Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 51(8): 1005-1015, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37142426

ABSTRACT

Pharmacokinetic variability in drug plasma exposure between different studies within the same species is not unexpected due to a variety of factors (such as differences in formulation, active pharmaceutical ingredient salt form and solid-state, genetic strain, sex, environmental, disease status, bioanalysis methods, circadian rhythms, etc.) although variability from within the same research group typically does not occur to a great degree because these variables are commonly controlled. Surprisingly, a pharmacology proof of concept study with a previously validated tool compound from the literature failed to show expected response in murine glucose-6-phosphate isomerase-induced arthritis model which was tied to compound plasma exposure unexpectedly 10-fold lower than exposure observed from early pharmacokinetic study confirming adequate exposure prior to proof of concept. A systematic series of studies were conducted to investigate causes for exposure difference between pharmacology and pharmacokinetic studies identifying the presence or absence of soy protein in animal chow as the causative variable. Cyp3a11 expression in intestine and liver was determined to increase in a time dependent manner in mice switched to diets containing soybean meal compared with mice on diets without soybean meal. The repeated pharmacology experiments using the soybean meal free diet achieved plasma exposures that were maintained above the EC50 and showed efficacy and proof of concept for the target. This effect was further confirmed with marker CYP3A4 substrates in follow on mouse studies. The role of soy protein containing diets on CYP expression necessitates the inclusion of controlling rodent diet as a variable for preventing possible exposure differences between studies. SIGNIFICANCE STATEMENT: The presence of soybean meal protein in murine diet increased clearance and decreased oral exposure for select cytochrome 3A4 substrates. Related effects were also observed on select liver enzyme expression.


Subject(s)
Diet , Soybean Proteins , Mice , Animals , Soybean Proteins/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , Intestines
2.
Environ Mol Mutagen ; 63(7): 336-350, 2022 08.
Article in English | MEDLINE | ID: mdl-36176055

ABSTRACT

Hydrazine is a rodent carcinogen and is classified as a probable human carcinogen by IARC. Though hydrazine is positive in both in vitro and in vivo DNA strand break (comet) assays, hydrazine was reported to be negative in an in vitro mutation Muta Mouse lung epithelial cell (FE1) test, as well as in a regulatory-compliant, in vivo Big Blue mouse mutation test. In this article, mechanistic studies explored the cellular response to hydrazine. When tested in a regulatory-compliant mouse lymphoma assay, hydrazine yielded unusual, weakly positive results. This prompted an investigation into the transcriptional response to hydrazine in FE1 cells via RNA sequencing. Amongst the changes identified was a dose-dependent increase in G2/M DNA damage checkpoint activation associated genes. Flow cytometric experiments in FE1 cells revealed that hydrazine exposure led to S-phase cell cycle arrest. Clonogenic assays in a variety of cell lines harboring key DNA repair protein deficiencies indicated that hydrazine could sensitize cells lacking homology dependent repair proteins (Brca2 and Fancg). Lastly, hprt assays with hydrazine were conducted to determine whether a lack of DNA repair could lead to mutagenicity. However, no robust, dose-dependent induction of mutations was noted. The transcriptional and cell cycle response to hydrazine, coupled with functional investigations of DNA repair-deficient cell lines support the inconsistencies noted in the genetic toxicology regulatory battery. In summary, while hydrazine may be genotoxic, transcriptional and functional processes involved in cell cycle regulation and DNA repair appear to play a nuanced role in mediating the mutagenic potential.


Subject(s)
DNA Repair , Transcriptome , Humans , Mice , Animals , Transcriptome/genetics , DNA Repair/genetics , DNA Damage , Mutagens/toxicity , Cell Line , Carcinogens/toxicity , Hydrazines/toxicity
3.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884436

ABSTRACT

Cancer therapies targeting the vascular endothelial growth factor (VEGF) signaling pathway can lead to renal damage by disrupting the glomerular ultrafiltration apparatus. The objective of the current study was to identify sensitive biomarkers for VEGF inhibition-induced glomerular changes in rats. Male Sprague-Dawley rats were administered an experimental VEGF receptor (VEGFR) inhibitor, ABT-123, for seven days to investigate the correlation of several biomarkers with microscopic and ultrastructural changes. Glomeruli obtained by laser capture microdissection were also subjected to gene expression analysis to investigate the underlying molecular events of VEGFR inhibition in glomerulus. ABT-123 induced characteristic glomerular ultrastructural changes in rats, including fusion of podocyte foot processes, the presence of subendothelial electron-dense deposits, and swelling and loss of fenestrations in glomerular endothelium. The subtle morphological changes cannot be detected with light microscopy or by changes in standard clinical chemistry and urinalysis. However, urinary albumin increased 44-fold as early as Day three. Urinary ß2-microglobulin levels were also increased. Other urinary biomarkers that are typically associated with tubular injury were not significantly impacted. Such patterns in urinary biomarkers can provide valuable diagnostic insight to VEGF inhibition therapy-induced glomeruli injuries.


Subject(s)
Kidney Diseases/urine , Protein Kinase Inhibitors/adverse effects , Signal Transduction/drug effects , beta 2-Microglobulin/urine , Albumins/metabolism , Animals , Biomarkers/urine , Disease Models, Animal , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Laser Capture Microdissection , Male , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
4.
Pharmacol Ther ; 200: 110-125, 2019 08.
Article in English | MEDLINE | ID: mdl-31028836

ABSTRACT

Antibody-drug conjugates (ADCs) are a promising therapeutic modality for oncology indications. The concept of an ADC platform is to increase the therapeutic index (TI) of chemotherapeutics through more selective delivery of cytotoxic agents to tumor cells while limiting exposure to healthy normal cells. Despite the use of antibodies targeting antigens abundantly and/or exclusively expressed on cancer cells (i.e., target cells), dose limiting toxicities (DLTs) in normal cells/tissues are frequently reported even at suboptimal therapeutic doses. Although advancement of ADC technology has helped to optimize all three key components (i.e., mAb, linker, and payload), DLTs remain a key challenge for ADC development. Mechanisms of ADC toxicity in normal cells/tissues are not clearly understood, but the majority of DLTs are considered to be target-independent. In addition to linker-drug instability contributing to the premature release of cytotoxic drug (payload) in circulation, uptake/trafficking of intact ADCs by both receptor-dependent (FcγRs, FcRn and C-type lectin receptors), and-independent (non-specific endocytosis) mechanisms may contribute to off-target toxicity in normal cells. In this article, we review potential mechanisms of target-independent ADC uptake and toxicity in normal cells, as well as discuss components of ADCs which may influence these mechanisms. This information will provide a deeper understanding of the underlying mechanisms of ADC off-target toxicity and prove helpful toward improving the overall TI of the next generation of ADCs.


Subject(s)
Immunoconjugates/adverse effects , Immunoconjugates/pharmacokinetics , Animals , Biological Transport , Humans
5.
Front Genet ; 9: 636, 2018.
Article in English | MEDLINE | ID: mdl-30723492

ABSTRACT

Gene expression profiling is a useful tool to predict and interrogate mechanisms of toxicity. RNA-Seq technology has emerged as an attractive alternative to traditional microarray platforms for conducting transcriptional profiling. The objective of this work was to compare both transcriptomic platforms to determine whether RNA-Seq offered significant advantages over microarrays for toxicogenomic studies. RNA samples from the livers of rats treated for 5 days with five tool hepatotoxicants (α-naphthylisothiocyanate/ANIT, carbon tetrachloride/CCl4, methylenedianiline/MDA, acetaminophen/APAP, and diclofenac/DCLF) were analyzed with both gene expression platforms (RNA-Seq and microarray). Data were compared to determine any potential added scientific (i.e., better biological or toxicological insight) value offered by RNA-Seq compared to microarrays. RNA-Seq identified more differentially expressed protein-coding genes and provided a wider quantitative range of expression level changes when compared to microarrays. Both platforms identified a larger number of differentially expressed genes (DEGs) in livers of rats treated with ANIT, MDA, and CCl4 compared to APAP and DCLF, in agreement with the severity of histopathological findings. Approximately 78% of DEGs identified with microarrays overlapped with RNA-Seq data, with a Spearman's correlation of 0.7 to 0.83. Consistent with the mechanisms of toxicity of ANIT, APAP, MDA and CCl4, both platforms identified dysregulation of liver relevant pathways such as Nrf2, cholesterol biosynthesis, eiF2, hepatic cholestasis, glutathione and LPS/IL-1 mediated RXR inhibition. RNA-Seq data showed additional DEGs that not only significantly enriched these pathways, but also suggested modulation of additional liver relevant pathways. In addition, RNA-Seq enabled the identification of non-coding DEGs that offer a potential for improved mechanistic clarity. Overall, these results indicate that RNA-Seq is an acceptable alternative platform to microarrays for rat toxicogenomic studies with several advantages. Because of its wider dynamic range as well as its ability to identify a larger number of DEGs, RNA-Seq may generate more insight into mechanisms of toxicity. However, more extensive reference data will be necessary to fully leverage these additional RNA-Seq data, especially for non-coding sequences.

6.
Front Genet ; 3: 213, 2012.
Article in English | MEDLINE | ID: mdl-23112805

ABSTRACT

Xenobiotic-mediated induction of cytochrome P450 (CYP) drug metabolizing enzymes (DMEs) is frequently encountered in drug discovery and can influence disposition, pharmacokinetic, and toxicity profiles. The CYP1A subfamily of DMEs plays a central role in the biotransformation of several drugs and environmental chemicals. Autoinduction of drugs through CYP3A enzymes is a common mechanism for their enhanced clearance. However, autoinduction via CYP1A is encountered less frequently. In this report, an experimental compound, A-998679 [3-(5-pyridin-3-yl-1,2,4-oxadiazol-3-yl) benzonitrile], was shown to enhance its own clearance via induction of Cyp1a1 and Cyp1a2. Rats were dosed for 5 days with 30, 100, and 200 mg/kg/day A-998679. During the dosing period, the compound's plasma AUC decreased at 30 mg/kg (95%) and 100 mg/kg (80%). Gene expression analysis and immunohistochemistry of the livers showed a large increase in the mRNA and protein levels of Cyp1a, which was involved in the biotransformation of A-998679. Induction of CYP1A was confirmed in primary rat, human, and dog hepatocytes. The compound also weakly inhibited CYP1A2 in human liver microsomes. A-998679 activated the aryl hydrocarbon receptor (AhR) in a luciferase gene reporter assay in HepG2 cells, upregulated expression of genes associated with AhR activation in rat liver and enhanced nuclear migration of AhR in HepG2 cells. Collectively these results demonstrate that A-998679 is an AhR activator that induces Cyp1a1 and Cyp1a2 expression, resulting in an autoinduction phenomenon. The unique properties of A-998679, along with its novel structure distinct from classical polycyclic aromatic hydrocarbons (PAHs), may warrant its further evaluation as a tool compound for use in studies involving AhR biology and CYP1A-related mechanisms of drug metabolism and toxicity.

7.
Toxicol Lett ; 208(1): 82-91, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22037397

ABSTRACT

N-vinylpyrrolidone dimer (VPD) is a novel experimental formulation excipient intended for preclinical toxicology studies. In a previous 4-week toxicity study, VPD induced dose-dependent hepatocellular and thyroid gland hypertrophy in Sprague-Dawley (SD) rats. The objectives of the current investigation were to define the underlying molecular mechanisms of these changes. Two separate studies were conducted using male SD rats, daily doses of 300, 1000 or 3,000 mg/kg of VPD, and a positive control (phenobarbital at 75 mg/kg/day): (1) a 28-day study to monitor thyroid hormone levels after 7 and 28 days of dosing; (2) a 5-day study to evaluate hepatic and thyroid gland transcriptomic changes, as well as hepatic UGT activity levels. At VPD dosages of 300 mg/kg/day and higher, 2-fold increases of serum thyroid stimulating hormone (TSH) levels were observed in male SD rats after 28 days of dosing, while serum thyroxine (T4) and triiodothyronine (T3) levels were unchanged. Liver UGT enzyme activity levels were increased in VPD-treated rats after 5 days. In addition, in the 5-day study, VPD caused increased hepatic mRNA levels of a panel of drug metabolizing enzymes (DMEs) and transporters, including Cyp3a1, Cyp2b1, Ugt 2b1, and Abcc3. Similar patterns of induction were observed in primary rat hepatocytes exposed to VPD. Transcriptomic changes in the thyroid gland were identified for genes involved in thyroid hormone biosynthesis and in the FAK, PTEN, and Wnt/ß-catenin signaling pathways. Collectively, these data indicate that VPD acts as an inducer of hepatic DMEs in SD rats and that this likely leads to enhanced peripheral metabolism of T3/T4, resulting in a feedback response characterized by increased serum TSH levels, and thyroid gland hypertrophy and hyperplasia.


Subject(s)
Excipients/toxicity , Hepatomegaly/chemically induced , Microsomes, Liver/enzymology , Pyrrolidinones/toxicity , Thyroid Diseases/chemically induced , Animals , Chemistry, Pharmaceutical , Constitutive Androstane Receptor , Dose-Response Relationship, Drug , Gene Expression Profiling , Hepatocytes/enzymology , Hepatocytes/pathology , Hepatomegaly/metabolism , Hypertrophy/chemically induced , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Microsomes, Liver/drug effects , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Thyroid Diseases/metabolism , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyroid Hormones/blood , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Transcriptome/drug effects
8.
Arch Toxicol ; 85(5): 513-23, 2011 May.
Article in English | MEDLINE | ID: mdl-21046364

ABSTRACT

Dibromoacetic acid (DBAA), a by-product formed during disinfection of drinking water, alters spermatogenesis in rats through defective spermiation. The mechanism underlying this toxicity is not fully understood. In this study, gene expression data generated with microarrays from testes were used to generate a mechanistic understanding of DBAA-induced testicular toxicity. Testes were collected from male Sprague-Dawley rats dosed orally for 1 and 4 days with DBAA at 250 mg/kg/day. At both time points, DBAA administration induced delayed spermiation in Stage X tubules and regulated the expression of a small number of genes, including a mild but consistent downregulation of cytochrome P450c17α (CYP17) mRNA, an enzyme expressed by Leydig cells and essential for the production of testicular androgens. Downregulation of CYP17 was confirmed at the protein level and its biological significance was substantiated by demonstrating reduced testicular testosterone levels in DBAA-dosed rats. Furthermore, testosterone production by human chorionic gonadotrophin (hCG)-stimulated rat primary Leydig cells was reduced following treatment with 100 µM DBAA. Collectively, these results indicate that DBAA can directly target rat Leydig cells and downregulate testicular CYP17 expression with a resulting decreased testicular testosterone production. This disruption of testicular steroidogenesis is likely to contribute to the mechanism of failed spermiation observed in rats following exposure to DBAA.


Subject(s)
Acetates/toxicity , Steroid 17-alpha-Hydroxylase/metabolism , Testicular Diseases/pathology , Testis/pathology , Animals , Chorionic Gonadotropin/metabolism , Down-Regulation , Gene Expression Profiling , Humans , Leydig Cells/metabolism , Male , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spermatogenesis/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Testicular Diseases/chemically induced , Testosterone/biosynthesis
9.
Arch Toxicol ; 84(4): 263-70, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20213465

ABSTRACT

Patients treated with highly active antiretroviral therapy may develop metabolic side effects such as hyperlipidemia, insulin resistance, lipoatrophy and lactic acidosis. The pathophysiology of these metabolic abnormalities is unknown, although some, e.g., lactic acidosis and lipoatrophy, are more associated with nucleoside use while protease inhibitors (PIs) have been shown to contribute to hyperlipidemia and insulin resistance. Identifying new PIs that are not associated with dyslipidemia has been hindered by the lack of mechanistic information and the unavailability of relevant animal models. In order to understand the molecular mechanism behind the hyperlipidemia associated with other protease inhibitors, and to develop a more effective, faster screen for compounds with this liability, we have analyzed expression profiles from PI-treated animals. Previously, we have shown that treatment of rats with ritonavir results in increases in the expression of proteasomal subunit genes in the liver. We show this increase is similar in rats treated with bortezomib, a proteasome inhibitor. In addition, we have treated rats with additional protease inhibitors, including atazanavir, which is associated with lower rates of lipid elevations in the clinic when administered in the absence of ritonavir. Our results indicate a strong correlation between proteasomal induction and lipid elevations, and have allowed us to develop a rapid screen for identifying novel PIs that do not induce the proteasome.


Subject(s)
Gene Expression Regulation, Enzymologic , HIV Protease Inhibitors/toxicity , Hyperlipidemias/chemically induced , Proteasome Endopeptidase Complex/genetics , Animals , Atazanavir Sulfate , Carbamates/toxicity , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Furans , Gene Expression Profiling , Male , Oligonucleotide Array Sequence Analysis , Oligopeptides/toxicity , Proteasome Endopeptidase Complex/metabolism , Pyridines/toxicity , Rats , Rats, Sprague-Dawley , Ritonavir/toxicity , Sulfonamides/toxicity
10.
Toxicol Lett ; 176(2): 138-48, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18164877

ABSTRACT

Preclinical pharmacokinetic (PK) evaluations are typically conducted in rats before in vivo toxicologic evaluations. It is unclear how the serial bleeding procedures in PK studies affect tissue homeostasis or sensitivity to toxicity. In this study, our objective was to evaluate the impact of serial bleeding on the transcriptome of various major tissues (kidney, heart, liver, spleen) and their response to two well-characterized molecules, doxorubicin and cisplatin. Rats received single i.v. injections of saline, doxorubicin (8 mg/kg) or cisplatin (4 mg/kg). In each group, half of the rats were serially bled by tail vein. Serial bleeding was associated with slight decreases of red blood cell parameters, but did not result in histopathological changes or in increased sensitivity to doxorubicin and cisplatin toxicity based on clinical pathology and histopathology evaluation. In addition, serial bleeding did not induce significant gene expression changes in either vehicle- or compound-treated rats when compared to their respective control groups. Overall, these results suggest that the serial bleeding procedure used in PK studies in our institution minimally affects the tissue response to toxicants, and support the use of these studies to generate early toxicology data in drug discovery.


Subject(s)
Cisplatin/toxicity , Doxorubicin/toxicity , Gene Expression Profiling/methods , Phlebotomy/methods , Animals , Blood Proteins/analysis , Cisplatin/administration & dosage , Doxorubicin/administration & dosage , Erythrocyte Count , Gene Expression/drug effects , Heart/drug effects , Hematocrit , Hemoglobins/analysis , Injections, Intravenous , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Leukocyte Count , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Myocardium/metabolism , Myocardium/pathology , Pathology, Clinical/methods , Rats , Rats, Sprague-Dawley , Regression Analysis , Spleen/drug effects , Spleen/metabolism , Spleen/pathology
11.
Pharmacogenomics ; 7(2): 177-86, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16515397

ABSTRACT

In vitro toxicogenomics represents a useful approach for evaluating the toxic properties of new drug candidates early in the drug discovery process using minimal amounts of compounds. The aim of this study was to develop in vitro-based gene expression assays for two prototypical toxicological classes: aryl hydrocarbon receptor (AhR) agonists and peroxisome proliferator activated receptor alpha (PPAR alpha) agonists. Primary rat hepatocytes were exposed to a number of class-specific compounds, including 3-methylcholanthrene, aroclor, and beta-napthoflavone as AhR agonists, bezafibrate, clofibrate, and Wy-14643 as peroxisome proliferators, and chlorpheniramine, penicillin and spectinomycin as negative controls. Global gene expression profiles were generated with microarrays for each class of compounds. Using linear discriminant analysis coupled with permutation-based t-test, gene signatures were established to classify compounds according to a discriminant score. The final gene signatures consist of eight genes for AhR agonism and 11 genes for PPAR alpha agonism, and were further validated using additional compounds. The assay was initially developed using a microarray platform. The authors then evaluated whether it could be transferred to a more cost-effective platform with higher throughput. The results indicate that a small set of genes can be used to quantitatively assess the degree to which a compound falls into a certain mechanistic toxicological class. While this study only focused on two classes, it could be expanded to encompass other toxicological mechanistic classes as well. Furthermore, by adapting this type of assay to a higher throughput platform, in vitro toxicogenomics can represent an effective approach to generate robust toxicological data early in the drug discovery process.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Toxicogenetics/methods , Animals , Data Interpretation, Statistical , Gene Expression , Hepatocytes/chemistry , Hepatocytes/metabolism , Oligonucleotide Array Sequence Analysis , PPAR alpha/biosynthesis , PPAR alpha/genetics , Peroxisome Proliferators/agonists , RNA/isolation & purification , Rats , Receptors, Aryl Hydrocarbon/antagonists & inhibitors
12.
Nucleic Acids Res ; 33(22): e187, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16377776

ABSTRACT

The comparability and reliability of data generated using microarray technology would be enhanced by use of a common set of standards that allow accuracy, reproducibility and dynamic range assessments on multiple formats. We designed and tested a complex biological reagent for performance measurements on three commercial oligonucleotide array formats that differ in probe design and signal measurement methodology. The reagent is a set of two mixtures with different proportions of RNA for each of four rat tissues (brain, liver, kidney and testes). The design provides four known ratio measurements of >200 reference probes, which were chosen for their tissue-selectivity, dynamic range coverage and alignment to the same exemplar transcript sequence across all three platforms. The data generated from testing three biological replicates of the reagent at eight laboratories on three array formats provides a benchmark set for both laboratory and data processing performance assessments. Close agreement with target ratios adjusted for sample complexity was achieved on all platforms and low variance was observed among platforms, replicates and sites. The mixed tissue design produces a reagent with known gene expression changes within a complex sample and can serve as a paradigm for performance standards for microarrays that target other species.


Subject(s)
Gene Expression Profiling/standards , Oligonucleotide Array Sequence Analysis/standards , RNA, Messenger/standards , Animals , Gene Expression Profiling/methods , Male , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotide Probes , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results , Tissue Distribution
13.
Toxicol Pathol ; 32 Suppl 2: 19-25, 2004.
Article in English | MEDLINE | ID: mdl-15503661

ABSTRACT

Hepatotoxicity characterized by microvesicular steatosis (MVS) is characterized by an abnormal accumulation of numerous small cytoplasmic lipid droplets in hepatocytes. Fulminant or progressive cases of microvesicular steatosis may lead to liver failure and death. Experimentally, short-chain carboxylic acids are known to induce microvesicular steatosis. The identification of gene changes that correlate with MVS concomitant with biochemical and histological indices could provide a better understanding of how this toxicity occurs as well as biomarkers that could be used to avoid this toxicity in the future. Sprague-Dawley rats were dosed days with cyclopropane carboxylic acid (CPCA) a short-chain fatty acid that can induce microvesicular steatosis, and with butyrate, a short chain fatty acid that served as a negative control. CPCA initiated microvesicular steatosis while butyrate did not. In addition, CPCA inhibited beta-oxidation in a concentration-dependent manner in vitro and caused a reduction in mitochondrial respiration ex vivo; no inhibition was evident with butyrate. Microarray results showed that gene expression changes with CPCA resulted in regulation of genes involved in beta-oxidation, as well as other genes associated with mitochondrial function. Overall, these results support altered hepatic mitochondrial function as a mechanism of the toxicity induced by a short-chain fatty acid and may provide potential biomarkers for this toxicity.


Subject(s)
Cyclopropanes/administration & dosage , Fatty Acids/administration & dosage , Fatty Liver/chemically induced , Gene Expression/drug effects , Mitochondria, Liver/drug effects , Animals , Cells, Cultured , Cyclopropanes/metabolism , Cyclopropanes/pharmacology , Cyclopropanes/toxicity , Dose-Response Relationship, Drug , Fatty Acids/chemistry , Fatty Acids/metabolism , Fatty Acids/pharmacology , Fatty Acids/toxicity , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Profiling , Hepatocytes/drug effects , Hepatocytes/metabolism , Mitochondria, Liver/metabolism , Molecular Structure , Molecular Weight , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Time Factors
14.
Toxicol In Vitro ; 17(5-6): 693-701, 2003.
Article in English | MEDLINE | ID: mdl-14599465

ABSTRACT

In vitro human hepatocyte cultures are a key tool in the investigation of xenobiotic toxicity and metabolism. In most in vitro hepatocyte studies, the cells are allowed to adhere to an extracellular matrix, such as collagen. Unfortunately, the ability of freshly isolated hepatocytes to adhere to collagen varies from donor to donor. We used microarray analysis to determine what gene expression differences exist between hepatocytes in suspension and hepatocytes attached to collagen. Results from different donors showed a considerable difference in gene expression patterns between the two hepatocyte populations. In addition, we also compared the gene expression profiles of hepatocytes in culture with liver tissue. The results showed that both hepatocytes in suspension and hepatocytes attached to collagen display significant gene expression differences compared with liver tissue. Finally, we show that both populations of hepatocytes are responsive to dexamethasone and regulate some of the same genes. Overall, our results suggest that either significant gene expression changes occur in isolated hepatocytes or that suspended and attached cells represent different populations of hepatocytes found in intact livers.


Subject(s)
Cell Adhesion/physiology , Gene Expression Profiling , Gene Expression , Hepatocytes/metabolism , Adult , Aged , Albumins/genetics , Albumins/metabolism , Branched DNA Signal Amplification Assay/methods , Cells, Cultured , Collagen/metabolism , Dexamethasone/pharmacology , Female , Hepatocytes/cytology , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Mol Cell Endocrinol ; 203(1-2): 155-68, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12782412

ABSTRACT

Protein tyrosine phosphatases are important regulators of insulin signal transduction. Our studies have shown that in insulin resistant and diabetic ob/ob and db/db mice, reducing the levels of protein tyrosine phosphatase 1B (PTP1B) protein by treatment with a PTP1B antisense oligonucleotide resulted in improved insulin sensitivity and normalized plasma glucose levels. The mechanism by which PTP1B inhibition improves insulin sensitivity is not fully understood. We have used microarray analysis to compare gene expression changes in adipose tissue, liver and muscle of PTP1B antisense-treated ob/ob mice. Our results show that treatment with PTP1B antisense resulted in the downregulation of genes involved in lipogenesis in both fat and liver, and a downregulation of genes involved in adipocyte differentiation in fat, suggesting that PTP1B antisense acts through a different mechanism than thiazolidinedione (TZD) treatment. In summary, microarray results suggest that reduction of PTP1B may alleviate hyperglycemia and enhance insulin sensitivity by a different mechanism than TZD treatment.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Lipids/biosynthesis , Liver/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Tyrosine Phosphatases/physiology , Adipose Tissue/cytology , Animals , Blood Glucose/drug effects , Cell Differentiation/drug effects , Down-Regulation/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Insulin Resistance , Mice , Mice, Obese , Muscles/metabolism , Oligonucleotides, Antisense/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/antagonists & inhibitors
16.
EHP Toxicogenomics ; 111(1T): 53-60, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12735110

ABSTRACT

Toxicogenomics is an emerging field combining genomics and bioinformatics to identify and characterize mechanisms of toxicity of compounds. One of the main tools used in toxicogenomics is DNA microarrays. We have used a novel strategy to create a library highly enriched for genes expressed in the liver under hepatotoxic conditions. Using this library, we have created a new oligonucleotide microarray dedicated to the study of rat liver function. Oligonucleotide probes for these genes were designed and used in experimental hybridization studies to deduce the correct sequence orientation and to determine those sequences exhibiting differential regulation under a variety of toxicity-related treatments and conditions. The final array was benchmarked on treatments with 3-methylcholanthrene, Aroclor 1254, and cyclopropane carboxylic acid. Our results showed that the subtractive hybridization greatly enriched for genes regulated during a hepatotoxic response. Overall, our strategy for design of a new rat toxicology microarray can be applied to other systems as well and should aid greatly in the development of microarrays targeted for specific scientific areas.


Subject(s)
/toxicity , Cyclopropanes/toxicity , Liver/drug effects , Methylcholanthrene/toxicity , Oligonucleotide Array Sequence Analysis/methods , Pharmacogenetics , Animals , Liver/metabolism , Male , Nucleic Acid Hybridization , Oligonucleotide Probes , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
17.
Toxicology ; 181-182: 537-50, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12505364

ABSTRACT

Microarray analysis of gene expression has become a powerful approach for exploring the biological effects of drugs and other chemicals. In toxicology research, gene expression profiling may help identify hazards by comparing results for an experimental compound with a database, and establish mechanistic hypotheses through examination of discrete gene changes. Here we examine the hepatic effects of a thienopyridine inhibitor of NF-kappa B-mediated expression of cellular adhesion proteins. In a 3-day toxicity study in Sprague-Dawley rats, A-277249 induced hypertrophy of the liver and elevated serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP). To investigate mechanism, microarray analysis was done on RNA from livers of A-277249-treated rats. Gene expression profiles from A-277249 were compared with a database of profiles from fifteen known hepatotoxins. Agglomerative hierarchical cluster analysis showed A-277249 to have a profile most similar to the aromatic hydrocarbons Aroclor 1254 and 3-methylcholanthrene (3MC), two known activators of the aryl hydrocarbon nuclear receptor (AhR). Several genes regulated by the AhR, including cytochrome P450 1A1, were upregulated by A-277249. In addition, several genes involved in apoptosis and cell cycle were differentially expressed consistent with cell turnover, hypertrophy and hyperplasia observed by histology. Results from this study indicate that A-277249 hepatic toxicity is mediated by the AhR either directly or through effects on NF-kappa B, and demonstrate the utility of microarray analysis for the rapid identification of toxic hazards for new chemical entities.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/biosynthesis , Oligonucleotide Array Sequence Analysis/methods , Receptors, Aryl Hydrocarbon/physiology , Signal Transduction/drug effects , Toxicology/instrumentation , Animals , Blotting, Western , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Gene Expression/drug effects , Liver/pathology , Liver Function Tests , NF-kappa B/genetics , NF-kappa B/metabolism , Proteins/metabolism , Pyridines/toxicity , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...