53 results on '"Tryndyak VP"'
Search Results
2. A preclinical model of severe NASH-like liver injury by chronic administration of a high-fat and high-sucrose diet in mice.
- Author
-
Willett RA, Tryndyak VP, Hughes Hanks JM, Elkins L, Nagumalli SK, Avigan MI, Ross SA, da Costa GG, Beland FA, Rusyn I, and Pogribny IP
- Subjects
- Animals, Male, Female, Mice, Disease Progression, Dietary Sucrose adverse effects, Lipid Metabolism drug effects, Liver Cirrhosis pathology, Liver Cirrhosis chemically induced, Non-alcoholic Fatty Liver Disease pathology, Diet, High-Fat adverse effects, Disease Models, Animal, Liver pathology, Liver metabolism, Liver drug effects
- Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive liver disease, affecting 38% of adults globally. If left untreated, NAFLD may progress to more advanced forms of the disease, including non-alcoholic steatohepatitis (NASH), liver cirrhosis, and fibrosis. Early NAFLD detection is critical to prevent disease progression. Using an obesogenic high-fat and high-sucrose (HF/HS) diet, we characterized the progression of NAFLD in male and female Collaborative Cross CC042 mice after 20-, 40-, and 60-week intervals of chronic HF/HS diet feeding. The incidence and severity of liver steatosis, inflammation, and fibrosis increased in both sexes over time, with male mice progressing to a NASH-like disease state faster than female mice, as indicated by earlier and more pronounced changes in liver steatosis. Histopathological indication of macrovesicular steatosis and gene expression changes of key lipid metabolism genes were found to be elevated in both sexes after 20 weeks of HF/HS diet. Measurement of circulating markers of inflammation (CXCL10 and TNF-α), histopathological analysis of immune cell infiltrates, and gene expression changes in inflammation-related genes indicated significant liver inflammation after 40 and 60 weeks of HF/HS diet exposure in both sexes. Liver fibrosis, as assessed by Picosirius red and Masson's trichrome staining and changes in expression of key fibrosis related genes indicated significant changes after 40 and 60 weeks of HF/HS diet exposure. In conclusion, we present a preclinical animal model of dietary NAFLD progression, which recapitulates human pathophysiological and pathomorphological changes, that could be used to better understand the progression of NAFLD and support development of new therapeutics., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
3. Cellular and molecular alterations in a human hepatocellular in vitro model of nonalcoholic fatty liver disease development and stratification.
- Author
-
Willett RA, Tryndyak VP, Beland FA, and Pogribny IP
- Subjects
- Humans, Liver Cirrhosis genetics, Liver Cirrhosis metabolism, Liver Cirrhosis pathology, Fatty Acids, Nonesterified metabolism, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease metabolism, Non-alcoholic Fatty Liver Disease pathology, Carcinoma, Hepatocellular, Liver Neoplasms genetics, Liver Neoplasms pathology
- Abstract
The rapidly increasing incidence of nonalcoholic fatty liver disease (NAFLD) is a growing health crisis worldwide. If not detected early, NAFLD progression can lead to irreversible pathological states, including liver fibrosis and cirrhosis. Using in vitro models to understand the molecular pathogenesis has been extremely beneficial; however, most studies have utilized only short-term exposures, highlighting a limitation in current research to model extended fat-induced liver injury. We treated Hep3B cells continuously with a low dose of oleic and palmitic free fatty acids (FFAs) for 7 or 28 days. Transcriptomic analysis identified dysregulated molecular pathways and differential expression of 984 and 917 genes after FFA treatment for 7 and 28 days respectively. DNA methylation analysis of altered DNA methylated regions (DMRs) found 7 DMRs in common. Pathway analysis of differentially expressed genes (DEGs) revealed transcriptomic changes primarily involved in lipid metabolism, small molecule biochemistry, and molecular transport. Western blot analysis revealed changes in PDK4 and CPT1A protein levels, indicative of mitochondrial stress. In line with this, there was mitochondrial morphological change demonstrating breakdown of the mitochondrial network. This in vitro model of human NAFL mimics results observed in human patients and may be used as a pre-clinical model for drug intervention.
- Published
- 2024
- Full Text
- View/download PDF
4. Evaluating the toxicokinetics of some metabolites of a C6 polyfluorinated compound, 6:2 fluorotelomer alcohol in pregnant and nonpregnant rats after oral exposure to the parent compound.
- Author
-
Rice PA, Kabadi SV, Doerge DR, Vanlandingham MM, Churchwell MI, Tryndyak VP, Fisher JW, Aungst J, and Beland FA
- Subjects
- Rats, Humans, Animals, Pregnancy, Female, Toxicokinetics, Biological Transport, Carboxylic Acids, Fluorocarbons toxicity, Fluorocarbons chemistry
- Abstract
The 6:2 fluorotelomer alcohol (6:2 FTOH) is a common impurity in per- and polyfluoroalkyl substances (PFASs) used in many applications. Our previous toxicokinetic (TK) evaluation of 6:2 FTOH calculated times to steady state (tss) of one of its metabolites, 5:3 fluorotelomer carboxylic acid (5:3A), in the plasma and tissues of up to a year after oral exposure to rats. Our current work further elucidated the TK of 5:3A and other metabolites of 6:2 FTOH in pregnant and nonpregnant rats after repeated oral exposure and examined the role of renal transporters in the biopersistence of 5:3A. The tss values for 5:3A in serum and tissues of adult nonpregnant animals ranged from 150 days to over a year. 4:3 fluorotelomer carboxylic acid (4:3A) was an additional potentially-biopersistent metabolite. 5:3A was the major metabolite of 6:2 FTOH in serum of pregnant dams and fetuses at each time interval. 5:3A was not a substrate for renal transporters in a human kidney cell line in vitro, indicating that renal reuptake of 5:3A is unlikely contribute to its biopersistence. Further research is needed to identify the underlying processes and evaluate the impact of these 6:2 FTOH metabolites on human health., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
5. Gene expression analyses reveal potential mechanism of inorganic arsenic-induced apoptosis in zebrafish.
- Author
-
Silva CS, Kudlyk T, Tryndyak VP, Twaddle NC, Robinson B, Gu Q, Beland FA, Fitzpatrick SC, and Kanungo J
- Subjects
- Animals, Humans, Zebrafish genetics, Tumor Suppressor Protein p53, Hedgehog Proteins pharmacology, Gene Expression Profiling, Apoptosis, Arsenic toxicity, Arsenites toxicity
- Abstract
Our previous study showed that sodium arsenite (200 mg/L) affected the nervous system and induced motor neuron development via the Sonic hedgehog pathway in zebrafish larvae. To gain more insight into the effects of arsenite on other signaling pathways, including apoptosis, we have performed quantitative polymerase chain reaction array-based gene expression analyses. The 96-well array plates contained primers for 84 genes representing 10 signaling pathways that regulate several biological functions, including apoptosis. We exposed eggs at 5 h postfertilization until the 72 h postfertilization larval stage to 200 mg/L sodium arsenite. In the Janus kinase/signal transducers and activators of transcription, nuclear factor κ-light-chain-enhancer of activated B cells, and Wingless/Int-1 signaling pathways, the expression of only one gene in each pathway was significantly altered. The expression of multiple genes was altered in the p53 and oxidative stress pathways. Sodium arsenite induced excessive apoptosis in the larvae. This compelled us to analyze specific genes in the p53 pathway, including cdkn1a, gadd45aa, and gadd45ba. Our data suggest that the p53 pathway is likely responsible for sodium arsenite-induced apoptosis. In addition, sodium arsenite significantly reduced global DNA methylation in the zebrafish larvae, which may indicate that epigenetic factors could be dysregulated after arsenic exposure. Together, these data elucidate potential mechanisms of arsenic toxicity that could improve understanding of arsenic's effects on human health., (Published 2023. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2023
- Full Text
- View/download PDF
6. Effect of an obesogenic high-fat and high-sucrose diet on hepatic gene expression signatures in male Collaborative Cross mice.
- Author
-
Tryndyak VP, Willett RA, Nagumalli SK, Li D, Avigan MI, Beland FA, Rusyn I, and Pogribny IP
- Subjects
- Male, Humans, Mice, Animals, Transcriptome, Collaborative Cross Mice genetics, Sucrose metabolism, Liver metabolism, Diet, High-Fat, Lipids, Mice, Inbred C57BL, Lipid Metabolism, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Nonalcoholic fatty liver disease (NAFLD), the most prevalent chronic liver disease, is characterized by substantial variations in case-level severity. In this study, we used a genetically diverse Collaborative Cross (CC) mouse population model to analyze the global transcriptome and clarify the molecular mechanisms involved in hepatic fat accumulation that determine the level and severity of NAFLD. Twenty-four strains of male CC mice were maintained on a high-fat/high-sucrose (HF/HS) diet for 12 wk, and their hepatic gene expression profiles were determined by next-generation RNA sequencing. We found that the development of the nonalcoholic fatty liver (NAFL) phenotype in CC mice coincided with significant changes in the expression of hepatic genes at the population level, evidenced by the presence of 724 differentially expressed genes involved in lipid and carbohydrate metabolism, cell morphology, vitamin and mineral metabolism, energy production, and DNA replication, recombination, and repair. Importantly, expression of 68 of these genes strongly correlated with the extent of hepatic lipid accumulation in the overall population of HF/HS diet-fed male CC mice. Results of partial least squares (PLS) modeling showed that these derived hepatic gene expression signatures help to identify the individual mouse strains that are highly susceptible to the development of NAFLD induced by an HF/HS diet. These findings imply that gene expression profiling, combined with a PLS modeling approach, may be a useful tool to predict NAFLD severity in genetically diverse patient populations. NEW & NOTEWORTHY Feeding male Collaborative Cross mice an obesogenic diet allows modeling NAFLD at the population level. The development of NAFLD coincided with significant hepatic transcriptomic changes in this model. Genes (724) were differentially expressed and expression of 68 genes strongly correlated with the extent of hepatic lipid accumulation. Partial least squares modeling showed that derived hepatic gene expression signatures may help to identify individual mouse strains that are highly susceptible to the development of NAFLD.
- Published
- 2023
- Full Text
- View/download PDF
7. Temporal dynamics of SARS-CoV-2 genome and detection of variants of concern in wastewater influent from two metropolitan areas in Arkansas.
- Author
-
Silva CS, Tryndyak VP, Camacho L, Orloff MS, Porter A, Garner K, Mullis L, and Azevedo M
- Subjects
- Arkansas epidemiology, COVID-19 Testing, Humans, Membrane Glycoproteins, Phosphoproteins, Polyproteins, RNA, Viral genetics, Wastewater, Wastewater-Based Epidemiological Monitoring, COVID-19, SARS-CoV-2 genetics
- Abstract
Although SARS-CoV-2 can cause severe illness and death, a percentage of the infected population is asymptomatic. This, along with other factors, such as insufficient diagnostic testing and underreporting due to self-testing, contributes to the silent transmission of SARS-CoV-2 and highlights the importance of implementing additional surveillance tools. The fecal shedding of the virus from infected individuals enables its detection in community wastewater, and this has become a valuable public health tool worldwide as it allows the monitoring of the disease on a populational scale. Here, we monitored the presence of SARS-CoV-2 and its dynamic genomic changes in wastewater sampled from two metropolitan areas in Arkansas during major surges of COVID-19 cases and assessed how the viral titers in these samples related to the clinical case counts between late April 2020 and January 2022. The levels of SARS-CoV-2 RNA were quantified by reverse-transcription quantitative polymerase chain reaction (RT-qPCR) using a set of TaqMan assays targeting three different viral genes (encoding ORF1ab polyprotein, surface glycoprotein, and nucleocapsid phosphoprotein). An allele-specific RT-qPCR approach was used to screen the samples for SARS-CoV-2 mutations. The identity and genetic diversity of the virus were further investigated through amplicon-based RNA sequencing, and SARS-CoV-2 variants of concern were detected in wastewater samples throughout the duration of this study. Our data show how changes in the virus genome can affect the sensitivity of specific RT-qPCR assays used in COVID-19 testing with the surge of new variants. A significant association was observed between viral titers in wastewater and recorded number of COVID-19 cases in the areas studied, except when assays failed to detect targets due to the presence of particular variants. These findings support the use of wastewater surveillance as a reliable complementary tool for monitoring SARS-CoV-2 and its genetic variants at the community level., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Published by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
8. Non-alcoholic fatty liver disease-associated DNA methylation and gene expression alterations in the livers of Collaborative Cross mice fed an obesogenic high-fat and high-sucrose diet.
- Author
-
Tryndyak VP, Willett RA, Avigan MI, Sanyal AJ, Beland FA, Rusyn I, and Pogribny IP
- Subjects
- Humans, Male, Female, Mice, Animals, DNA Methylation, Collaborative Cross Mice genetics, Sucrose metabolism, Liver metabolism, Diet, DNA metabolism, Gene Expression, Diet, High-Fat adverse effects, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent chronic liver disease, and patient susceptibility to its onset and progression is influenced by several factors. In this study, we investigated whether altered hepatic DNA methylation in liver tissue correlates with the degree of severity of NAFLD-like liver injury induced by a high-fat and high-sucrose (HF/HS) diet in Collaborative Cross (CC) mice. Using genome-wide targeted bisulphite DNA methylation next-generation sequencing, we found that mice with different non-alcoholic fatty liver (NAFL) phenotypes could be distinguished by changes in hepatic DNA methylation profiles. Specifically, NAFL-prone male CC042 mice exhibited more prominent DNA methylation changes compared with male CC011 mice and female CC011 and CC042 mice that developed only a mild NAFL phenotype. Moreover, these mouse strains demonstrated different patterns of DNA methylation. While the HF/HS diet induced both DNA hypomethylation and DNA hypermethylation changes in all the mouse strains, the NAFL-prone male CC042 mice demonstrated a global predominance of DNA hypermethylation, whereas a more pronounced DNA hypomethylation pattern developed in the mild-NAFL phenotypic mice. In a targeted analysis of selected genes that contain differentially methylated regions (DMRs), we identified NAFL phenotype-associated differences in DNA methylation and gene expression of the Apoa4, Gls2 , and Apom genes in severe NAFL-prone mice but not in mice with mild NAFL phenotypes. These changes in the expression of Apoa4 and Gls2 coincided with similar findings in a human in vitro cell model of diet-induced steatosis and in patients with NAFL. These results suggest that changes in the expression and DNA methylation status of these three genes may serve as a set of predictive markers for the development of NAFLD.
- Published
- 2022
- Full Text
- View/download PDF
9. Lipidomic profiling of the hepatic esterified fatty acid composition in diet-induced nonalcoholic fatty liver disease in genetically diverse Collaborative Cross mice.
- Author
-
Nagumalli SK, Willett RA, de Conti A, Tryndyak VP, Avigan MI, da Costa GG, Beland FA, Rusyn I, and Pogribny IP
- Subjects
- Animals, Choline, Collaborative Cross Mice, Diet, High-Fat adverse effects, Disease Models, Animal, Fatty Acids, Fatty Acids, Monounsaturated, Fatty Acids, Nonesterified, Fatty Acids, Unsaturated, Female, Folic Acid, Lipidomics, Liver, Male, Mice, Sucrose, Non-alcoholic Fatty Liver Disease etiology
- Abstract
Non-alcoholic fatty liver disease (NAFLD), one of the most common forms of chronic liver disease, is characterized by the excessive accumulation of lipid species in hepatocytes. Recent studies have indicated that in addition to the total lipid quantities, changes in lipid composition are a determining factor in hepatic lipotoxicity. Using ultra-high performance liquid chromatography coupled with electrospray tandem mass spectrometry, we analyzed the esterified fatty acid composition in 24 strains of male and female Collaborative Cross (CC) mice fed a high fat/high sucrose (HF/HS) diet for 12 weeks. Changes in lipid composition were found in all strains after the HF/HS diet, most notably characterized by increases in monounsaturated fatty acids (MUFA) and decreases in polyunsaturated fatty acids (PUFA). Similar changes in MUFA and PUFA were observed in a choline- and folate-deficient (CFD) mouse model of NAFLD, as well as in hepatocytes treated in vitro with free fatty acids. Analysis of fatty acid composition revealed that alterations were accompanied by an increase in the estimated activity of MUFA generating SCD1 enzyme and an estimated decrease in the activity of PUFA generating FADS1 and FADS2 enzymes. PUFA/MUFA ratios were inversely correlated with lipid accumulation in male and female CC mice fed the HF/HS diet and with morphological markers of hepatic injury in CFD diet-fed mouse model of NAFLD. These results demonstrate that different models of NAFLD are characterized by similar changes in the esterified fatty acid composition and that alterations in PUFA/MUFA ratios may serve as a diagnostic marker for NAFLD severity., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
10. Epigenetic changes induced in mice liver by methionine-supplemented and methionine-deficient diets.
- Author
-
Aissa AF, Tryndyak VP, de Conti A, Rita Thomazela Machado A, Tuttis K, da Silva Machado C, Hernandes LC, Wellington da Silva Santos P, Mara Serpeloni J, P Pogribny I, and Antunes LMG
- Subjects
- Animals, Chromatin metabolism, DNA Methylation, Diet, Epigenesis, Genetic, Female, Liver, Mice, Methionine, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
A diet deficient in donors of methyl group, such as methionine, affects DNA methylation and hepatic lipid metabolism. Methionine also affects other epigenetic mechanisms, such as microRNAs. We investigated the effects of methionine-supplemented or methionine-deficient diets on the expression of chromatin-modifying genes, global DNA methylation, the expression and methylation of genes related to lipid metabolism, and the expression of microRNAs in mouse liver. Female Swiss albino mice were fed a control diet (0.3% methionine), a methionine-supplemented diet (2% methionine), and a methionine-deficient diet (0% methionine) for 10 weeks. The genes most affected by the methionine-supplemented diet were associated with histone and DNA methyltransferases activity, while the methionine-deficient diet mostly altered the expression of histone methyltransferases genes. Both diets altered the global DNA methylation and the expression and gene-specific methylation of the lipid metabolism gene Apoa5. Both diets altered the expression of several liver homeostasis-related microRNAs, including miR-190b-5p, miR-130b-3p, miR-376c-3p, miR-411-5p, miR-29c-3p, miR-295-3p, and miR-467d-5p, with the methionine-deficient diet causing a more substantial effect. The effects of improper amounts of methionine in the diet on liver pathologies may involve a cooperative action of chromatin-modifying genes, which results in an aberrant pattern of global and gene-specific methylation, and microRNAs responsible for liver homeostasis., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
11. Epigenetic effects of low-level sodium arsenite exposure on human liver HepaRG cells.
- Author
-
Tryndyak VP, Borowa-Mazgaj B, Steward CR, Beland FA, and Pogribny IP
- Subjects
- Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Claudins genetics, Claudins metabolism, DNA Damage, Epithelial-Mesenchymal Transition drug effects, Gene Expression Regulation, Neoplastic, Hepatocytes metabolism, Hepatocytes pathology, Histones genetics, Histones metabolism, Humans, Liver Neoplasms genetics, Liver Neoplasms metabolism, Liver Neoplasms pathology, Mutation, Arsenites toxicity, Cell Transformation, Neoplastic chemically induced, DNA Methylation drug effects, Epigenesis, Genetic drug effects, Hepatocytes drug effects, Liver Neoplasms chemically induced, Sodium Compounds toxicity
- Abstract
Chronic exposure to inorganic arsenic is associated with a variety of adverse health effects, including lung, bladder, kidney, and liver cancer. Several mechanisms have been proposed for arsenic-induced tumorigenesis; however, insufficient knowledge and many unanswered questions remain to explain the integrated molecular pathogenesis of arsenic carcinogenicity. In the present study, using non-tumorigenic human liver HepaRG cells, we investigated epigenetic alterations upon prolonged exposure to a noncytotoxic concentration of sodium arsenite (NaAsO
2 ). We demonstrate that continuous exposure of HepaRG cells to 1 µM sodium arsenite (NaAsO2 ) for 14 days resulted in substantial cytosine DNA demethylation and hypermethylation across the genome, among which the claudin 14 (CLDN14) gene was hypermethylated and the most down-regulated gene. Another important finding was a profound loss of histone H3 lysine 36 (H3K36) trimethylation, which was accompanied by increased damage to genomic DNA and an elevated de novo mutation frequency. These results demonstrate that continuous exposure of HepaRG cells to a noncytotoxic concentration of NaAsO2 results in substantial epigenetic abnormalities accompanied by several carcinogenesis-related events, including induction of epithelial-to-mesenchymal transition, damage to DNA, inhibition of DNA repair genes, and induction of de novo mutations. Importantly, this study highlights the intimate mechanistic link and interplay between two fundamental cancer-associated events, epigenetic and genetic alterations, in arsenic-associated carcinogenesis.- Published
- 2020
- Full Text
- View/download PDF
12. From the Cover: Aloin, a Component of the Aloe Vera Plant Leaf, Induces Pathological Changes and Modulates the Composition of Microbiota in the Large Intestines of F344/N Male Rats.
- Author
-
Boudreau MD, Olson GR, Tryndyak VP, Bryant MS, Felton RP, and Beland FA
- Subjects
- Animals, Colon microbiology, Colon pathology, Dose-Response Relationship, Drug, Emodin administration & dosage, Emodin toxicity, Feces microbiology, Growth, Male, Rats, Rats, Inbred F344, Aloe chemistry, Colon drug effects, Emodin analogs & derivatives, Gastrointestinal Microbiome drug effects, Plant Extracts toxicity, Plant Leaves chemistry
- Abstract
In a previous study, the oral administration of an Aloe vera whole leaf extract induced dose-related mucosal and goblet cell hyperplasia in the rat colon after 13 weeks and colon cancer after 2 years. The primary goal of this study was to determine whether or not the administration of aloin, a component of the Aloe vera plant leaf, would replicate the pathophysiological effects that were observed in rats in the previous study with an Aloe vera whole leaf extract. Groups of 10 male F344/N rats were administered aloin at 0, 6.95, 13.9, 27.8, 55.7, 111, 223, and 446 mg/kg drinking water for 13 weeks. At the end of study, rat feces were collected, and the composition of fecal bacteria was investigated by next generation sequencing of the PCR-amplified V3/V4 region of the 16S rRNA gene. At necropsy, blood was collected by cardiac puncture and organs and sections of the large intestine were collected for histopathology. Aloin induced dose-related increased incidences and severities of mucosal and goblet cell hyperplasia that extended from the cecum to the rectum, with increased incidences and severities detected at aloin doses ≥55.7 mg/kg drinking water. Analysis of the 16S rRNA metagenomics sequencing data revealed marked shifts in the structure of the gut microbiota in aloin-treated rats at each taxonomic rank. This study highlights the similarities in effects observed for aloin and the Aloe vera whole leaf extract, and points to a potential mechanism of action to explain the observed pathological changes via modulation of the gut microbiota composition., (Published by Oxford University Press on behalf of the Society of Toxicology 2017. This work is written by US Government employees and is in the public domain in the US.)
- Published
- 2017
- Full Text
- View/download PDF
13. Status of hepatic DNA methylome predetermines and modulates the severity of non-alcoholic fatty liver injury in mice.
- Author
-
Tryndyak VP, Han T, Fuscoe JC, Ross SA, Beland FA, and Pogribny IP
- Subjects
- Animals, Choline, CpG Islands, Cytosine chemistry, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases metabolism, DNA Methyltransferase 3A, Diet, Folic Acid, Histones metabolism, Mice, Mice, Inbred A, Mice, Inbred Strains, DNA Methylation, Epigenesis, Genetic, Liver physiopathology, Non-alcoholic Fatty Liver Disease genetics
- Abstract
Background: Nonalcoholic fatty liver disease (NAFLD) is a major health problem and a leading cause of chronic liver disease in the United States and Western countries. In humans, genetic factors greatly influence individual susceptibility to NAFLD; nonetheless, the effect of inter-individual differences in the normal liver epigenome with regard to the susceptibility to NAFLD has not been determined., Results: In the present study, we investigated the association between the DNA methylation status in the livers of A/J and WSB/EiJ mice and the severity of NAFLD-associated liver injury. We demonstrate that A/J and WSB/EiJ mice, which are characterized by significant differences in the severity of liver injury induced by a choline- and folate-deficient (CFD) diet exhibit substantial differences in cytosine DNA methylation in their normal livers. Furthermore, feeding A/J and WSB/EiJ mice a CFD diet for 12 weeks resulted in different trends and changes in hepatic cytosine DNA methylation., Conclusion: Our findings indicate a primary role of hepatic DNA methylation in the pathogenesis of NAFLD and suggest that individual variations in DNA methylation across the genome may be a factor determining and influencing the vulnerability to NAFLD.
- Published
- 2016
- Full Text
- View/download PDF
14. MicroRNA changes, activation of progenitor cells and severity of liver injury in mice induced by choline and folate deficiency.
- Author
-
Tryndyak VP, Marrone AK, Latendresse JR, Muskhelishvili L, Beland FA, and Pogribny IP
- Subjects
- Animals, Choline Deficiency genetics, Choline Deficiency pathology, Folic Acid Deficiency genetics, Folic Acid Deficiency pathology, Mice, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease pathology, Choline Deficiency metabolism, Folic Acid Deficiency metabolism, MicroRNAs metabolism, Non-alcoholic Fatty Liver Disease metabolism, Stem Cells metabolism
- Abstract
Dietary deficiency in methyl-group donors and cofactors induces liver injury that resembles many pathophysiological and histopathological features of human nonalcoholic fatty liver disease (NAFLD), including an altered expression of microRNAs (miRNAs). We evaluated the consequences of a choline- and folate-deficient (CFD) diet on the expression of miRNAs in the livers of male A/J and WSB/EiJ mice. The results demonstrate that NAFLD-like liver injury induced by the CFD diet in A/J and WSB/EiJ mice was associated with marked alterations in hepatic miRNAome profiles, with the magnitude of miRNA expression changes being greater in WSB/EiJ mice, the strain characterized by the greatest severity of liver injury. Specifically, WSB/EiJ mice exhibited more prominent changes in the expression of common miRNAs as compared to A/J mice and distinct miRNA alterations, including the overexpression of miR-134, miR-409-3p, miR-410 and miR-495 miRNAs that were accompanied by an activation of hepatic progenitor cells and fibrogenesis. This in vivo finding was further confirmed by in vitro experiments showing an overexpression of these miRNAs in undifferentiated progenitor hepatic HepaRG cells compared to in fully differentiated HepaRG cells. Additionally, a marked elevation of miR-134, miR-409-3p, miR-410 and miR-495 was found in plasma of WSB/EiJ mice fed the CFD diet, while none of the miRNAs was changed in plasma of A/J mice. These findings suggest that miRNAs may be crucial regulators responsible for the progression of NAFLD and may be useful as noninvasive diagnostic indicators of the severity and progression of NAFLD., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
15. Antiproliferative and proapoptotic effects of a pyrrole containing arylthioindole in human Jurkat leukemia cell line and multidrug-resistant Jurkat/A4 cells.
- Author
-
Philchenkov AA, Zavelevich MP, Tryndyak VP, Kuiava LM, Blokhin DY, Miura K, Silvestri R, and Pogribny IP
- Subjects
- Antineoplastic Agents chemistry, Caspase 3 metabolism, Cell Cycle drug effects, Cell Proliferation drug effects, Gene Expression Profiling, Humans, Jurkat Cells, Leukemia genetics, Leukemia metabolism, Poly(ADP-ribose) Polymerases metabolism, Tubulin Modulators chemistry, Antineoplastic Agents pharmacology, Apoptosis drug effects, Drug Resistance, Neoplasm, Tubulin Modulators pharmacology
- Abstract
Recently, a series of novel arylthioindole compounds, potent inhibitors of tubulin polymerization and cancer cell growth, were synthesized. In the present study the effects of 2-(1H-pyrrol-3-yl)-3-((3,4,5-trimethoxyphenyl)thio)-1H-indole (ATI5 compound) on cell proliferation, cell cycle progression, and induction of apoptosis in human T-cell acute leukemia Jurkat cells and their multidrug resistant Jurkat/A4 subline were investigated. Treatment of the Jurkat cells with the ATI5 compound for 48 hrs resulted in a strong G2/M cell cycle arrest and p53-independent apoptotic cell death accompanied by the induction of the active form of caspase-3 and poly(ADP-ribose) polymerase-1 (PARP-1) cleavage. ATI5 treatment also caused non-cell death related mitotic arrest in multidrug resistant Jurkat/A4 cells after 48 hrs of treatment suggesting promising opportunities for the further design of pyrrole-containing ATI compounds as anticancer agents. Cell death resistance of Jurkat/A4 cells to ATI5 compound was associated with alterations in the expression of pro-survival and anti-apoptotic protein-coding and microRNA genes. More importantly, findings showing that ATI5 treatment induced p53-independent apoptosis are of great importance from a therapeutic point of view since p53 mutations are common genetic alterations in human neoplasms.
- Published
- 2015
- Full Text
- View/download PDF
16. Interstrain differences in the progression of nonalcoholic steatohepatitis to fibrosis in mice are associated with altered hepatic iron metabolism.
- Author
-
Shpyleva S, Pogribna M, Cozart C, Bryant MS, Muskhelishvili L, Tryndyak VP, Ross SA, Beland FA, and Pogribny IP
- Subjects
- Animals, Apoferritins genetics, Apoferritins metabolism, Cation Transport Proteins genetics, Cation Transport Proteins metabolism, Choline Deficiency complications, Choline Deficiency pathology, Disease Progression, Down-Regulation, Folic Acid Deficiency complications, Folic Acid Deficiency pathology, Hepatocytes cytology, Hepatocytes metabolism, Iron Regulatory Protein 1 genetics, Iron Regulatory Protein 1 metabolism, Liver pathology, Male, Mice, Mice, Inbred C57BL, Mice, Inbred Strains, MicroRNAs genetics, MicroRNAs metabolism, Receptors, Transferrin genetics, Receptors, Transferrin metabolism, Iron metabolism, Liver metabolism, Liver Cirrhosis pathology, Non-alcoholic Fatty Liver Disease pathology
- Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major health problem worldwide. Currently, there is a lack of conclusive information to clarify the molecular events and mechanisms responsible for the progression of NAFLD to fibrosis and cirrhosis and, more importantly, for differences in interindividual disease severity. The aim of this study was to investigate a role of interindividual differences in iron metabolism among inbred mouse strains in the pathogenesis and severity of fibrosis in a model of NAFLD. Feeding male A/J, 129S1/SvImJ and WSB/EiJ mice a choline- and folate-deficient diet caused NAFLD-associated liver injury and iron metabolism abnormalities, especially in WSB/EiJ mice. NAFLD-associated fibrogenesis was correlated with a marked strain- and injury-dependent increase in the expression of iron metabolism genes, especially transferrin receptor (Tfrc), ferritin heavy chain (Fth1), and solute carrier family 40 (iron-regulated transporter), member 1 (Slc40a1, Fpn1) and their related proteins, and pronounced down-regulation of the iron regulatory protein 1 (IRP1), with the magnitude being A/J<129S1/SvImJ
- Published
- 2014
- Full Text
- View/download PDF
17. Iron metabolism disturbances in the MCF-7 human breast cancer cells with acquired resistance to doxorubicin and cisplatin.
- Author
-
Chekhun VF, Lukyanova NY, Burlaka CA, Bezdenezhnykh NA, Shpyleva SI, Tryndyak VP, Beland FA, and Pogribny IP
- Subjects
- Antibiotics, Antineoplastic pharmacology, Antineoplastic Agents pharmacology, Apoferritins antagonists & inhibitors, Apoferritins genetics, Blotting, Western, Breast Neoplasms pathology, Electron Spin Resonance Spectroscopy, Female, Gene Expression Regulation, Neoplastic, Humans, Immunoenzyme Techniques, MicroRNAs genetics, RNA, Small Interfering genetics, Tumor Cells, Cultured, Apoferritins metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cisplatin pharmacology, Doxorubicin pharmacology, Drug Resistance, Neoplasm, Iron metabolism
- Abstract
The development of resistance of cancer cells to therapeutic agents is the major obstacle in the successful treatment of breast cancer and the main cause of breast cancer recurrence. The results of several studies have demonstrated an important role of altered cellular iron metabolism in the progression of breast cancer and suggested that iron metabolism may be involved in the acquisition of a cancer cell drug-resistant phenotype. In the present study, we show that human MCF-7 breast cancer cells with an acquired resistance to the chemotherapeutic drugs doxorubicin (MCF-7/DOX) and cisplatin (MCF-7/CDDP) exhibited substantial alterations in the intracellular iron content and levels of iron-regulatory proteins involved in the cellular uptake, storage and export of iron, especially in profoundly increased levels of ferritin light chain (FTL) protein. The increased levels of FTL in breast cancer indicate that FTL may be used as a diagnostic and prognostic marker for breast cancer. Additionally, we demonstrate that targeted downregulation of FTL protein by the microRNA miR-133a increases sensitivity of MCF-7/DOX and MCF-7/CDDP cells to doxorubicin and cisplatin. These results suggest that correction of iron metabolism abnormalities may substantially improve the efficiency of breast cancer treatment.
- Published
- 2013
- Full Text
- View/download PDF
18. Modulation of intracellular iron metabolism by iron chelation affects chromatin remodeling proteins and corresponding epigenetic modifications in breast cancer cells and increases their sensitivity to chemotherapeutic agents.
- Author
-
Pogribny IP, Tryndyak VP, Pogribna M, Shpyleva S, Surratt G, Gamboa da Costa G, and Beland FA
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis genetics, Breast Neoplasms genetics, Breast Neoplasms pathology, Cisplatin pharmacology, Down-Regulation, Female, Gene Expression Regulation, Neoplastic drug effects, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Histones genetics, Histones metabolism, Humans, Iron Chelating Agents pharmacology, MCF-7 Cells, Up-Regulation, Breast Neoplasms metabolism, Chromatin Assembly and Disassembly genetics, Epigenesis, Genetic, Iron metabolism
- Abstract
Iron plays a vital role in the normal functioning of cells via the regulation of essential cellular metabolic reactions, including several DNA and histone-modifying proteins. The metabolic status of iron and the regulation of epigenetic mechanisms are well-balanced and tightly controlled in normal cells; however, in cancer cells these processes are profoundly disturbed. Cancer-related abnormalities in iron metabolism have been corrected through the use of iron-chelating agents, which cause an inhibition of DNA synthesis, G₁-S phase arrest, an inhibition of epithelial-to-mesenchymal transition, and the activation of apoptosis. In the present study, we show that, in addition to these well-studied molecular mechanisms, the treatment of wild-type TP53 MCF-7 and mutant TP53 MDA-MB-231 human breast cancer cells with desferrioxamine (DFO), a model iron chelator, causes significant epigenetic alterations at the global and gene-specific levels. Specifically, DFO treatment decreased the protein levels of the histone H3 lysine 9 demethylase, Jumonji domain-containing protein 2A (JMJD2A), in the MCF-7 and MDA-MB-231 cells and down-regulated the levels of the histone H3 lysine 4 demethylase, lysine-specific demethylase 1 (LSD1), in the MDA-MB-231 cells. These changes were accompanied by alterations in corresponding metabolically sensitive histone marks. Additionally, we demonstrate that DFO treatment activates apoptotic programs in MCF-7 and MDA-MB-231 cancer cells and enhances their sensitivity to the chemotherapeutic agents, doxorubicin and cisplatin; however, the mechanisms underlying this activation differ. The induction of apoptosis in wild-type TP53 MCF-7 cells was p53-dependent, triggered mainly by the down-regulation of the JMJD2A histone demethylase, while in mutant TP53 MDA-MB-231 cells, the activation of the p53-independent apoptotic program was driven predominantly by the epigenetic up-regulation of p21.
- Published
- 2013
- Full Text
- View/download PDF
19. Acetaminophen-induced acute liver injury in HCV transgenic mice.
- Author
-
Uehara T, Kosyk O, Jeannot E, Bradford BU, Tech K, Macdonald JM, Boorman GA, Chatterjee S, Mason RP, Melnyk SB, Tryndyak VP, Pogribny IP, and Rusyn I
- Subjects
- Acetaminophen administration & dosage, Acute Disease, Analgesics, Non-Narcotic administration & dosage, Animals, Disease Models, Animal, Disease Progression, Disease Susceptibility, Dose-Response Relationship, Drug, Fasting, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mitochondria, Liver pathology, Time Factors, Acetaminophen toxicity, Analgesics, Non-Narcotic toxicity, Chemical and Drug Induced Liver Injury etiology, Hepatitis C complications, Mitochondria, Liver drug effects
- Abstract
The exact etiology of clinical cases of acute liver failure is difficult to ascertain and it is likely that various co-morbidity factors play a role. For example, epidemiological evidence suggests that coexistent hepatitis C virus (HCV) infection increased the risk of acetaminophen-induced acute liver injury, and was associated with an increased risk of progression to acute liver failure. However, little is known about possible mechanisms of enhanced acetaminophen hepatotoxicity in HCV-infected subjects. In this study, we tested a hypothesis that HCV-Tg mice may be more susceptible to acetaminophen hepatotoxicity, and also evaluated the mechanisms of acetaminophen-induced liver damage in wild type and HCV-Tg mice expressing core, E1 and E2 proteins. Male mice were treated with a single dose of acetaminophen (300 or 500 mg/kg in fed animals; or 200 mg/kg in fasted animals; i.g.) and liver and serum endpoints were evaluated at 4 and 24h after dosing. Our results suggest that in fed mice, liver toxicity in HCV-Tg mice is not markedly exaggerated as compared to the wild-type mice. In fasted mice, greater liver injury was observed in HCV-Tg mice. In fed mice dosed with 300 mg/kg acetaminophen, we observed that liver mitochondria in HCV-Tg mice exhibited signs of dysfunction showing the potential mechanism for increased susceptibility., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
20. Modifying metabolically sensitive histone marks by inhibiting glutamine metabolism affects gene expression and alters cancer cell phenotype.
- Author
-
Simpson NE, Tryndyak VP, Pogribna M, Beland FA, and Pogribny IP
- Subjects
- Apoptosis drug effects, Apoptosis genetics, Benzophenanthridines pharmacology, Breast Neoplasms pathology, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases genetics, DNA-Binding Proteins genetics, Enzyme Inhibitors pharmacology, Epigenesis, Genetic drug effects, Female, Gene Expression Regulation, Neoplastic, Glutaminase antagonists & inhibitors, Glutamine genetics, Histone-Lysine N-Methyltransferase genetics, Histones genetics, Humans, Nuclear Proteins genetics, Promoter Regions, Genetic, Sirtuin 1 genetics, Transcription Factors genetics, DNA Methyltransferase 3B, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Glutamine metabolism, Histones metabolism
- Abstract
The interplay of metabolism and epigenetic regulatory mechanisms has become a focal point for a better understanding of cancer development and progression. In this study, we have acquired data supporting previous observations that demonstrate glutamine metabolism affects histone modifications in human breast cancer cell lines. Treatment of non-invasive epithelial (T-47D and MDA-MB-361) and invasive mesenchymal (MDA-MB-231 and Hs-578T) breast cancer cell lines with the glutaminase inhibitor, Compound 968, resulted in cytotoxicity in all cell lines, with the greatest effect being observed in MDA-MB-231 breast cancer cells. Compound 968-treatment induced significant downregulation of 20 critical cancer-related genes, the majority of which are anti-apoptotic and/or promote metastasis, including AKT, BCL2, BCL2L1, CCND1, CDKN3, ERBB2, ETS1, E2F1, JUN, KITLG, MYB, and MYC. Histone H3K4me3, a mark of transcriptional activation, was reduced at the promoters of all but one of these critical cancer genes. The decrease in histone H3K4me3 at global and gene-specific levels correlated with reduced expression of SETD1 and ASH2L, genes encoding the histone H3K4 methyltransferase complex. Further, the expression of other epigenetic regulatory genes, known to be downregulated during apoptosis (e.g., DNMT1, DNMT3B, SETD1 and SIRT1), was also downregulated by Compound 968. These changes in gene expression and histone modifications were accompanied by the activation of apoptosis, and decreased invasiveness and resistance of MDA-MB-231 cells to chemotherapeutic drug doxorubicin. The results of this study provide evidence to a link between cytotoxicity caused by inhibiting glutamine metabolism with alterations of the epigenome of breast cancer cells and suggest that modification of intracellular metabolism may enhance the efficiency of epigenetic therapy.
- Published
- 2012
- Full Text
- View/download PDF
21. Plasma microRNAs are sensitive indicators of inter-strain differences in the severity of liver injury induced in mice by a choline- and folate-deficient diet.
- Author
-
Tryndyak VP, Latendresse JR, Montgomery B, Ross SA, Beland FA, Rusyn I, and Pogribny IP
- Subjects
- Animals, Biomarkers blood, Disease Models, Animal, Fatty Liver etiology, Fatty Liver pathology, Genetic Predisposition to Disease, Male, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Mice, Inbred Strains, Non-alcoholic Fatty Liver Disease, Severity of Illness Index, Species Specificity, Choline Deficiency complications, Fatty Liver genetics, Folic Acid Deficiency complications, MicroRNAs blood
- Abstract
MicroRNAs (miRNAs) are a class of small, conserved, tissue-specific regulatory non-coding RNAs that modulate a variety of biological processes and play a fundamental role in the pathogenesis of major human diseases, including nonalcoholic fatty liver disease (NAFLD). However, the association between inter-individual differences in susceptibility to NAFLD and altered miRNA expression is largely unknown. In view of this, the goals of the present study were (i) to determine whether or not individual differences in the extent of NAFLD-induced liver injury are associated with altered miRNA expression, and (ii) assess if circulating blood miRNAs may be used as potential biomarkers for the noninvasive evaluation of the severity of NAFLD. A panel of seven genetically diverse strains of inbred male mice (A/J, C57BL/6J, C3H/HeJ, 129S/SvImJ, CAST/EiJ, PWK/PhJ, and WSB/EiJ) were fed a choline- and folate-deficient (CFD) diet for 12weeks. This diet induced liver injury in all mouse strains; however, the extent of NAFLD-associated pathomorphological changes in the livers was strain-specific, with A/J, C57BL/6J, and C3H/HeJ mice being the least sensitive and WSB/EiJ mice being the most sensitive. The morphological changes in the livers were accompanied by differences in the levels of hepatic and plasma miRNAs. The levels of circulating miR-34a, miR-122, miR-181a, miR-192, and miR-200b miRNAs were significantly correlated with a severity of NAFLD-specific liver pathomorphological features, with the strongest correlation occurring with miR-34a. These observations suggest that the plasma levels of miRNAs may be used as biomarkers for noninvasive monitoring the extent of NAFLD-associated liver injury and susceptibility to NAFLD., (Published by Elsevier Inc.)
- Published
- 2012
- Full Text
- View/download PDF
22. An in vitro investigation of metabolically sensitive biomarkers in breast cancer progression.
- Author
-
Simpson NE, Tryndyak VP, Beland FA, and Pogribny IP
- Subjects
- Acetylation drug effects, Cell Line, Tumor, Disease Progression, Female, Gene Expression Regulation, Neoplastic, Glutamine metabolism, Histone Acetyltransferases genetics, Histone Acetyltransferases metabolism, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases genetics, Histone Deacetylases metabolism, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Humans, Methylation drug effects, Repressor Proteins genetics, Repressor Proteins metabolism, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism
- Abstract
Epigenetic biomarkers are emerging as determinants of breast cancer prognosis. Breast cancer cells display unique alterations in major cellular metabolic pathways and it is becoming widely recognized that enzymes that regulate epigenetic alterations are metabolically sensitive. In this study, we used microarray data from the GEO database to compare gene expression for regulators of metabolism and epigenetic alterations among non-invasive epithelial (MCF-7, MDA-MB-361, and T-47D) and invasive mesenchymal (MDA-MB-231, Hs-578T, and BT-549) breast cancer cell lines. The expression of genes, including GLS1, GFPT2, LDHA, HDAC9, MYST2, and SUV420H2, was assessed using RT-PCR. There was differential expression between epithelial and mesenchymal cell lines. MYST2 and SUV420H2 regulate the levels of the epigenetic biomarkers histone H4 lysine 16 acetylation (H4K16ac) and histone H4 lysine 20 trimethylation (H4K20me3), respectively. Reduced amounts of H4K16ac and H4K20me3 correlated with lower levels of MYST2 and SUV420H2 in mesenchymal cells and, along with reduced amounts of histone H3 lysine 9 acetylation (H3K9ac), were found to distinguish epithelial from mesenchymal cells. In addition, both GLS1 and GFPT2 play roles in glutamine metabolism and were observed to be more highly expressed in mesenchymal cell lines, and when glutamine and glutamate levels reported in the NCI-60 metabolomics dataset were compared, the ratio of glutamate/glutamine was found to be higher in mesenchymal cells. Blocking the conversion of glutamine to glutamate using an allosteric inhibitor, Compound 968, against GLS1, increased H4K16ac in T-47D and MDA-MB-231 cells, linking glutamine metabolism to a particular histone modification in breast cancer. These findings support the concept that metabolically sensitive histone modifications and corresponding histone modifying enzymes can be used as diagnostic and prognostic biomarkers for breast cancer. It also further emphasizes the importance of glutamine metabolism in tumor progression and that inhibitors of cellular metabolic pathways may join histone deacetylase inhibitors as a form of epigenetic therapy.
- Published
- 2012
- Full Text
- View/download PDF
23. Chronic administration of 2-acetylaminofluorene alters the cellular iron metabolism in rat liver.
- Author
-
Shpyleva SI, Muskhelishvili L, Tryndyak VP, Koturbash I, Tokar EJ, Waalkes MP, Beland FA, and Pogribny IP
- Subjects
- Animals, Antimicrobial Cationic Peptides genetics, Antimicrobial Cationic Peptides metabolism, Cation Transport Proteins genetics, Cation Transport Proteins metabolism, Cell Line, Tumor, Disease Progression, Female, Gene Expression Regulation, Neoplastic drug effects, Hepatocytes metabolism, Hepcidins, Homeostasis drug effects, Liver drug effects, Liver metabolism, Liver Neoplasms genetics, Liver Neoplasms metabolism, Male, Precancerous Conditions genetics, Precancerous Conditions metabolism, Rats, Rats, Sprague-Dawley, 2-Acetylaminofluorene toxicity, Carcinogens toxicity, Hepatocytes drug effects, Iron metabolism, Liver Neoplasms chemically induced, Precancerous Conditions chemically induced
- Abstract
Dysregulated intracellular iron homeostasis has been found not only in rodent and human hepatocellular carcinomas but also in several preneoplastic pathological states associated with hepatocarcinogenesis; however, the precise underlying mechanisms of metabolic iron disturbances in preneoplastic liver and the role of these disturbances remain unexplored. In the present study, using an in vivo model of rat hepatocarcinogenesis induced by 2-acetylaminofluorene, we found extensive alterations in cellular iron metabolism at preneoplastic stages of liver carcinogenesis. These were characterized by a substantial decrease in the levels of cytoplasmic non-heme iron in foci of initiated hepatocytes and altered expression of the major genes responsible for the proper maintenance of intracellular iron homeostasis. Gene expression analysis revealed that the decreased intracellular levels of iron in preneoplastic foci might be attributed to increased iron export from the cells, driven by upregulation of ferroportin (Fpn1), the only known non-heme iron exporter. Likewise, increased Fpn1 gene expression was found in vitro in TRL1215 rat liver cells with an acquired malignant phenotype, suggesting that upregulation of Fpn1 might be a specific feature of neoplastically transformed cells. Other changes observed in vivo included the downregulation of hepcidin (Hamp) gene, a key regulator of Fpn1, and this was accompanied by decreased levels of CCAAT/enhancer binding proteins alpha and beta, especially at the Hamp promoter. In conclusion, our results demonstrate the significance of altered intracellular iron metabolism in the progression of liver carcinogenesis and suggest that correction of these alterations could possibly affect liver cancer development.
- Published
- 2011
- Full Text
- View/download PDF
24. The role of epigenetic events in genotoxic hepatocarcinogenesis induced by 2-acetylaminofluorene.
- Author
-
Pogribny IP, Muskhelishvili L, Tryndyak VP, and Beland FA
- Subjects
- 2-Acetylaminofluorene pharmacology, Animals, Apoptosis drug effects, Cell Proliferation drug effects, DNA Methylation, Histones metabolism, Male, MicroRNAs metabolism, Rats, Rats, Sprague-Dawley, 2-Acetylaminofluorene toxicity, DNA Damage, Epigenesis, Genetic, Liver Neoplasms, Experimental chemically induced, Liver Neoplasms, Experimental genetics, Mutagens toxicity
- Abstract
It is well established that genotoxic reactivity of chemical carcinogens or their metabolites is a critical event in the initiation of tumorigenesis. However, the underlying mechanisms of events following initiation are less well understood, and with respect to genotoxic liver carcinogenesis, it is largely unknown how the initiated cells progress to form preneoplastic hepatic foci. In the present study, we investigated the underlying events associated with tumor-promoting activity of 2-acetylaminofluorene (2-AAF), a powerful complete genotoxic rat liver carcinogen. Male Sprague-Dawley rats were fed NIH-31 diet containing 0.02% of 2-AAF for 24 weeks, and the status of cytosine DNA methylation, histone methylation, and microRNA expression was determined in the livers of control and 2-AAF-fed rats. The results demonstrate that stages of multistage carcinogenesis following the initiation are driven primarily by carcinogen-induced epigenetic alterations. This was evidenced by altered global histone lysine methylation patterns, increased histone H3 lysine 9 and histone H3 lysine 27 trimethylation in the promoter regions of Rassf1a, p16(INK4a), Socs1, Cdh1, and Cx26 tumor suppressor genes, early Rassf1a and p16(INK4a) promoter CpG island hypermethylation, and altered microRNA expression in preneoplastic livers of rats exposed to 2-AAF. These changes were accompanied by dysregulation of the balance between cell proliferation and apoptosis, a fundamental pro-tumorigenic event in hepatocarcinogenesis. These results signify the fundamental role of epigenetic alterations in genotoxic liver carcinogenesis., (Published by Elsevier B.V.)
- Published
- 2011
- Full Text
- View/download PDF
25. Coupling global methylation and gene expression profiles reveal key pathophysiological events in liver injury induced by a methyl-deficient diet.
- Author
-
Tryndyak VP, Han T, Muskhelishvili L, Fuscoe JC, Ross SA, Beland FA, and Pogribny IP
- Subjects
- Animals, Carcinoma, Hepatocellular etiology, Carcinoma, Hepatocellular genetics, CpG Islands, DNA Damage, Diet, Fatty Liver etiology, Fatty Liver physiopathology, Gene Expression Regulation, Neoplastic, Liver Neoplasms, Experimental etiology, Liver Neoplasms, Experimental genetics, Male, Mice, Mice, Inbred C57BL, Non-alcoholic Fatty Liver Disease, Oligonucleotide Array Sequence Analysis, DNA Methylation, Gene Expression Profiling, Malnutrition complications
- Abstract
Scope: A methyl-deficient diet induces liver injury similar to human nonalcoholic steatohepatitis, one of the main risk factors for the development of hepatocellular carcinoma. Previous studies have demonstrated that this diet perturbs DNA methylation by causing a profound loss of global cytosine methylation, predominantly at heavily methylated repetitive sequences. However, whether methyl deficiency affects the methylation status of gene promoters has not been explored., Methods and Results: Mouse gene expression and CpG island microarrays were used to characterize the gene expression and CpG island methylation profiles in the livers of C57BL/6J mice fed a methyl-deficient diet. We detected 164 genes that were differentially expressed and exhibited an inverse relationship between the gene expression and the extent of CpG island methylation. Furthermore, these genes were associated with altered lipid and glucose metabolism, DNA damage and repair, apoptosis, the development of fibrosis, and liver tissue remodeling. Although there were both increased and decreased levels of CpG island methylation, the number of hypomethylated genes was substantially greater than the number of hypermethylated genes., Conclusion: The results this study demonstrate that pairing methylation profiles with gene expression profiles is a powerful approach to identify dysregulated high-priority fundamental pathophysiological pathways associated with disease development., (Copyright © 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2011
- Full Text
- View/download PDF
26. Role of ferritin alterations in human breast cancer cells.
- Author
-
Shpyleva SI, Tryndyak VP, Kovalchuk O, Starlard-Davenport A, Chekhun VF, Beland FA, and Pogribny IP
- Subjects
- Antibiotics, Antineoplastic pharmacology, Blotting, Western, Breast Neoplasms genetics, Cell Nucleus drug effects, Cell Nucleus metabolism, Female, Ferritins genetics, Fluorescent Antibody Technique, Humans, Luciferases metabolism, MicroRNAs genetics, MicroRNAs metabolism, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Doxorubicin pharmacology, Ferritins metabolism, Gene Expression Regulation, Neoplastic
- Abstract
Breast cancer is the most common malignancy in women. Successful treatment of breast cancer relies on a better understanding of the molecular mechanisms involved in breast cancer initiation and progression. Recent studies have suggested a crucial role of perturbations in ferritin levels and tightly associated with this, the deregulation of intracellular iron homeostasis; however, the underlying molecular mechanisms for the cancer-linked ferritin alterations remain largely unknown and often with conflicting conclusions. Therefore, this study was undertaken to define the role of ferritin in breast cancer. We determined that human breast cancer cells with an epithelial phenotype, such as MCF-7, MDA-MB-361, T-47D, HCC70 and cells, expressed low levels of ferritin light chain, ferritin heavy chain, transferrin, transferring receptor, and iron-regulatory proteins 1 and 2. In contrast, expression of these proteins was substantially elevated in breast cancer cells with an aggressive mesenchymal phenotype, such as Hs-578T, BT-549, and especially MDA-MB-231 cells. The up-regulation of ferritin light chain and ferritin heavy chain in MDA-MB-231 cells was accompanied by alterations in the subcellular distribution of these proteins as characterized by an increased level of nuclear ferritin and a lower level of the cellular labile iron pool as compared to MCF-7 cells. We established that ferritin heavy chain is a target of miRNA miR-200b, suggesting that its up-regulation in MDA-MB-231 cells may be triggered by the low expression of miR-200b. Ectopic up-regulation of miR-200b by transfection of MDA-MB-231 cells with miR-200b substantially decreased the level of ferritin heavy chain. More importantly, miR-200b-induced down-regulation of ferritin was associated with an increased sensitivity of the MDA-MB-231 cells to the chemotherapeutic agent doxorubicin. These results suggest that perturbations in ferritin levels are associated with the progression of breast cancer toward a more advanced malignant phenotype.
- Published
- 2011
- Full Text
- View/download PDF
27. Alterations of microRNAs and their targets are associated with acquired resistance of MCF-7 breast cancer cells to cisplatin.
- Author
-
Pogribny IP, Filkowski JN, Tryndyak VP, Golubov A, Shpyleva SI, and Kovalchuk O
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Blotting, Western, Breast Neoplasms pathology, Female, Gene Expression Profiling, Humans, Luciferases metabolism, Multidrug Resistance-Associated Proteins genetics, Multidrug Resistance-Associated Proteins metabolism, Oligonucleotide Array Sequence Analysis, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Cisplatin pharmacology, Drug Resistance, Neoplasm, MicroRNAs physiology
- Abstract
Cancer cells that develop resistance to chemotherapeutic agents are a major clinical obstacle in the successful treatment of breast cancer. Acquired cancer chemoresistance is a multifactorial phenomenon, involving various mechanisms and processes. Recent studies suggest that chemoresistance may be linked to drug-induced dysregulation of microRNA function. Furthermore, mounting evidence indicates the existence of similarities between drug-resistant and metastatic cancer cells in terms of resistance to apoptosis and enhanced invasiveness. We studied the role of miRNA alterations in the acquisition of cisplatin-resistant phenotype in MCF-7 human breast adenocarcinoma cells. We identified a total of 103 miRNAs that were overexpressed or underexpressed (46 upregulated and 57 downregulated) in MCF-7 cells resistant to cisplatin. These differentially expressed miRNAs are involved in the control of cell signaling, cell survival, DNA methylation and invasiveness. The most significantly dysregulated miRNAs were miR-146a, miR-10a, miR-221/222, miR-345, miR-200b and miR-200c. Furthermore, we demonstrated that miR-345 and miR-7 target the human multidrug resistance-associated protein 1. These results suggest that dysregulated miRNA expression may underlie the abnormal functioning of critical cellular processes associated with the cisplatin-resistant phenotype.
- Published
- 2010
- Full Text
- View/download PDF
28. Difference in expression of hepatic microRNAs miR-29c, miR-34a, miR-155, and miR-200b is associated with strain-specific susceptibility to dietary nonalcoholic steatohepatitis in mice.
- Author
-
Pogribny IP, Starlard-Davenport A, Tryndyak VP, Han T, Ross SA, Rusyn I, and Beland FA
- Subjects
- Animals, Chronic Disease, Diet, Disease Models, Animal, Fatty Liver chemically induced, Fatty Liver pathology, Gene Expression Regulation, Lipogenesis, Liver pathology, Male, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, MicroRNAs genetics, Species Specificity, Disease Susceptibility, Fatty Liver genetics, Liver metabolism, MicroRNAs biosynthesis
- Abstract
The importance of dysregulation of microRNA (miRNA) expression in nonalcoholic steatohepatitis (NASH) has been increasingly recognized; however, the association between altered expression of miRNAs and pathophysiological features of NASH and whether there is a connection between susceptibility to NASH and altered expression of miRNAs are largely unknown. In this study, male inbred C57BL/6J and DBA/2J mice were fed a lipogenic methyl-deficient diet that causes liver injury similar to human NASH, and the expression of miRNAs and the level of proteins targeted by these miRNAs in the livers were determined. Administration of the methyl-deficient diet triggered NASH-specific changes in the livers of C57BL/6J and DBA/2J mice, with the magnitude being more severe in DBA/2J mice. This was evidenced by a greater extent of expression of fibrosis-related genes in the livers of methyl-deficient DBA/2J mice. The development of NASH was accompanied by prominent changes in the expression of miRNAs, including miR-29c, miR-34a, miR-155, and miR-200b. Interestingly, changes in the expression of these miRNAs and protein levels of their targets, including Cebp-β, Socs 1, Zeb-1, and E-cadherin, in the livers of DBA/2J mice fed a methyl-deficient diet were more pronounced as compared with those in C57BL/6J mice. These results show that alterations in the expression of miRNAs are a prominent event during development of NASH induced by methyl deficiency and strongly suggest that severity of NASH and susceptibility to NASH may be determined by variations in miRNA expression response. More important, our data provide a mechanistic link between alterations in miRNA expression and pathophysiological and pathomorphological features of NASH.
- Published
- 2010
- Full Text
- View/download PDF
29. E-cadherin transcriptional down-regulation by epigenetic and microRNA-200 family alterations is related to mesenchymal and drug-resistant phenotypes in human breast cancer cells.
- Author
-
Tryndyak VP, Beland FA, and Pogribny IP
- Subjects
- Blotting, Western, Breast Neoplasms enzymology, Breast Neoplasms physiopathology, Cadherins deficiency, Cell Line, Tumor, DNA Methylation genetics, DNA Primers, DNA, Neoplasm genetics, Down-Regulation, Female, Gene Expression Regulation, Neoplastic, Histone Deacetylases genetics, Homeodomain Proteins genetics, Humans, Phenotype, Promoter Regions, Genetic, Reverse Transcriptase Polymerase Chain Reaction, Transcription Factors genetics, Transfection, Tumor Cells, Cultured, Zinc Finger E-box-Binding Homeobox 1, Breast Neoplasms genetics, Cadherins genetics, MicroRNAs genetics, Neoplasm Invasiveness genetics, RNA, Neoplasm genetics, Transcription, Genetic
- Abstract
The conversion of early stage tumors into invasive malignancies with an aggressive phenotype has been associated with the irreversible loss of E-cadherin expression. The loss of E-cadherin expression in human tumors, including breast cancer, has been attributed to promoter CpG island hypermethylation and direct inhibition by transcriptional repressors. Recent evidence demonstrates that up-regulation of E-cadherin by microRNA-200b (miR-200b) and miR-200c through direct targeting of transcriptional repressors of E-cadherin, ZEB1, and ZEB2, inhibits epithelial-to-mesenchymal transition (EMT), a crucial process in the tumor progression. We demonstrate that microRNA miR-200 family-mediated transcriptional up-regulation of E-cadherin in mesenchymal MDA-MB-231 and BT-549 cells is associated directly with translational repression of ZEB1 and indirectly with increased acetylation of histone H3 at the E-cadherin promoter. The increase in histone H3 acetylation may be attributed to the disruption of repressive complexes between ZEB1 and histone deacetylases and to the inhibition of SIRT1, a class III histone deacetylase. These events inhibit EMT and reactivate a less aggressive epithelial phenotype in cancer cells. Additionally, disruption of ZEB1-histone deacetylase repressor complexes and down-regulation of SIRT1 histone deacetylase up-regulate proapoptotic genes in the p53 apoptotic pathway resulting in the increased sensitivity of cancer cells to the chemotherapeutic agent doxorubicin.
- Published
- 2010
- Full Text
- View/download PDF
30. Mechanisms of epigenetic silencing of the Rassf1a gene during estrogen-induced breast carcinogenesis in ACI rats.
- Author
-
Starlard-Davenport A, Tryndyak VP, James SR, Karpf AR, Latendresse JR, Beland FA, and Pogribny IP
- Subjects
- Animals, CpG Islands, DNA Methylation, Exons genetics, Female, Histone Methyltransferases, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Hyperplasia, Mammary Glands, Animal drug effects, Mammary Glands, Animal metabolism, Mammary Glands, Animal pathology, Mammary Neoplasms, Experimental chemically induced, Neoplasm Proteins metabolism, Precancerous Conditions genetics, Precancerous Conditions metabolism, Promoter Regions, Genetic genetics, Random Allocation, Rats, Rats, Inbred ACI, Tumor Suppressor Proteins biosynthesis, Tumor Suppressor Proteins genetics, Cell Transformation, Neoplastic genetics, Estradiol toxicity, Gene Expression Regulation, Neoplastic, Gene Silencing, Genes, Tumor Suppressor, Mammary Neoplasms, Experimental genetics
- Abstract
Breast cancer, the most common malignancy in women, emerges through a multistep process, encompassing the progressive sequential evolution of morphologically distinct stages from a normal cell to hyperplasia (with and without atypia), carcinoma in situ, invasive carcinoma and metastasis. The success of treatment of breast cancer could be greatly improved by the detection at early stages of cancer. In the present study, we investigated the underlying molecular mechanisms involved in breast carcinogenesis in Augustus and Copenhagen-Irish female rats, a cross between the ACI strains, induced by continuous exposure to 17beta-estradiol. The results of our study demonstrate that early stages of estrogen-induced breast carcinogenesis are characterized by altered global DNA methylation, aberrant expression of proteins responsible for the proper maintenance of DNA methylation pattern and epigenetic silencing of the critical Rassf1a (Ras-association domain family 1, isoform A) tumor suppressor gene. Interestingly, transcriptional repression of the Rassf1a gene in mammary glands during early stages of breast carcinogenesis was associated with an increase in trimethylation of histones H3 lysine 9 and H3 lysine 27 and de novo CpG island methylation and at the Rassf1a promoter and first exon. In conclusion, we demonstrate that epigenetic alterations precede formation of preneoplastic lesions indicating the significance of epigenetic events in induction of oncogenic pathways in early stages of carcinogenesis.
- Published
- 2010
- Full Text
- View/download PDF
31. Hepatic epigenetic phenotype predetermines individual susceptibility to hepatic steatosis in mice fed a lipogenic methyl-deficient diet.
- Author
-
Pogribny IP, Tryndyak VP, Bagnyukova TV, Melnyk S, Montgomery B, Ross SA, Latendresse JR, Rusyn I, and Beland FA
- Subjects
- Animals, Choline Deficiency complications, DNA Methylation, Disease Susceptibility, Fatty Liver genetics, Fatty Liver pathology, Folic Acid Deficiency complications, Histones metabolism, Liver pathology, Male, Methionine deficiency, Mice, Mice, Inbred Strains, Phenotype, Species Specificity, Epigenesis, Genetic, Fatty Liver etiology, Liver metabolism
- Abstract
Background/aims: The importance of epigenetic changes in etiology and pathogenesis of disease has been increasingly recognized. However, the role of epigenetic alterations in the genesis of hepatic steatosis and cause of individual susceptibilities to this pathological state are largely unknown., Methods: Male inbred C57BL/6J and DBA/2J mice were fed a lipogenic methyl-deficient diet (MDD) that causes liver injury similar to human non-alcoholic steatohepatitis (NASH) for 6, 12, or 18 weeks, and the status of global and repetitive elements cytosine methylation, histone modifications, and expression of proteins responsible for those epigenetic modifications in livers was determined., Results: The development of hepatic steatosis in inbred C57BL/6J and DBA/2J mice was accompanied by prominent epigenetic abnormalities. This was evidenced by pronounced loss of genomic and repetitive sequences cytosine methylation, especially at major and minor satellites, accompanied by increased levels of repeat-associated transcripts, aberrant histone modifications, and alterations in expression of the maintenance DNA methyltransferase 1 (DNMT1) and de novo DNMT3A proteins in the livers of both mouse strains. However, the DBA/2J mice, which were characterized by an initially lower degree of methylation of repetitive elements and lower extent of histone H3 lysine 9 (H3K9) and H3 lysine 27 (H3K27) trimethylation in the normal livers, as compared to those in the C57BL/6J mice, developed more prominent NASH-specific pathomorphological changes., Conclusions: These results mechanistically link epigenetic alterations to the pathogenesis of hepatic steatosis and strongly suggest that differences in the cellular epigenetic status may be a predetermining factor to individual susceptibilities to hepatic steatosis.
- Published
- 2009
- Full Text
- View/download PDF
32. Down-regulation of the microRNAs miR-34a, miR-127, and miR-200b in rat liver during hepatocarcinogenesis induced by a methyl-deficient diet.
- Author
-
Tryndyak VP, Ross SA, Beland FA, and Pogribny IP
- Subjects
- Animals, Apoptosis, Blotting, Western, Liver Neoplasms, Experimental etiology, Liver Neoplasms, Experimental pathology, Male, Rats, Rats, Inbred F344, Reverse Transcriptase Polymerase Chain Reaction, Diet, Down-Regulation, Liver Neoplasms, Experimental genetics, MicroRNAs genetics
- Abstract
Altered expression of microRNAs (miRNAs) has been reported in diverse human cancers; however, the down-regulation or up-regulation of any particular miRNAs in cancer is not sufficient to address the role of these changes in carcinogenesis. In this study, using the rat model of liver carcinogenesis induced by a methyl-deficient diet, which is relevant to the hepatocarcinogenesis in humans associated with viral hepatitis C and B infections, alcohol exposure and metabolic liver diseases, we showed that the development of hepatocellular carcinoma (HCC) is characterized by prominent early changes in expression of miRNA genes, specifically by inhibition of expression of microRNAs miR-34a, miR-127, miR-200b, and miR-16a involved in the regulation of apoptosis, cell proliferation, cell-to-cell connection, and epithelial-mesenchymal transition. The mechanistic link between these alterations in miRNAs expression and the development of HCC was confirmed by the corresponding changes in the levels of E2F3, NOTCH1, BCL6, ZFHX1B, and BCL2 proteins targeted by these miRNAs. The significance of miRNAs expression dysregulation in respect to hepatocarcinogenesis was confirmed by the persistence of these miRNAs alterations in the livers of methyl-deficient rats re-fed a methyl-adequate diet. Altogether, the early occurrence of alterations in miRNAs expression and their persistence during the entire process of hepatocarcinogenesis indicate that the dysregulation of microRNAs expression may be an important contributing factor in the development of HCC.
- Published
- 2009
- Full Text
- View/download PDF
33. The tumor-promoting activity of 2-acetylaminofluorene is associated with disruption of the p53 signaling pathway and the balance between apoptosis and cell proliferation.
- Author
-
Pogribny IP, Muskhelishvili L, Tryndyak VP, and Beland FA
- Subjects
- 2-Acetylaminofluorene administration & dosage, Animals, Apoptosis physiology, Carcinogens administration & dosage, Liver Neoplasms, Experimental chemically induced, Liver Neoplasms, Experimental metabolism, Liver Neoplasms, Experimental pathology, Male, MicroRNAs genetics, Rats, Rats, Sprague-Dawley, Signal Transduction physiology, Tumor Suppressor Protein p53 metabolism, 2-Acetylaminofluorene toxicity, Apoptosis drug effects, Carcinogens toxicity, Cell Proliferation drug effects, Signal Transduction drug effects, Tumor Suppressor Protein p53 antagonists & inhibitors
- Abstract
The aromatic amine 2-acetylaminofluore (2-AAF) is a powerful complete genotoxic rat liver carcinogen that induces tumors without any additional interventions. While the tumor-initiating genotoxic activity of 2-AAF is well established, its tumor-promotion activity is far less understood. It is believed that the tumor-promoting property of 2-AAF is associated with selective enhancement of cell replication and sustained suppression of apoptosis in initiated cells. In the present study, we investigated the underlying mechanisms of tumor-promoting events induced by 2-AAF-exposure. Male Sprague-Dawley rats were fed NIH-31 diet containing 0.02% of 2-AAF for 12 and 24 weeks, and the expression pattern of genes associated with the p53-signaling pathway and microRNA genes was determined in the livers of control and 2-AAF-fed rats. The results indicate that the tumor-promoting property of 2-AAF during hepatocarcinogenesis is associated predominantly with the up-regulation of anti-apoptotic growth-related genes and down-regulation of expression of pro-apoptotic genes. This disrupts the balance between cell proliferation and apoptosis, which leads to consequential unrestricted cell proliferation, especially of initiated cells. Also, the long-term-administration of 2-AAF resulted in disruption of regulatory miR-34a-p53 feed-back loop that mediates apoptosis. This was evidenced by an increased expression of miR-34a in response to genotoxic effects of 2-AAF in the absence of p53 up-regulation, and loss of regulatory control of mir-34a on SIRT1 function. Additionally, the livers of 2-AAF-exposed rats were characterized by the substantial deregulation of expression of miR-18, miR-21, miR-182, and miR-200 family, microRNAs involved in control of apoptosis/cell proliferation and cell-cell contact pathways, two major pathways disrupted during the promotion stage of hepatocarcinogenesis.
- Published
- 2009
- Full Text
- View/download PDF
34. Epigenetic alterations in the brains of Fisher 344 rats induced by long-term administration of folate/methyl-deficient diet.
- Author
-
Pogribny IP, Karpf AR, James SR, Melnyk S, Han T, and Tryndyak VP
- Subjects
- Animals, DNA Modification Methylases classification, DNA Modification Methylases genetics, DNA Modification Methylases metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Homocysteine metabolism, Male, Methionine deficiency, Proliferating Cell Nuclear Antigen genetics, Proliferating Cell Nuclear Antigen metabolism, Rats, Rats, Inbred F344, Regulatory Factor X Transcription Factors, S-Adenosylhomocysteine metabolism, S-Adenosylmethionine metabolism, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, Brain metabolism, Diet, Reducing adverse effects, Epigenesis, Genetic physiology, Folic Acid Deficiency etiology, Folic Acid Deficiency pathology
- Abstract
The maintenance of the cellular epigenomic landscape, which depends on the status of the one-carbon metabolic pathway, is essential for normal central nervous system development and function. In the present study, we examined the epigenetic alterations in the brains of Fisher 344 rats induced by the long-term administration of a diet lacking of essential one-carbon nutrients, methionine, choline, and folic acid. The results demonstrated that feeding a folate/methyl-deficient diet causes global DNA hypermethylation as indicated by an increase of genomic 5-methyl-2'-deoxycytidine (5mdC) content and more importantly, by an increase of methylation within unmethylated CpG-rich DNA domains. Interestingly, these epigenetic changes were opposite to those observed in the livers of the same folate/methyl-deficient rats. The hypermethylation changes were associated with an increased protein expression of de novo DNA methyltransferase DNMT3a and methyl-CpG-binding protein 2. Additionally, the gene expression profiling identified 33 significantly up- or down-regulated genes (fold change > or =1.5 and p< or =0.05) in the brains of rats fed a folate/methyl-deficient diet for 36 weeks. Interestingly, we detected an up-regulation of regulatory factor X, 3 (Rfx3) gene, a sequence-specific DNA-binding protein, that mediates the transcriptional activation of silenced by methylation genes, which may be an adaptive protective brain response to hypermethylation. Together, these data suggest that the proper maintenance of the epigenomic landscape in normal brain depends on the adequate supply of essential nutrients involved in the metabolism of methyl groups.
- Published
- 2008
- Full Text
- View/download PDF
35. Induction of oxidative stress and DNA damage in rat brain by a folate/methyl-deficient diet.
- Author
-
Bagnyukova TV, Powell CL, Pavliv O, Tryndyak VP, and Pogribny IP
- Subjects
- Animals, Antioxidants metabolism, Apoptosis physiology, Diet, Reducing adverse effects, Folic Acid Deficiency physiopathology, Gene Expression Regulation physiology, Glutathione metabolism, Male, Rats, Rats, Inbred F344, Reactive Oxygen Species metabolism, Superoxide Dismutase metabolism, Time Factors, Brain metabolism, Brain physiopathology, DNA Damage physiology, Folic Acid Deficiency pathology, Oxidative Stress physiology
- Abstract
The age-associated decline in cellular antioxidant defenses and resultant accumulation of DNA damage in central nervous system has been mechanistically implicated in the etiology and pathogenesis of neurodegenerative diseases. Neurons possess a high metabolic activity and are especially vulnerable to the long-term effects of continuous exposure to endogenous reactive oxygen species. It is well recognized that adequate availability of essential nutrients involved in cellular one-carbon metabolism is essential for normal brain development and function. Additionally, the synthesis of the primary low-molecular cellular antioxidant glutathione is inter-dependently linked to one-carbon metabolic pathway. Thus, any aberrant disruptions in one-carbon metabolism can result in potentially deleterious effects including cell death as a result of an imbalance in the cellular redox state. Hence, in the present study, we examined the long-term effects of a folate/methyl-deficient (FMD) diet on cellular antioxidant defenses and DNA damage in the rat brain. Feeding male Fisher 344 rats a FMD diet resulted in perturbations in the levels of one-carbon metabolites along with induction of oxidative stress and oxidative DNA damage in the brain. This was evidenced by a decrease in the reduced oxidized/glutathione ratio, imbalance of cellular antioxidant defense system; specifically, altered activity and expression of antioxidant enzymes Mn-containing superoxide dismutase (Mn-SOD), catalase, and glutathione peroxidase (GPX), increased accumulation of oxidative DNA lesions, 8-hydroxydeoxyguanosine (8-OH-dG) and DNA single-strand breaks, even in the presence of increased expression of critical DNA repair genes apurinic/apyrimidinic endonuclease 1 (Apex1) and DNA polymerase beta (Polbeta), and apoptosis in the brains of folate/methyl-deficient rats. These results indicate that chronic methyl group deficiency leads to an imbalance in cellular antioxidant defense systems, increased oxidative stress, and apoptosis. Any of these events may compromise normal central nervous system function and contribute to the development of various neurological, behavioral, and neurocognitive dysfunctions.
- Published
- 2008
- Full Text
- View/download PDF
36. Epigenetic downregulation of the suppressor of cytokine signaling 1 (Socs1) gene is associated with the STAT3 activation and development of hepatocellular carcinoma induced by methyl-deficiency in rats.
- Author
-
Bagnyukova TV, Tryndyak VP, Muskhelishvili L, Ross SA, Beland FA, and Pogribny IP
- Subjects
- Animals, Diet, Down-Regulation, Gene Expression Regulation, Humans, Liver metabolism, Liver pathology, Male, Methylation, Platelet-Derived Growth Factor genetics, Platelet-Derived Growth Factor metabolism, Promoter Regions, Genetic, Random Allocation, Rats, Rats, Inbred F344, Receptors, Platelet-Derived Growth Factor genetics, Receptors, Platelet-Derived Growth Factor metabolism, STAT3 Transcription Factor genetics, Signal Transduction physiology, Suppressor of Cytokine Signaling 1 Protein, Suppressor of Cytokine Signaling Proteins genetics, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Carcinoma, Hepatocellular metabolism, Epigenesis, Genetic, Liver Neoplasms metabolism, Methionine deficiency, STAT3 Transcription Factor metabolism, Suppressor of Cytokine Signaling Proteins metabolism
- Abstract
The members of the platelet-derived growth factor (PDGF) and the transforming growth factor-beta (TGFbeta) pathways are important in the induction of liver fibrosis and cirrhosis; however, their role in the subsequent progression to hepatocellular carcinoma (HCC) remains elusive. Our study provides new insights into mechanisms of dysregulation of PDGFs, TGFbeta and signal transducer and activator of transcription (STAT) pathways in the pathogenesis of methyl-deficient rodent liver carcinogenesis, a remarkably relevant model to the development of HCC in humans. We demonstrated a progressive increase in the Pdgfs and TGFbeta expression in preneoplastic tissue and liver tumors indicating their promotional role in carcinogenesis, particularly in progression of liver fibrosis and cirrhosis. However, activation of the STAT3 occurred only in fully developed HCC and was associated with downregulation of the Socs1 gene. The inhibition of the Socs1 expression in HCC was associated with an increase in histone H3 lysine 9, H3 lysine 27, and H4 lysine 20 trimethylation at the Socs1 promoter, but not with promoter methylation. The results of our study suggest the following model of events in hepatocarcinogenesis: during early stages, overexpression of the Socs1 effectively inhibits TGFbeta- and PDGF-induced STAT3 activation, whereas, during the advanced stages of hepatocarcinogenesis, the Socs1 downregulation resulted in loss of its ability to attenuate the signal from the upregulated TGFbeta and PDGFs leading to oncogenic STAT3 activation and malignant cell transformation. This model illustrates that the Socs1 acts as classic tumor suppressor by preventing activation of the STAT3 and downregulation of Socs1 and consequent activation of STAT3 may be a crucial events leading to formation of HCC.
- Published
- 2008
- Full Text
- View/download PDF
37. Mechanisms of peroxisome proliferator-induced DNA hypomethylation in rat liver.
- Author
-
Pogribny IP, Tryndyak VP, Boureiko A, Melnyk S, Bagnyukova TV, Montgomery B, and Rusyn I
- Subjects
- Animals, Cell Proliferation drug effects, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases metabolism, DNA Breaks, Single-Stranded, Diethylhexyl Phthalate toxicity, Histones chemistry, Histones metabolism, Male, Methylation, Mutagens toxicity, Proliferating Cell Nuclear Antigen metabolism, Pyrimidines toxicity, Rats, Rats, Inbred F344, DNA Methylation drug effects, Liver drug effects, Liver metabolism, Peroxisome Proliferators toxicity
- Abstract
Genomic hypomethylation is a consistent finding in both human and animal tumors and mounting experimental evidence suggests a key role for epigenetic events in tumorigenesis. Furthermore, it has been suggested that early changes in DNA methylation and histone modifications may serve as sensitive predictive markers in animal testing for carcinogenic potency of environmental agents. Alterations in metabolism of methyl donors, disturbances in activity and/or expression of DNA methyltransferases, and presence of DNA single-strand breaks could contribute to the loss of cytosine methylation during carcinogenesis; however, the precise mechanisms of genomic hypomethylation induced by chemical carcinogens remain largely unknown. This study examined the mechanism of DNA hypomethylation during hepatocarcinogenesis induced by peroxisome proliferators WY-14,643 (4-chloro-6-(2,3-xylidino)-pyrimidynylthioacetic acid) and DEHP (di-(2-ethylhexyl)phthalate), agents acting through non-genotoxic mode of action. In the liver of male Fisher 344 rats exposed to WY-14,643 (0.1% (w/w), 5 months), the level of genomic hypomethylation increased by approximately 2-fold, as compared to age-matched controls, while in the DEHP group (1.2% (w/w), 5 months) DNA methylation did not change. Global DNA hypomethylation in livers from WY-14,643 group was accompanied by the accumulation of DNA single-strand breaks, increased cell proliferation, and diminished expression of DNA methyltransferase 1, while the metabolism of methyl donors was not affected. In contrast, none of these parameters changed significantly in rats fed DEHP. Since WY-14,643 is much more potent carcinogen than DEHP, we conclude that the extent of loss of DNA methylation may be related to the carcinogenic potential of the chemical agent, and that accumulation of DNA single-strand breaks coupled to the increase in cell proliferation and altered DNA methyltransferase expression may explain genomic hypomethylation during peroxisome proliferator-induced carcinogenesis.
- Published
- 2008
- Full Text
- View/download PDF
38. Differential expression of microRNAs during hepatocarcinogenesis induced by methyl deficiency in rats.
- Author
-
Pogribny IP, Tryndyak VP, Ross SA, and Beland FA
- Subjects
- Animals, Diet, Down-Regulation, Genes, Tumor Suppressor, Liver Neoplasms, Experimental etiology, Methylation, Rats, Reverse Transcriptase Polymerase Chain Reaction, Gene Expression genetics, Liver Neoplasms, Experimental genetics, MicroRNAs genetics
- Published
- 2008
- Full Text
- View/download PDF
39. Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin.
- Author
-
Kovalchuk O, Filkowski J, Meservy J, Ilnytskyy Y, Tryndyak VP, Chekhun VF, and Pogribny IP
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Argonaute Proteins, Base Sequence, Breast Neoplasms enzymology, Breast Neoplasms genetics, Cell Line, Tumor, Drug Screening Assays, Antitumor, Eukaryotic Initiation Factor-2 metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, MicroRNAs genetics, Molecular Sequence Data, Ribonuclease III metabolism, Breast Neoplasms pathology, Doxorubicin pharmacology, Drug Resistance, Neoplasm drug effects, MicroRNAs metabolism
- Abstract
Many chemotherapy regiments are successfully used to treat breast cancer; however, often breast cancer cells develop drug resistance that usually leads to a relapse and worsening of prognosis. We have shown recently that epigenetic changes such as DNA methylation and histone modifications play an important role in breast cancer cell resistance to chemotherapeutic agents. Another mechanism of gene expression control is mediated via the function of small regulatory RNA, particularly microRNA (miRNA); its role in cancer cell drug resistance still remains unexplored. In the present study, we investigated the role of miRNA in the resistance of human MCF-7 breast adenocarcinoma cells to doxorubicin (DOX). Here, we for the first time show that DOX-resistant MCF-7 cells (MCF-7/DOX) exhibit a considerable dysregulation of the miRNAome profile and altered expression of miRNA processing enzymes Dicer and Argonaute 2. The mechanistic link of miRNAome deregulation and the multidrug-resistant phenotype of MCF-7/DOX cells was evidenced by a remarkable correlation between specific miRNA expression and corresponding changes in protein levels of their targets, specifically those ones that have a documented role in cancer drug resistance. Furthermore, we show that microRNA-451 regulates the expression of multidrug resistance 1 gene. More importantly, transfection of the MCF-7/DOX-resistant cells with microRNA-451 resulted in the increased sensitivity of cells to DOX, indicating that correction of altered expression of miRNA may have significant implications for therapeutic strategies aiming to overcome cancer cell resistance.
- Published
- 2008
- Full Text
- View/download PDF
40. Genetic and epigenetic changes in rat preneoplastic liver tissue induced by 2-acetylaminofluorene.
- Author
-
Bagnyukova TV, Tryndyak VP, Montgomery B, Churchwell MI, Karpf AR, James SR, Muskhelishvili L, Beland FA, and Pogribny IP
- Subjects
- Animals, Chromatography, High Pressure Liquid, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases metabolism, DNA Adducts metabolism, Female, Immunohistochemistry, Kidney metabolism, Liver metabolism, Liver Neoplasms, Experimental chemically induced, Liver Neoplasms, Experimental metabolism, Male, Polymerase Chain Reaction, Precancerous Conditions chemically induced, Precancerous Conditions metabolism, Proliferating Cell Nuclear Antigen metabolism, Rats, Rats, Sprague-Dawley, Tandem Mass Spectrometry, 2-Acetylaminofluorene toxicity, Carcinogens toxicity, Epigenesis, Genetic, Liver Neoplasms, Experimental genetics, Precancerous Conditions genetics
- Abstract
Genotoxic carcinogens, including 2-acetylaminofluorene (2-AAF), in addition to exerting their genotoxic effects, often cause a variety of non-genotoxic alterations in cells. It is believed that these non-genotoxic effects may be indispensable events in tumorigenesis; however, there is insufficient knowledge to clarify the role of carcinogens in both the genetic and epigenetic changes in premalignant tissues and a lack of conclusive information on the link between epigenetic alterations and carcinogenic exposure. In the current study, we investigated whether or not the mechanism of 2-AAF-induced hepatocarcinogenesis consists of both genotoxic (genetic) and non-genotoxic (epigenetic) alterations. Male and female Sprague-Dawley rats were fed NIH-31 diet containing 0.02% of 2-AAF for 6, 12, 18 or 24 weeks. The levels of DNA adducts obtained from 2-AAF in liver and kidney tissues were assessed by high-performance liquid chromatography combined with electrospray tandem mass spectrometry (HPLC-ES-MS/MS). N-(Deoxyguanosine-8-yl)-2-aminofluorene was the major adduct detected at all time points in both tissues. Global DNA methylation in the livers and kidneys, as determined by an HpaII-based cytosine extension assay and by HPLC-ES-MS/MS, did not change over the 24-week period. In the livers of male rats, there was a progressive decrease of global and long interspersed nucleotide element-1-associated histone H4 lysine 20 trimethylation, as well as hypermethylation of the p16(INK4A) gene. These epigenetic changes were not observed in the livers of female rats or the kidneys of both sexes. Importantly, morphological evidence of formation and progression of neoplastic process was observed in the liver of male rats only. In conclusion, we have demonstrated that exposure of rats to genotoxic hepatocarcinogen 2-AAF, in addition to formation of 2-AAF-specific DNA lesions, resulted in substantial alterations in cellular epigenetic status.
- Published
- 2008
- Full Text
- View/download PDF
41. Epigenetic effects of the continuous exposure to peroxisome proliferator WY-14,643 in mouse liver are dependent upon peroxisome proliferator activated receptor alpha.
- Author
-
Pogribny IP, Tryndyak VP, Woods CG, Witt SE, and Rusyn I
- Subjects
- Animals, Base Sequence, Carcinogens toxicity, CpG Islands drug effects, DNA Methylation drug effects, DNA Primers genetics, Epigenesis, Genetic physiology, Histones metabolism, Long Interspersed Nucleotide Elements drug effects, Male, Mice, Mice, Knockout, PPAR alpha deficiency, PPAR alpha genetics, Retroelements drug effects, Terminal Repeat Sequences drug effects, Epigenesis, Genetic drug effects, Liver drug effects, Liver metabolism, PPAR alpha metabolism, Peroxisome Proliferators toxicity, Pyrimidines toxicity
- Abstract
Peroxisome proliferators are potent rodent liver carcinogens that act via a non-genotoxic mechanism. The mode of action of these agents in rodent liver includes increased cell proliferation, decreased apoptosis, secondary oxidative stress and other events; however, it is not well understood how peroxisome proliferators are triggering the plethora of the molecular signals leading to cancer. Epigenetic changes have been implicated in the mechanism of liver carcinogenesis by a number of environmental agents. Short-term treatment with peroxisome proliferators and other non-genotoxic carcinogens leads to global and locus-specific DNA hypomethylation in mouse liver, events that were suggested to correlate with a burst of cell proliferation. In the current study, we investigated the effects of long-term exposure to a model peroxisome proliferator WY-14,643 on DNA and histone methylation. Male SV129mice were fed a control or WY-14,643-containing (1000ppm) diet for one week, five weeks or five months. Treatment with WY-14,643 led to progressive global hypomethylation of liver DNA as determined by an HpaII-based cytosine extension assay with the maximum effect reaching over 200% at five months. Likewise, trimethylation of histone H4 lysine 20 and H3 lysine 9 was significantly decreased at all time points. The majority of cytosine methylation in mammals resides in repetitive DNA sequences. In view of this, we measured the effect of WY-14,643 on the methylation status of major and minor satellites, as well as in IAP, LINE1 and LINE2 elements in liver DNA. Exposure to WY-14,643 resulted in a gradual loss of cytosine methylation in major and minor satellites, IAP, LINE1 and LINE2 elements. The epigenetic changes correlated with the temporal effects of WY-14,643 on cell proliferation rates in liver, but no sustained effect on c-Myc promoter methylation was observed. Finally, WY-14,643 had no effect on DNA and histone methylation status in Pparalpha-null mice at any of the time points considered in this study. These data indicate the importance of epigenetic alterations in the mechanism of action of peroxisome proliferators and the key role of Pparalpha.
- Published
- 2007
- Full Text
- View/download PDF
42. Gene expression profiling reveals underlying molecular mechanisms of the early stages of tamoxifen-induced rat hepatocarcinogenesis.
- Author
-
Pogribny IP, Bagnyukova TV, Tryndyak VP, Muskhelishvili L, Rodriguez-Juarez R, Kovalchuk O, Han T, Fuscoe JC, Ross SA, and Beland FA
- Subjects
- Animals, Apoptosis drug effects, Carcinogens metabolism, Carcinogens toxicity, Cell Cycle drug effects, Cell Proliferation drug effects, Estrogen Antagonists metabolism, Female, G1 Phase drug effects, G1 Phase genetics, Gene Expression Profiling, Lipid Metabolism drug effects, Liver drug effects, Liver pathology, Oligonucleotide Array Sequence Analysis, Rats, Rats, Inbred F344, S Phase drug effects, S Phase genetics, Signal Transduction drug effects, Tamoxifen metabolism, Time Factors, Estrogen Antagonists toxicity, Gene Expression Regulation, Neoplastic drug effects, Liver Neoplasms etiology, Tamoxifen toxicity
- Abstract
Tamoxifen is a widely used anti-estrogenic drug for chemotherapy and, more recently, for the chemoprevention of breast cancer. Despite the indisputable benefits of tamoxifen in preventing the occurrence and re-occurrence of breast cancer, the use of tamoxifen has been shown to induce non-alcoholic steatohepatitis, which is a life-threatening fatty liver disease with a risk of progression to cirrhosis and hepatocellular carcinoma. In recent years, the high-throughput microarray technology for large-scale analysis of gene expression has become a powerful tool for increasing the understanding of the molecular mechanisms of carcinogenesis and for identifying new biomarkers with diagnostic and predictive values. In the present study, we used the high-throughput microarray technology to determine the gene expression profiles in the liver during early stages of tamoxifen-induced rat hepatocarcinogenesis. Female Fisher 344 rats were fed a 420 ppm tamoxifen containing diet for 12 or 24 weeks, and gene expression profiles were determined in liver of control and tamoxifen-exposed rats. The results indicate that early stages of tamoxifen-induced liver carcinogenesis are characterized by alterations in several major cellular pathways, specifically those involved in the tamoxifen metabolism, lipid metabolism, cell cycle signaling, and apoptosis/cell proliferation control. One of the most prominent changes during early stages of tamoxifen-induced hepatocarcinogenesis is dysregulation of signaling pathways in cell cycle progression from the G(1) to S phase, evidenced by the progressive and sustained increase in expression of the Pdgfc, Calb3, Ets1, and Ccnd1 genes accompanied by the elevated level of the PI3K, p-PI3K, Akt1/2, Akt3, and cyclin B, D1, and D3 proteins. The early appearance of these alterations suggests their importance in the mechanism of neoplastic cell transformation induced by tamoxifen.
- Published
- 2007
- Full Text
- View/download PDF
43. Estrogen-induced rat breast carcinogenesis is characterized by alterations in DNA methylation, histone modifications and aberrant microRNA expression.
- Author
-
Kovalchuk O, Tryndyak VP, Montgomery B, Boyko A, Kutanzi K, Zemp F, Warbritton AR, Latendresse JR, Kovalchuk I, Beland FA, and Pogribny IP
- Subjects
- Animals, Blotting, Western, Cell Transformation, Neoplastic chemically induced, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, DNA (Cytosine-5-)-Methyltransferases metabolism, Electrophoresis, Polyacrylamide Gel, Estradiol toxicity, Female, Gene Expression Regulation drug effects, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental metabolism, Methylation drug effects, Rats, DNA Methylation drug effects, Estrogens toxicity, Histones metabolism, Mammary Neoplasms, Experimental chemically induced, MicroRNAs genetics
- Abstract
Breast cancer is the most common malignancy in women continuing to rise worldwide. Breast cancer emerges through a multi-step process, encompassing progressive changes from a normal cell to hyperplasia (with and without atypia), carcinoma in situ, invasive carcinoma, and metastasis. In the current study, we analyzed the morphological changes and alterations of DNA methylation, histone methylation and microRNA expression during estradiol-17beta (E(2))-induced mammary carcinogenesis in female August Copenhagen Irish (ACI) rats. E(2)-induced breast carcinogenesis in ACI rats provides a physiologically relevant and genetically defined animal model for studying human sporadic breast cancer. The pattern of morphological changes in mammary glands during E(2)-induced carcinogenesis was characterized by transition from normal appearing alveolar and ductular hyperplasia to focal hyperplastic areas of atypical glands and ducts accompanied by a rapid and sustained loss of global DNA methylation, LINE-1 hypomethylation, loss of histone H3 lysine 9 and histone H4 lysine 20 trimethylation, and altered microRNAs expression. More importantly, these alterations in the mammary tissue occurred after six weeks of E(2)-treatment, whereas the atypical hyperplasia, which represents a putative precursor lesion to mammary carcinoma in this model, was detected only after twelve weeks of exposure, demonstrating clearly that these events are directly associated with the effects of E(2) and are not a consequence of the preexisting preneoplastic lesions. The results of this study show that deregulation of cellular epigenetic processes plays a crucial role in the mechanism of E(2)-induced mammary carcinogenesis in ACI rats, especially in the tumor initiation process.
- Published
- 2007
- Full Text
- View/download PDF
44. Role of epigenetic effectors in maintenance of the long-term persistent bystander effect in spleen in vivo.
- Author
-
Koturbash I, Boyko A, Rodriguez-Juarez R, McDonald RJ, Tryndyak VP, Kovalchuk I, Pogribny IP, and Kovalchuk O
- Subjects
- Animals, Brain physiology, Brain radiation effects, DNA Methyltransferase 3A, Humans, Male, Models, Animal, Rats, Rats, Long-Evans, Time Factors, Whole-Body Irradiation, Bystander Effect physiology, Bystander Effect radiation effects, Epigenesis, Genetic radiation effects, Spleen physiology, Spleen radiation effects
- Abstract
Radiation therapy is a primary treatment modality for brain tumors, yet it has been linked to the increased incidence of secondary, post-radiation therapy cancers. These cancers are thought to be linked to indirect radiation-induced bystander effect. Bystander effect occurs when irradiated cells communicate damage to nearby, non-irradiated 'bystander' cells, ultimately contributing to genome destabilization in the non-exposed cells. Recent evidence suggests that bystander effect may be epigenetic in nature; however, characterization of epigenetic mechanisms involved in bystander effect generation and its long-term persistence has yet to be defined. To investigate the possibility that localized X-ray irradiation induces persistent bystander effects in distant tissue, we monitored the induction of epigenetic changes (i.e. alterations in DNA methylation, histone methylation and microRNA (miRNA) expression) in the rat spleen tissue 24 h and 7 months after localized cranial exposure to 20 Gy of X-rays. We found that localized cranial radiation exposure led to the induction of bystander effect in lead-shielded, distant spleen tissue. Specifically, this exposure caused the profound epigenetic dysregulation in the bystander spleen tissue that manifested as a significant loss of global DNA methylation, alterations in methylation of long interspersed nucleotide element-1 (LINE-1) retrotransposable elements and down-regulation of DNA methyltransferases and methyl-binding protein methyl CpG binding protein 2 (MeCP2). Further, irradiation significantly altered expression of miR-194, a miRNA putatively targeting both DNA methyltransferase-3a and MeCP2. This study is the first to report conclusive evidence of the long-term persistence of bystander effects in radiation carcinogenesis target organ (spleen) upon localized distant exposure using the doses comparable with those used for clinical brain tumor treatments.
- Published
- 2007
- Full Text
- View/download PDF
45. Induction of microRNAome deregulation in rat liver by long-term tamoxifen exposure.
- Author
-
Pogribny IP, Tryndyak VP, Boyko A, Rodriguez-Juarez R, Beland FA, and Kovalchuk O
- Subjects
- Animals, Antineoplastic Agents, Hormonal administration & dosage, Antineoplastic Agents, Hormonal toxicity, Apoptosis drug effects, Apoptosis genetics, Carcinogens administration & dosage, Carcinogens toxicity, Cell Cycle drug effects, Cell Cycle genetics, DNA Replication drug effects, DNA Replication genetics, Female, Gene Expression Profiling, Liver cytology, Liver Neoplasms, Experimental chemically induced, Liver Neoplasms, Experimental genetics, Oligonucleotide Array Sequence Analysis, Rats, Rats, Inbred F344, Tamoxifen administration & dosage, Liver drug effects, Liver metabolism, MicroRNAs genetics, MicroRNAs metabolism, Tamoxifen toxicity
- Abstract
Micro RNAs (miRNAs) are small non-coding RNA molecules that function as negative regulators of gene expression. They play a crucial role in the regulation of genes involved in the control of development, cell proliferation, apoptosis, and stress response. Although miRNA levels are substantially altered in tumors, their role in carcinogenesis, specifically at the early pre-cancerous stages, has not been established. Here we report that exposure of Fisher 344 rats to tamoxifen, a potent hepatocarcinogen in rats, for 24 weeks leads to substantial changes in the expression of miRNA genes in the liver. We noted a significant up-regulation of known oncogenic miRNAs, such as the 17-92 cluster, miR-106a, and miR-34. Furthermore, we confirmed the corresponding changes in the expression of proteins targeted by these miRNAs, which include important cell cycle regulators, chromatin modifiers, and expression regulators implicated in carcinogenesis. All these miRNA changes correspond to previously reported alterations in full-fledged tumors, including hepatocellular carcinomas. Thus, our findings indicate that miRNA changes occur prior to tumor formation and are not merely a consequence of a transformed state.
- Published
- 2007
- Full Text
- View/download PDF
46. Epigenetic profiling of multidrug-resistant human MCF-7 breast adenocarcinoma cells reveals novel hyper- and hypomethylated targets.
- Author
-
Chekhun VF, Lukyanova NY, Kovalchuk O, Tryndyak VP, and Pogribny IP
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma metabolism, Antibiotics, Antineoplastic pharmacology, Antineoplastic Agents pharmacology, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cisplatin pharmacology, Doxorubicin pharmacology, Gene Expression Regulation, Neoplastic drug effects, Histone Methyltransferases, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Humans, Protein Glutamine gamma Glutamyltransferase 2, Protein Methyltransferases, Tumor Cells, Cultured drug effects, Adenocarcinoma genetics, Breast Neoplasms genetics, DNA Methylation drug effects, Drug Resistance, Multiple genetics, Drug Resistance, Neoplasm, Epigenesis, Genetic
- Abstract
The successful treatment of cancer requires a clear understanding of multiple interacting factors involved in the development of drug resistance. Presently, two hypotheses, genetic and epigenetic, have been proposed to explain mechanisms of acquired cancer drug resistance. In the present study, we examined the alterations in epigenetic mechanisms in the drug-resistant MCF-7 human breast cancer cells induced by doxorubicin (DOX) and cisplatin (cisDDP), two chemotherapeutic drugs with different modes of action. Despite this difference, both of the drug-resistant cell lines displayed similar pronounced changes in the global epigenetic landscape showing loss of global DNA methylation, loss of histone H4 lysine 20 trimethylation, increased phosporylation of histone H3 serine 10, and diminished expression of Suv4-20h2 histone methyltransferase compared with parental MCF-7 cells. In addition to global epigenetic changes, the MCF-7/DOX and MCF-7/cisDDP drug-resistant cells are characterized by extensive alterations in region-specific DNA methylation, as indicated by the appearance of the number of differentially methylated DNA genes. A detailed analysis of hypo- and hypermethylated DNA sequences revealed that the acquisition of drug-resistant phenotype of MCF-7 cells to DOX and cisDDP, in addition to specific alterations induced by a particular drug only, was characterized by three major common mechanisms: dysfunction of genes involved in estrogen metabolism (sulfatase 2 and estrogen receptor alpha), apoptosis (p73, alpha-tubulin, BCL2-antagonist of cell death, tissue transglutaminase 2 and forkhead box protein K1), and cell-cell contact (leptin, stromal cell-derived factor receptor 1, activin A receptor E-cadherin) and showed that two opposing hypo- and hypermethylation processes may enhance and complement each other in the disruption of these pathways. These results provided evidence that epigenetic changes are an important feature of cancer cells with acquired drug-resistant phenotype and may be a crucial contributing factor to its development. Finally, deregulation of similar pathways may explain the existence and provide mechanism of cross-resistance of cancer cells to different types of chemotherapeutic agents.
- Published
- 2007
- Full Text
- View/download PDF
47. Epigenetic reprogramming of liver cells in tamoxifen-induced rat hepatocarcinogenesis.
- Author
-
Tryndyak VP, Kovalchuk O, Muskhelishvili L, Montgomery B, Rodriguez-Juarez R, Melnyk S, Ross SA, Beland FA, and Pogribny IP
- Subjects
- Animals, Antigens, Nuclear genetics, Antigens, Nuclear metabolism, CpG Islands, DNA Methylation, DNA Polymerase beta genetics, DNA Polymerase beta metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Female, Hepatocytes metabolism, Immunoenzyme Techniques, Ku Autoantigen, Liver Neoplasms drug therapy, Liver Neoplasms pathology, Liver Neoplasms, Experimental chemically induced, Long Interspersed Nucleotide Elements genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Rats, Rats, Inbred F344, Reverse Transcriptase Polymerase Chain Reaction, Cell Transformation, Neoplastic, Epigenesis, Genetic drug effects, Estrogen Antagonists toxicity, Hepatocytes drug effects, Liver Neoplasms etiology, Liver Neoplasms, Experimental genetics, Tamoxifen toxicity
- Abstract
Tamoxifen, a nonsteroidal anti-estrogen, is a potent genotoxic hepatocarcinogen in rats, with both tumor initiating and promoting properties. Recently it has been demonstrated that genotoxic carcinogens, in addition to exerting genotoxic effects, often cause epigenetic alterations and these induced epigenetic changes may play important mechanistic role in carcinogenesis. In the present study, we investigated the role of tamoxifen-induced epigenetic changes in hepatocarcinogenic process. The results of the study showed that exposure of female F344 rats to tamoxifen resulted in progressive loss of CpG methylation in regulatory sequences of long interspersed nucleotide elements (LINE-1) and prominent increase in expression of LINE-1 elements and c-myc proto-oncogene. The accumulation of tamoxifen-induced DNA lesions was accompanied by the decreased level of Rad51, Ku70, and DNA polymerase beta (Polbeta) proteins that play a crucial role in maintenance of genomic stability. Furthermore, feeding rats with tamoxifen-containing diet led to increased regenerative cell proliferation, as indicated by the increased level of Ki-67 and proliferating cell nuclear antigen (PCNA) proteins. These data indicate that exposure of animals to genotoxic hepatocarcinogen tamoxifen led to early phenotypical alterations in livers characterized by emergence of epigenetically reprogrammed cells with a specific cancer-related epigenetic phenotype prior to tumor formation., ((c) 2006 Wiley-Liss, Inc.)
- Published
- 2007
- Full Text
- View/download PDF
48. Methyl deficiency, alterations in global histone modifications, and carcinogenesis.
- Author
-
Pogribny IP, Tryndyak VP, Muskhelishvili L, Rusyn I, and Ross SA
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Methylation, Rats, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Histones genetics, Histones metabolism
- Abstract
The methyl-deficient model of endogenous hepatocarcinogenesis in rodents is unique in that dietary omission rather than the addition of chemical carcinogens leads to tumor formation. Thus, the biochemical and molecular events predisposing to cancer in this model result from chronic metabolic stress and provide an ideal model system to study progressive alterations that occur during carcinogenesis. Moreover, epigenetic alterations imposed by this diet are believed to be 1 of the main mechanisms responsible for malignant transformation of rat liver cells. In this study we examined the changes in global histone modification patterns in liver during hepatocarcinogenesis induced by methyl deficiency. Feeding animals the methyl-deficient diet (MDD) led to progressive loss of histone H4 lysine 20 trimethylation (H4K20me3), H3 lysine 9 trimethylation (H3K9me3), and histone H3 lysine 9 (H3K9ac) and histone H4 lysine 16 (H4K16ac) acetylation. A considerable decrease of H4K20me3 and H3K9ac was also detected in liver tumors induced by MDD. In contrast, liver tumors displayed an increase in H3K9me3 and H4K16ac. To determine the possible mechanism of alterations of histone modifications, we analyzed the expression of histone-modifying enzymes in liver during hepatocarcinogenesis. The expression of Suv4-20h2 and RIZ1 histone methyltransferases (HMTs) steadily decreased along with the development of liver tumors and reached its lowest level in tumor tissue, whereas the expression of Suv39-h1 HMT and histone acetyltransferase 1 (HAT1) substantially increased in tumors. These results illustrate the complexity and importance of histone modification changes in the etiology of hepatocarcinogenesis induced by MDD.
- Published
- 2007
- Full Text
- View/download PDF
49. Effect of long-term tamoxifen exposure on genotoxic and epigenetic changes in rat liver: implications for tamoxifen-induced hepatocarcinogenesis.
- Author
-
Tryndyak VP, Muskhelishvili L, Kovalchuk O, Rodriguez-Juarez R, Montgomery B, Churchwell MI, Ross SA, Beland FA, and Pogribny IP
- Subjects
- Animals, Antineoplastic Agents, Hormonal chemistry, Chromatography, High Pressure Liquid, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases genetics, DNA (Cytosine-5-)-Methyltransferases metabolism, DNA Adducts chemistry, DNA Methyltransferase 3A, Female, Histones metabolism, Liver drug effects, Liver enzymology, Liver pathology, Rats, Rats, Inbred F344, Tamoxifen chemistry, Tissue Distribution, DNA Methyltransferase 3B, Antineoplastic Agents, Hormonal metabolism, DNA Adducts metabolism, DNA Methylation, Epigenesis, Genetic, Liver Neoplasms, Experimental etiology, Liver Neoplasms, Experimental metabolism, Tamoxifen metabolism
- Abstract
Tamoxifen is a non-steroidal anti-estrogen used for the treatment of breast cancer and, more recently, as a chemopreventive agent in healthy women at high risk of developing breast cancer. On the other hand, tamoxifen is a potent hepatocarcinogen in rats, with both tumor-initiating and tumor-promoting properties. There is substantial evidence that hepatic tumors in rats are initiated as a result of formation of tamoxifen-DNA adducts; however, events subsequent to DNA adduct formation are not clear. Recently, it has been demonstrated that genotoxic carcinogens, in addition to exerting genotoxic effects, often cause epigenetic alterations. In the current study, we investigated whether or not the mechanism of tamoxifen-induced hepatocarcinogenesis includes both genotoxic and epigenetic components. Female Fisher 344 rats were fed a 420 p.p.m. tamoxifen diet for 6, 12, 18 or 24 weeks. Hepatic tamoxifen-DNA adduct levels, as assessed by high-performance liquid chromatography and electrospray tandem mass spectrometry, were 580 adducts/10(8) nt at 6 weeks, and increased to approximately 1700 adducts/10(8) nt by 18 weeks. Global liver DNA hypomethylation, as determined by an HpaII-based cytosine extension assay, was increased at all time points, with the maximum increase (approximately 200%) occurring at 6 weeks. Protein expressions of maintenance (DNMT1) DNA methyltransferase and de novo DNA methyltransferases DNMT3a and DNMT3b were decreased at all time points. Likewise, trimethylation of histone H4 lysine 20 was significantly decreased at all time points. In contrast, non-target tissues (i.e. mammary gland, pancreas and spleen) did not show any changes in global DNA methylation or DNA methyltransferase activity. These data indicate the importance of genotoxic and epigenetic alterations in the etiology of tamoxifen-induced hepatocarcinogenesis.
- Published
- 2006
- Full Text
- View/download PDF
50. Histone H3 lysine 9 and H4 lysine 20 trimethylation and the expression of Suv4-20h2 and Suv-39h1 histone methyltransferases in hepatocarcinogenesis induced by methyl deficiency in rats.
- Author
-
Pogribny IP, Ross SA, Tryndyak VP, Pogribna M, Poirier LA, and Karpinets TV
- Subjects
- Animals, DNA Methylation, Epigenesis, Genetic, Histone Methyltransferases, Histone-Lysine N-Methyltransferase chemistry, Histones metabolism, Long Interspersed Nucleotide Elements, Male, Methylation, Methyltransferases chemistry, Mutation, Protein Methyltransferases, Rats, Rats, Inbred F344, Repressor Proteins chemistry, Histone-Lysine N-Methyltransferase biosynthesis, Histone-Lysine N-Methyltransferase metabolism, Histones chemistry, Liver Neoplasms metabolism, Methyltransferases biosynthesis, Repressor Proteins biosynthesis
- Abstract
The field of cancer epigenetics has received much attention in recent years. However, the relationship of cancer epigenetics with cancer etiology is not clear. Recent studies suggest the involvement of altered DNA methylation and histone modifications in the emergence of epigenetically reprogrammed cells with specific tumor-related phenotypes at premalignant stages of tumor development. In this study, we used a methyl-deficient model of rodent hepatocarcinogenesis to examine the roles of DNA, histone H3 lysine 9 and histone H4 lysine 20 methylation, and the level of the expression of Suv39h1 and Suv4-20h2 histone methyltransferases in the carcinogenic process. We demonstrated that the development of liver tumors was characterized by progressive demethylation of DNA repeats, decrease in histone H4 lysine 20 trimethylation, and a gradual decrease in the expression of Suv4-20h2 histone methyltransferase. A prominent increase in the trimethylation of histone H3 lysine 9 and in the expression of Suv39h1 histone methyltransferase was observed in preneoplastic nodules and liver tumors indicating the promotional role of these epigenetic alterations at later stages of carcinogenesis. The appearance of tumor-specific epigenetic alterations (demethylation of repetitive elements, loss of histone H4 lysine 20 trimethylation, altered expression of Suv4-20h2 and Suv39h1 histone methyltransferases) at preneoplastic stages of hepatocarcinogenesis provides experimental support for the epigenetic hypothesis of tumorigenesis that considers stress-induced epigenetic reprogramming of the cell as an important prerequisite to succeeding mutations.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.