangue , Feminino , Furanos/administração & dosagem , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Testes de Mutagenicidade , Mutagênicos/administração & dosagem , Ratos , Ratos Endogâmicos F344 , Fatores Sexuais , Fatores de TempoRESUMO
The mouse liver is a frequent target organ for chemical carcinogenesis (Huff et al., 1988, 1991; Gold et al., 1989) and tumor development exhibits preferential strain sensitivity (Dragani et al., 1992; Drinkwater and Bennett, 1991). In some reports a positive correlation has been observed between the degree of spontaneous liver tumor incidence and the propensity to develop liver tumors after treatment with chemical carcinogens (Della Porta et al., 1967; Flaks, 1968; Dragani et al., 1984, 1987; Diwan et al., 1986; Drinkwater and Ginsler, 1986), but this is not always the case (Grasso and Hardy, 1975; Hanigan et al., 1988; Dragani et al., 1992). Thus, the interpretation of this endpoint in assessing potential health hazards to humans continues to be the subject of active debate. Studies of molecular and genetic factors that modulate the genesis of mouse liver tumors should enhance our understanding of the relevance of this response following exposure to genotoxic as well as nongenotoxic chemicals. To utilize intelligently animal models as surrogates for human carcinogenesis, the validity of rodent tumor endpoints in assessing potential human health hazards from chemical exposure remains an important issue. One approach has been to understand the animal system itself and the mechanisms by which chemicals induce tumors in the animal model. Information regarding the molecular events associated with tumor induction should make the relevance of results from rodent carcinogenicity studies to human risk easier to assess. Results to date have identified activation of ras proto-oncogenes as one early event and an important factor associated with chemical induction of mouse liver neoplasia (Reynolds et al., 1986, 1987; Wiseman et al., 1986), although ras-independent pathways appear to account for an appreciable proportion of some chemically induced mouse liver tumors (Fox et al., 1990; Buchmann et al., 1991). Available data emphasize the complexity of H-ras activation in murine hepatocarcinogenesis. Not only the genetic background of the mouse but also the dose of the carcinogen may influence significantly the number of tumors containing activated H-ras. Both high sensitivity and low sensitivity strains of mice can develop liver tumors which contain activated H-ras oncogenes, showing that the ability to activate this gene does not in itself determine susceptibility to hepatocarcinogenesis. Ras gene mutational profiles in chemically induced liver tumors may be different and distinguishable from those in spontaneous tumors. Since multiple genetic as well as nongenetic events are associated with tumor development, defining a precise role for ras gene mutations when they occur in mouse liver tumors is often difficult.(ABSTRACT TRUNCATED AT 400 WORDS)
Assuntos
Carcinógenos/toxicidade , Genes ras , Neoplasias Hepáticas Experimentais/genética , Animais , Códon , Regulação da Expressão Gênica/efeitos dos fármacos , Genes ras/efeitos dos fármacos , Genes ras/genética , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Mutação , Proto-Oncogene Mas , Especificidade da EspécieRESUMO
Furan is a potent rodent hepatocarcinogen that probably acts through non-genotoxic mechanisms involving hepatotoxicity and regenerative hepatocyte proliferation. In addition to inducing necrosis, cytotoxicants like furan may also induce cytolethality through apoptosis which has been suggested to play a key role in carcinogenesis. Hepatocyte proliferation and apoptosis were studied in female B6C3F1 mice exposed to furan by oral gavage for 3 weeks at National Toxicology Program (NTP) bioassay doses (8 and 15 mg/kg body weight) and lower (4 mg/kg). Furan treatment led to a 2- to 3-fold significant increase in liver-related enzymes and bile acids in blood serum as compared to the control group. These changes were accompanied by minor subcapsular inflammation and minimal necrosis at 8 and 15 mg furan/kg. A dose-related increase in bromodeoxyuridine-labeling index (1.4- to 1.7-fold) and hematoxylin- and eosin-defined apoptotic index (6- to 15-fold) was observed at 8 and 15 mg/kg. Co-treatment of mice with aminobenzotriazole, an irreversible inhibitor of cytochromes P-450, prevented the observed hepatotoxic effects induced by furan. These results indicate that furan elicits hepatotoxicity in a dose-related manner through a toxic metabolite and, furthermore, suggest that apoptosis is an important form of cell death at hepatocarinogenic doses under short-term conditions.
Assuntos
Apoptose/efeitos dos fármacos , Ácidos e Sais Biliares/sangue , Carcinógenos/toxicidade , Furanos/toxicidade , Fígado/efeitos dos fármacos , Administração Oral , Alanina Transaminase/sangue , Análise de Variância , Animais , Bromodesoxiuridina/química , Carcinógenos/administração & dosagem , Contagem de Células , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Furanos/administração & dosagem , L-Iditol 2-Desidrogenase/sangue , Fígado/citologia , Camundongos , NecroseRESUMO
The availability of in vivo transgenic mouse mutagenesis assays provides the opportunity to study the induction of mutations that reflects the integration of pharmacokinetics, biotransformation. DNA repair responses, and tissue susceptibilities. Transgenic mice containing in vivo marker genes for mutagenicity within shuttle vectors also permit the determination of the resulting mutational spectra in tissues of mice following exposure to mutagens and carcinogens. Exposure of B6C3F1 lacI transgenic mice to the rodent carcinogen 1,3-butadiene by inhalation results in an increase in the overall lacI mutant frequency in bone marrow with an increased frequency of point mutations at A:T base pairs. These studies demonstrate the utilization of transgenic mouse models for establishing linkages between exposure to carcinogens, internal tissue dose, and in vivo mutational events.
Assuntos
Butadienos/toxicidade , Carcinógenos/toxicidade , Mutagênicos/toxicidade , Animais , Masculino , Camundongos , Camundongos Transgênicos , Mutação PuntualRESUMO
A class of genes, the so-called tumor suppressor genes or anti-oncogenes, was originally identified as being responsible for germ-line transmission of cancer susceptibility in humans. Tumor suppressor genes are recessive at the cellular level with respect to oncogenesis but often manifest as dominantly inherited familial cancer syndromes. This type of cancer syndrome arises in the Eker rat due to a genetic defect in the tuberous sclerosis 2 (Tsc2) gene. The Eker rat familial cancer syndrome serves as a unique animal model in which to study the molecular pathways of renal tubular epithelial carcinogenesis as well as a valuable tool for studies that examine how chemical carcinogens interact with cancer susceptibility genes.
Assuntos
Carcinoma de Células Renais/genética , Genes Supressores de Tumor , Neoplasias Renais/genética , Proteínas Repressoras/genética , Animais , Modelos Animais de Doenças , Ratos , Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de TumorRESUMO
The Tg.AC mouse carrying the v-Ha-ras structural gene is a useful model for the study of chemical carcinogens, especially those acting via non-genotoxic mechanisms. This study evaluated the efficacy of the non-toxic, water-soluble antioxidant from spinach, natural antioxidant (NAO), in reducing skin papilloma induction in female hemizygous Tg.AC mice treated dermally five times over 2.5 weeks with 2.5 microg 12-O-tetradecanoylphorbol-13-acetate (TPA). The TPA-only group was considered as a control; the other two groups received, additionally, NAO topically (2 mg) or orally (100 mg/kg), 5 days/week for 5 weeks. Papilloma counts made macroscopically during the clinical observations showed a significant decrease in multiplicity (P<0.01) in the NAO topically treated group. According to histological criteria, papilloma multiplicity were lower in both topical-NAO and oral-NAO groups, but significantly so only in the oral-NAO mice (P<0.01). The beneficial effect of NAO in the Tg.AC mouse is reported.