Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Toxicol Pathol ; 28(5): 705-15, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11026607

RESUMEN

Using zebrafish, Danio rerio, initial pioneering work in the 1960s revealed carcinogen responsiveness of fish, yet very few subsequent tumorigenesis investigations have utilized this species. We exposed embryos (60 hours postfertilization) and fry (3 week posthatch) to 7,12-dimethylbenz[a]anthracene (DMBA) by immersion in aqueous solutions for 24 hours, at concentrations of 0-1 or 0-5 ppm (mg/L), respectively. Juvenile zebrafish 2 months posthatch were fed a diet containing 0-1,000 ppm DMBA for 4 months. Fish were sampled for histologic evaluation at 7-12 months after the onset of carcinogen treatment. Fry were most responsive to DMBA and showed the widest diversity of target tissues and histologic types of neoplasia, having several types of epithelial, mesenchymal, and neural neoplasia. The principal target tissues for carcinogenic response were liver following embryo or fry exposure, with gill and blood vessel the second and third most responsive tissues in fry. Intestine was the primary target and gill a secondary target in fish that received dietary DMBA as juveniles. These studies indicate that young zebrafish are most responsive to DMBA, showing a greater diversity of neoplasm types than rainbow trout. Thus, zebrafish are a valuable model system in which to study mechanistic aspects of the carcinogenesis process.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/toxicidad , Carcinógenos/toxicidad , Neoplasias Experimentales/inducido químicamente , Pez Cebra/embriología , 9,10-Dimetil-1,2-benzantraceno/administración & dosificación , Animales , Peso Corporal , Carcinógenos/administración & dosificación , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/patología , Branquias/efectos de los fármacos , Branquias/patología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/patología , Mesodermo/efectos de los fármacos , Mesodermo/patología , Neoplasias de los Tejidos Conjuntivo y Blando/inducido químicamente , Neoplasias de los Tejidos Conjuntivo y Blando/patología , Neoplasias Experimentales/epidemiología , Neoplasias Experimentales/patología , Neoplasias Glandulares y Epiteliales/inducido químicamente , Neoplasias Glandulares y Epiteliales/patología , Neoplasias de Tejido Vascular/inducido químicamente , Neoplasias de Tejido Vascular/patología , Cresta Neural/efectos de los fármacos , Cresta Neural/patología , Razón de Masculinidad
2.
Toxicol Pathol ; 28(5): 716-25, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11026608

RESUMEN

We exposed embryos (83 hours postfertilizaton) and fry (3 weeks posthatch) to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) by immersion in aqueous solutions of 0-10 ppm for 1 hour (embryo) or 0-2 ppm for 24 hours (fry). Zebrafish embryos were microinjected with MNNG at levels of 0 or 96 ng/egg. Diets containing 0-2,000 ppm MNNG were fed to juvenile zebrafish for 3 months beginning at 2 months posthatch. Fish were sampled for histopathologic study at 6-12 months after initiation of carcinogen exposure. Embryos and fry were both quite responsive to MNNG; however, juvenile zebrafish were remarkably refractory to MNNG-induced neoplasia. Principal target organs in zebrafish treated as embryos with MNNG were liver and testis, with hepatocellular adenoma the most prevalent hepatic neoplasm. A variety of mesenchymal neoplasms occurred in zebrafish following embryo exposure to MNNG, including chondroma, hemangioma, hemangiosarcoma, leiomyosarcoma, and rhabdomyosarcoma. Testis and blood vessels were primary target organs for MNNG following fry exposure, with seminoma, hemangioma, hemangiosarcoma, and various other epithelial and mesenchymal neoplasms occurring. The zebrafish is a responsive, cost-effective lower vertebrate model system in which to study mechanisms of carcinogenesis.


Asunto(s)
Metilnitronitrosoguanidina/toxicidad , Neoplasias Experimentales/inducido químicamente , Pez Cebra/embriología , Animales , Peso Corporal , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/patología , Branquias/efectos de los fármacos , Branquias/patología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/patología , Masculino , Mesodermo/efectos de los fármacos , Mesodermo/patología , Metilnitronitrosoguanidina/administración & dosificación , Neoplasias de los Tejidos Conjuntivo y Blando/inducido químicamente , Neoplasias de los Tejidos Conjuntivo y Blando/patología , Neoplasias Experimentales/epidemiología , Neoplasias Experimentales/patología , Neoplasias Glandulares y Epiteliales/inducido químicamente , Neoplasias Glandulares y Epiteliales/patología , Neoplasias de Tejido Vascular/inducido químicamente , Neoplasias de Tejido Vascular/patología , Razón de Masculinidad , Neoplasias Testiculares/inducido químicamente , Neoplasias Testiculares/patología
3.
Carcinogenesis ; 20(10): 1919-26, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10506105

RESUMEN

Cancer chemoprevention by dietary chlorophyllin (CHL) was investigated in a rainbow trout multi-organ tumor model. In study 1, duplicate groups of 130 juvenile trout were treated for 2 weeks with control diet, 500 p.p.m. dibenzo[a,l]pyrene (DB[a,l]P) or 500 p.p.m. DB[a,l]P + 2052 p.p.m. CHL, then returned to control diet. DB[a,l]P alone proved somewhat toxic but induced high tumor incidences in liver (61%), stomach (91%) and swimbladder (53%) 11 months after initiation. CHL co-feeding abrogated DB[a,l]P acute toxicity and reduced tumor incidences to 18% in liver, 34% in stomach and 3% in swimbladder (P

Asunto(s)
Anticarcinógenos/farmacología , Benzopirenos/toxicidad , Carcinógenos/toxicidad , Clorofilidas/farmacología , Neoplasias Experimentales/prevención & control , Animales , Anticarcinógenos/administración & dosificación , Clorofilidas/administración & dosificación , Dieta , Pruebas de Mutagenicidad , Neoplasias Experimentales/inducido químicamente , Oncorhynchus mykiss
4.
Carcinogenesis ; 20(3): 453-8, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10190561

RESUMEN

Indole-3-carbinol (I3C), a metabolite of glucobrassicin found in cruciferous vegetables, is documented as acting as a modulator of carcinogenesis and, depending on timing and dose of administration, it may promote hepatocarcinogenesis in some animal models. In this study we demonstrate that, when given post-initiation, dietary I3C promotes aflatoxin B1 (AFB1)-induced hepatocarcinogenesis in the rainbow trout model at levels as low as 500 p.p.m. Trout embryos (approximately 9000) were initiated with 0, 25, 50, 100, 175 or 250 p.p.b. AFB1 by a 30 min immersion. Experimental diets containing 0, 250, 500, 750, 1000 or 1250 p.p.m. I3C were administered starting at 3 months and fish were sampled for liver tumors at 11-13 months. Promotion at the level of tumor incidence was statistically significant for all dietary levels, except 250 p.p.m. Relative potency for promotion markedly increased at dietary levels >750 p.p.m. We propose that more than one mechanism could be involved in promotion and that both estrogenic and Ah receptor-mediated pathways could be active. The estrogenicity of I3C, measured as its ability to induce vitellogenin (an estrogen biomarker in oviparous vertebrates) was evident at the lowest dietary level (250 p.p.m.), whereas CYPIA (a P450 isozyme induced through the Ah receptor pathway) was not induced until dietary levels of 1000 p.p.m. Therefore, at lower dietary levels, promotion by I3C in this model could be explained by estrogenic activities of I3C acid derivatives, as it is known that estrogens promote hepatocarcinogenesis in trout. Much stronger promotion was observed at high dietary I3C levels (1000 and 1250 p.p.m.), at which levels both CYP1A and vitellogenin were induced.


Asunto(s)
Aflatoxina B1/toxicidad , Carcinógenos/toxicidad , Dieta , Indoles/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Animales , Cocarcinogénesis , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Oncorhynchus mykiss
5.
J Environ Pathol Toxicol Oncol ; 18(4): 261-9, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-15281236

RESUMEN

There is an evident need of low-cost vertebrates to be used in experimental carcinogenesis. Medaka (Oryzias latipes) provide a useful vertebrate model system for investigating tissue tropism of carcinogens and the action mechanisms of environmental contaminants posing a potential risk to human health. Juvenile medaka 2 months of age fed diets containing 100 ppm (dry weight basis) dibenzo[a,l]pyrene (DBP) for 28 days responded with hepatic neoplasia predominately of hepatocellular origin. When sampled 9 months following the termination of carcinogen exposure, medaka showed 26% incidence of neoplasia and 25% hepatic neoplasia, compared with 8% total neoplasia and 0% hepatic neoplasia in control fish. The predominant spontaneous neoplasms in this group of medaka were ovarian germ cell tumors. Hepatic neoplasia occurred at a higher incidence in female DBP-treated medaka than in males (11/29 vs 5/36). Nonneoplastic lesions observed in the livers of DBP-exposed fish included spongiosis hepatis, globular hyaline eosinophilic cytoplasmic inclusions in hepatocytes, foci of hepatocellular degeneration, extensive cytomegaly, and karyomegaly of hepatocytes. No activating exon I mutations in the one ras protooncogene examined were detected among six liver neoplasms. These results indicate that medaka are sensitive to the tumorigenic effects of the environmental carcinogen DBP, administered by dietary exposure.


Asunto(s)
Adenoma de Células Hepáticas/inducido químicamente , Benzopirenos/toxicidad , Carcinógenos Ambientales/toxicidad , Carcinoma Hepatocelular/inducido químicamente , Neoplasias Hepáticas/inducido químicamente , Oryzias , Adenoma de Células Hepáticas/genética , Adenoma de Células Hepáticas/patología , Animales , Benzopirenos/administración & dosificación , Carcinógenos Ambientales/administración & dosificación , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , ADN de Neoplasias/análisis , Dieta , Modelos Animales de Enfermedad , Genes ras/genética , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Longevidad/efectos de los fármacos , Mutación , Reacción en Cadena de la Polimerasa
6.
Mutat Res ; 399(2): 233-44, 1998 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-9672662

RESUMEN

Rainbow trout, a species highly sensitive to aflatoxins, was used to investigate the relative carcinogenicities of four structurally related aflatoxins in terms of their target organ DNA binding characteristics. Tritiated syntheses were carried out, DNA binding dose-response curves were established, and liver DNA binding indices were calculated for the four aflatoxins following a 2-week dietary fry exposure protocol. The results indicated that adduct levels increased linearly with dietary dose concentration, with relative DNA binding indices of 20.7, 20.3, 2.35, and 2.22 x 10(3) (pmoles aflatoxin mg-1 DNA)/(pmoles aflatoxin g-1 diet) for aflatoxin B1 (AFB1), aflatoxicol (AFL), aflatoxin M1 (AFM1), and aflatoxicol M1 (AFLM1), respectively. A similar protocol used over 7200 trout fry averaging 1.2 g initial body weight to establish full carcinogen dose-response curves for each aflatoxin, along with a single-dose estimate of DNA binding index within the tumor study animals. Owing to trout sensitivity a total of 180 micrograms or less of each aflatoxin was required. Data analyzed on logit incidence vs. Ln dose coordinates generated four curves which were modeled as parallel in slope over most or all dose ranges studied. By this analysis, relative tumorigenic potencies were: AFB1 1.00; AFL 0.936; AFM1 0.086; and AFLM1 0.041. When data were plotted as logit incidence vs. Ln adducts (effective dose received), all aflatoxin adducts described the same dose-response curve; that is, they were equally tumorigenic, except those from AFLM1, which were 2-3 fold less potent. Therefore, by these molecular dose studies, differences in tumorigenicity among the four dietary aflatoxins are largely or entirely accounted for by differences in uptake and metabolism leading to DNA adduction, rather than any inherent differences in tumor initiating potency per DNA adduct.


Asunto(s)
Aflatoxinas/toxicidad , Carcinógenos/toxicidad , Neoplasias Hepáticas/inducido químicamente , Aflatoxinas/química , Animales , ADN/metabolismo , Aductos de ADN , Dieta , Relación Dosis-Respuesta a Droga , Hígado/metabolismo , Neoplasias Hepáticas/patología , Oncorhynchus mykiss
7.
J Toxicol Environ Health A ; 54(3): 227-42, 1998 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-9643874

RESUMEN

The present study examined whether modified xenobiotic transport, resulting from chlordecone (CD) or dieldrin pretreatment, would alter polycyclic aromatic hydrocarbon (PAH) or organochlorine (OC) target organ doses and subsequent tumor organospecificity or incidence rates in rainbow trout. Additionally, the potential for exposure to dieldrin or CD, following PAH exposure, to enhance tumor incidence was assessed. Evaluation of CD pretreatment effects on [14C]CD disposition in trout was conducted following two i.p. (0-15 mg/kg) and two dietary (0-0.4 mg/kg/d) pretreatment regimes. To assess the influence of OC pretreatment on cancer induced by the PAH 7,12-dimethylbenz[a]anthracene (DMBA), juvenile trout were fed control, CD (0.1, 0.4 mg/kg/d), or dieldrin (0.1, 0.3 mg/kg/d) diets for 9 wk, received a waterborne [3H]DMBA exposure (1 mg/L, 20 h), and resumed control, CD, or dieldrin diets for 33 wk. [3H]DMBA disposition and hepatic [3H]DMBA binding were examined immediately and 24 h after exposure. Hepatic and stomach tumor incidences were determined 33 wk after DMBA exposure. CD pretreatment did not influence [14C]CD or [3H]DMBA hepatic concentrations, hepatic [3H]DMBA DNA binding, or hepatic/stomach tumor incidence. It did, however, elevate bile [14C]CD and [3H]DMBA concentrations. Postinitiation exposure to CD weakly enhanced DMBA-induced hepatic tumor incidence at the low but not the high CD dose. Dieldrin pretreatment did not influence stomach [3H]DMBA equivalents or stomach tumor incidence but did cause an elevation in biliary and hepatic concentrations of [3H]DMBA equivalents. [3H]DMBA binding to liver DNA was significantly increased and hepatic tumor incidence was elevated by dieldrin pretreatment. Dieldrin treatment following DMBA initiation did not enhance hepatic or stomach tumor incidence. Ecoepidemiology studies, to date, have reported correlations between the co-occurrence of PAHs and OCs and elevated tumor incidence in feral fish, but cause-and-effect relationships have been difficult to establish. The results of the present study confirm that OCs, such as dieldrin and CD, play a role in modifying PAH-induced carcinogenesis in fish.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/farmacocinética , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Carcinógenos/farmacocinética , Carcinógenos/toxicidad , Clordecona/farmacología , Dieldrín/farmacología , Insecticidas/farmacología , Neoplasias Hepáticas Experimentales/inducido químicamente , Oncorhynchus mykiss/metabolismo , Animales , Peso Corporal/efectos de los fármacos , ADN/metabolismo , Dieta , Interacciones Farmacológicas , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/patología , Distribución Tisular
8.
Carcinogenesis ; 19(1): 161-7, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9472708

RESUMEN

Previously, we demonstrated that dehydroepiandrosterone (DHEA) enhances aflatoxin B1 (AFB1) hepatocarcinogenesis in trout when administered following AFB1 exposure. This paper examines the effect of DHEA on tumor latency and the comparative potency for DHEA to modulate AFB1 carcinogenesis when administered prior to and concurrent with AFB1 compared with a post-initiation exposure. Trout were initiated by a 30 min water bath exposure to 10 p.p.b. AFB1. At 3 months post-initiation, animals were started on either control diet or a diet containing 444 p.p.m. dehydroepiandrosterone (DHEA). Fifty trout per treatment were sampled prior to the start of experimental diets, and then at monthly intervals for the next 7 months and examined for the presence of tumors. Tumors were not detected in initiated controls until 7 months after initiation. In initiated trout fed DHEA, the first tumor was detected 5 months after initiation (after just 2 months of dietary DHEA). At 6 months post-initiation, 20% of the AFB1-initiated trout fed DHEA had tumors, while no tumors were visible in either AFB1-initiated controls or noninitiated trout fed DHEA. A second experiment was designed to determine if the enhancing effect of DHEA on AFB1 carcinogenesis is dependent on the time of DHEA administration relative to the time of AFB1 exposure, and if DHEA could be chemopreventive if administered prior to and concurrent with AFB1. Trout were fed one of two levels of DHEA (888 or 1776 p.p.m.) either prior to and during a 4-week initiation period of dietary AFB1 administration, or for 8 weeks following initiation with AFB1. At 9 months after initiation the livers were examined for tumors. Neither exposure protocol provided protection towards AFB1 hepatocarcinogenesis. The strongest enhancement occurred when DHEA was fed during the post-initiation period. Levels of p53 and p34cdc2 were decreased by DHEA treatment, indicating that DHEA may act through alterations in cell-cycle control.


Asunto(s)
Aflatoxina B1/toxicidad , Carcinógenos/toxicidad , Deshidroepiandrosterona/toxicidad , Neoplasias Hepáticas Experimentales/patología , Hígado/patología , Aflatoxina B1/administración & dosificación , Animales , Proteína Quinasa CDC2/análisis , Carcinógenos/administración & dosificación , Ciclo Celular , Deshidroepiandrosterona/administración & dosificación , Dieta , Esquema de Medicación , Sinergismo Farmacológico , Hígado/efectos de los fármacos , Neoplasias Hepáticas Experimentales/inducido químicamente , Factores de Tiempo , Trucha , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/biosíntesis
9.
Cancer Lett ; 118(1): 87-94, 1997 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-9310264

RESUMEN

Indole-3-carbinol (I3C), a natural component from cruciferous vegetables, has been demonstrated to be a modulator of carcinogenesis in various animal models. Along with the promising perspectives of I3C as a possible chemopreventive agent for human breast cancer, some concerns have been raised regarding the tumor-promotional potency of this compound in other target organs. In this study we examined the hepatic tumor-modulatory properties of I3C fed to C57BL/6J mice, initiated with diethylnitrosamine (DEN). Infant male mice were initiated with 0, 2 or 5 mg/kg DEN (i.p. injection) at 15 days of age. Mice were weaned 9 days later and immediately put on AIN76A semipurified diet (with no antioxidants) containing 0 or 0.15% (1500 ppm) I3C. In addition, at the age of 2 months, one group of mice initiated with 2 mg/kg DEN was injected i.p. with a single dose (20 mg/kg) of 3,4,5,3',4',5'-hexachlorobiphenyl (HCB), to serve as a positive control group for promotion. Mice were sampled for hepatic tumors at the age of 6 or 8 months. Each sampled group contained 11-12 mice except the HCB group (nine animals). After 8 months, there was a statistically significant (P < 0.0005) inhibition of hepatocarcinogenesis observed for I3C-fed animals initiated with the high dose of DEN. A single injection of HCB at 2 months of age significantly (P = 0.0003) enhanced hepatocarcinogenesis in mice initiated with 2 mg/kg DEN. There was no statistically significant difference between groups sampled at 6 months of age. Our observations indicate that long term administration of I3C in the diet inhibits DEN-initiated hepatocarcinogenesis in the infant mouse model.


Asunto(s)
Anticarcinógenos/farmacología , Indoles/farmacología , Neoplasias Hepáticas Experimentales/prevención & control , Hígado/efectos de los fármacos , Administración Oral , Envejecimiento , Animales , Neoplasias de la Mama/prevención & control , Carcinógenos , Dietilnitrosamina , Femenino , Alimentos Fortificados , Humanos , Hígado/crecimiento & desarrollo , Hígado/patología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
10.
Toxicol Appl Pharmacol ; 146(1): 69-78, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9299598

RESUMEN

This study investigated the inductive response of cytochrome P4501A (CYP1A) in the zebrafish (Danio rerio) following exposure to Aroclor 1254, beta-naphthoflavone (betaNF), and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and then investigated TCDD modulation of aflatoxin B1 (AFB1) metabolism and hepatic AFB1-DNA adduction. Aroclor 1254 fed at 500 ppm for 1 to 9 days or intraperitoneal (ip) injection of 75-200 mg Aroclor 1254/kg body weight failed to induce CYP1A protein or associated 7-ethoxyresorufin-O-deethylase (EROD) activity. By contrast, dietary betaNF at 500 ppm for 3 or 7 days induced CYP1A protein and EROD activity approximately threefold above controls. A single ip injection of 150 mg/kg betaNF showed maximal induction of CYP1A protein and EROD activity near 24 hr, both of which decreased to control levels during the next 6 days. Single ip administration of 25, 50, 100, or 150 mg betaNF/kg body weight provided dose-responsive increases in CYP1A and EROD activity. Dietary exposure to 0.75 ppm TCDD for 3 days also significantly induced CYP1A and EROD. The effect of TCDD on the metabolism of [3H]AFB1 in zebrafish was then investigated. The major [3H]AFB1 metabolites excreted in water over 24 hr in the control group were aflatoxicol, aflatoxicol-glucuronide, and parent AFB1. By contrast, the predominant metabolites in the TCDD-pretreated group were aflatoxicol-M1-glucuronide, aflatoxicol, aflatoxin M1 plus aflatoxicol-M1 (unresolved), aflatoxicol-glucuronide, and parent AFB1. Surprisingly, hepatic AFB1-DNA adduction was approximately fourfold higher in the TCDD treated group than in controls. This significant difference could not be explained by increased capacity for bioactivation of AFB1 as measured by an in vitro AFB1-exo-8, 9-epoxide trapping assay. However, it was demonstrated that both control and induced zebrafish have high capacity to bioactivate aflatoxin M1 to a reactive intermediate, such that secondary bioactivation of this genotoxic intermediate may be responsible for the increased DNA binding.


Asunto(s)
Aflatoxina B1/metabolismo , Arocloros/farmacología , Sistema Enzimático del Citocromo P-450/biosíntesis , Aductos de ADN/metabolismo , Dibenzodioxinas Policloradas/farmacología , beta-naftoflavona/farmacología , Aflatoxina M1/metabolismo , Animales , Citocromo P-450 CYP1A1/biosíntesis , Inducción Enzimática/efectos de los fármacos , Pez Cebra
11.
Mol Mar Biol Biotechnol ; 6(2): 88-97, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9200835

RESUMEN

Three methods were used in succession to screen a whole adult zebrafish cDNA library for expressed p53-like genes. The sequences of the resultant clones describe an open reading frame 1122 nucleotides in length, with another 43 and 940 bases of 5' and 3' untranslated sequence, respectively. The deduced amino acid sequence of the zebrafish p53 protein is 63% identical to that of trout and 48% identical to that of human p53. Two of the three zebrafish clones overlap to span the entire reported cDNA sequence and are identical in their deduced amino acid sequence over their coincident length. The third clone contains a conservative amino acid change, as well as an inserted amino acid subsequently found to be at the junction of exons 2 and 3, suggestive of alternative splicing in the p53 mRNA for this species. Northern analysis demonstrated a zebrafish p53-related transcript to be present and most abundant in zygotes and early-cleavage embryos less than 1 hour after fertilization, thereafter declining to barely detectable levels at 48 hours. A similar temporal expression was detected for the zebrafish L-myc, known to be present in maternally derived RNA, whereas zebrafish N-myc and the zebrafish homologue of the murine T gene were not detectable prior to the onset of zygotic transcription.


Asunto(s)
Genes p53 , Proteína p53 Supresora de Tumor/biosíntesis , Pez Cebra/embriología , Pez Cebra/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN Complementario/genética , Expresión Génica , Biblioteca de Genes , Ratones , Datos de Secuencia Molecular , Precursores del ARN/metabolismo , ARN Mensajero/análisis , Proteínas Recombinantes/biosíntesis , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Proteína p53 Supresora de Tumor/genética
12.
Toxicol Appl Pharmacol ; 143(1): 213-20, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9073610

RESUMEN

The zebrafish (Danio rerio) is assuming prominence in developmental genetics research. By comparison, little is known of tumorigenesis and nothing is known of carcinogen metabolism in this species. This study evaluated the ability of zebrafish to metabolize a well-characterized human carcinogen, aflatoxin B1 (AFB1), to phase I and phase II metabolites and assessed hepatic AFB1-DNA adduction in vivo. Fish i.p. injected with 50-400 micrograms [3H]AFB1/kg body wt displayed a linear dose response for hepatic DNA binding at 24 hr. AFB1-DNA adduct levels among treatments showed no statistical difference over the period from 1 to 21 days after injection, suggesting poor adduct repair in this species. DNA binding in female fish was 1-7-fold higher than that in males (p < 0.01). An in vitro AFB1 metabolism assay verified that zebrafish liver extracts oxidize AFB1 to the 8,9-epoxide proximate electrophile (Km = 79.0 +/- 16.4 microM, Vmax = 11.7 +/- 1.4 pmol/min/mg protein at 28 degrees C). The excretion of AFB1 and its metabolites was also examined by HPLC. As is typical of other fish studied, major metabolites excreted were aflatoxicol (AFL) and aflatoxicol-glucuronide (AFL-g), followed by unreacted AFB1. AFL appeared as early as 5 min after injection, whereas AFL-g was a significant metabolite after 18 hr. This study shows that in vivo administration of AFB1 to zebrafish results in moderate adduction of the carcinogen to liver DNA and that zebrafish have the capacity for both phase I and phase II metabolism of AFB1. The approximate fourfold difference between rainbow trout and zebrafish AFB1-DNA covalent binding index appears insufficient to explain the relative resistance of zebrafish to dietary AFB1 hepatocarcinogenicity.


Asunto(s)
Aflatoxina B1/metabolismo , Carcinógenos/metabolismo , Aductos de ADN/metabolismo , Hígado/metabolismo , Pez Cebra/metabolismo , Aflatoxina B1/toxicidad , Animales , Bioensayo , Carcinógenos/toxicidad , Aductos de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Inmunidad Innata , Hígado/efectos de los fármacos , Masculino , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/fisiopatología , Oncorhynchus mykiss/metabolismo , Especificidad de la Especie
13.
Toxicol Appl Pharmacol ; 141(2): 548-54, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8975780

RESUMEN

Dehydroepiandrosterone (DHEA) and its sulfate conjugate are the major circulating steroids in human plasma. Low levels of these adrenal steroids are associated with a number of human diseases including certain cancers. In animal studies, DHEA is chemopreventive toward both spontaneous and chemically induced cancers. A potential concern for long-term usage of DHEA in humans is the finding that DHEA is hepatocarcinogenic in rats. The human health risk has been thought to be minimal, however, as the mechanism of DHEA hepatocarcinogenesis is assumed to be due to its properties as a peroxisome proliferator, a class of compounds to which humans are relatively insensitive. Recently, we have found DHEA to be a potent promoter of aflatoxin B1-initiation as well as a complete hepatocarcinogen in the rainbow trout, a species which is also insensitive to peroxisome proliferators. In order to determine the initiator- and tissue-specificity of DHEA promotion, we examined the effects of DHEA on N-methyl-N'-nitro-nitrosoguanidine (MNNG)-initiated carcinogenesis. Trout fry were initiated by a bath exposure (30 min at 35 ppm) to MNNG and then fed DHEA at levels of 0, 55, 111, 222, 444, or 888 ppm for 7 months. DHEA increased liver tumor incidence, multiplicity, and size in a dose-dependent manner. The liver tumor incidence ranged from 0 in the MNNG-initiated controls to 99% in initiated trout fed 888 ppm DHEA. The latter represents a potential synergistic interaction in liver between MNNG and DHEA, as tumor incidence in sham-initiated trout fed this level of DHEA was 41%. The kidney tumor incidence was also enhanced two- and threefold over initiated controls by 111 and 888 ppm DHEA, respectively. In contrast, the total number of stomach and swim bladder tumors was reduced by DHEA treatment. This study demonstrates differential effects of DHEA on MNNG-initiated carcinogenesis in liver, kidney, stomach, and swim bladder.


Asunto(s)
Carcinógenos/toxicidad , Deshidroepiandrosterona/farmacología , Metilnitronitrosoguanidina/toxicidad , Neoplasias Experimentales/inducido químicamente , Animales , Neoplasias Renales/inducido químicamente , Neoplasias Hepáticas Experimentales/inducido químicamente , Oncorhynchus mykiss , Neoplasias Gástricas/inducido químicamente , Neoplasias de la Vejiga Urinaria/inducido químicamente
14.
Fundam Appl Toxicol ; 34(1): 132-40, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8937900

RESUMEN

Dehydroepiandrosterone (DHEA) is an adrenal steroid with chemoprotective effects against a wide variety of conditions including cancer, obesity, diabetes, and cardiovascular disease. However, DHEA is also a carcinogen in laboratory animals, possibly through its function as a precursor of sex steroids or peroxisome proliferation. The structural analog 16 alpha-fluoro-5-androsten-17-one (8354) has been reported to have enhanced chemopreventive activity without the steroid precursor and peroxisome proliferating effects of DHEA. This study compares DHEA and 8354 in rainbow trout, a species that is resistant to peroxisome proliferation but is highly susceptible to the carcinogenic and tumor enhancing effects of DHEA. Trout were exposed as fry to aflatoxin B1 (AFB1) or given a sham exposure, then were fed diets containing 444 ppm DHEA or 8354 for 6 months. Postinitiation treatment with DHEA significantly increased liver tumor incidence, multiplicity, and size compared to initiated controls. The analog 8354 slightly increased tumor incidence (p = 0.06) but had no effect on multiplicity or size. Six percent of trout treated with DHEA alone developed tumors, whereas no tumors occurred in noninitiated trout fed control or 8354-containing diets. Serum levels of androstenedione were elevated by DHEA (48-fold) or 8354 (6-fold) treatment. Serum beta-estradiol titers were increased in DHEA- but not 8354-treated trout. Vitellogenin was induced significantly by either DHEA (434-fold) or 8354 (21-fold). Peroxisomal beta-oxidation was not increased by either compound and catalase activity was decreased in DHEA-treated animals. Both steroids were potent inhibitors in vitro of trout liver glucose-6-phosphate dehydrogenase with IC50s of 24 and 0.5 microM for DHEA and 8354, respectively. This research suggests that in trout the tumor enhancing effects of DHEA may be due to its function as a sex steroid precursor and are unrelated to peroxisome proliferation. These carcinogenic properties are reduced in the analog 8354 which has been advocated as an alternative to DHEA for chemoprevention.


Asunto(s)
Aflatoxina B1/toxicidad , Androstenos/farmacología , Anticarcinógenos/farmacología , Carcinógenos/toxicidad , Deshidroepiandrosterona/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Animales , Sinergismo Farmacológico , Neoplasias Hepáticas Experimentales/sangre , Oncorhynchus mykiss
15.
Carcinogenesis ; 17(11): 2403-9, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8968055

RESUMEN

This study investigated pre-initiation and post-initiation effects of dietary ellagic acid (EA) on 7,12-dimethylbenz[a]anthracene (DMBA) multi-organ carcinogenesis in rainbow trout (Oncorhynchus mykiss). EA at 100, 250 (study 2), 1000 and 2000 (study 1) p.p.m. suppressed stomach adenopapilloma incidence by 33, 60, 70 and 78% (P < or = 0.001), respectively, as well as tumor multiplicity (P < 0.01) and size (P < 0.001) when fed continuously following DMBA initiation. However, continuous EA feeding also produced modest (250 p.p.m.) to extensive (1000, 2000 p.p.m.) growth rate suppression in these studies. Retrospective logistic regression modeling of the data allowed separation of growth-related from non-growth-related inhibitory effects. By this approach: (i) tumor development showed a similarly strong dependence (same regression slope) on animal growth rate in all treatment groups; (ii) EA-mediated reduction in mean population growth contributed to suppressed stomach tumor response above 250 p.p.m. EA; and (iii) even at high, toxic doses EA displayed inhibitory mechanisms additional to, and distinct from, growth suppression effect. The effects of post-initiation EA were organ specific. Chronic EA treatment significantly suppressed swim-bladder as well as stomach tumor incidence at doses > or = 1000 p.p.m., but increased liver tumor incidence at doses > or = 250 p.p.m. Three protocols examined EA effects on the initiation process. EA fed at 1000 p.p.m. concurrently with 750 p.p.m. dietary DMBA for 7 weeks modestly reduced stomach tumor incidence (from 85 to 78%, P < 0.05) and multiplicity (from 6.3 +/- 4.3 to 4.9 +/- 2.9, P < 0.01), but did not alter swim-bladder or liver response. The effect of EA pretreatment prior to DMBA single-dose initiation by gill uptake was also examined. When fed for 1 week prior to initiation, 2000 p.p.m. EA again imposed a small reduction in stomach adenoma incidence (from 88 to 78%; P < 0.05) and multiplicity (from 5.5 +/- 3.2 to 4.4 +/- 3.2; P < 0.01). However, when EA was pre-fed for 3 weeks instead of 1 week, protection in the stomach was lost and response in liver and swim-bladder significantly increased. In sum, these studies demonstrate that EA influence on DMBA tumorigenesis in this multi-organ model is highly protocol dependent and organ specific. Post-initiation dietary EA consistently suppressed stomach tumor development in trout, at EA doses far lower than those required for protection in rodents. At higher doses, however, EA also displayed toxicity and a potential in some protocols to enhance tumor response in other organs.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno , Anticarcinógenos/uso terapéutico , Carcinógenos , Ácido Elágico/uso terapéutico , Neoplasias Experimentales/prevención & control , Especificidad de Órganos , Animales , Dieta , Interacciones Farmacológicas , Neoplasias Experimentales/inducido químicamente , Oncorhynchus mykiss , Papiloma/inducido químicamente , Papiloma/prevención & control
16.
Environ Health Perspect ; 104 Suppl 1: 5-21, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8722107

RESUMEN

Progress over the past 30 years has revealed many strengths of the rainbow trout as an alternative model for environmental carcinogenesis research. These include low rearing costs, an early life-stage ultrasensitive bioassay, sensitivity to many classes of carcinogen, a well-described tumor pathology, responsiveness to tumor promoters and inhibitors, and a mechanistically informative nonmammalian comparative status. Low-cost husbandry, for example, has permitted statistically challenging tumor study designs with up to 10,000 trout to investigate the quantitative interrelationships among carcinogen dose, anticarcinogen dose, DNA adduct formation, and final tumor outcome. The basic elements of the trout carcinogen bioassay include multiple exposure routes, carcinogen response, husbandry requirements, and pathology. The principal known neoplasms occur in liver (mixed hepatocellular/cholangiocellular adenoma and carcinoma, hepatocellular carcinoma), kidney (nephroblastoma), swim bladder (adenopapilloma), and stomach (adenopapilloma). Trout possess a complex but incompletely characterized array of cytochromes P450, transferases, and other enzymic systems for phase I and phase II procarcinogen metabolism. In general, trout exhibit only limited capacity for DNA repair, especially for removal of bulky DNA adducts. This factor, together with a high capacity for P450 bioactivation and negligible glutathione transferase-mediated detoxication of the epoxide, accounts for the exceptional sensitivity of trout to aflatoxin B1 carcinogenesis. At the gene level, all trout tumors except nephroblastoma exhibit variable and often high incidences of oncogenic Ki-ras gene mutations. Mutations in the trout p53 tumor suppressor gene have yet to be described. There are many aspects of the trout model, especially the lack of complete organ homology, that limit its application as a surrogate for human cancer research. Within these limitations, however, it is apparent that trout and other fish models can serve as highly useful adjuncts to conventional rodent models in the study of environmental carcinogenesis and its modulation. For some problems, fish models can provide wholly unique approaches.


Asunto(s)
Carcinógenos Ambientales/toxicidad , Neoplasias Hepáticas/inducido químicamente , Oncorhynchus mykiss , Neoplasias Gástricas/inducido químicamente , Animales , Pruebas de Carcinogenicidad , Carcinógenos Ambientales/administración & dosificación , Carcinógenos Ambientales/metabolismo , Cocarcinogénesis , Sistema Enzimático del Citocromo P-450/metabolismo , Modelos Animales de Enfermedad
17.
Carcinogenesis ; 16(12): 2893-8, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8603461

RESUMEN

Dehydroepiandrosterone (DHEA), fed for 30 weeks to rainbow trout after initiation with the hepatocarcinogen aflatoxin B1 (AFB1), produced a dose-dependent enhancement of carcinogenesis as measured by increased tumor incidence, multiplicity and size. Significant enhancement was observed at 222 p.p.m., which corresponds to a daily dosage one-half that previously administered to humans in clinical trials. DHEA was also capable of acting as a complete carcinogen in this model, producing liver tumors at doses as low as 222-444 p.p.m. Tumors isolated from trout treated with DHEA alone contained mutations in Ki-ras, primarily codon 12[1] G-->A transitions, providing the first suggestive evidence that DHEA could be a genotoxic carcinogen. The carcinogenicity of DHEA in trout is independent of peroxisome proliferation, as measurements of peroxisomal beta-oxidation and catalase activity support previous observations that trout, like humans, are weak responders to peroxisome proliferators.


Asunto(s)
Carcinógenos/toxicidad , Deshidroepiandrosterona/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Microcuerpos/efectos de los fármacos , Aflatoxina B1/toxicidad , Animales , Cocarcinogénesis , Genes ras , Neoplasias Hepáticas Experimentales/genética , Mutación , Oncorhynchus mykiss
18.
Toxicol Pathol ; 23(4): 447-57, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7501957

RESUMEN

Eighteen-mo feeding trials of rainbow trout were used to test the carcinogenicity of 5 chemicals in this species. A single exposure level was used for each substance. The doses and chemicals tested were 1,556 ppm 2,6-dimethylnitrosomorpholine (DMNM), 500 ppm N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), 2,000 ppm 1,2-dibromoethane (DBE), 2,000 ppm 1,1-dichloroethylene (DCE), and 200 ppm cyclophosphamide (CP). Liver and/or glandular stomach neoplasms were produced by DMNM (liver and stomach), MNNG (stomach), and DBE (chiefly, stomach tumors). In addition, DMNM produced a low incidence of swimbladder papillomas and caused testicular atrophy in 50% of treated males. DCE and CP produced no neoplasms at the exposure levels used. No evidence of other chronic toxicity was seen for any of the 5 compounds.


Asunto(s)
Carcinógenos , Dibromuro de Etileno/toxicidad , Metilnitronitrosoguanidina/toxicidad , Nitrosaminas/toxicidad , Alimentación Animal , Animales , Ciclofosfamida/toxicidad , Dicloroetilenos/toxicidad , Femenino , Masculino , Oncorhynchus mykiss , Caracteres Sexuales
19.
Fundam Appl Toxicol ; 25(1): 146-53, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7601322

RESUMEN

Rainbow trout (initial weight of 4 or 5 g) were acclimated at a cool, 11.0 degrees C (C), a warm, 18.0 degrees C (W), or an intermediate temperature 14.5 degrees C (I) for 1 month. There was a slight difference in hepatic microsomal content of one of six cytochrome P450 isozymes between acclimation groups. Monounsaturated fatty acids in hepatic phosphotidylethanolamine but not phosphotidylcholine increased at lower acclimation temperatures. Saturated fatty acid content decreased with temperature for both phospholipid classes. Fish were exposed to 0.08-0.12 ppm waterborne aflatoxin B1 (AFB1) for 30 min at respective acclimation temperatures or after acute temperature shifts (24 hr) and reared for 9 months at C, I, or W. With exposure concentrations which delivered equivalent target organ doses, trout acclimated, exposed, and reared at C, I, or W had liver tumor incidences of 4, 35, and 61%, respectively. The average number of tumors per liver increased from 1.25-1.34 at C to 2.46-2.66 at W. There were no temperature-dependent differences in tumor diameter. When C- and W-acclimated fish were AFB1 exposed and reared at I, tumor incidence was 12.5% for W-I-shifted fish and 26.5% for C-I-shifted fish. This was consistent with previous work which demonstrated acute downward temperature shift reduced [3H]AFB1 adduction to hepatic DNA. Tumor incidence and multiplicity data suggested manipulation of temperature permitted selective modulation of cancer initiation and promotion in rainbow trout.


Asunto(s)
Aflatoxina B1/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Animales , Colesterol/análisis , Ácidos Grasos/análisis , Hígado/química , Microsomas Hepáticos , Oncorhynchus mykiss , Fosfatidilcolinas/análisis , Fosfolípidos/análisis , Temperatura
20.
Food Chem Toxicol ; 32(8): 707-15, 1994 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8070735

RESUMEN

The effectiveness of ammonia in inactivating aflatoxins in contaminated cottonseed was investigated. Two aflatoxin-contaminated cottonseed lots were treated separately using an atmospheric pressure, ambient temperature ammoniation procedure (APAT) or a high pressure, high temperature ammoniation procedure (HPHT), and incorporated into dairy cow rations. Isocalorific diets containing 25% defatted, dried milk from cows fed aflatoxin-contaminated cottonseed without or with APAT or HPHT treatment, or an aflatoxin-free human grade commercial milk powder, were then fed for 12 months to rainbow trout (Oncorhynchus mykiss). Aflatoxin M1 (AFM1) concentrations in milk powders without and with seed treatment were: APAT, 85 and < 0.05 microgram/kg; HPHT, 32 and < 0.05 microgram/kg. In the APAT experiment, trout consuming the diet containing milk from cows fed the aflatoxin-contaminated cottonseed had a 42% incidence of hepatic tumours; APAT cottonseed treatment reduced this to 2.5%. Positive controls were included to demonstrate trout responsiveness. AFB1 fed continuously for 12 months at 4 micrograms/kg resulted in a 34% tumour incidence, whereas positive controls fed 20 micrograms AFB1/kg, 80 micrograms AFM1/kg, or 800 micrograms AFM1/kg for 2 wk and killed 9 months later had a 37, 5.7 and 50% incidence of tumours, respectively. These data demonstrate that APAT ammonia treatment of aflatoxin-contaminated dairy cattle cottonseed feedstock abolished the detectable transfer of AFM1 or AFB1 into milk powder, and greatly reduced the carcinogenic risk posed by any carry-over of aflatoxins or their derivatives into milk. In addition, the results confirm AFM1 to be a lower level hepatocarcinogen in comparison with AFB1 in the trout carcinogenicity assay. In the separate HPHT experiment, no tumours were observed in the livers of trout fed diets containing milk from either the ammonia-treated or untreated source, or the control diet containing 8 micrograms AFM1/kg. Positive controls fed 64 micrograms AFB1/kg for 2 wk exhibited a 29% tumour incidence 12 months later. Thus in this experiment, neither AFM1 at 8 micrograms/kg nor any HPHT-derived aflatoxin derivatives that might have been carried over into milk, represented a detectably carcinogenic hazard to trout.


Asunto(s)
Aflatoxina B1/toxicidad , Aflatoxina M1/análisis , Amoníaco/farmacología , Alimentación Animal/toxicidad , Contaminación de Alimentos , Neoplasias Hepáticas Experimentales/inducido químicamente , Leche/química , Aflatoxina B1/análisis , Aflatoxina M1/toxicidad , Animales , Bovinos , Aceite de Semillas de Algodón , Femenino , Oncorhynchus mykiss
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...