Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Cancer ; 135(7): 1574-85, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24535843

RESUMEN

The process of hepatocarcinogenesis in the diethylnitrosamine (DEN) initiation/phenobarbital (PB) promotion mouse model involves the selective clonal outgrowth of cells harboring oncogene mutations in Ctnnb1, while spontaneous or DEN-only-induced tumors are often Ha-ras- or B-raf-mutated. The molecular mechanisms and pathways underlying these different tumor sub-types are not well characterized. Their identification may help identify markers for xenobiotic promoted versus spontaneously occurring liver tumors. Here, we have characterized mouse liver tumors harboring either Ctnnb1 or Ha-ras mutations via integrated molecular profiling at the transcriptional, translational and post-translational levels. In addition, metabolites of the intermediary metabolism were quantified by high resolution (1)H magic angle nuclear magnetic resonance. We have identified tumor genotype-specific differences in mRNA and miRNA expression, protein levels, post-translational modifications, and metabolite levels that facilitate the molecular and biochemical stratification of tumor phenotypes. Bioinformatic integration of these data at the pathway level led to novel insights into tumor genotype-specific aberrant cell signaling and in particular to a better understanding of alterations in pathways of the cell intermediary metabolism, which are driven by the constitutive activation of the ß-Catenin and Ha-ras oncoproteins in tumors of the two genotypes.


Asunto(s)
Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica , Genes ras/genética , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Metabolómica , Mutación/genética , beta Catenina/genética , Animales , Biomarcadores de Tumor/metabolismo , Western Blotting , Redes y Vías Metabólicas , Ratones , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
2.
Toxicol Rep ; 8: 155-161, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33473352

RESUMEN

Species differences in hepatic metabolism of thyroxine (T4) by uridine diphosphate glucuronosyl transferase (UGT) and susceptibility to thyroid hormone imbalance could underlie differences in thyroid carcinogenesis caused by hepatic enzyme inducers in rats and humans. To investigate this hypothesis we examined profiles of hepatic UGT induction by the prototypical CAR activator phenobarbital (PB) in rat and human liver 3D microtissues. The rationale for this approach was that 3D microtissues would generate data more relevant to humans. Rat and human liver 3D microtissues were exposed to PB over a range of concentrations (500 u M - 2000 u M) and times (24-96 hr). Microarray and proteomics analyses were performed on parallel samples to generate integrated differentially expressed gene (DEG) datasets. Bioinformatics analysis of DEG data, including CAR response element (CRE) sequence analysis of UGT promoters, was used to assess species differences in UGT induction relative to CAR-mediated transactivation potential. A higher proportion of human UGT promoters were found to contain consensus CREs compared to the rat homologs. UGTs 1a6, 2b17 and 2b37 were upregulated by PB in rat liver 3D microtissues, but unaltered in human liver 3D microtissues. By contrast, human UGTs 1A8, 1A10 and 2B10 showed higher levels of induction (RNA and /or protein) compared to the rat homologs. There was general concordance between the presence of CREs and the induction of UGT RNA. As UGT1A and 2B isoforms metabolise T4, these results suggest that differences in UGT induction could contribute to differential susceptibility to CAR-mediated thyroid carcinogenesis in rats and humans.

3.
Carcinogenesis ; 30(9): 1571-80, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19608619

RESUMEN

To better understand the role of transcription factor NF-E2-related factor (NRF) 2 in the human and its contribution to cancer chemoprevention, we have knocked down its negative regulators, Kelch-like ECH-associated protein 1 (KEAP1) and broad-complex, tramtrack and bric à brac and cap'n'collar homology 1 (BACH1), in HaCaT keratinocytes. Whole-genome microarray revealed that knockdown of KEAP1 resulted in 23 messenger RNAs (mRNAs) being up-regulated > or = 2.0-fold. mRNA for aldo-keto reductase (AKR) 1B10, AKR1C1, AKR1C2 and AKR1C3 were induced to the greatest extent, showing increases of between 12- and 16-fold, whereas mRNA for glutamate-cysteine ligase catalytic and modifier subunits, NAD(P)H:quinone oxidoreductase-1 and haem oxygenase-1 (HMOX1) were induced between 2.0- and 4.8-fold. Knockdown of BACH1 increased HMOX1 135-fold but induced the other genes examined to a maximum of only 2.7-fold. Activation of NRF2, by KEAP1 knockdown, caused a 75% increase in the amount of glutathione in HaCaT cells and a 1.4- to 1.6-fold increase in their resistance to the electrophiles acrolein, chlorambucil and cumene hydroperoxide (CuOOH), as well as the redox-cycling agent menadione. Inhibition of glutathione synthesis during KEAP1 knockdown, by treatment with buthionine sulfoximine, abrogated resistance to acrolein, chlorambucil and CuOOH, but not to menadione. In contrast, knockdown of BACH1 did not increase glutathione levels or resistance to xenobiotics. Knockdown of NRF2 in HaCaT cells decreased glutathione to approximately 80% of normal homeostatic levels and similarly reduced their tolerance of electrophiles. Thus, the KEAP1-NRF2 pathway determines resistance to electrophiles and redox-cycling compounds in human keratinocytes through glutathione-dependent and glutathione-independent mechanisms. This study also shows that AKR1B10, AKR1C1 and AKR1C2 proteins have potential utility as biomarkers for NRF2 activation in the human.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Citoprotección , Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Queratinocitos/metabolismo , Factor 2 Relacionado con NF-E2/fisiología , Neoplasias/prevención & control , 20-Hidroxiesteroide Deshidrogenasas/genética , Células Cultivadas , Perfilación de la Expresión Génica , Glutatión/metabolismo , Hemo-Oxigenasa 1/genética , Humanos , Hidroxiesteroide Deshidrogenasas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Queratinas/genética , Elementos de Respuesta , Transducción de Señal
4.
Toxicol Rep ; 6: 998-1005, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31673501

RESUMEN

Characterisation of the mode of action (MOA) of constitutive androstane receptor (CAR)-mediated rodent liver tumours involves measurement 5 key events including activation of the CAR receptor, altered gene expression, hepatocellular proliferation, clonal expansion and increased hepatocellular adenomas/carcinomas. To test whether or not liver 3D microtissues (LiMTs) recapitulate CAR- mediated procarcinogenic key events in response to the prototypical CAR activator phenobarbital (PB) we performed hepatocyte proliferation (LI%) analysis in rat and human LiMTs using a microTMA technology in conjunction with integrated transcriptomics (microarray) and proteomics analysis. The rationale for this approach was that LiMTs containing parenchymal and non-parenchymal cells (NPCs) are more physiologically representative of liver and thus would generate data more relevant to the in vivo situation. Rat and human LiMTs were treated with PB over a range of concentrations (500 uM - 2000 uM) and times (24 h - 96 h) in a dose-response/time-course analysis. There was a dose-dependent induction of LI% in rat LiMTs, however there was little or no effect of PB on LI% in human LiMTs. ATP levels in the rat and human LiMTs were similar to control in all of the PB treatments. There was also a dose- and time-dependent PB-mediated RNA induction of CAR regulated genes CYP2B6/Cyp2b2, CYP3A7/Cyp3a9 and UGT1A6/Ugt1a6 in human and rat LiMTs, respectively. These CAR regulated genes were also upregulated at the protein level. Ingenuity pathways analysis (IPA) indicated that there was a significant (Z score >2.0;-log p value >) activation of CAR by PB in both human and rat LiMTs. These results indicate that human and rat LiMTs showed the expected responses at the level of PB-induced hepatocyte proliferation and enzyme induction with rat LiMTs showing significant dose-dependent effects while human LiMTs showed no proliferation response but did show dose-dependent enzyme induction at the RNA and protein levels. In conclusion LiMTs serve as a model to provide mechanistic data for 3 of the 5 key events considered necessary to establish a CAR-mediated MOA for liver tumourigenesis and thus can potentially reduce the use of animals when compiling mechanistic data packages.

5.
Sci Rep ; 9(1): 1407, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723234

RESUMEN

A high throughput histology (microTMA) platform was applied for testing drugs against tumors in a novel 3D heterotypic glioblastoma brain sphere (gBS) model consisting of glioblastoma tumor cells, iPSC-derived neurons, glial cells and astrocytes grown in a spheroid. The differential responses of gBS tumors and normal neuronal cells to sustained treatments with anti-cancer drugs temozolomide (TMZ) and doxorubicin (DOX) were investigated. gBS were exposed to TMZ or DOX over a 7-day period. Untreated gBS tumors increased in size over a 4-week culture period, however, there was no increase in the number of normal neuronal cells. TMZ (100 uM) and DOX (0.3 uM) treatments caused ~30% (P~0.07) and ~80% (P < 0.001) decreases in the size of the tumors, respectively. Neither treatment altered the number of normal neuronal cells in the model. The anti-tumor effects of TMZ and DOX were mediated in part by selective induction of apoptosis. This platform provides a novel approach for screening new anti-glioblastoma agents and evaluating different treatment options for a given patient.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Glioblastoma/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Modelos Biológicos , Medicina de Precisión/métodos , Esferoides Celulares/efectos de los fármacos , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Glioblastoma/patología , Humanos , Temozolomida/farmacología
6.
Toxicol Sci ; 97(2): 520-32, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17379624

RESUMEN

We undertook transcription profiling of fetal testis RNA on gestational days e15.5, 17.5, and 19.5 in offspring from dams treated daily from e12.5 with 500 mg/kg di(n-butyl) phthalate (DBP). At e17.5-19.5, reduced expression of genes involved in cholesterol uptake/metabolism and steroidogenesis was identified in DBP-exposed animals, including scavenger receptor B1 (SCARB1), HMGCoA synthase, steroidogenic acute regulatory protein, Cyp11a, and Cyp17. Genes encoding inhibin-alpha, phosphatidylethanolamine-binding protein (PEBP), and cellular retinoic acid-binding protein 2 (CRABP2) were also downregulated. Most of the aforementioned genes are regulated by steroidogenic factor 1 (SF1) but no consistent change in SF1 mRNA or protein expression was detected. Expression of the aforementioned genes was unaffected at e15.5, but expression of other genes was significantly altered (mostly upregulated). To gain further insight, RNA from interstitial (INT) and seminiferous cord (CORD) tissue obtained by laser capture microdissection (e19.5) was used for transcription profiling. This confirmed most gene expression changes identified for whole testes, but some were remarkably compartment specific. Inhibin-alpha, PEBP, and CRABP2 gene expression were all downregulated in INT but not in CORD, as confirmed by immunohistochemistry; similarly, SCARB1 was downregulated 4.6-fold in INT but only 2.3-fold in CORD. DBP-induced gene expression changes specific to CORD involved small magnitude (less than twofold) reductions or upregulation. These results extend earlier findings and point to the Leydig cells as a primary target of DBP-induced dysfunction. The observed gene expression changes, and their compartmentalization, suggest a possible role for peroxisome proliferator-mediated alteration of cofactor availability as a mechanism underlying DBP-induced Leydig cell dysfunction.


Asunto(s)
Dibutil Ftalato/toxicidad , Feto/metabolismo , Perfilación de la Expresión Génica , Expresión Génica/efectos de los fármacos , Testículo/metabolismo , Animales , Western Blotting , Biología Computacional , Feto/efectos de los fármacos , Feto/ultraestructura , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Rayos Láser , Células Intersticiales del Testículo/efectos de los fármacos , Masculino , Microdisección , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/análisis , ARN/biosíntesis , ARN/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/ultraestructura
7.
Clin Cancer Res ; 8(6): 1857-62, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12060628

RESUMEN

Malignant mesothelioma (MM) is a fatal tumor of increasing incidence, which is resistant to current therapy. Cyclooxygenase-2 (COX-2) plays an important role in solid tumor growth, invasiveness, and angiogenesis, in part through the synthesis of prostaglandins such as prostaglandin E(2) (PGE(2)). In a prospective study, we evaluated COX-2 expression in snap-frozen, surgically resected MM tissue specimens using immunohistochemistry and semiquantitative Western blotting. PGE(2) was assessed by enzyme immunoassay. Thirty epithelioid, 10 biphasic, and 8 sarcomatoid tumors were evaluated. Immunohistochemistry demonstrated strong cytoplasmic tumor cell and variable stromal staining in all of the cases. COX-2 protein levels were correlated with clinicopathological prognostic factors using Kaplan-Meier and Cox proportional hazards models. High COX-2 band densitometry values correlated with poor survival (P = 0.008). In multivariate analysis, high COX-2 expression (P = 0.0005), nonepithelioid cell type (P = 0.002), and chest pain (P = 0.04) were independent predictors of poor prognosis. Furthermore, COX-2 expression contributed in multivariate analysis to both European Organization for Research and Treatment of Cancer (P = 0.001) and Cancer and Leukemia Group B (P = 0.003) prognostic scoring systems. The presence of PGE(2) was demonstrated in all of the samples. These results suggest that COX-2 expression is a prognostic factor in MM. COX-2 is a potential therapeutic target in MM, and trials are required of COX-2 inhibitors alone or in combination with existing treatment modalities.


Asunto(s)
Isoenzimas/metabolismo , Mesotelioma/enzimología , Neoplasias Pleurales/enzimología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Biomarcadores de Tumor/metabolismo , Ciclooxigenasa 2 , Dinoprostona/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Isoenzimas/antagonistas & inhibidores , Masculino , Proteínas de la Membrana , Mesotelioma/patología , Mesotelioma/cirugía , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacología , Neoplasias Pleurales/patología , Neoplasias Pleurales/cirugía , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia
8.
Clin Cancer Res ; 10(20): 6847-54, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15501961

RESUMEN

Curcumin, a polyphenolic antioxidant derived from a dietary spice, exhibits anticancer activity in rodents and in humans. Its efficacy appears to be related to induction of glutathione S-transferase enzymes, inhibition of prostaglandin E(2) (PGE(2)) production, or suppression of oxidative DNA adduct (M(1)G) formation. We designed a dose-escalation study to explore the pharmacology of curcumin in humans. Fifteen patients with advanced colorectal cancer refractory to standard chemotherapies consumed capsules compatible with curcumin doses between 0.45 and 3.6 g daily for up to 4 months. Levels of curcumin and its metabolites in plasma, urine, and feces were analyzed by high-pressure liquid chromatography and mass spectrometry. Three biomarkers of the potential activity of curcumin were translated from preclinical models and measured in patient blood leukocytes: glutathione S-transferase activity, levels of M(1)G, and PGE(2) production induced ex vivo. Dose-limiting toxicity was not observed. Curcumin and its glucuronide and sulfate metabolites were detected in plasma in the 10 nmol/L range and in urine. A daily dose of 3.6 g curcumin engendered 62% and 57% decreases in inducible PGE(2) production in blood samples taken 1 hour after dose on days 1 and 29, respectively, of treatment compared with levels observed immediately predose (P < 0.05). A daily oral dose of 3.6 g of curcumin is advocated for Phase II evaluation in the prevention or treatment of cancers outside the gastrointestinal tract. PGE(2) production in blood and target tissue may indicate biological activity. Levels of curcumin and its metabolites in the urine can be used to assess general compliance.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Curcumina/uso terapéutico , Cooperación del Paciente , Administración Oral , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Curcumina/administración & dosificación , Curcumina/farmacocinética , Curcumina/farmacología , Dinoprostona/sangre , Femenino , Glutatión Transferasa/farmacología , Humanos , Masculino , Persona de Mediana Edad
9.
PLoS One ; 9(5): e97640, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24830643

RESUMEN

In the area of omics profiling in toxicology, i.e. toxicogenomics, characteristic molecular profiles have previously been incorporated into prediction models for early assessment of a carcinogenic potential and mechanism-based classification of compounds. Traditionally, the biomarker signatures used for model construction were derived from individual high-throughput techniques, such as microarrays designed for monitoring global mRNA expression. In this study, we built predictive models by integrating omics data across complementary microarray platforms and introduced new concepts for modeling of pathway alterations and molecular interactions between multiple biological layers. We trained and evaluated diverse machine learning-based models, differing in the incorporated features and learning algorithms on a cross-omics dataset encompassing mRNA, miRNA, and protein expression profiles obtained from rat liver samples treated with a heterogeneous set of substances. Most of these compounds could be unambiguously classified as genotoxic carcinogens, non-genotoxic carcinogens, or non-hepatocarcinogens based on evidence from published studies. Since mixed characteristics were reported for the compounds Cyproterone acetate, Thioacetamide, and Wy-14643, we reclassified these compounds as either genotoxic or non-genotoxic carcinogens based on their molecular profiles. Evaluating our toxicogenomics models in a repeated external cross-validation procedure, we demonstrated that the prediction accuracy of our models could be increased by joining the biomarker signatures across multiple biological layers and by adding complex features derived from cross-platform integration of the omics data. Furthermore, we found that adding these features resulted in a better separation of the compound classes and a more confident reclassification of the three undefined compounds as non-genotoxic carcinogens.


Asunto(s)
Pruebas de Carcinogenicidad , Carcinógenos/química , Neoplasias Hepáticas Experimentales/metabolismo , Toxicogenética/métodos , Algoritmos , Animales , Área Bajo la Curva , Inteligencia Artificial , Análisis por Conglomerados , Biología Computacional , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas Experimentales/inducido químicamente , Masculino , Modelos Estadísticos , Análisis por Matrices de Proteínas , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
10.
Toxicol Sci ; 132(2): 443-57, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23358192

RESUMEN

Previous analysis of in utero dibutylphthalate (DBP)-exposed fetal rat testes indicated that DBP's antiandrogenic effects were mediated, in part, by indirect inhibition of steroidogenic factor 1 (SF1), suggesting that peroxisome proliferator-activated receptor alpha (PPARα) might be involved through coactivator (CREB-binding protein [CBP]) sequestration. To test this hypothesis, we have performed chromatin immunoprecipitation (ChIP) microarray analysis to assess the DNA binding of PPARα, SF1, CBP, and RNA polymerase II in DBP-induced testicular maldevelopment target genes. Pathway analysis of expression array data in fetal rat testes examined at gestational day (GD) 15, 17, or 19 indicated that lipid metabolism genes regulated by SF1 and PPARα, respectively, were overrepresented, and the time dependency of changes to PPARα-regulated lipid metabolism genes correlated with DBP-mediated repression of SF1-regulated steroidogenesis genes. ChIP microarrays were used to investigate whether DBP-mediated repression of SF1-regulated genes was associated with changes in SF1 binding to genes involved in DBP-induced testicular maldevelopment. DBP treatment caused reductions in SF1 binding in CYP11a, StAR, and CYP17a. Follicle-stimulating hormone receptor (FSHR), regulated by SF1 but unaffected by DBP-treatment, also contained SF1-binding peaks, but DBP did not change this compared with control. GD15 and GD19 fetal testes contained PPARα protein-binding peaks in CYP11a, StAR, and CYP17a regulatory regions. In contrast to its repressive effect on SF1, DBP treatment caused increases in these peaks compared with control. PPARα-binding peaks in the FSHR promoter were not detected in GD15 samples. Hence, the repressive effect of DBP on SF1-regulated steroidogenic genes correlates with inhibition of SF1-DNA binding and increased PPARα-DNA binding. The data indicate that PPARα may act as an indirect transrepressor of SF1 on steroidogenic genes in fetal rat testes in response to DBP treatment.


Asunto(s)
Dibutil Ftalato/toxicidad , Testículo/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Inmunoprecipitación de Cromatina , Femenino , Inmunoquímica , Masculino , Ratas , Ratas Wistar , Testículo/metabolismo
11.
PLoS One ; 6(11): e27172, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22087261

RESUMEN

Exposure to phthalates in utero alters fetal rat testis gene expression and testosterone production, but much remains to be done to understand the mechanisms underlying the direct action of phthalate within the fetal testis. We aimed to investigate the direct mechanisms of action of mono-(2-ethylhexyl) phthalate (MEHP) on the rat fetal testis, focusing on Leydig cell steroidogenesis in particular. We used an in vitro system based on the culture for three days, with or without MEHP, of rat fetal testes obtained at 14.5 days post-coitum.Exposure to MEHP led to a dose-dependent decrease in testosterone production. Moreover, the production of 5 alpha-dihydrotestosterone (5α-DHT) (-68%) and androstenedione (-54%) was also inhibited by 10 µM MEHP, whereas 17 alpha-hydroxyprogesterone (17α-OHP) production was found to increase (+41%). Testosterone synthesis was rescued by the addition of androstenedione but not by any of the other precursors used. Thus, the hormone data suggested that steroidogenesis was blocked at the level of the 17,20 lyase activity of the P450c17 enzyme (CYP17), converting 17α-OHP to androstenedione. The subsequent gene expression and protein levels supported this hypothesis. In addition to Cyp17a1, microarray analysis showed that several other genes important for testes development were affected by MEHP. These genes included those encoding insulin-like factor 3 (INSL3), which is involved in controlling testicular descent, and Inha, which encodes the alpha subunit of inhibin B.These findings indicate that under in vitro conditions known to support normal differentiation of the fetal rat testis, the exposure to MEHP directly inhibits several important Leydig cell factors involved in testis function and that the Cyp17a1 gene is a specific target to MEHP explaining the MEHP-induced suppression of steroidogenesis observed.


Asunto(s)
Dietilhexil Ftalato/análogos & derivados , Regulación de la Expresión Génica/efectos de los fármacos , Células Intersticiales del Testículo/efectos de los fármacos , Liasas/metabolismo , Esteroide 17-alfa-Hidroxilasa/metabolismo , Testículo/efectos de los fármacos , 17-alfa-Hidroxiprogesterona/metabolismo , Animales , Células Cultivadas , Dietilhexil Ftalato/farmacología , Dihidrotestosterona/metabolismo , Feto/patología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratas , Esteroides/biosíntesis , Testículo/metabolismo , Testículo/patología , Testosterona/biosíntesis
12.
Toxicol Sci ; 116(2): 452-66, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20403969

RESUMEN

Mouse nongenotoxic hepatocarcinogens phenobarbital (PB) and chlordane induce hepatomegaly characterized by hypertrophy and hyperplasia. Increased cell proliferation is implicated in the mechanism of tumor induction. The relevance of these tumors to human health is unclear. The xenoreceptors, constitutive androstane receptors (CARs), and pregnane X receptor (PXR) play key roles in these processes. Novel "humanized" and knockout models for both receptors were developed to investigate potential species differences in hepatomegaly. The effects of PB (80 mg/kg/4 days) and chlordane (10 mg/kg/4 days) were investigated in double humanized PXR and CAR (huPXR/huCAR), double knockout PXR and CAR (PXRKO/CARKO), and wild-type (WT) C57BL/6J mice. In WT mice, both compounds caused increased liver weight, hepatocellular hypertrophy, and cell proliferation. Both compounds caused alterations to a number of cell cycle genes consistent with induction of cell proliferation in WT mice. However, these gene expression changes did not occur in PXRKO/CARKO or huPXR/huCAR mice. Liver hypertrophy without hyperplasia was demonstrated in the huPXR/huCAR animals in response to both compounds. Induction of the CAR and PXR target genes, Cyp2b10 and Cyp3a11, was observed in both WT and huPXR/huCAR mouse lines following treatment with PB or chlordane. In the PXRKO/CARKO mice, neither liver growth nor induction of Cyp2b10 and Cyp3a11 was seen following PB or chlordane treatment, indicating that these effects are CAR/PXR dependent. These data suggest that the human receptors are able to support the chemically induced hypertrophic responses but not the hyperplastic (cell proliferation) responses. At this time, we cannot be certain that hCAR and hPXR when expressed in the mouse can function exactly as the genes do when they are expressed in human cells. However, all parameters investigated to date suggest that much of their functionality is maintained.


Asunto(s)
Clordano/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Fenobarbital/toxicidad , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Esteroides/fisiología , Animales , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Proliferación Celular/efectos de los fármacos , Receptor de Androstano Constitutivo , Citocromo P-450 CYP3A/biosíntesis , Familia 2 del Citocromo P450 , Humanos , Hiperplasia , Hipertrofia , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas Experimentales/patología , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Receptor X de Pregnano , Especificidad de la Especie , Esteroide Hidroxilasas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA