Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 642, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802535

ABSTRACT

Alterations in the experience-dependent and autonomous elaboration of neural circuits are assumed to underlie autism spectrum disorder (ASD), though it is unclear what synaptic traits are responsible. Here, utilizing a valproic acid-induced ASD marmoset model, which shares common molecular features with idiopathic ASD, we investigate changes in the structural dynamics of tuft dendrites of upper-layer pyramidal neurons and adjacent axons in the dorsomedial prefrontal cortex through two-photon microscopy. In model marmosets, dendritic spine turnover is upregulated, and spines are generated in clusters and survived more often than in control marmosets. Presynaptic boutons in local axons, but not in commissural long-range axons, demonstrate hyperdynamic turnover in model marmosets, suggesting alterations in projection-specific plasticity. Intriguingly, nasal oxytocin administration attenuates clustered spine emergence in model marmosets. Enhanced clustered spine generation, possibly unique to certain presynaptic partners, may be associated with ASD and be a potential therapeutic target.


Subject(s)
Callithrix , Disease Models, Animal , Neuronal Plasticity , Oxytocin , Animals , Oxytocin/metabolism , Male , Synapses/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Dendritic Spines/drug effects , Autism Spectrum Disorder/metabolism , Autistic Disorder/metabolism , Autistic Disorder/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Prefrontal Cortex/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Valproic Acid/pharmacology , Presynaptic Terminals/metabolism , Female , Axons/metabolism
2.
Nat Commun ; 12(1): 5388, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526497

ABSTRACT

Autism spectrum disorder (ASD) is a multifactorial disorder with characteristic synaptic and gene expression changes. Early intervention during childhood is thought to benefit prognosis. Here, we examined the changes in cortical synaptogenesis, synaptic function, and gene expression from birth to the juvenile stage in a marmoset model of ASD induced by valproic acid (VPA) treatment. Early postnatally, synaptogenesis was reduced in this model, while juvenile-age VPA-treated marmosets showed increased synaptogenesis, similar to observations in human tissue. During infancy, synaptic plasticity transiently increased and was associated with altered vocalization. Synaptogenesis-related genes were downregulated early postnatally. At three months of age, the differentially expressed genes were associated with circuit remodeling, similar to the expression changes observed in humans. In summary, we provide a functional and molecular characterization of a non-human primate model of ASD, highlighting its similarity to features observed in human ASD.


Subject(s)
Autism Spectrum Disorder/physiopathology , Disease Models, Animal , Evoked Potentials/physiology , Neurons/physiology , Prefrontal Cortex/physiology , Synaptic Transmission/physiology , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/genetics , Callithrix , Dendritic Spines/physiology , Electric Stimulation , Gene Expression Profiling/methods , Humans , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurons/metabolism , Oligonucleotide Array Sequence Analysis/methods , Patch-Clamp Techniques/methods , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Valproic Acid
3.
Neuroimage ; 195: 243-251, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30953832

ABSTRACT

In autism spectrum disorder (ASD), disrupted functional and structural connectivity in the social brain has been suggested as the core biological mechanism underlying the social recognition deficits of this neurodevelopmental disorder. In this study, we aimed to identify genetic and neurostructural abnormalities at birth in a non-human primate model of ASD, the common marmoset with maternal exposure to valproic acid (VPA), which has been reported to display social recognition deficit in adulthood. Using a comprehensive gene expression analysis, we found that 20 genes were significantly downregulated in VPA-exposed neonates. Of these, Frizzled3 (FZD3) and PIK3CA were identified in an axon guidance signaling pathway. FZD3 is essential for the normal development of the anterior commissure (AC) and corpus callosum (CC); hence, we performed diffusion tensor magnetic resonance imaging with a 7-Tesla scanner to measure the midsagittal sizes of these structures. We found that the AC size in VPA-exposed neonates was significantly smaller than that in age-matched controls, while the CC size did not differ. These results suggest that downregulation of the genes related to axon guidance and decreased AC size in neonatal primates may be linked to social brain dysfunctions that can happen later in life.


Subject(s)
Anterior Commissure, Brain/pathology , Autism Spectrum Disorder/pathology , Axon Guidance/physiology , Animals , Animals, Newborn , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/metabolism , Axon Guidance/drug effects , Callithrix , Class I Phosphatidylinositol 3-Kinases/biosynthesis , Disease Models, Animal , Frizzled Receptors/biosynthesis , GABA Agents/toxicity , Transcriptome/drug effects , Valproic Acid/toxicity
4.
J Appl Toxicol ; 39(7): 1030-1042, 2019 07.
Article in English | MEDLINE | ID: mdl-30883854

ABSTRACT

Tadpoles during metamorphosis are sensitive to chemical exposure as shown in the amphibian metamorphosis assay, which is a method to detect effects of chemicals on the functions of hypothalamus-pituitary-thyroid axis. The present study reports existence of different modes of action between pyriproxyfen (PYR) and 6-propyl-2-thiouracil (PTU) under different feeding conditions based on gene expression profiles (transcriptomics) in the thyroid glands of tadpoles of the African clawed frog, Xenopus laevis. PTU and PYR were exposed to the tadpoles during metamorphosis under normal (fed groups, both of PTU and PRY) and restricted feeding (fasted groups, PTU only) conditions; and effects were compared to control groups. Delayed development based on decreased Nieuwkoop and Faber developmental stage number without any histopathological changes was observed in the control of restricted feeding (control-fasted) group, and the PYR group with reduced food consumption. Clear developmental retardation with typical thyroid histopathological changes was observed in the PTU groups. To find clusters of all samples based on their similarity of expression patterns, hierarchical clustering analysis using selected gene probes was conducted. It revealed gene profiles from samples of the PYR group were quite similar to those of the control-fasted group, followed by the control group with normal feeding (control-fed). The results suggest that key events in the thyroid glands of tadpoles induced by PYR should be quite similar to those of control-fasted, and quite different from those of the PTU groups. Our findings demonstrated the usefulness of transcriptomics, which enabled recognition of the different modes of actions.


Subject(s)
Food Deprivation , Larva/drug effects , Metamorphosis, Biological/drug effects , Thyroid Gland/drug effects , Transcriptome/drug effects , Animal Feed , Animals , Biological Assay , Endocrine Disruptors/pharmacology , Gene Expression Profiling , Larva/genetics , Larva/growth & development , Metamorphosis, Biological/genetics , Propylthiouracil/pharmacology , Pyridines/pharmacology , Thyroid Gland/growth & development , Thyroid Gland/metabolism , Xenopus laevis
5.
Toxicol Sci ; 168(2): 572-596, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30629241

ABSTRACT

The nongenotoxic pyrethroid insecticide permethrin produced hepatocellular tumors in CD-1 mice but not in Wistar rats. Recently, based on findings of a Pathology Working Group involving an expert panel of pathologists, it was concluded that permethrin increased liver tumors at 2500 and 5000 ppm in female mice, but no treatment-related tumorigenic response occurred in male mice at dose levels examined in the 2-year bioassay. To evaluate a possible mode of action (MOA) for the permethrin female CD-1 mouse hepatocellular tumors, a number of investigative studies were conducted. In time-course studies in female CD-1 mice, permethrin increased relative liver weight and enhanced hepatocyte proliferation within 1 week. Treatment with permethrin resulted in marked increases in CYP4A enzyme activities and mRNA levels, but only slightly increased CYP2B markers, suggesting that permethrin primarily activates the peroxisome proliferator-activated receptor alpha (PPARα) and to a much lesser extent the constitutive androstane receptor. The effects of permethrin on relative liver weight, hepatocyte proliferation and CYP4A enzyme activities and mRNA levels were dose-dependent and were reversible within 5 weeks after cessation of treatment. The hepatic effects of permethrin observed in wild-type female mice were markedly reduced in PPARα knockout female mice. These results demonstrate that the MOA for hepatocellular tumor formation by permethrin in female mice involves activation of PPARα resulting in a mitogenic effect. The MOA for permethrin-induced mouse liver tumor formation due to PPARα activation is considered to be not plausible for humans. This conclusion is strongly supported by available epidemiological data for permethrin.


Subject(s)
Carcinogens/toxicity , Hepatocytes/drug effects , Liver Neoplasms/chemically induced , Liver/drug effects , PPAR alpha/metabolism , Permethrin/toxicity , Animals , Cell Proliferation/drug effects , Cytochrome P-450 CYP4A/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Hepatocytes/pathology , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred ICR , Mice, Knockout , PPAR alpha/genetics
6.
Stem Cells ; 37(1): 89-101, 2019 01.
Article in English | MEDLINE | ID: mdl-30270488

ABSTRACT

Granulocyte colony stimulating factor (G-CSF) has been reported to ameliorate impaired liver function in patients with advanced liver diseases through mobilization and proliferation of hepatic progenitor cells (HPCs). However, the underlying mechanisms remain unknown. We previously showed that G-CSF treatment increased the number of bone marrow (BM)-derived cells migrating to the fibrotic liver following repeated carbon tetrachloride (CCl4 ) injections into mice. In this study, we identified opioid growth factor receptor-like 1 (OGFRL1) as a novel BM cell-derived accelerator of fibrotic liver regeneration in response to G-CSF treatment. Endogenous Ogfrl1 was highly expressed in the hematopoietic organs such as the BM and spleen, whereas the liver contained a relatively small amount of Ogfrl1 mRNA. Among the peripheral blood cells, monocytes were the major sources of OGFRL1. Endogenous Ogfrl1 expression in both the peripheral blood monocytes and the liver was decreased following repeated CCl4 injections. An intrasplenic injection of cells overexpressing OGFRL1 into CCl4 -treated fibrotic mice increased the number of HPC and stimulated proliferation of hepatic parenchymal cells after partial resection of the fibrotic liver. Furthermore, overexpression of OGFRL1 in cultured HPC accelerated their differentiation as estimated by increased expression of liver-specific genes such as hepatocyte nuclear factor 4α, cytochrome P450, and fatty acid binding protein 1, although it did not affect the colony forming ability of HPC. These results indicate a critical role of OGFRL1 in the mobilization and differentiation of HPC in the fibrotic liver, and administration of OGFRL1-expressing cells may serve as a potential regenerative therapy for advanced liver fibrosis. Stem Cells 2019;37:89-101.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Liver Cirrhosis/genetics , Liver Cirrhosis/therapy , Liver Regeneration/genetics , Regenerative Medicine/methods , Stem Cells/metabolism , Animals , Cell Differentiation , Humans , Male , Mice , Transfection
7.
J Appl Toxicol ; 38(12): 1529-1537, 2018 12.
Article in English | MEDLINE | ID: mdl-30047161

ABSTRACT

Transcriptomics technologies have been used for risk assessment of chemicals, mainly to predict the modes of action (MOAs) of chemicals or identify biomarkers. Transcriptomics data may also be helpful to understand MOAs of chemicals at the molecular level in more detail. As an example of the known MOAs, there are two MOAs of thyroid toxicity: inhibition of thyroid hormone synthesis ("direct" effect) and hypermetabolism of thyroid hormone by enzyme induction in liver ("indirect" effect). In the present study, global profiles of gene expression were analyzed in rats treated with chemicals acting directly on the thyroid (thyroid peroxidase inhibitors such as propylthiouracil and methimazole) and chemicals acting indirectly on the thyroid (hepatic enzyme inducers such as phenobarbital and pregnenolone-16α-carbonitrile) using microarrays. Using a subtraction method between these two types of chemicals, we identified characteristic gene expression changes on the thyroid hormone synthesis pathway by direct-acting chemicals. Based on the functions of these genes, alterations of their expression seem to indicate the results of thyroid peroxidase inhibition, and might be helpful in more accurate evaluation of MOAs for thyroid toxicity.


Subject(s)
Antithyroid Agents/toxicity , Liver/drug effects , Thyroid Gland/drug effects , Thyroid Hormones/biosynthesis , Transcriptome/drug effects , Animals , Enzyme Induction/drug effects , Gene Expression Profiling , Iodide Peroxidase/antagonists & inhibitors , Liver/enzymology , Male , Methimazole/toxicity , Microarray Analysis , Phenobarbital/toxicity , Propylthiouracil/toxicity , Rats, Wistar , Thyroid Gland/metabolism
8.
Regul Toxicol Pharmacol ; 91 Suppl 1: S3-S13, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28958911

ABSTRACT

Prevailing knowledge gaps in linking specific molecular changes to apical outcomes and methodological uncertainties in the generation, storage, processing, and interpretation of 'omics data limit the application of 'omics technologies in regulatory toxicology. Against this background, the European Centre for Ecotoxicology and Toxicology of Chemicals (ECETOC) convened a workshop Applying 'omics technologies in chemicals risk assessment that is reported herein. Ahead of the workshop, multi-expert teams drafted frameworks on best practices for (i) a Good-Laboratory Practice-like context for collecting, storing and curating 'omics data; (ii) the processing of 'omics data; and (iii) weight-of-evidence approaches for integrating 'omics data. The workshop participants confirmed the relevance of these Frameworks to facilitate the regulatory applicability and use of 'omics data, and the workshop discussions provided input for their further elaboration. Additionally, the key objective (iv) to establish approaches to connect 'omics perturbations to phenotypic alterations was addressed. Generally, it was considered promising to strive to link gene expression changes and pathway perturbations to the phenotype by mapping them to specific adverse outcome pathways. While further work is necessary before gene expression changes can be used to establish safe levels of substance exposure, the ECETOC workshop provided important incentives towards achieving this goal.


Subject(s)
Congresses as Topic , Ecotoxicology/methods , Education/methods , Genomics/methods , Metabolomics/methods , Research Report , Animals , Congresses as Topic/trends , Ecotoxicology/trends , Education/trends , Europe , Genomics/trends , Humans , Metabolomics/trends , Proteomics/methods , Proteomics/trends , Research Report/trends , Risk Assessment , Spain
9.
J Proteome Res ; 16(10): 3634-3645, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28825834

ABSTRACT

Transcriptomic, proteomic, phosphoproteomic, and metabolomic analyses were combined to determine the role of pregnane X receptor (PXR) in nongenotoxic signaling and energy homeostasis in liver after rats were repeatedly orally dosed with the PXR agonist pregnenolone carbonitrile (PCN) for 7 days. Analyses of mRNAs and proteins in the supernatant, membrane, and cytosolic fractions of enlarged liver homogenates showed diverse expression profiles. Gene set enrichment analysis showed that the synchronous increase in mRNAs and proteins involved in chemical carcinogenesis and the response to drug was possibly mediated by the PXR pathway and proteasome core complex assembly was possibly mediated by the Nrf2 pathway. In addition, levels of proteins in the endoplasmic reticulum lumen and involved in the acute-phase response showed specific increase with no change in mRNA level, and those composed of the mitochondrial inner membrane showed specific decrease. The analysis of phosphorylated peptides of poly(A) RNA binding proteins showed a decrease in phosphorylation, possibly by casein kinase 2, which may be related to the regulation of protein expression. Proteins involved in insulin signaling pathways showed an increase in phosphorylation, possibly by protein kinase A, and those involved in apoptosis showed a decrease. Metabolomic analysis suggested the activation of the pentose phosphate and anaerobic glycolysis pathways and the increase of amino acid and fatty acid levels, as occurs in the Warburg effect. In conclusion, the results of combined analyses suggest that PXR's effects are due to transcriptional and post-transcriptional regulation with alteration of nongenotoxic signaling pathways and energy homeostasis.


Subject(s)
Genomics , Proteomics , Receptors, Steroid/metabolism , Transcriptome/genetics , Animals , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Homeostasis/genetics , Humans , Liver/metabolism , Phosphorylation , Pregnane X Receptor , Pregnenolone Carbonitrile/administration & dosage , Rats , Receptors, Steroid/agonists , Receptors, Steroid/genetics , Signal Transduction/drug effects
10.
Toxicol Sci ; 158(2): 412-430, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28520973

ABSTRACT

High dietary levels of momfluorothrin, a nongenotoxic synthetic pyrethroid, induced hepatocellular tumors in male and female Wistar rats in a 2-year bioassay. The mode of action (MOA) for rat hepatocellular tumors was postulated to occur via activation of the constitutive androstane receptor (CAR), as momfluorothrin is a close structural analogue of the pyrethroid metofluthrin, which is known to produce rat liver tumors through a CAR-mediated MOA. To elucidate the MOA for rat hepatocellular tumor formation by momfluorothrin, this study was conducted to examine effects on key and associative events of the CAR-mediated MOA for phenobarbital based on the International Programme on Chemical Safety framework. A 2-week in vivo study in Wistar rats revealed that momfluorothrin induced CYP2B activities, increased liver weights, produced hepatocyte hypertrophy and increased hepatocyte replicative DNA synthesis. These effects correlated with the dose-response relationship for liver tumor formation and also showed reversibility upon cessation of treatment. Moreover, momfluorothrin did not increase CYP2B1/2 mRNA expression and hepatocyte replicative DNA synthesis in CAR knockout rats. Using cultured Wistar rat hepatocytes and the RNA interference technique, knockdown of CAR resulted in a suppression of induction of CYP2B1/2 mRNA levels by momfluorothrin. Alternative MOAs for liver tumor formation were excluded. A global gene expression profile analysis of the liver of male Wistar rats treated with momfluorothrin for 2 weeks also showed similarity to the prototypic CAR activator phenobarbital. Overall, these data strongly support that the postulated MOA for momfluorothrin-induced rat hepatocellular tumors as being mediated by CAR activation.


Subject(s)
Liver Neoplasms, Experimental/chemically induced , Pyrethrins/toxicity , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , DNA Replication/drug effects , Dose-Response Relationship, Drug , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Liver/pathology , Liver Neoplasms, Experimental/metabolism , Male , Microsomes, Liver/enzymology , Mitogens/pharmacology , Mitosis/drug effects , Organ Size/drug effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/genetics
11.
Toxicol Sci ; 157(2): 465-486, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28431163

ABSTRACT

Permethrin increased the incidence of bronchiolo-alveolar adenomas in female mice but not male mice or female or male rats. Studies were conducted to determine whether permethrin has mitogenic activity in Club cells in mouse lung as the basis for the mode of action (MOA) for the lung adenoma induction. Several short-term experiments focusing on time-course, dose-response, reversibility, sex difference, strain difference, and species difference were evaluated for Club cell proliferation and morphology. The findings demonstrated that permethrin slightly and continuously enhanced Club cell proliferation at tumor-associated dose levels in female mice, but did not increase proliferation in male mice or in female rats. Electron microscopic examination demonstrated that permethrin produced morphological alterations in Club cells prior to increasing the Club cell proliferation. There was no evidence of increased cell death. These alterations in Club cells were also observed with a close structural analog cypermethrin. Taken together, the present studies provide evidence that the MOA for induction of mouse lung adenomas by permethrin involves slight morphological effects on Club cells, sustained Club cell proliferation, and eventually hyperplasia and bronchiolo-alveolar adenoma in susceptible mice. The potential human carcinogenic hazard of permethrin based on the tumorigenic MOA for lung tumors in mice was evaluated using the International Programme on Chemical Safety Human Relevance Framework. As humans are quantitatively much less sensitive to agents that increase Club cell proliferation and tumor formation in mice, it is not likely permethrin will lead to an increase in susceptibility to lung tumor development in humans. Epidemiological data for permethrin strongly supports this conclusion.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Insecticides/toxicity , Lung Neoplasms/chemically induced , Permethrin/toxicity , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Rats, Wistar , Sex Factors , Species Specificity , Time Factors , Transcriptome/drug effects
12.
Toxicol Res (Camb) ; 6(6): 795-813, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-30090543

ABSTRACT

Phenobarbital (PB) is a nongenotoxic hepatocellular carcinogen in rodents. PB induces hepatocellular tumors by activating the constitutive androstane receptor (CAR). Some previous research has suggested the possible involvement of epigenetic regulation in PB-promoted hepatocellular tumorigenesis, but the details of its molecular mechanism are not fully understood. In the present study, comprehensive analyses of DNA methylation, hydroxymethylation and gene expression using microarrays were performed in mouse hepatocellular adenomas induced by a single 90 mg kg-1 intraperitoneal injection dose of diethylnitrosamine (DEN) followed by 500 ppm PB in the diet for 27 weeks. DNA modification and expression of hundreds of genes are coordinately altered in PB-induced mouse hepatocellular adenomas. Of these, gene network analysis showed alterations of CAR signaling and tumor development-related genes. Pathway enrichment analysis revealed that differentially methylated or hydroxymethylated genes belong mainly to pathways involved in development, immune response and cancer cells in contrast to differentially expressed genes belonging primarily to the cell cycle. Furthermore, overlap was evaluated between the genes with altered expression levels with 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) alterations in mouse hepatocellular adenoma induced by DEN/PB and the genes with altered expression levels in the liver of CD-1 mice or humanized chimeric mice treated with PB for 7 days. With the integration of transcriptomic and epigenetic approaches, we detected candidate genes responsible for early key events of PB-promoted mouse hepatocellular tumorigenesis. Interestingly, these genes did not overlap with genes altered by the PB treatment of humanized chimeric mice, thus suggesting a species difference between the effects of PB in mouse and human hepatocytes.

13.
J Toxicol Pathol ; 29(4): 229-236, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27821907

ABSTRACT

Although 3,3'-iminodipropionitrile (IDPN) is widely used as a neurotoxicant to cause axonopathy due to accumulation of neurofilaments in several rodent models, its mechanism of neurotoxicity has not been fully understood. In particular, no information regarding microRNA (miRNA) alteration associated with IDPN is available. This study was conducted to reveal miRNA alteration related to IDPN-induced neurotoxicity. Rats were administered IDPN (20, 50, or 125 mg/kg/day) orally for 3, 7, and 14 days. Histopathological features were investigated using immunohistochemistry for neurofilaments and glial cells, and miRNA alterations were analyzed by microarray and reverse transcription polymerase chain reaction. Nervous symptoms such as ataxic gait and head bobbing were observed from Day 9 at 125 mg/kg. Axonal swelling due to accumulation of neurofilaments was observed especially in the pons, medulla, and spinal cord on Day 7 at 125 mg/kg and on Day 14 at 50 and 125 mg/kg. Furthermore, significant upregulation of miR-547* was observed in the pons and medulla in treated animals only on Day 14 at 125 mg/kg. This is the first report indicating that miR-547* is associated with IDPN-induced neurotoxicity, especially in an advanced stage of axonopathy.

14.
J Appl Toxicol ; 36(2): 296-308, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26198598

ABSTRACT

Carcinogenicity studies using animals are expensive and time consuming. Therefore, the development of a highly accurate carcinogenicity prediction system to interpret short-term test results would be beneficial. The Ames test is popular for mutagens; however, it cannot detect non-genotoxic carcinogens. Previously, we reported a prediction system using gene expression data obtained from a short-term (28-day) study that screened candidate compounds for testing in long-term carcinogenicity studies. In this study, our system was improved by adding more gene expression data. To establish our new system, we used the data of 93 test compounds (41 hepatocarcinogens and 52 non-hepatocarcinogens). Analysis of liver gene expression data by dividing compounds into 'for training' and 'for test' categories (20 cases assigned randomly) using Support Vector Machine (SVM) identified a set of marker probe sets that could be used to predict hepatocarcinogenicity. The assigned 42 probe sets have included the cancer- or c-Myc-related genes such as Hsp90, Pink1, Hspc111, Fbx29, Hepsin, Syndecan2 and Synbindin. Compared with the older version, the improved system had a higher concordance rate with the training data and a good performance with the external test data.


Subject(s)
Carcinogenesis/genetics , Carcinogenicity Tests/methods , Carcinogens/toxicity , DNA Damage/genetics , Gene Expression Profiling/methods , Liver Neoplasms, Experimental/genetics , Toxicogenetics/methods , Animals , Carcinogenesis/drug effects , DNA Damage/drug effects , Male , Rats
16.
Toxicol Appl Pharmacol ; 286(3): 188-97, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25933445

ABSTRACT

To determine miRNAs and their predicted target proteins regulatory networks which are potentially involved in onset of pulmonary fibrosis in the bleomycin rat model, we conducted integrative miRNA microarray and iTRAQ-coupled LC-MS/MS proteomic analyses, and evaluated the significance of altered biological functions and pathways. We observed that alterations of miRNAs and proteins are associated with the early phase of bleomycin-induced pulmonary fibrosis, and identified potential target pairs by using ingenuity pathway analysis. Using the data set of these alterations, it was demonstrated that those miRNAs, in association with their predicted target proteins, are potentially involved in canonical pathways reflective of initial epithelial injury and fibrogenic processes, and biofunctions related to induction of cellular development, movement, growth, and proliferation. Prediction of activated functions suggested that lung cells acquire proliferative, migratory, and invasive capabilities, and resistance to cell death especially in the very early phase of bleomycin-induced pulmonary fibrosis. The present study will provide new insights for understanding the molecular pathogenesis of idiopathic pulmonary fibrosis.


Subject(s)
Bleomycin/toxicity , Disease Models, Animal , MicroRNAs/biosynthesis , Proteomics/methods , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Animals , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/physiology , Male , MicroRNAs/genetics , Pulmonary Fibrosis/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
17.
Toxicol Pathol ; 43(2): 198-208, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24777749

ABSTRACT

Circulating microRNAs (miRNAs) show promise as biomarkers due to their tissue-specific expression and high stability. This study was conducted to investigate whether nervous system-enriched miR-9* and hippocampus-enriched miR-384-5p could be indicators of neurotoxicity in serum. Rats were given a single administration of trimethyltin (TMT) chloride at 6, 9, or 12 mg/kg by gavage, and brain and serum were collected 1, 4, and 7 days after administration. MiR-9* and miR-384-5p levels in serum and hippocampus were analyzed by reverse transcriptase polymerase chain reaction (RT-PCR), and their neurotoxicity detection sensitivities were compared with nervous symptoms, auditory response, and histopathology. TMT caused tremor, hypersensitivity, and decreased auditory response at 12 mg/kg on day 1 and at 9 mg/kg on day 4. Histopathologically, neural cell death and glial reaction were observed in brain (mainly hippocampus) at 12 mg/kg on day 1, 4, and 7 and at 6 and 9 mg/kg on day 4 and 7. MiR-9* and miR-384-5p levels were elevated in serum at 9 and 12 mg/kg on days 4 and 7 (at 9 mg/kg on day 7, miR-9* only) but were not changed in hippocampus. These miRNAs were considered to be elevated with the evolution of neural cell death and were thus considered possible novel indicators of neurotoxicity.


Subject(s)
MicroRNAs/blood , Neurotoxicity Syndromes/blood , Trimethyltin Compounds/toxicity , Animals , Apoptosis/drug effects , Body Weight/drug effects , Brain/drug effects , Brain/pathology , Hearing/drug effects , Hippocampus/metabolism , MicroRNAs/metabolism , Motor Activity/drug effects , Neuroglia/pathology , Neurons/pathology , Neurotoxicity Syndromes/psychology , Organ Size/drug effects , Rats
18.
Toxicol Sci ; 142(1): 137-57, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25145657

ABSTRACT

High doses of sodium phenobarbital (NaPB), a constitutive androstane receptor (CAR) activator, have been shown to produce hepatocellular tumors in rodents by a mitogenic mode of action (MOA) involving CAR activation. The effect of 1-week dietary treatment with NaPB on liver weight and histopathology, hepatic CYP2B enzyme activity and CYP2B/3A mRNA expression, replicative DNA synthesis and selected genes related to cell proliferation, and functional transcriptomic and metabolomic analyses was studied in male CD-1 mice, Wistar Hannover (WH) rats, and chimeric mice with human hepatocytes. The treatment of chimeric mice with 1000-1500-ppm NaPB resulted in plasma levels around 3-5-fold higher than those observed in human subjects given therapeutic doses of NaPB. NaPB produced dose-dependent increases in hepatic CYP2B activity and CYP2B/3A mRNA levels in all animal models. Integrated functional metabolomic and transcriptomic analyses demonstrated that the responses to NaPB in the human liver were clearly different from those in rodents. Although NaPB produced a dose-dependent increase in hepatocyte replicative DNA synthesis in CD-1 mice and WH rats, no increase in replicative DNA synthesis was observed in human hepatocyte-originated areas of chimeric mice. In addition, treatment with NaPB had no effect on Ki-67, PCNA, GADD45ß, and MDM2 mRNA expression in chimeric mice, whereas significant increases were observed in CD-1 mice and/or WH rats. However, increases in hepatocyte replicative DNA synthesis were observed in chimeric mice both in vivo and in vitro after treatment epidermal growth factor. Thus, although NaPB could activate CAR in both rodent and human hepatocytes, NaPB did not increase replicative DNA synthesis in human hepatocytes of chimeric mice, whereas it was mitogenic to rat and mouse hepatocytes. As human hepatocytes are refractory to the mitogenic effects of NaPB, the MOA for NaPB-induced rodent liver tumor formation is thus not relevant for humans.


Subject(s)
Chimera , Cytochrome P-450 CYP2B6/metabolism , Hepatocytes/drug effects , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Phenobarbital/toxicity , Animals , Cells, Cultured , Constitutive Androstane Receptor , DNA Replication/drug effects , Gene Expression Profiling , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Liver/enzymology , Liver/pathology , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , Male , Mice, Inbred Strains , Organ Size/drug effects , Phenobarbital/pharmacokinetics , Rats, Inbred Strains , Receptors, Cytoplasmic and Nuclear/metabolism , Species Specificity , Transcriptome/drug effects
19.
J Biol Chem ; 289(37): 25833-43, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25074924

ABSTRACT

Human embryonic stem cells (hESCs) are pluripotent stem cells from early embryos, and their self-renewal capacity depends on the sustained expression of hESC-specific molecules and the suppressed expression of differentiation-associated genes. To discover novel molecules expressed on hESCs, we generated a panel of monoclonal antibodies against undifferentiated hESCs and evaluated their ability to mark cancer cells, as well as hESCs. MAb7 recognized undifferentiated hESCs and showed a diffuse band with molecular mass of >239 kDa in the lysates of hESCs. Although some amniotic epithelial cells expressed MAb7 antigen, its expression was barely detected in normal human keratinocytes, fibroblasts, or endothelial cells. The expression of MAb7 antigen was observed only in pancreatic and gastric cancer cells, and its levels were elevated in metastatic and poorly differentiated cancer cell lines. Analyses of MAb7 antigen suggested that the clustered NeuAcα2-3Galß O-linked oligosaccharides on DMBT1 (deleted in malignant brain tumors 1) were critical for MAb7 binding in cancer cells. Although features of MAb7 epitope were similar with those of TRA-1-60, distribution of MAb7 antigen in cancer cells was different from that of TRA-1-60 antigen. Exposure of a histone deacetylase inhibitor to differentiated gastric cancer MKN74 cells evoked the expression of MAb7 antigen, whereas DMBT1 expression remained unchanged. Cell sorting followed by DNA microarray analyses identified the down-regulated genes responsible for the biosynthesis of MAb7 antigen in MKN74 cells. In addition, treatment of metastatic pancreatic cancer cells with MAb7 significantly abrogated the adhesion to endothelial cells. These results raised the possibility that MAb7 epitope is a novel marker for undifferentiated cells such as hESCs and cancer stem-like cells and plays a possible role in the undifferentiated cells.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/immunology , Oligosaccharides/immunology , Receptors, Cell Surface/genetics , Antibodies, Monoclonal/genetics , Calcium-Binding Proteins , Cell Differentiation/immunology , DNA-Binding Proteins , Embryonic Stem Cells/cytology , Endothelial Cells/metabolism , Epitopes/immunology , Fibroblasts/metabolism , Flow Cytometry , Humans , Keratinocytes/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Oligosaccharides/genetics , Receptors, Cell Surface/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Tumor Suppressor Proteins
20.
J Toxicol Sci ; 39(3): 383-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24849673

ABSTRACT

The widely used antiepileptic drug valproic acid (VPA) is known to exhibit teratogenicity in the form of a failure of the neural tube in humans. Embryonic stem cells (ESCs) are reported to be a promising cell source for evaluating chemical teratogenicity, because they are capable of reproducing embryonic developmental model and enable reduction in the number of experimental animals used. We previously investigated 22 genes for which expressions are altered by teratogens, specifically focusing on neural differentiation of mouse ESCs. In the present study, expressions of the investigated genes were evaluated by quantitative real-time PCR and compared during differentiation of human ESCs into neurons with or without VPA. Under the conditions, almost all gene expressions significantly changed in VPA-containing culture. Specifically, in neural development-related genes such as DCX, ARX, MAP2, and NNAT, more than 2-fold expression was observed. The findings suggest that the genes focused on in this study may help to elucidate the teratogenic effects of VPA and might be a useful tool to analyze embryotoxic potential of chemicals in humans.


Subject(s)
Anticonvulsants/toxicity , Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/drug effects , Gene Expression/drug effects , Neurons/cytology , Valproic Acid/toxicity , Animals , Cell Differentiation/drug effects , Cells, Cultured , Doublecortin Domain Proteins , Doublecortin Protein , Embryonic Stem Cells/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Real-Time Polymerase Chain Reaction , Teratogenesis/drug effects , Teratogenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL