Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Appl Toxicol ; 42(5): 864-882, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34779009

RESUMEN

We have previously reported that the valproic acid (VPA)-induced disruption pattern of hippocampal adult neurogenesis differs between developmental and 28-day postpubertal exposure. In the present study, we performed brain region-specific global gene expression profiling to compare the profiles of VPA-induced neurotoxicity between developmental and postpubertal exposure. Offspring exposed to VPA at 0, 667, and 2000 parts per million (ppm) via maternal drinking water from gestational day 6 until weaning (postnatal day 21) were examined, along with male rats orally administered VPA at 0, 200, and 900 mg/kg body weight for 28 days starting at 5 weeks old. Four brain regions-the hippocampal dentate gyrus, corpus callosum, cerebral cortex, and cerebellar vermis-were subjected to expression microarray analysis. Profiled data suggested a region-specific pattern of effects after developmental VPA exposure, and a common pattern of effects among brain regions after postpubertal VPA exposure. Developmental VPA exposure typically led to the altered expression of genes related to nervous system development (Msx1, Xcl1, Foxj1, Prdm16, C3, and Kif11) in the hippocampus, and those related to nervous system development (Neurod1) and gliogenesis (Notch1 and Sox9) in the corpus callosum. Postpubertal VPA exposure led to the altered expression of genes related to neuronal differentiation and projection (Cd47, Cyr61, Dbi, Adamts1, and Btg2) in multiple brain regions. These findings suggested that neurotoxic patterns of VPA might be different between developmental and postpubertal exposure, which was consistent with our previous study. Of note, the hippocampal dentate gyrus might be a sensitive target of developmental neurotoxicants after puberty.


Asunto(s)
Síndromes de Neurotoxicidad , Efectos Tardíos de la Exposición Prenatal , Animales , Encéfalo , Hipocampo , Masculino , Neurogénesis , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Maduración Sexual , Transcriptoma , Ácido Valproico/metabolismo , Ácido Valproico/toxicidad
2.
J Appl Toxicol ; 40(11): 1467-1479, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32596862

RESUMEN

We previously found downregulation of low-density lipoprotein receptor class A domain-containing protein 4 (LDLRAD4), a negative regulator of transforming growth factor (TGF)-ß signaling, in glutathione S-transferase placental form (GST-P) expressing (+ ) pre-neoplastic lesions produced by treatment with nongenotoxic hepatocarcinogens for up to 90 days in rats. Here, we investigated the relationship between LDLRAD4 downregulation and TGFß signaling in nongenotoxic hepatocarcinogenesis. The transcripts of Tgfb and Hb-egf increased after ≥28 days of treatment. After 84 or 90 days, Snai1 increased transcripts and the subpopulation of GST-P+ foci downregulating LDLRAD4 co-expressed TGFß1, phosphorylated EGFR, or phosphorylated AKT2, and downregulated PTEN, showing higher incidences than those in GST-P+ foci expressing LDLRAD4. The subpopulation of GST-P+ foci downregulating LDLRAD4 also co-expressed caveolin-1 or TACE/ADAM17, suggesting that disruptive activation of TGFß signaling through a loss of LDLRAD4 enhances EGFR and PTEN/AKT-dependent pathways via caveolin-1-dependent activation of TACE/ADAM17 during nongenotoxic hepatocarcinogenesis. The numbers of c-MYC+ cells and PCNA+ cells were higher in LDLRAD4-downregulated GST-P+ foci than in LDLRAD4-expressing GST-P+ foci, suggesting a preferential proliferation of pre-neoplastic cells by LDLRAD4 downregulation. Nongenotoxic hepatocarcinogens markedly downregulated Nox4 after 28 days and later decreased cleaved caspase 3+ cells in LDLRAD4-downregulated GST-P+ foci, suggesting an attenuation of apoptosis by LDLRAD4 downregulation through activation of the EGFR pathway. At the late hepatocarcinogenesis stage in a two-stage model, LDLRAD4 downregulation was higher in adenoma and carcinoma than in pre-neoplastic cell foci, suggesting a role of LDLRAD4 downregulation in tumor development. Our results suggest that nongenotoxic hepatocarcinogens cause disruptive activation of TGFß signaling through downregulating LDLRAD4 toward carcinogenesis in the rat liver.


Asunto(s)
Apoptosis , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/metabolismo , Lesiones Precancerosas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Tetracloruro de Carbono , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Dietilnitrosamina , Modelos Animales de Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Metapirileno , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Ratas Endogámicas F344 , Transducción de Señal , Tioacetamida , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética
3.
Yakugaku Zasshi ; 140(4): 491-498, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-32238630

RESUMEN

Because the liver is the primary target organ for chemicals and pharmaceuticals, evaluation of these substances' liver toxicity is of critical importance. New evaluation methods without animal testing (i.e., in vitro and/or in silico) are eagerly anticipated, both for animal welfare and for decreasing cost. Also, the importance of mechanistic interpretation of the output derived from non-animal testing has been increasing. Accordingly, we investigated the potential for evaluating liver toxicity by applying the adverse outcome pathway (AOP) concept using gene set enrichment analysis (GSEA) from gene expression (GEx) data. A case study targeting hepatocellular fatty degeneration (HFD) is reported and discussed. We first identified the events detectable in an in vitro system by comparing the GEx data from the rat primary hepatocyte (in vitro) and rat liver (in vivo) treated with a chemical with the ability to induce HFD as one of the phenotypes in a 28-day repeated-dose toxicity test. Then, the scores based on GSEA were calculated after establishing the gene sets for each event leading to HFD. As a result, the mechanistic information leading to HFD was obtained from the score calculated based on the GSEA and the usefulness of the transcriptome-driven evaluation using AOP was demonstrated.


Asunto(s)
Rutas de Resultados Adversos , Hígado/efectos de los fármacos , Hígado/metabolismo , Pruebas de Toxicidad/métodos , Transcriptoma , Tejido Adiposo/metabolismo , Alternativas a las Pruebas en Animales , Bienestar del Animal , Animales , Simulación por Computador , Expresión Génica , Hepatocitos/metabolismo , Humanos , Técnicas In Vitro , Ratas
4.
J Toxicol Pathol ; 32(4): 261-274, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31719753

RESUMEN

To clarify difference in the responses on the reprogramming of metabolism toward carcinogenesis between genotoxic and non-genotoxic hepatocarcinogens in the liver, rats were repeatedly administered genotoxic hepatocarcinogens (N-nitrosodiethylamine, aflatoxin B1, N-nitrosopyrrolidine, or carbadox) or non-genotoxic hepatocarcinogens (carbon tetrachloride, thioacetamide, or methapyrilene hydrochloride) for 28, 84, or 90 days. Non-genotoxic hepatocarcinogens revealed transcript expression changes suggestive of suppressed mitochondrial oxidative phosphorylation (OXPHOS) after 28 days and increased glutathione S-transferase placental form-positive (GST-P+) foci downregulating adenosine triphosphate (ATP) synthase subunit beta, mitochondrial precursor (ATPB), compared with genotoxic hepatocarcinogens after 84 or 90 days, suggesting that non-genotoxic hepatocarcinogens are prone to suppress OXPHOS from the early stage of treatment, which is in contrast to genotoxic hepatocarcinogens. Both genotoxic and non-genotoxic hepatocarcinogens upregulated glycolytic enzyme genes and increased cellular membrane solute carrier family 2, facilitated glucose transporter member 1 (GLUT1) expression in GST-P+ foci for up to 90 days, suggesting induction of a metabolic shift from OXPHOS to glycolysis at early hepatocarcinogenesis by hepatocarcinogens unrelated to genotoxic potential. Non-genotoxic hepatocarcinogens increased c-MYC+ cells after 28 days and downregulated Tp53 after 84 or 90 days, suggesting a commitment to enhanced metabolic shift and cell proliferation. Genotoxic hepatocarcinogens also enhanced c-MYC activation-related metabolic shift until 84 or 90 days. In addition, both genotoxic and non-genotoxic hepatocarcinogens upregulated glutaminolysis-related Slc1a5 or Gls, or both, after 28 days and induced liver cell foci immunoreactive for neutral amino acid transporter B(0) (SLC1A5) in the subpopulation of GST-P+ foci after 84 or 90 days, suggesting glutaminolysis-mediated facilitation of cell proliferation toward hepatocarcinogenesis. These results suggest differential responses between genotoxic and non-genotoxic hepatocarcinogens on reprogramming of energy metabolic pathways toward carcinogenesis in liver cells from the early stage of hepatocarcinogen treatment.

5.
Regul Toxicol Pharmacol ; 107: 104403, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31195068

RESUMEN

In silico toxicology (IST) approaches to rapidly assess chemical hazard, and usage of such methods is increasing in all applications but especially for regulatory submissions, such as for assessing chemicals under REACH as well as the ICH M7 guideline for drug impurities. There are a number of obstacles to performing an IST assessment, including uncertainty in how such an assessment and associated expert review should be performed or what is fit for purpose, as well as a lack of confidence that the results will be accepted by colleagues, collaborators and regulatory authorities. To address this, a project to develop a series of IST protocols for different hazard endpoints has been initiated and this paper describes the genetic toxicity in silico (GIST) protocol. The protocol outlines a hazard assessment framework including key effects/mechanisms and their relationships to endpoints such as gene mutation and clastogenicity. IST models and data are reviewed that support the assessment of these effects/mechanisms along with defined approaches for combining the information and evaluating the confidence in the assessment. This protocol has been developed through a consortium of toxicologists, computational scientists, and regulatory scientists across several industries to support the implementation and acceptance of in silico approaches.


Asunto(s)
Modelos Teóricos , Mutágenos/toxicidad , Proyectos de Investigación , Toxicología/métodos , Animales , Simulación por Computador , Humanos , Pruebas de Mutagenicidad , Medición de Riesgo
6.
Toxicol Lett ; 312: 195-203, 2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31085223

RESUMEN

Developmental exposure to valproic acid (VPA), a model compound for experimental autism, has shown to primarily target GABAergic interneuron subpopulations in hippocampal neurogenesis of rat offspring. The VPA-exposed animals had revealed late effects on granule cell lineages, involving progenitor cell proliferation and synaptic plasticity. To investigate the possibility whether hippocampal neurogenesis in postpubertal rats in a protocol of 28-day repeated exposure is affected in relation with the property of a developmental neurotoxicant by developmental exposure, VPA was orally administered to 5-week-old male rats at 0, 200, 800 and 900 mg/kg body weight/day for 28 days. At 900 mg/kg, GFAP+ cells increased in number, but DCX+ cells decreased in number in the granule cell lineages. Moreover, CHRNB2+ cells and NeuN+ postmitotic neurons decreased in number in the hilus of the dentate gyrus. Transcript level examined at 900 mg/kg in the dentate gyrus was increased with Kit, but decreased with Dpsyl3, Btg2, Pvalb and Chrnb2. These results suggest that VPA increased type-1 stem cells in relation to the activation of SCF-KIT signaling and suppression of BTG2-mediated antiproliferative effect on stem cells. VPA also decreased type-3 progenitor cells and immature granule cells probably in relation with PVALB+ interneuron hypofunction and reduced CHRNB2+ interneuron subpopulation in the hilus, as well as with suppression of BTG2-mediated terminal differentiation of progenitor cells. Thus, the disruption pattern of VPA by postpubertal exposure was different from developmental exposure. However, disruption itself can be detected, suggesting availability of hippocampal neurogenesis in detecting developmental neurotoxicants in a 28-day toxicity study.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Hipocampo/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Apoptosis/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteína Doblecortina , Esquema de Medicación , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Ratas , Maduración Sexual
7.
Toxicol Sci ; 169(1): 122-136, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30690589

RESUMEN

This study examined hypermethylated and downregulated genes specific to carbon tetrachloride (CCl4) by Methyl-Seq analysis combined with expression microarray analysis in the liver of rats treated with CCl4 or N-nitrosodiethylamine (DEN) for 28 days, by excluding those with DEN. Among 52 genes, Ldlrad4, Proc, Cdh17, and Nfia were confirmed to show promoter-region hypermethylation by methylation-specific quantitative PCR analysis on day 28. The transcript levels of these 4 genes decreased by real-time reverse transcription-PCR analysis in the livers of rats treated with nongenotoxic hepatocarcinogens for up to 90 days compared with untreated controls and genotoxic hepatocarcinogens. Immunohistochemically, LDLRAD4 and PROC showed decreased immunoreactivity, forming negative foci, in glutathione S-transferase placental form (GST-P)+ foci, and incidences of LDLRAD4- and PROC- foci in GST-P+ foci induced by treatment with nongenotoxic hepatocarcinogens for 84 or 90 days were increased compared with those with genotoxic hepatocarcinogens. In contrast, CDH17 and NFIA responded to hepatocarcinogens without any relation to the genotoxic potential of carcinogens. All 4 genes did not respond to renal carcinogens after treatment for 28 days. Considering that Ldlrad4 is a negative regulator of transforming growth factor-ß signaling, Proc participating in p21WAF1/CIP1 upregulation by activation, Cdh17 inducing cell cycle arrest by gene knockdown, and Nfia playing a role in a tumor-suppressor, all these genes may be potential in vivo epigenetic markers of nongenotoxic hepatocarcinogens from the early stages of treatment in terms of gene expression changes. LDLRAD4 and PROC may have a role in the development of preneoplastic lesions produced by nongenotoxic hepatocarcinogens.


Asunto(s)
Tetracloruro de Carbono/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Metilación de ADN/efectos de los fármacos , Dietilnitrosamina/toxicidad , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Hígado/efectos de los fármacos , Lesiones Precancerosas/inducido químicamente , Animales , Cadherinas/genética , Cadherinas/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación hacia Abajo , Gutatión-S-Transferasa pi/genética , Gutatión-S-Transferasa pi/metabolismo , Hígado/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteína C/genética , Proteína C/metabolismo , Ratas Endogámicas F344 , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Tiempo
8.
Regul Toxicol Pharmacol ; 96: 1-17, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29678766

RESUMEN

The present publication surveys several applications of in silico (i.e., computational) toxicology approaches across different industries and institutions. It highlights the need to develop standardized protocols when conducting toxicity-related predictions. This contribution articulates the information needed for protocols to support in silico predictions for major toxicological endpoints of concern (e.g., genetic toxicity, carcinogenicity, acute toxicity, reproductive toxicity, developmental toxicity) across several industries and regulatory bodies. Such novel in silico toxicology (IST) protocols, when fully developed and implemented, will ensure in silico toxicological assessments are performed and evaluated in a consistent, reproducible, and well-documented manner across industries and regulatory bodies to support wider uptake and acceptance of the approaches. The development of IST protocols is an initiative developed through a collaboration among an international consortium to reflect the state-of-the-art in in silico toxicology for hazard identification and characterization. A general outline for describing the development of such protocols is included and it is based on in silico predictions and/or available experimental data for a defined series of relevant toxicological effects or mechanisms. The publication presents a novel approach for determining the reliability of in silico predictions alongside experimental data. In addition, we discuss how to determine the level of confidence in the assessment based on the relevance and reliability of the information.


Asunto(s)
Simulación por Computador , Pruebas de Toxicidad/métodos , Toxicología/métodos , Animales , Humanos
9.
Environ Sci Pollut Res Int ; 25(15): 15265-15267, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29079977

RESUMEN

In the original article wrong unites were quoted in Table 3 (page 508) and Table 4 (page 510) as well as in the paragraph 3.2 Core chemical exposure experiments on page 509. Also in paragraph 2.3 Selection and testing of chemicals the link to the Supplemental Materials (ESM) was missing.

10.
Toxicology ; 389: 55-66, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28688903

RESUMEN

This study was performed to compare the exposure effects of N-methyl-N-nitrosourea (MNU), a cytocidal agent of proliferating cells, on rat hippocampal neurogenesis between developmental and postpubertal periods. Developmental exposure through maternal drinking water from gestational day 6 to day 21 after delivery on weaning decreased GFAP-immunoreactive (+) stem cells and increased immunoreactive cells indicative of subsequent progenitor and postmitotic immature neuronal populations, TUNEL+ or p21Cip1/Waf1+ stem/progenitor cells and COX2+ granule cells, on postnatal day (PND) 21. On PND 77 after cessation of developmental exposure, NeuN+ postmitotic granule cells decreased in number. Postpubertal exposure by oral gavage for 28days decreased the numbers of all granule cell lineage populations and ARC+ or COX2+ granule cells and increased the number of TUNEL+ stem/progenitor cells. These results suggested that both developmental and postpubertal exposure caused apoptosis of stem/progenitor cells. However, developmental exposure increased COX2 expression to facilitate intermediate progenitor cell proliferation and increased neuronal plasticity. This effect was concurrent with the induction of p21Cip1/Waf1 that causes cell cycle arrest of stem/progenitor cells in response to accumulating DNA damage on weaning, resulting in a subsequent reduction of postmitotic granule cells. In contrast, postpubertal exposure suppressed neuronal plasticity as evidenced by downregulation of ARC and COX2. The COX2 downregulation was responsible for the lack of facilitating stem/progenitor cell proliferation. Induction of apoptosis and the lack of cell proliferation facilitation may be responsible for the overall reduction of neurogenesis caused by postpubertal exposure. Thus, the disrupted pattern of hippocampal neurogenesis induced by MNU is different between developmental and postpubertal exposure.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Hipocampo/efectos de los fármacos , Exposición Materna/efectos adversos , Metilnitrosourea/toxicidad , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Factores de Edad , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Hipocampo/enzimología , Hipocampo/patología , Masculino , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Plasticidad Neuronal/efectos de los fármacos , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
11.
Neurotox Res ; 31(1): 46-62, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27566479

RESUMEN

Valproic acid (VPA) is used to establish models of experimental autism. The present study investigated the developmental exposure effect of VPA on postnatal hippocampal neurogenesis in accordance with the exposure scheme of OECD Test Guideline 426 adopted for developmental neurotoxicity. Pregnant rats were administered drinking water containing 0, 667, or 2000 ppm VPA from gestational day 6 until day 21 post-delivery. In the subgranular zone (SGZ) and granule cell layer (GCL) of offspring, the number of granule cell lineage subpopulations remained unchanged upon weaning. However, in the hilus of the dentate gyrus, the number of reelin+ interneurons decreased at ≥667 ppm, and the number of PVALB+ or GAD67+ interneurons decreased at 2000 ppm. Conversely, Reln and Gad1 transcript levels increased at 2000 ppm, but Pvalb and Grin2d decreased, in the dentate gyrus. At the adult stage, PCNA+ proliferating SGZ cells, NeuN+ postmitotic SGZ/GCL neurons, and ARC+ or COX2+ GCL neurons increased at ≥667 ppm. In the dentate hilus, decreases in GAD67+ interneuron subpopulations and Grin2d transcript levels sustained at 2000 ppm. These results suggested that VPA primarily targets interneurons by developmental exposure, and this is followed by late effects on granule cell lineages, likely by influencing SGZ cell proliferation and synaptic plasticity. A reduced population of reelin+ or PVALB+ interneurons did not affect distribution of granule cell lineage subpopulations upon weaning. The late effect on neurogenesis, which resulted in increased GCL neurons, might be the result of a sustained decrease in GAD67+ interneurons expressing NR2D encoded by Grin2d.


Asunto(s)
Giro Dentado/efectos de los fármacos , Interneuronas/efectos de los fármacos , Exposición Materna , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Ácido Valproico/toxicidad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Movimiento Celular/efectos de los fármacos , Corteza Cerebelosa/efectos de los fármacos , Corteza Cerebelosa/crecimiento & desarrollo , Corteza Cerebelosa/metabolismo , Corteza Cerebelosa/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Giro Dentado/crecimiento & desarrollo , Giro Dentado/metabolismo , Giro Dentado/patología , Modelos Animales de Enfermedad , Agua Potable , Femenino , Interneuronas/metabolismo , Interneuronas/patología , Masculino , Neurogénesis/fisiología , Embarazo , Distribución Aleatoria , Ratas , Proteína Reelina , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/fisiología
12.
Toxicol Lett ; 261: 59-71, 2016 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-27553673

RESUMEN

6-Propyl-2-thiouracil (PTU)-induced hypothyroidism disrupts neuronal/glial development. This study sought to identify the sensitive immunohistochemical parameters of developmental neurotoxicity (DNT) following PTU-exposure, as well as their responses in a 28-day toxicity study in adults. In the developmental exposure study, pregnant rats were treated with 0, 1, 3, and 10ppm PTU in drinking water from gestational day 6 to postnatal day (PND) 21 and pups were examined on PNDs 21 and 77. In the adult-stage exposure study, 5-week-old male rats were treated with 0, 0.1 and 10mg PTU/kg by oral gavage for 28 days. In the developmental exposure study on PND 21, there were fewer GFAP+, PAX6+, and DCX+ cells in the subgranular zone (SGZ) of the hippocampal dentate gyrus at ≥3 or 10ppm. Regarding synaptic plasticity-related molecules, there were fewer EPHA4+ and ARC+ cells in the dentate granule cell layer. Regarding GABAergic interneuron subpopulations, there were more RELN+, CALB2+, and SST+ cells and fewer PVALB+ cells in the dentate hilus. There were also differences in the numbers of RELN+, PVALB+, CALB2+, and NPY+ cells in the cerebral cortex, and RELN+, PVALB+, and SST+ cells in the cerebellar cortex. Most of these changes were sustained until PND 77. Following adult-stage exposure (10mg/kg), there were fewer SGZ DCX+ cells, but more RELN+ and SST+ cells in the dentate hilus. Results suggest that GABAergic interneuron populations in cortical tissues, hippocampal neurogenesis, and synaptic plasticity are sensitive to PTU-induced DNT during development. In contrast, only hippocampal neurogenesis was sensitive to adult-stage exposure.


Asunto(s)
Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Propiltiouracilo/toxicidad , Animales , Biomarcadores , Proteína Doblecortina , Femenino , Hipotiroidismo , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Proteína Reelina
13.
Toxicol Appl Pharmacol ; 310: 20-31, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27523638

RESUMEN

Both developmental and postpubertal cuprizone (CPZ) exposure impairs hippocampal neurogenesis in rats. We previously found that developmental CPZ exposure alters the expression of genes related to neurogenesis, myelination, and synaptic transmission in specific brain regions of offspring. Here, we examined neuronal and glial toxicity profiles in response to postpubertal CPZ exposure by using expression microarray analysis in the hippocampal dentate gyrus, corpus callosum, cerebral cortex, and cerebellar vermis of 5-week-old male rats exposed to 0, 120, and 600mg/kg CPZ for 28days. Genes showing transcript upregulation were subjected to immunohistochemical analysis. We found transcript expression alterations at 600mg/kg for genes related to synaptic transmission, Ache and Prima1, and cell cycle regulation, Tfap4 and Cdkn1a, in the dentate gyrus, which showed aberrant neurogenesis in the subgranular zone. This dose downregulated myelination-related genes in multiple brain regions, whereas KLOTHO+ oligodendrocyte density was decreased only in the corpus callosum. The corpus callosum showed an increase in transcript levels for inflammatory response-related genes and in the number of CD68+ microglia, MT+ astrocytes, and TUNEL+ apoptotic cells. These results suggest that postpubertal CPZ exposure targets synaptic transmission and cell cycle regulation to affect neurogenesis in the dentate gyrus. CPZ suppressed myelination in multiple brain regions and KLOTHO-mediated oligodendrocyte maturation only in the corpus callosum. The increased number of CD68+ microglia, MT+ astrocytes, and TUNEL+ apoptotic cells in the corpus callosum may be involved in the induction of KLOTHO+ oligodendrocyte death and be a protective mechanism against myelin damage following CPZ exposure.


Asunto(s)
Encéfalo/metabolismo , Cuprizona/farmacología , Perfilación de la Expresión Génica , Vaina de Mielina/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Masculino , Ratas , Ratas Sprague-Dawley
14.
J Toxicol Sci ; 41(3): 383-90, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27193730

RESUMEN

Carcinogenicity of chemicals in our environment is one of the most important health hazards to humans. Recently, a microarray-based short-term prediction system for the hepatocarcinogenicity of chemicals, named CARCINOscreen(®), was developed. Although the system is a promising tool reported to have an ability to predict hepatocarcinogenicity in rats with 92.9% accuracy, it requires specialized equipment and skilled bioinformatics approaches for data analysis. Therefore, we attempted to develop a quantitative PCR (qPCR)-based system as an alternative to microarray-based CARCINOscreen(®). Finally, an optimized gene set consisting of four predictive genes (Abcb1b, Eprs, Map3K8, and Igh-6) was selected from among 3,150 combinations of candidate gene sets. The results of training- and validation-phase trials showed that the qPCR-based alternative to the microarray-based CARCINOscreen(®) could predict the hepatocarcinogenicity of chemicals in rats with 82.8%-86.4% accuracy. One of the predictive genes, Abcb1b, a member of the ATP-binding cassette protein superfamily and multi-drug resistance-associated protein, and the results of this study may indicate a close relation of this gene to the carcinogenicity of chemicals. The prediction performance of the qPCR-based CARCINOscreen(®), as well as its user-friendliness and cost efficiency, suggests that this method is promising for application to primary health hazard assessment. Thus, the qPCR-based CARCINOscreen(®) is considered as a promising tool for predicting the carcinogenicity of chemicals.


Asunto(s)
Biomarcadores de Tumor/genética , Pruebas de Carcinogenicidad/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias Hepáticas/inducido químicamente , Reacción en Cadena de la Polimerasa , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Perfilación de la Expresión Génica/métodos , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Endogámicas F344 , Reproducibilidad de los Resultados , Medición de Riesgo
15.
Toxicol Appl Pharmacol ; 290: 10-20, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26577399

RESUMEN

Developmental cuprizone (CPZ) exposure impairs rat hippocampal neurogenesis. Here, we captured the developmental neurotoxicity profile of CPZ using a region-specific expression microarray analysis in the hippocampal dentate gyrus, corpus callosum, cerebral cortex and cerebellar vermis of rat offspring exposed to 0, 0.1, or 0.4% CPZ in the maternal diet from gestation day 6 to postnatal day (PND) 21. Transcripts of those genes identified as altered were subjected to immunohistochemical analysis on PNDs 21 and 77. Our results showed that transcripts for myelinogenesis-related genes, including Cnp, were selectively downregulated in the cerebral cortex by CPZ at ≥0.1% or 0.4% on PND 21. CPZ at 0.4% decreased immunostaining intensity for 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase) and CNPase(+) and OLIG2(+) oligodendrocyte densities in the cerebral cortex, whereas CNPase immunostaining intensity alone was decreased in the corpus callosum. By contrast, a striking transcript upregulation for Klotho gene and an increased density of Klotho(+) oligodendrocytes were detected in the corpus callosum at ≥0.1%. In the dentate gyrus, CPZ at ≥0.1% or 0.4% decreased the transcript levels for Gria1, Grin2a and Ptgs2, genes related to the synapse and synaptic transmission, and the number of GRIA1(+) and GRIN2A(+) hilar γ-aminobutyric acid (GABA)-ergic interneurons and cyclooxygenase-2(+) granule cells. All changes were reversed at PND 77. Thus, developmental CPZ exposure reversibly decreased mature oligodendrocytes in both cortical and white matter tissues, and Klotho protected white matter oligodendrocyte growth. CPZ also reversibly targeted glutamatergic signals of GABAergic interneuron to affect dentate gyrus neurogenesis and synaptic plasticity in granule cells.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Cuprizona/toxicidad , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Glucuronidasa/genética , Glucuronidasa/metabolismo , Proteínas Klotho , Masculino , Oligodendroglía/citología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica , Sustancia Blanca/citología , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/metabolismo , Ácido gamma-Aminobutírico/metabolismo
16.
J Appl Toxicol ; 36(1): 24-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25825206

RESUMEN

We previously found that developmental hypothyroidism changed the expression of genes in the rat hippocampal dentate gyrus, a brain region where adult neurogenesis is known to occur. In the present study, we performed brain region-specific global gene expression profiling in an adult rat hypothyroidism model to see if it reflected the developmental neurotoxicity we saw in the developmental hypothyroidism model. Starting when male rats were 5 weeks old, we administered 6-propyl-2-thiouracil at a doses of 0, 0.1 and 10 mg kg(-1) body weight by gavage for 28 days. We selected four brain regions to represent both cerebral and cerebellar tissues: hippocampal dentate gyrus, cerebral cortex, corpus callosum and cerebellar vermis. We observed significant alterations in the expression of genes related to neural development (Eph family genes and Robo3) in the cerebral cortex and hippocampal dentate gyrus and in the expression of genes related to myelination (Plp1 and Mbp) in the hippocampal dentate gyrus. We observed only minor changes in the expression of these genes in the corpus callosum and cerebellar vermis. We used real-time reverse-transcription polymerase chain reaction to confirm Chrdl1, Hes5, Mbp, Plp1, Slit1, Robo3 and the Eph family transcript expression changes. The most significant changes in gene expression were found in the dentate gyrus. Considering that the gene expression profile of the adult dentate gyrus closely related to neurogenesis, 28-day toxicity studies looking at gene expression changes in adult hippocampal dentate gyrus may also detect possible developmental neurotoxic effects.


Asunto(s)
Giro Dentado/metabolismo , Perfilación de la Expresión Génica , Hipotiroidismo/fisiopatología , Neurogénesis , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Pruebas de Toxicidad
17.
Toxicol Appl Pharmacol ; 287(3): 210-21, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26057786

RESUMEN

Developmental exposure to cuprizone (CPZ), a demyelinating agent, impairs intermediate-stage neurogenesis in the hippocampal dentate gyrus of rat offspring. To investigate the possibility of alterations in adult neurogenesis following postpubertal exposure to CPZ in a framework of general toxicity studies, CPZ was orally administered to 5-week-old male rats at 0, 120, or 600mg/kg body weight/day for 28days. In the subgranular zone (SGZ), 600mg/kg CPZ increased the number of cleaved caspase-3(+) apoptotic cells. At ≥120mg/kg, the number of SGZ cells immunoreactive for TBR2, doublecortin, or PCNA was decreased, while that for SOX2 was increased. In the granule cell layer, CPZ at ≥120mg/kg decreased the number of postmitotic granule cells immunoreactive for NEUN, CHRNA7, ARC or FOS. In the dentate hilus, CPZ at ≥120mg/kg decreased phosphorylated TRKB(+) interneurons, although the number of reelin(+) interneurons was unchanged. At 600mg/kg, mRNA levels of Bdnf and Chrna7 were decreased, while those of Casp4, Casp12 and Trib3 were increased in the dentate gyrus. These data suggest that CPZ in a scheme of 28-day toxicity study causes endoplasmic reticulum stress-mediated apoptosis of granule cell lineages, resulting in aberrations of intermediate neurogenesis and late-stage neurogenesis and following suppression of immediate early gene-mediated neuronal plasticity. Suppression of BDNF signals to interneurons caused by decreased cholinergic signaling may play a role in these effects of CPZ. The effects of postpubertal CPZ on neurogenesis were similar to those observed with developmental exposure, except for the lack of reelin response, which may contribute to a greater decrease in SGZ cells.


Asunto(s)
Cuprizona/toxicidad , Hipocampo/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Administración Oral , Factores de Edad , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores/metabolismo , Linaje de la Célula , Cuprizona/administración & dosificación , Relación Dosis-Respuesta a Droga , Proteína Doblecortina , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Interneuronas/patología , Masculino , Vaina de Mielina/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Ratas Sprague-Dawley , Proteína Reelina , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
18.
Toxicol Lett ; 234(3): 180-93, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25704630

RESUMEN

The exposure to cuprizone (CPZ) leads to demyelination in the central nervous system in rodents. To examine the developmental effects of CPZ exposure on hippocampal neurogenesis, pregnant rats were treated with 0, 0.1 or 0.4% CPZ in the diet from gestational day 6 to day 21 after delivery. On postnatal day 21, male offspring had a decreased density of new glue2(+) oligodendrocyte progenitor cells in the dentate hilus and in the area of the cerebellar medulla in the presence of 0.4% CPZ. With regard to neurogenesis-related parameters, offspring had decreased T box brain 2(+) progenitor cells and increased apoptotic cells, as detected by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling, which was accompanied by the up-regulation of Casp12 and Bcl2l11 in the subgranular zone, and increased reelin(+) interneurons in the dentate hilus. In addition, the density of phosphorylated TrkB(+) interneurons decreased in the dentate hilus, which was accompanied by transcript down-regulation of Bdnf and Chrna7 in the dentate gyrus. Moreover, granule cells expressing gene products of immediate-early genes, i.e., Arc and Fos, decreased. These results suggest that maternal exposure to 0.4% CPZ decreases proliferative type-2 progenitor cells via endoplasmic reticulum stress-mediated apoptosis and inhibition of cholinergic signals to intermediate-stage progenitor cells following reduced oligodendrocyte production and suppression of the brain-derived neurotrophic factor signaling cascade. Increases in reelin-expressing interneurons may compensate for impaired granule cell migration and/or correct positioning due to decreased immediate-early gene-mediated neuronal plasticity. However, all observed fluctuations disappeared at the adult stage, suggesting that CPZ-induced developmental neurotoxicity was reversible.


Asunto(s)
Quelantes/toxicidad , Neuronas Colinérgicas/efectos de los fármacos , Cuprizona/toxicidad , Hipocampo/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/etiología , Animales , Apoptosis/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Masculino , Embarazo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Reelina
19.
J Appl Toxicol ; 34(12): 1389-99, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24395379

RESUMEN

We previously found that exposure to glycidol at 1000 ppm in drinking water caused axonopathy in maternal rats and aberrations in late-stage hippocampal neurogenesis, targeting the process of neurite extension in offspring. To identify the profile of developmental neurotoxicity of glycidol, pregnant Sprague-Dawley rats were given drinking water containing glycidol from gestational day 6 until weaning on day 21 after delivery, and offspring at 0, 300 and 1000 ppm were subjected to region-specific global gene expression profiling. Four brain regions were selected to represent both cerebral and cerebellar tissues, i.e., the cingulate cortex, corpus callosum, hippocampal dentate gyrus and cerebellar vermis. Downregulated genes in the dentate gyrus were related to axonogenesis (Nfasc), myelination (Mal, Mrf and Ugt8), and cell proliferation (Aurkb and Ndc80) at ≥ 300 ppm, and upregulated genes were related to neural development (Frzb and Fzd6) at 1000 ppm. Upregulation was observed for genes related to myelination (Kl, Igf2 and Igfbp2) in the corpus callosum and axonogenesis and neuritogenesis (Efnb3, Tnc and Cd44) in the cingulate cortex, whereas downregulation was observed for genes related to synaptic transmission (Thbs2 and Ccl2) in the cerebellar vermis; all of these changes were mostly observed at 1000 ppm. Altered gene expression of Cntn3, which functions on neurite outgrowth-promotion, was observed in all four brain regions at 1000 ppm. Gene expression profiles suggest that developmental exposure to glycidol affected plasticity of neuronal networks in the broad brain areas, and dentate gyrus neurogenesis may be the sensitive target of this type of toxicity.


Asunto(s)
Encéfalo/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Compuestos Epoxi/toxicidad , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/genética , Propanoles/toxicidad , Transcriptoma/efectos de los fármacos , Animales , Animales Recién Nacidos , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Toxicol Lett ; 224(3): 424-32, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24185127

RESUMEN

Developmental exposure to glycidol induces aberrations of late-stage neurogenesis in the hippocampal dentate gyrus of rat offspring, whereas maternal animals develop axonopathy. To investigate the possibility whether similar effects on adult neurogenesis could be induced by exposure in a framework of 28-day toxicity study, glycidol was orally administered to 5-week-old male Sprague-Dawley rats by gavage at 0, 30 or 200 mg/kg for 28 days. At 200 mg/kg, animals revealed progressively worsening gait abnormalities as well as histopathological and immunohistochemical changes suggestive of axonal injury as evidenced by generation of neurofilament-L(+) spheroids in the cerebellar granule layer and dorsal funiculus of the medulla oblongata, central chromatolysis in the trigeminal nerve ganglion cells and axonal degeneration in the sciatic nerves. At the same dose, animals revealed aberrations in neurogenesis at late-stage differentiation as evidenced by decreases of both doublecortin(+) and dihydropyrimidinase-like 3(+) cells in the subgranular zone (SGZ) and increased reelin(+) or calbindin-2(+) γ-aminobutyric acid-ergic interneurons and neuron-specific nuclear protein(+) mature neurons in the dentate hilus. These effects were essentially similar to that observed in offspring after maternal exposure to glycidol. These results suggest that glycidol causes aberrations in adult neurogenesis in the SGZ at the late stage involving the process of neurite extension similar to the developmental exposure study in a standard 28-day toxicity study.


Asunto(s)
Axones/patología , Carcinógenos/toxicidad , Diferenciación Celular/efectos de los fármacos , Compuestos Epoxi/toxicidad , Hipocampo/citología , Hipocampo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Propanoles/toxicidad , Animales , Antígenos Nucleares/metabolismo , Apoptosis/efectos de los fármacos , Recuento de Células , Giro Dentado/efectos de los fármacos , Giro Dentado/crecimiento & desarrollo , Proteína Doblecortina , Femenino , Trastornos Neurológicos de la Marcha/inducido químicamente , Trastornos Neurológicos de la Marcha/patología , Hipocampo/crecimiento & desarrollo , Hormonas/sangre , Inmunohistoquímica , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Reelina , Células Madre/efectos de los fármacos , Hormonas Tiroideas/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...