Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(52): 33414-33425, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33318186

RESUMO

The ability to accurately measure mutations is critical for basic research and identifying potential drug and chemical carcinogens. Current methods for in vivo quantification of mutagenesis are limited because they rely on transgenic rodent systems that are low-throughput, expensive, prolonged, and do not fully represent other species such as humans. Next-generation sequencing (NGS) is a conceptually attractive alternative for detecting mutations in the DNA of any organism; however, the limit of resolution for standard NGS is poor. Technical error rates (∼1 × 10-3) of NGS obscure the true abundance of somatic mutations, which can exist at per-nucleotide frequencies ≤1 × 10-7 Using duplex sequencing, an extremely accurate error-corrected NGS (ecNGS) technology, we were able to detect mutations induced by three carcinogens in five tissues of two strains of mice within 31 d following exposure. We observed a strong correlation between mutation induction measured by duplex sequencing and the gold-standard transgenic rodent mutation assay. We identified exposure-specific mutation spectra of each compound through trinucleotide patterns of base substitution. We observed variation in mutation susceptibility by genomic region, as well as by DNA strand. We also identified a primordial marker of carcinogenesis in a cancer-predisposed strain of mice, as evidenced by clonal expansions of cells carrying an activated oncogene, less than a month after carcinogen exposure. These findings demonstrate that ecNGS is a powerful method for sensitively detecting and characterizing mutagenesis and the early clonal evolutionary hallmarks of carcinogenesis. Duplex sequencing can be broadly applied to basic mutational research, regulatory safety testing, and emerging clinical applications.


Assuntos
Carcinogênese/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutagênese/genética , Animais , Carcinógenos/toxicidade , Análise por Conglomerados , DNA/genética , Genes ras , Loci Gênicos , Genoma , Humanos , Camundongos Transgênicos , Mutação/genética , Neoplasias/genética , Oncogenes , Fenótipo , Transcrição Gênica
2.
Environ Mol Mutagen ; 61(8): 770-785, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32078182

RESUMO

Genome instability is a hallmark of most human cancers and is exacerbated following replication stress. However, the effects that drugs/xenobiotics have in promoting genome instability including chromosomal structural rearrangements in normal cells are not currently assessed in the genetic toxicology battery. Here, we show that drug-induced replication stress leads to increased genome instability in vitro using proliferating primary human cells as well as in vivo in rat bone marrow (BM) and duodenum (DD). p53-binding protein 1 (53BP1, biomarker of DNA damage repair) nuclear bodies were increased in a dose-dependent manner in normal proliferating human mammary epithelial fibroblasts following treatment with compounds traditionally classified as either genotoxic (hydralazine) and nongenotoxic (low-dose aphidicolin, duvelisib, idelalisib, and amiodarone). Comparatively, no increases in 53BP1 nuclear bodies were observed in nonproliferating cells. Negative control compounds (mannitol, alosteron, diclofenac, and zonisamide) not associated with cancer risk did not induce 53BP1 nuclear bodies in any cell type. Finally, we studied the in vivo genomic consequences of drug-induced replication stress in rats treated with 10 mg/kg of cyclophosphamide for up to 14 days followed by polymerase chain reaction-free whole genome sequencing (30X coverage) of BM and DD cells. Cyclophosphamide induced chromosomal structural rearrangements at an average of 90 genes, including 40 interchromosomal/intrachromosomal translocations, within 2 days of treatment. Collectively, these data demonstrate that this drug-induced genome instability test (DiGIT) can reveal potential adverse effects of drugs not otherwise informed by standard genetic toxicology testing batteries. These efforts are aligned with the food and drug administration's (FDA's) predictive toxicology roadmap initiative.


Assuntos
Replicação do DNA/efeitos dos fármacos , Genoma/efeitos dos fármacos , Instabilidade Genômica , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Biomarcadores/metabolismo , Aberrações Cromossômicas , Ciclofosfamida/toxicidade , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Sequenciamento Completo do Genoma
3.
Trends Pharmacol Sci ; 39(3): 232-247, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29242029

RESUMO

Cancer risk assessment of therapeutics is plagued by poor translatability of rodent models of carcinogenesis. In order to overcome this fundamental limitation, new approaches are needed that enable us to evaluate cancer risk directly in humans and human-based cellular models. Our enhanced understanding of the mechanisms of carcinogenesis and the influence of human genome sequence variation on cancer risk motivates us to re-evaluate how we assess the carcinogenic risk of therapeutics. This review will highlight new opportunities for applying this knowledge to the development of a battery of human-based in vitro models and biomarkers for assessing cancer risk of novel therapeutics.


Assuntos
Carcinógenos/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Neoplasias/prevenção & controle , Farmacovigilância , Biomarcadores Farmacológicos/análise , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Predisposição Genética para Doença , Humanos
4.
Int J Toxicol ; 35(3): 294-308, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26941242

RESUMO

Etelcalcetide is a novel d-amino acid peptide that functions as an allosteric activator of the calcium-sensing receptor and is being developed as an intravenous calcimimetic for the treatment of secondary hyperparathyroidism in patients with chronic kidney disease on hemodialysis. To support clinical development and marketing authorization, a comprehensive nonclinical safety package was generated. Primary adverse effects included hypocalcemia, tremoring, and convulsions. Other adverse effects were considered sequelae of stress associated with hypocalcemia. Cardiovascular safety evaluations in the dog revealed an anticipated prolongation of the corrected QT interval that was related to reductions in serum calcium. Etelcalcetide did not affect the human ether-a-go-go gene ion channel current. Etelcalcetide was mutagenic in some strains of Salmonella, however, based on the negative results in 2 in vitro and 2 in vivo mammalian genotoxicity assays, including a 28-day Muta mouse study, etelcalcetide is considered nongenotoxic. Further support for a lack of genotoxicity was provided due to the fact that etelcalcetide was not carcinogenic in a 6-month transgenic rasH2 mouse model or a 2-year study in rats. There were no effects on fertility, embryo-fetal development, and prenatal and postnatal development. All of the adverse effects observed in both rat and dog were considered directly or secondarily related to the pharmacologic activity of etelcalcetide and the expected sequelae associated with dose-related reductions in serum calcium due to suppression of parathyroid hormone secretion. These nonclinical data indicate no safety signal of concern for human risk beyond that associated with hypocalcemia and associated QT prolongation.


Assuntos
Peptídeos/toxicidade , Animais , Pressão Sanguínea/efeitos dos fármacos , Cálcio/sangue , Cães , Canal de Potássio ERG1/fisiologia , Feminino , Células HEK293 , Frequência Cardíaca/efeitos dos fármacos , Humanos , Hiperparatireoidismo Secundário/tratamento farmacológico , Hipocalcemia/induzido quimicamente , Masculino , Camundongos Transgênicos , Testes de Mutagenicidade , Peptídeos/farmacocinética , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Coelhos , Ratos Sprague-Dawley , Reprodução/efeitos dos fármacos , Convulsões/induzido quimicamente , Tremor/induzido quimicamente
5.
Regul Toxicol Pharmacol ; 70(1): 87-97, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24932799

RESUMO

Large molecule therapeutics (MW>1000daltons) are not expected to enter the cell and thus have reduced potential to interact directly with DNA or related physiological processes. Genotoxicity studies are therefore not relevant and typically not required for large molecule therapeutic candidates. Regulatory guidance supports this approach; however there are examples of marketed large molecule therapeutics where sponsors have conducted genotoxicity studies. A retrospective analysis was performed on genotoxicity studies of United States FDA approved large molecule therapeutics since 1998 identified through the Drugs@FDA website. This information was used to provide a data-driven rationale for genotoxicity evaluations of large molecule therapeutics. Fifty-three of the 99 therapeutics identified were tested for genotoxic potential. None of the therapeutics tested showed a positive outcome in any study except the peptide glucagon (GlucaGen®) showing equivocal in vitro results, as stated in the product labeling. Scientific rationale and data from this review indicate that testing of a majority of large molecule modalities do not add value to risk assessment and support current regulatory guidance. Similarly, the data do not support testing of peptides containing only natural amino acids. Peptides containing non-natural amino acids and small molecules in conjugated products may need to be tested.


Assuntos
Testes de Mutagenicidade/métodos , Preparações Farmacêuticas/administração & dosagem , Medição de Risco/métodos , Aprovação de Drogas , Rotulagem de Medicamentos , Glucagon/toxicidade , Humanos , Peso Molecular , Peptídeos/toxicidade , Preparações Farmacêuticas/química , Estudos Retrospectivos , Estados Unidos , United States Food and Drug Administration
7.
Toxicol Sci ; 124(1): 54-74, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21813463

RESUMO

Evaluating the risk of chemical carcinogenesis has long been a challenge owing to the protracted nature of the pathology and the limited translatability of animal models. Although numerous short-term in vitro and in vivo assays have been developed, they have failed to reliably predict the carcinogenicity of nongenotoxic compounds. Extending upon previous microarray work (Fielden, M. R., Nie, A., McMillian, M., Elangbam, C. S., Trela, B. A., Yang, Y., Dunn, R. T., II, Dragan, Y., Fransson-Stehen, R., Bogdanffy, M., et al. (2008). Interlaboratory evaluation of genomic signatures for predicting carcinogenicity in the rat. Toxicol. Sci. 103, 28-34), we have developed and extensively evaluated a quantitative PCR-based signature to predict the potential for nongenotoxic compounds to induce liver tumors in the rat as a first step in the safety assessment of potential nongenotoxic carcinogens. The training set was derived from liver RNA from rats treated with 72 compounds and used to develop a 22-gene signature on the TaqMan array platform, providing an economical and standardized assay protocol. Independent testing on over 900 diverse samples (66 compounds) confirmed the interlaboratory precision of the assay and its ability to predict known nongenotoxic hepatocarcinogens (NGHCs). When tested under different experimental designs, strains, time points, dose setting criteria, and other preanalytical processes, the signature sensitivity and specificity was estimated to be 67% (95% confidence interval [CI] = 38-88%) and 59% (95% CI = 44-72%), respectively, with an area under the receiver operating characteristic curve of 0.65 (95% CI = 0.46-0.83%). Compounds were best classified using expression data from short-term repeat dose studies; however, the prognostic expression changes appeared to be preserved after longer term treatment. Exploratory evaluations also revealed that different modes of action for nongenotoxic and genotoxic compounds can be discriminated based on the expression of specific genes. These results support a potential early preclinical testing paradigm to catalyze broader understanding of putative NGHCs.


Assuntos
Carcinógenos/toxicidade , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/induzido quimicamente , Fígado/efeitos dos fármacos , Modelos Genéticos , Animais , Carcinógenos/classificação , Marcadores Genéticos , Genômica , Fígado/metabolismo , Neoplasias Hepáticas Experimentais/genética , Masculino , Valor Preditivo dos Testes , Ratos , Ratos Sprague-Dawley
8.
Chem Biol Interact ; 176(2-3): 252-60, 2008 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19086139

RESUMO

Carcinogenicity of chemicals can currently only be evaluated in 2-year rodent bioassays. Therefore, the development of early biomarkers for carcinogenesis would result in substantial savings in time and expense. The current study investigates whether early changes in gene expression may be developed as markers for cancer. Animals were treated for 1 or 5 days with either non-genotoxic carcinogens or non-carcinogens and gene expression was analyzed by quantitative PCR (qPCR).We tested two gene signatures previously reported to detect non-genotoxic carcinogens. Using one gene signature it was confirmed that 3/3 nongenotoxic carcinogens and 2/2 non-carcinogens are correctly identified with data from 1 or 5 days of dosing. In contrast an alternative signature correctly identified 0/3 and 2/3 nongenotoxic carcinogens at 1 and 5 days of treatment, respectively and 2/2 non-carcinogens at both time-points. Additionally, we evaluated a novel panel of putative biomarker genes, from the literature, many of which have roles in cell growth and division, including myc, cdc2 and mcm6. These genes were significantly induced by non-genotoxic carcinogens and not by non-carcinogens. Using the average fold-induction across this panel, 2/3 non-genotoxic carcinogens were detected on both day 1 and day 5. These data support the idea that acute changes in gene expression may provide biomarkers for non-genotoxic carcinogenesis but also highlight interesting differences in the sensitivities of distinct gene signatures.


Assuntos
Bioensaio/métodos , Biomarcadores/análise , Carcinógenos/toxicidade , Regulação Neoplásica da Expressão Gênica/genética , Valor Preditivo dos Testes , Animais , Testes de Carcinogenicidade , Relação Dose-Resposta a Droga , Masculino , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Medição de Risco , Fatores de Tempo
9.
Toxicology ; 246(2-3): 91-100, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18289764

RESUMO

Drug-induced renal injury is a common finding in the early preclinical phase of drug development. But the specific genes responding to renal injury remain poorly defined. Identification of drug-induced gene changes is critical to provide insights into molecular mechanisms and detection of renal damage. To identify genes associated with the development of drug-induced nephrotoxicity, a literature survey was conducted and a panel of 48 genes was selected based on gene expression changes in multiple published studies. Male Sprague-Dawley rats were dosed daily for 1, 3 or 5 days to the known nephrotoxicants gentamicin, bacitracin, vancomycin and cisplatin, or the known hepatotoxicants ketoconazole, 1-naphthyl isothiocyanate and 4,4-diaminodiphenylmethane. Histopathological evaluation and clinical chemistry revealed renal proximal tubular necrosis in rats treated with the nephrotoxicants, but not from those treated with the hepatotoxicants. RNA was extracted from the kidney, and RT-PCR was performed to evaluate expression profiles of the selected genes. Among the genes examined, 24 genes are confirmed to be highly induced or repressed in rats treated with nephrotoxicants; further investigation identified that 5 of the 24 genes were also altered by hepatotoxicants. These data led to the identification of a set of genomic biomarker candidates whose expression in kidney is selectively regulated only by nephrotoxicants. Among those genes displaying the highest expression changes specifically in nephrotoxicant-treated rats were kidney injury molecule 1 (Kim1), lipocalin 2 (Lcn2), and osteopontin (Spp1). The establishment of such a genomic marker set offers a new tool in our ongoing quest to monitor nephrotoxicity.


Assuntos
Antibacterianos/toxicidade , Antineoplásicos/toxicidade , Marcadores Genéticos , Nefropatias/induzido quimicamente , Nefropatias/genética , Rim/efeitos dos fármacos , Animais , Bacitracina/toxicidade , Cisplatino/toxicidade , Expressão Gênica/genética , Perfilação da Expressão Gênica , Gentamicinas/toxicidade , Rim/metabolismo , Rim/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Toxicogenética , Vancomicina/toxicidade
10.
Toxicol Sci ; 103(1): 28-34, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18281259

RESUMO

The Critical Path Institute recently established the Predictive Safety Testing Consortium, a collaboration between several companies and the U.S. Food and Drug Administration, aimed at evaluating and qualifying biomarkers for a variety of toxicological endpoints. The Carcinogenicity Working Group of the Predictive Safety Testing Consortium has concentrated on sharing data to test the predictivity of two published hepatic gene expression signatures, including the signature by Fielden et al. (2007, Toxicol. Sci. 99, 90-100) for predicting nongenotoxic hepatocarcinogens, and the signature by Nie et al. (2006, Mol. Carcinog. 45, 914-933) for predicting nongenotoxic carcinogens. Although not a rigorous prospective validation exercise, the consortium approach created an opportunity to perform a meta-analysis to evaluate microarray data from short-term rat studies on over 150 compounds. Despite significant differences in study designs and microarray platforms between laboratories, the signatures proved to be relatively robust and more accurate than expected by chance. The accuracy of the Fielden et al. signature was between 63 and 69%, whereas the accuracy of the Nie et al. signature was between 55 and 64%. As expected, the predictivity was reduced relative to internal validation estimates reported under identical test conditions. Although the signatures were not deemed suitable for use in regulatory decision making, they were deemed worthwhile in the early assessment of drugs to aid decision making in drug development. These results have prompted additional efforts to rederive and evaluate a QPCR-based signature using these samples. When combined with a standardized test procedure and prospective interlaboratory validation, the accuracy and potential utility in preclinical applications can be ascertained.


Assuntos
Testes de Carcinogenicidade/métodos , Genômica , Animais , Perfilação da Expressão Gênica , Masculino , Ratos , Ratos Sprague-Dawley
11.
Toxicol Sci ; 99(1): 90-100, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17557906

RESUMO

There are currently no accurate and well-validated short-term tests to identify nongenotoxic hepatic tumorigens, thus necessitating an expensive 2-year rodent bioassay before a risk assessment can begin. Using hepatic gene expression data from rats treated for 5 days with one of 100 structurally and mechanistically diverse nongenotoxic hepatocarcinogens and nonhepatocarcinogens, a novel multigenebiomarker (i.e., signature) was derived to predict the likelihood of nongenotoxic chemicals to induce liver tumors in longer term studies. Independent validation of the signature on 47 test chemicals indicates an assay sensitivity and specificity of 86% and 81%, respectively. Alternate short-term in vivo pathological and genomic biomarkers were evaluated in parallel for comparison, including liver weight, hepatocellular hypertrophy, hepatic necrosis, serum alanine aminotransferase activity, induction of cytochrome P450 genes, and repression of Tsc-22 or alpha2-macroglobulin messenger RNA. In contrast to these biomarkers, the gene expression-based signature was more accurate. Unlike existing tests, an understanding of potential modes of action for hepatic tumorigenicity can be derived by comparison of the signature profile of test chemicals to hepatic tumorigens of known mechanism, including regenerative proliferation, proliferation associated with xenobiotic receptor activation, peroxisome proliferation, and steroid hormone-mediated mechanisms. This signature is not only more accurate than current methods, but also facilitates the identification of mode of action to aid in the early assessment of human cancer risk.


Assuntos
Biomarcadores Tumorais/genética , Carcinógenos/toxicidade , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/genética , Alanina Transaminase/sangue , Animais , Biomarcadores Tumorais/metabolismo , Carcinógenos/classificação , Sistema Enzimático do Citocromo P-450/biossíntese , Indução Enzimática , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Necrose , Análise de Sequência com Séries de Oligonucleotídeos , Valor Preditivo dos Testes , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reprodutibilidade dos Testes , alfa-Macroglobulinas/genética , alfa-Macroglobulinas/metabolismo
12.
J Biotechnol ; 119(3): 219-44, 2005 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16005536

RESUMO

Successful drug discovery requires accurate decision making in order to advance the best candidates from initial lead identification to final approval. Chemogenomics, the use of genomic tools in pharmacology and toxicology, offers a promising enhancement to traditional methods of target identification/validation, lead identification, efficacy evaluation, and toxicity assessment. To realize the value of chemogenomics information, a contextual database is needed to relate the physiological outcomes induced by diverse compounds to the gene expression patterns measured in the same animals. Massively parallel gene expression characterization coupled with traditional assessments of drug candidates provides additional, important mechanistic information, and therefore a means to increase the accuracy of critical decisions. A large-scale chemogenomics database developed from in vivo treated rats provides the context and supporting data to enhance and accelerate accurate interpretation of mechanisms of toxicity and pharmacology of chemicals and drugs. To date, approximately 600 different compounds, including more than 400 FDA approved drugs, 60 drugs approved in Europe and Japan, 25 withdrawn drugs, and 100 toxicants, have been profiled in up to 7 different tissues of rats (representing over 3200 different drug-dose-time-tissue combinations). Accomplishing this task required evaluating and improving a number of in vivo and microarray protocols, including over 80 rigorous quality control steps. The utility of pairing clinical pathology assessments with gene expression data is illustrated using three anti-neoplastic drugs: carmustine, methotrexate, and thioguanine, which had similar effects on the blood compartment, but diverse effects on hepatotoxicity. We will demonstrate that gene expression events monitored in the liver can be used to predict pathological events occurring in that tissue as well as in hematopoietic tissues.


Assuntos
Biotecnologia/métodos , Desenho de Fármacos , Indústria Farmacêutica/métodos , 5-Aminolevulinato Sintetase/biossíntese , Animais , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Automação , Ductos Biliares/patologia , Carmustina/toxicidade , Biologia Computacional , Bases de Dados como Assunto , Relação Dose-Resposta a Droga , Regulação para Baixo , Expressão Gênica , Humanos , Hiperplasia/etiologia , Fígado/efeitos dos fármacos , Masculino , Metotrexato/toxicidade , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho do Órgão , Farmacologia/métodos , RNA/química , RNA Complementar/metabolismo , Ratos , Ratos Sprague-Dawley , Reticulócitos/citologia , Reticulócitos/metabolismo , Tioguanina/toxicidade , Fatores de Tempo , Distribuição Tecidual , Toxicologia/métodos
13.
Am J Pharmacogenomics ; 5(3): 161-71, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15952870

RESUMO

The economic hurdles of drug development and the emergence of genomic technologies such as chemogenomics are combining to shift the existing paradigms in preclinical drug development. Today, the information gleaned from high content molecular data has begun to augment traditional approaches to the assessment of drug safety. The optimal approach is a hybrid strategy employing chemogenomic data and gene expression-based biomarkers of drug efficacy and toxicity to supplement low content and insensitive methods for risk assessment and mechanistic evaluation of drug candidates. Large reference databases of chemogenomic data are essential to the derivation and validation of accurate and predictive gene expression biomarkers. An example of the development of a predictive biomarker for hepatic bile duct hyperplasia is described herein. As gene expression technologies improve, biomarkers will achieve higher throughput, and become more cost effective and increasingly accurate. This will elevate the value of chemogenomics in drug development, shift attrition to earlier in the process, and reduce the overall cost of drug development. Over the past 2 to 3 years, the transition of chemogenomics from a research tool to a decision-making tool has begun and regulatory agencies are anxiously awaiting implementation of this technology to make faster and more informed evaluations of potential drugs.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Fígado/efeitos dos fármacos , Farmacogenética/métodos , Animais , Ductos Biliares/metabolismo , Ductos Biliares/patologia , Biomarcadores , Carcinógenos/toxicidade , Bases de Dados Factuais , Perfilação da Expressão Gênica , Humanos , Hiperplasia , Técnicas In Vitro , Fígado/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oncogenes/efeitos dos fármacos , Farmacogenética/estatística & dados numéricos , Segurança
14.
Endocrinology ; 143(8): 3044-59, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12130571

RESUMO

The objective of the study was to determine the long-term effects of gestational and lactational exposure to diethylstilbestrol (DES; 0, 0.1, 1, and 10 microg/kg maternal body weight) on mouse testicular growth, epididymal sperm count, in vitro fertilizing ability, and testicular gene expression using cDNA microarrays and real-time PCR in mice on postnatal day (PND) 21, 105, and 315. In the high dose group there was a persistent decrease in the number of Sertoli cells, and sperm count was decreased on PND315 (P < 0.05). Sperm motion was unaffected; however, the in vitro fertilizing ability of epididymal sperm was decreased in the high dose group on both PND105 (P < 0.001) and PND315 (P < 0.05). Early and latent alterations in the expression of genes involved in estrogen signaling (estrogen receptor alpha), steroidogenesis (steroidogenic factor 1, 17alpha-hydroxylase/C17,20-lyase, P450 side chain cleavage, steroidogenic acute regulatory protein, and scavenger receptor class B1), lysosomal function (LGP85 and prosaposin), and regulation of testicular development (testicular receptor 2, inhibin/activin beta C, and Hoxa10) were confirmed by real-time PCR. The results demonstrate that early exposure to DES causes long-term adverse effects on testicular development and sperm function, and these effects are associated with changes in testicular gene expression, even long after the cessation of DES exposure.


Assuntos
Dietilestilbestrol/toxicidade , Fertilização in vitro , Feto/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Espermatozoides/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Feminino , Expressão Gênica/efeitos dos fármacos , Lactação , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Gravidez , Testículo/metabolismo
15.
Toxicol Lett ; 133(2-3): 181-91, 2002 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-12119126

RESUMO

The objective of the study was to determine the effect of in utero and lactational exposure to genistein (0, 0.1, 0.5, 2.5 and 10 mg/kg/day) on mammary gland morphology in female B6D2F1 mice at levels comparable to or greater than human exposures. The effect of diethylstilbestrol (DES; 0, 0.1, 1, 10 microg/kg/day) on the mammary gland was also examined as a positive estrogenic control. Pregnant females were treated by daily gavage from gestational day 12 to postnatal day (PND) 20. Female offspring were weaned on PND21 and mammary gland whole mounts were examined for growth (length and area of the epithelial tree), proliferation (number of terminal end buds (TEBs)), and differentiation (density of alveolar buds (ABs)) on PND49. The highest dose of DES induced a significant increase in mammary gland growth (P<0.05) and also decreased the number of TEBs (P<0.06). The density of ABs was not significantly affected by DES. By contrast to DES, genistein had no effect on mammary gland morphology at any dose. These results suggest that in utero and lactational exposure to genistein at levels comparable to or greater than human exposures do not adversely affect mammary gland development in pubertal female B6D2F1 mice.


Assuntos
Anticarcinógenos/farmacologia , Genisteína/farmacologia , Lactação/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Efeitos Tardios da Exposição Pré-Natal , Maturidade Sexual/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Dieta , Feminino , Crescimento/efeitos dos fármacos , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA