1
|
Bates CA, Haber LT, Moore MM, Schoeny R, Maier A. Development of a framework for risk assessment of dietary carcinogens. Food Chem Toxicol 2023; 180:114022. [PMID: 37716495 DOI: 10.1016/j.fct.2023.114022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 08/09/2023] [Accepted: 09/01/2023] [Indexed: 09/18/2023]
Abstract
Although there are a number of guidance documents and frameworks for evaluation of carcinogenicity, none of the current methods fully reflects the state of the science. Common limitations include the absence of dose-response assessment and not considering the impact of differing exposure patterns (e.g., intermittent, high peaks vs. lower, continuous exposures). To address these issues, we have developed a framework for risk assessment of dietary carcinogens. This framework includes an enhanced approach for weight of evidence (WOE) evaluation for genetic toxicology data, with a focus on evaluating studies based on the most recent testing guidance to determine whether a chemical is a mutagen. Included alongside our framework is a discussion of resources for evaluating tissue dose and the temporal pattern of internal dose, taking into account the chemical's toxicokinetics. The framework then integrates the mode of action (MOA) and associated dose metric category with the exposure data to identify the appropriate approach(es) to low-dose extrapolation and level of concern associated with the exposure scenario. This framework provides risk managers with additional flexibility in risk management and risk communication options, beyond the binary choice of linear low-dose extrapolation vs. application of uncertainty factors.
Collapse
Affiliation(s)
| | - Lynne T Haber
- Risk Science Center, University of Cincinnati College of Medicine, USA
| | | | | | | |
Collapse
|
2
|
Legacy and Emerging Per- and Polyfluoroalkyl Substances: Analytical Techniques, Environmental Fate, and Health Effects. Int J Mol Sci 2021; 22:ijms22030995. [PMID: 33498193 PMCID: PMC7863963 DOI: 10.3390/ijms22030995] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 01/24/2023] Open
Abstract
Due to their unique chemical properties, per- and polyfluoroalkyl substances (PFAS) have been used extensively as industrial surfactants and processing aids. While several types of PFAS have been voluntarily phased out by their manufacturers, these chemicals continue to be of ecological and public health concern due to their persistence in the environment and their presence in living organisms. Moreover, while the compounds referred to as “legacy” PFAS remain in the environment, alternative compounds have emerged as replacements for their legacy predecessors and are now detected in numerous matrices. In this review, we discuss the historical uses of PFAS, recent advances in analytical techniques for analysis of these compounds, and the fate of PFAS in the environment. In addition, we evaluate current biomonitoring studies of human exposure to legacy and emerging PFAS and examine the associations of PFAS exposure with human health impacts, including cancer- and non-cancer-related outcomes. Special focus is given to short-chain perfluoroalkyl acids (PFAAs) and ether-substituted, polyfluoroalkyl alternatives including hexafluoropropylene oxide dimer acid (HFPO-DA; tradename GenX), 4,8-dioxa-3H-perfluorononanoic acid (DONA), and 6:2 chlorinated polyfluoroethersulfonic acid (6:2 Cl-PFESA; tradename F-53B).
Collapse
|
3
|
Xi Y, Zhang Y, Zhu S, Luo Y, Xu P, Huang Z. PPAR-Mediated Toxicology and Applied Pharmacology. Cells 2020; 9:cells9020352. [PMID: 32028670 PMCID: PMC7072218 DOI: 10.3390/cells9020352] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 01/26/2020] [Accepted: 01/30/2020] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs), members of the nuclear hormone receptor family, attract wide attention as promising therapeutic targets for the treatment of multiple diseases, and their target selective ligands were also intensively developed for pharmacological agents such as the approved drugs fibrates and thiazolidinediones (TZDs). Despite their potent pharmacological activities, PPARs are reported to be involved in agent- and pollutant-induced multiple organ toxicity or protective effects against toxicity. A better understanding of the protective and the detrimental role of PPARs will help to preserve efficacy of the PPAR modulators but diminish adverse effects. The present review summarizes and critiques current findings related to PPAR-mediated types of toxicity and protective effects against toxicity for a systematic understanding of PPARs in toxicology and applied pharmacology.
Collapse
Affiliation(s)
- Yue Xi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yunhui Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Sirui Zhu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuping Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence: (P.X.); (Z.H.); Tel.: +1-412-708-4694(P.X.); +86-20-39943092 (Z.H.)
| | - Zhiying Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Correspondence: (P.X.); (Z.H.); Tel.: +1-412-708-4694(P.X.); +86-20-39943092 (Z.H.)
| |
Collapse
|
4
|
Permenter MG, McDyre BC, Ippolito DL, Stallings JD. Alterations in tissue microRNA after heat stress in the conscious rat: potential biomarkers of organ-specific injury. BMC Genomics 2019; 20:141. [PMID: 30770735 PMCID: PMC6377737 DOI: 10.1186/s12864-019-5515-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/07/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Heat illness remains a significant cause of morbidity in susceptible populations. Recent research elucidating the cellular mechanism of heat stress leading to heat illness may provide information to develop better therapeutic interventions, risk assessment strategies, and early biomarkers of organ damage. microRNA (miRNA) are promising candidates for therapeutic targets and biomarkers for a variety of clinical conditions since there is the potential for high specificity for individual tissues and unique cellular functions. The objective of this study was to identify differentially expressed microRNAs and their putative mRNA targets in the heart, liver, kidney, and lung in rats at three time points: during heat stress (i.e., when core temperature reached 41.8 °C), or following a 24 or 48 h recovery period. RESULTS Rats did not show histological evidence of tissue pathology until 48 h after heat stress, with 3 out of 6 rats showing cardiac inflammation and renal proteinosis at 48 h. The three rats with cardiac and renal pathology had 86, 7, 159, and 37 differentially expressed miRNA in the heart, liver, kidney, or lung, respectively compared to non-heat stressed control animals. During heat stress one differentially expressed miRNA was found in the liver and five in the lung, with no other modulated miRNA after 24 h or 48 h in animals with no evidence of organ injury. Pathway enrichment analysis revealed enrichment in functional pathways associated with heat stress, with the greatest effects observed in animals with histological evidence of cardiac and renal damage at 48 h. Inhibiting miR-21 in cultured cardiomyocytes increased the percent apoptotic cells five hours after heat stress from 70.9 ± 0.8 to 84.8 ± 2.2%. CONCLUSIONS Global microRNA and transcriptomics analysis suggested that perturbed miRNA due to heat stress are involved in biological pathways related to organ injury, energy metabolism, the unfolded protein response, and cellular signaling. These miRNA may serve as biomarkers of organ injury and potential pharmacological targets for preventing heat illness or organ injury.
Collapse
Affiliation(s)
- Matthew G. Permenter
- Excet, Inc., Fort Detrick, MD 21702-5010 USA
- U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland, MD 21702-5010 USA
| | - Bonna C. McDyre
- Oak Ridge Institute for Science and Education, Fort Detrick, MD 21702-5010 USA
- U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland, MD 21702-5010 USA
| | - Danielle L. Ippolito
- U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland, MD 21702-5010 USA
| | - Jonathan D. Stallings
- U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland, MD 21702-5010 USA
| |
Collapse
|
5
|
de la Rosa Rodriguez MA, Sugahara G, Hooiveld GJEJ, Ishida Y, Tateno C, Kersten S. The whole transcriptome effects of the PPARα agonist fenofibrate on livers of hepatocyte humanized mice. BMC Genomics 2018; 19:443. [PMID: 29879903 PMCID: PMC5991453 DOI: 10.1186/s12864-018-4834-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/29/2018] [Indexed: 01/08/2023] Open
Abstract
Background The role of PPARα in gene regulation in mouse liver is well characterized. However, less is known about the role of PPARα in human liver. The aim of the present study was to better characterize the impact of PPARα activation on gene regulation in human liver. To that end, chimeric mice containing hepatocyte humanized livers were given an oral dose of 300 mg/kg fenofibrate daily for 4 days. Livers were collected and analyzed by hematoxilin and eosin staining, qPCR, and transcriptomics. Transcriptomics data were compared with existing datasets on PPARα activation in normal mouse liver, human primary hepatocytes, and human precision cut liver slices. Results Of the different human liver models, the gene expression profile of hepatocyte humanized livers most closely resembled actual human liver. In the hepatocyte humanized mouse livers, the human hepatocytes exhibited excessive lipid accumulation. Fenofibrate increased the size of the mouse but not human hepatocytes, and tended to reduce steatosis in the human hepatocytes. Quantitative PCR indicated that induction of PPARα targets by fenofibrate was less pronounced in the human hepatocytes than in the residual mouse hepatocytes. Transcriptomics analysis indicated that, after filtering, a total of 282 genes was significantly different between fenofibrate- and control-treated mice (P < 0.01). 123 genes were significantly lower and 159 genes significantly higher in the fenofibrate-treated mice, including many established PPARα targets such as FABP1, HADHB, HADHA, VNN1, PLIN2, ACADVL and HMGCS2. According to gene set enrichment analysis, fenofibrate upregulated interferon/cytokine signaling-related pathways in hepatocyte humanized liver, but downregulated these pathways in normal mouse liver. Also, fenofibrate downregulated pathways related to DNA synthesis in hepatocyte humanized liver but not in normal mouse liver. Conclusion The results support the major role of PPARα in regulating hepatic lipid metabolism, and underscore the more modest effect of PPARα activation on gene regulation in human liver compared to mouse liver. The data suggest that PPARα may have a suppressive effect on DNA synthesis in human liver, and a stimulatory effect on interferon/cytokine signalling.
Collapse
Affiliation(s)
- Montserrat A de la Rosa Rodriguez
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708, WE, Wageningen, The Netherlands
| | - Go Sugahara
- Research and Development Department, PhoenixBio, Co., Ltd, 3-4-1 Kagamiyama, Higashi-, Hiroshima, Japan
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708, WE, Wageningen, The Netherlands
| | - Yuji Ishida
- Research and Development Department, PhoenixBio, Co., Ltd, 3-4-1 Kagamiyama, Higashi-, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Chise Tateno
- Research and Development Department, PhoenixBio, Co., Ltd, 3-4-1 Kagamiyama, Higashi-, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Sander Kersten
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708, WE, Wageningen, The Netherlands.
| |
Collapse
|
6
|
Differential actions of PPAR-α and PPAR-β/δ on beige adipocyte formation: A study in the subcutaneous white adipose tissue of obese male mice. PLoS One 2018; 13:e0191365. [PMID: 29351550 PMCID: PMC5774787 DOI: 10.1371/journal.pone.0191365] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/03/2018] [Indexed: 12/18/2022] Open
Abstract
Background and aims Obesity compromises adipocyte physiology. PPARs are essential to adipocyte plasticity, but its isolated role in the browning phenomenon is not clear. This study aimed to examine whether activation of PPAR-α or PPAR-β/δ could induce beige cell depots in the subcutaneous white adipose tissue of diet-induced obese mice. Material and methods Sixty animals were randomly assigned to receive a control diet (C, 10% lipids) or a high-fat diet (HF, 50% lipids) for ten weeks. Then each group was re-divided to begin the treatments that lasted 4 weeks, totalizing six groups: C, C-α (C plus PPAR-α agonist, 2.5 mg/kg BM), C-β (C plus PPAR-β/δ agonist, 1 mg/kg BM), HF, HF-α (HF plus PPAR-α agonist), HF-β (HF plus PPAR-β/δ agonist). Results HF animals presented with overweight, glucose intolerance and subcutaneous white adipocyte hypertrophy. Both treatments significantly attenuated these parameters. Browning, verified by UCP1 positive beige cells and enhanced body temperature, was just observed in PPAR-α treated groups. PPAR-α agonism also elicited an enhanced gene expression of the thermogenesis effector UCP1, the beige-selective gene TMEM26 and the PRDM16, an essential gene for brown-like phenotype maintenance in the beige adipocytes when compared to their counterparts. The enhanced CIDEA and the reduced UCP1 gene levels might justify the white phenotype predominance after the treatment with the PPAR-β/δ agonist. Conclusions This work provides evidence that the PPAR-β/δ agonist ameliorated metabolic disorders through enhanced beta-oxidation and better tolerance to glucose, whereas the PPAR-α agonism was confirmed as a promising therapeutic target for treating metabolic diseases via beige cell induction and enhanced thermogenesis.
Collapse
|
7
|
Chou CK, Yang YT, Yang HC, Liang SS, Wang TN, Kuo PL, Wang HMD, Tsai EM, Chiu CC. The Impact of Di(2-ethylhexyl)phthalate on Cancer Progression. Arch Immunol Ther Exp (Warsz) 2017; 66:183-197. [PMID: 29209738 DOI: 10.1007/s00005-017-0494-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 08/18/2017] [Indexed: 12/11/2022]
Abstract
Di(2-ethylhexyl)phthalate (DEHP), a widely used plasticizer, mainly serves as an additive to render polyvinyl chloride (PVC) soft and flexible. PVC plastics have become ubiquitous in our modern society. Yet, the leaching of DEHP from PVC-based consumables ultimately results in the deposition in certain tissues via inadvertent applications. Health risks for human populations exposed to DEHP has been assumed by studies on rodents and other species, including the DEHP-induced developmental dysregulation, reproductive impairments, tumorigenesis, and diseases in a transgenerational manner. In this review, we comprehensively summarize the accumulated literature regarding the multifaceted roles of DEHP in the activation of the nuclear receptors, the alteration of the redox homeostasis, epigenetic modifications and the acquisition of chemoresistance.
Collapse
Affiliation(s)
- Chon-Kit Chou
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Ya-Ting Yang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Ho-Chun Yang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Shih-Shin Liang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.,Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Tsu-Nai Wang
- Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.,Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 402, Taiwan
| | - Eing-Mei Tsai
- Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Headquarters of Research Centers, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan. .,Department of Medical Research, Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
8
|
Cichocki JA, Furuya S, Luo YS, Iwata Y, Konganti K, Chiu WA, Threadgill DW, Pogribny IP, Rusyn I. Nonalcoholic Fatty Liver Disease Is a Susceptibility Factor for Perchloroethylene-Induced Liver Effects in Mice. Toxicol Sci 2017; 159:102-113. [PMID: 28903486 PMCID: PMC5837635 DOI: 10.1093/toxsci/kfx120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent pathological liver condition in developed countries. NAFLD results in severe alterations in liver function, including xenobiotic metabolism. Perchloroethylene (PERC) is a ubiquitous environmental pollutant, a known hepatotoxicant in rodents, and a probable human carcinogen. It is known that PERC disposition and metabolism are affected by NAFLD in mice; here, we examined how NAFLD changes PERC-associated liver effects. Male C57Bl6/J mice were fed a low-fat diet (LFD), high-fat diet (HFD), or methionine/folate/choline-deficient diet (MCD) to model a healthy liver, or mild and severe forms of NAFLD, respectively. After 8 weeks on diets, mice were orally administered PERC (300 mg/kg/day) or vehicle (5% aqueous Alkamuls-EL620) for 5 days. PERC-induced liver effects were exacerbated in both NAFLD groups. PERC exposure was associated with up-regulation of genes involved in xenobiotic, lipid, and glutathione metabolism, and down-regulation of the complement and coagulation cascades, regardless of the diet. Interestingly, HFD-fed mice, not MCD-fed mice, were generally more sensitive to PERC-induced liver effects. This was indicated by histopathology and transcriptional responses, where induction of genes associated with cell cycle and inflammation were prominent. Liver effects positively correlated with diet-specific differences in liver concentrations of PERC. We conclude that NAFLD alters the toxicodynamics of PERC and that NAFLD is a susceptibility factor that should be considered in future risk management decisions for PERC and other chlorinated solvents.
Collapse
Affiliation(s)
- Joseph A. Cichocki
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - Shinji Furuya
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - Yu-Syuan Luo
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - Yasuhiro Iwata
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - Kranti Konganti
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas 77843
| | - Weihsueh A. Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - David W. Threadgill
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas 77843
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas 77843
| | - Igor P. Pogribny
- National Center for Toxicological Research, US FDA, Jefferson, Arkansas 72079
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
9
|
Davidson MA, Mattison DR, Azoulay L, Krewski D. Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol 2017; 48:52-108. [PMID: 28816105 DOI: 10.1080/10408444.2017.1351420] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thiazolidinedione (TZD) drugs used in the treatment of type 2 diabetes mellitus (T2DM) have proven effective in improving insulin sensitivity, hyperglycemia, and lipid metabolism. Though well tolerated by some patients, their mechanism of action as ligands of peroxisome proliferator-activated receptors (PPARs) results in the activation of several pathways in addition to those responsible for glycemic control and lipid homeostasis. These pathways, which include those related to inflammation, bone formation, and cell proliferation, may lead to adverse health outcomes. As treatment with TZDs has been associated with adverse hepatic, cardiovascular, osteological, and carcinogenic events in some studies, the role of TZDs in the treatment of T2DM continues to be debated. At the same time, new therapeutic roles for TZDs are being investigated, with new forms and isoforms currently in the pre-clinical phase for use in the prevention and treatment of some cancers, inflammatory diseases, and other conditions. The aims of this review are to provide an overview of the mechanism(s) of action of TZDs, a review of their safety for use in the treatment of T2DM, and a perspective on their current and future therapeutic roles.
Collapse
Affiliation(s)
- Melissa A Davidson
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada
| | - Donald R Mattison
- b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada
| | - Laurent Azoulay
- d Center for Clinical Epidemiology , Lady Davis Research Institute, Jewish General Hospital , Montreal , Canada.,e Department of Oncology , McGill University , Montreal , Canada
| | - Daniel Krewski
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada.,f Faculty of Medicine , University of Ottawa , Ottawa , Canada
| |
Collapse
|
10
|
Petkov PI, Schultz TW, Donner EM, Honma M, Morita T, Hamada S, Wakata A, Mishima M, Maniwa J, Todorov M, Kaloyanova E, Kotov S, Mekenyan OG. Integrated approach to testing and assessment for predicting rodent genotoxic carcinogenicity. J Appl Toxicol 2016; 36:1536-1550. [DOI: 10.1002/jat.3338] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Petko I. Petkov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Terry W. Schultz
- College of Veterinary Medicine; The University of Tennessee; Knoxville TN 37996-4500 USA
| | - E. Maria Donner
- DuPont Haskell Global Centers for Health and Environmental Sciences, Newark; DE USA
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis; National Institute of Health Sciences; Tokyo Japan
| | - Takeshi Morita
- Division of Risk Assessment; National Institute of Health Sciences; Tokyo Japan
| | | | | | - Masayuki Mishima
- Chugai Pharmaceutical Co., Ltd., Fuji Gotemba Research Labs; Shizuoka Japan
| | - Jiro Maniwa
- Clinical Science Division, Research & Development AstraZeneca KK; Osaka Japan
| | - Milen Todorov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Elena Kaloyanova
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Stefan Kotov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Ovanes G. Mekenyan
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| |
Collapse
|
11
|
Zhang P, Li L, Bao Z, Huang F. Role of BAF60a/BAF60c in chromatin remodeling and hepatic lipid metabolism. Nutr Metab (Lond) 2016; 13:30. [PMID: 27127533 PMCID: PMC4848843 DOI: 10.1186/s12986-016-0090-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/19/2016] [Indexed: 02/07/2023] Open
Abstract
The switching defective/sucrose non-fermenting (SWI/SNF) complexes play an important role in hepatic lipid metabolism regulating both transcriptional activation and repression. BAF60a is a core subunit of the SWI/SNF chromatin-remodeling complexes that activates the transcription of fatty acid oxidation genes during fasting/glucagon. BAF60c, another subunit of SWI/SNF complexes, is recruited to form the lipoBAF complex that activates lipogenic genes, promoting lipogenesis and increasing the triglyceride level in response to feeding/insulin. Interestingly, hepatocytes located in the periportal and perivenous zones of the liver display a remarkable heterogeneity in the activity of various enzymes, metabolic functions and gene expression. Especially, fatty-acid oxidation was shown to be mostly periportal, whereas lipogenesis was mostly perivenous. Therefore, the present review highlights the role of of SWI/SNF regulating lipid metabolism under nutritional and hormonal control, which may be associated with hepatocyte heterogeneity.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Lulu Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Zhengxi Bao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| |
Collapse
|
12
|
Kato H, Fujii S, Takahashi M, Matsumoto M, Hirata-Koizumi M, Ono A, Hirose A. Repeated dose and reproductive/developmental toxicity of perfluorododecanoic acid in rats. ENVIRONMENTAL TOXICOLOGY 2015; 30:1244-63. [PMID: 24753098 DOI: 10.1002/tox.21996] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 03/25/2014] [Accepted: 04/04/2014] [Indexed: 05/18/2023]
Abstract
Perfluoroalkyl carboxylic acids (PFCAs) are a series of environmental contaminants that have received attention because of their possible adverse effects on wildlife and human health. Although many toxicological studies have been performed on perfluorooctanoic acid with carbon chain length C8, available toxicity data on PFCAs with longer chains are still insufficient to evaluate their hazard. A combined repeated dose and reproductive/developmental toxicity screening study for perfluorododecanoic acid (PFDoA; C12) was conducted in accordance with OECD guideline 422 to fill these toxicity data gaps. PFDoA was administered by gavage to male and female rats at 0.1, 0.5, or 2.5 mg/kg/day. The administration of PFDoA at 0.5 and 2.5 mg/kg/day for 42-47 days mainly affected the liver, in which hypertrophy, necrosis, and inflammatory cholestasis were noted. Body weight gain was markedly inhibited in the 2.5 mg/kg/day group, and a decrease in hematopoiesis in the bone marrow and atrophic changes in the spleen, thymus, and adrenal gland were also observed. Regarding reproductive/developmental toxicity, various histopathological changes, including decreased spermatid and spermatozoa counts, were observed in the male reproductive organs, while continuous diestrous was observed in the females of the 2.5 mg/kg/day group. Seven of twelve females receiving 2.5 mg/kg/day died during late pregnancy while four other females in this group did not deliver live pups. No reproductive or developmental parameters changed at 0.1 or 0.5 mg/kg/day. Based on these results, the NOAELs of PFDoA were concluded to be 0.1 mg/kg/day for repeated dose toxicity and 0.5 mg/kg/day for reproductive/developmental toxicity.
Collapse
Affiliation(s)
- Hina Kato
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-Ku, Tokyo, 158-8501, Japan
| | - Sakiko Fujii
- Safety Research Institute for Chemical Compounds Co., Ltd., 363-24 Sin-ei, Kiyota-ku, Sapporo, Hokkaido, 004-0839, Japan
| | - Mika Takahashi
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-Ku, Tokyo, 158-8501, Japan
| | - Mariko Matsumoto
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-Ku, Tokyo, 158-8501, Japan
| | - Mutsuko Hirata-Koizumi
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-Ku, Tokyo, 158-8501, Japan
| | - Atsushi Ono
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-Ku, Tokyo, 158-8501, Japan
| | - Akihiko Hirose
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-Ku, Tokyo, 158-8501, Japan
| |
Collapse
|
13
|
Solleti SK, Simon DM, Srisuma S, Arikan MC, Bhattacharya S, Rangasamy T, Bijli KM, Rahman A, Crossno JT, Shapiro SD, Mariani TJ. Airway epithelial cell PPARγ modulates cigarette smoke-induced chemokine expression and emphysema susceptibility in mice. Am J Physiol Lung Cell Mol Physiol 2015; 309:L293-304. [PMID: 26024894 DOI: 10.1152/ajplung.00287.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/26/2015] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent, chronic inflammatory lung disease with limited existing therapeutic options. While modulation of peroxisome proliferator-activating receptor (PPAR)-γ activity can modify inflammatory responses in several models of lung injury, the relevance of the PPARG pathway in COPD pathogenesis has not been previously explored. Mice lacking Pparg specifically in airway epithelial cells displayed increased susceptibility to chronic cigarette smoke (CS)-induced emphysema, with excessive macrophage accumulation associated with increased expression of chemokines, Ccl5, Cxcl10, and Cxcl15. Conversely, treatment of mice with a pharmacological PPARγ activator attenuated Cxcl10 and Cxcl15 expression and macrophage accumulation in response to CS. In vitro, CS increased lung epithelial cell chemokine expression in a PPARγ activation-dependent fashion. The ability of PPARγ to regulate CS-induced chemokine expression in vitro was not specifically associated with peroxisome proliferator response element (PPRE)-mediated transactivation activity but was correlated with PPARγ-mediated transrepression of NF-κB activity. Pharmacological or genetic activation of PPARγ activity abrogated CS-dependent induction of NF-κB activity. Regulation of NF-κB activity involved direct PPARγ-NF-κB interaction and PPARγ-mediated effects on IKK activation, IκBα degradation, and nuclear translocation of p65. Our data indicate that PPARG represents a disease-relevant pathophysiological and pharmacological target in COPD. Its activation state likely contributes to NF-κB-dependent, CS-induced chemokine-mediated regulation of inflammatory cell accumulation.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Dawn M Simon
- Emory-Children's Center Pulmonary, Apnea, Cystic Fibrosis and Sleep Clinic, Atlanta, Georgia
| | - Sorachai Srisuma
- Faculty of Medicine, Department of Physiology, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Meltem C Arikan
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| | - Tirumalai Rangasamy
- Division of Pulmonary & Critical Care Medicine, University of Rochester Medical Center, Rochester, New York
| | - Kaiser M Bijli
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York; Atlanta VA and Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Arshad Rahman
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Joseph T Crossno
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, Colorado
| | - Steven D Shapiro
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| |
Collapse
|
14
|
Hirata-Koizumi M, Fujii S, Hina K, Matsumoto M, Takahashi M, Ono A, Hirose A. Repeated dose and reproductive/developmental toxicity of long-chain perfluoroalkyl carboxylic acids in rats: perfluorohexadecanoic acid and perfluorotetradecanoic acid. ACTA ACUST UNITED AC 2015. [DOI: 10.2131/fts.2.177] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Mutsuko Hirata-Koizumi
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences
| | - Sakiko Fujii
- Safety Research Institute for Chemical Compounds Co., Ltd
| | - Kato Hina
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences
| | - Mariko Matsumoto
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences
| | - Mika Takahashi
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences
| | - Atsushi Ono
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences
| | - Akihiko Hirose
- Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences
| |
Collapse
|
15
|
Yavaşoğlu NÜK, Köksal C, Dağdeviren M, Aktuğ H, Yavaşoğlu A. Induction of oxidative stress and histological changes in liver by subacute doses of butyl cyclohexyl phthalate. ENVIRONMENTAL TOXICOLOGY 2014; 29:345-353. [PMID: 22936646 DOI: 10.1002/tox.21813] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 08/02/2012] [Accepted: 08/04/2012] [Indexed: 06/01/2023]
Abstract
Phthalates are esters of phthalic acid and are mainly used as plasticizers in a wide variety of products and applications. There is no information on butyl cyclohexyl phthalate (BCP) toxicity. This study was performed to evaluate the histopathological effects and to determine oxidative stress inducing potential in liver by subacute exposure of BCP. The animals of the treatment groups were orally administered 100, 200, and 400 mg/kg/day BCP for 5 consecutive days per week during 28 days. As a result, no significant changes were observed in body weight gains, and absolute and relative liver weights of liver of BCP treated mice, when compared with control group. Although the degree of lipid peroxidation in the liver tissue of all BCP exposure groups were significantly higher than those of the control (p < 0.01), SOD and CAT activities in liver tissue of mice of 200 and 400 mg/kg exposure groups were significantly lower than those of the controls (p < 0.01). Moreover, BCP caused dose-dependent histological changes in the liver of mice such as congestions in vena centralis, an enlargement of the sinusoids, degeneration in hepatocytes, vacuole formations and presence of lipid droplets in hepatocytes, eosinophilic cytoplasm. While iNOS immunoreactivity was increased in all treatment groups, Type IV collagen and Connexin 43 immunoreactivities were decreased in all treatment groups compared with the control group. Significant decrease was observed in the number of TUNEL-positive liver cells of BCP treated mice. These results suggested that BCP exposure induces oxidative stress in liver and exposure of BCP during long time period could lead to hepatocarcinogenesis.
Collapse
|
16
|
Skrypnyk N, Chen X, Hu W, Su Y, Mont S, Yang S, Gangadhariah M, Wei S, Falck JR, Jat JL, Zent R, Capdevila JH, Pozzi A. PPARα activation can help prevent and treat non-small cell lung cancer. Cancer Res 2013; 74:621-31. [PMID: 24302581 DOI: 10.1158/0008-5472.can-13-1928] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Non-small cell lung cancer (NSCLC) not amenable to surgical resection has a high mortality rate, due to the ineffectiveness and toxicity of chemotherapy. Thus, there remains an urgent need of efficacious drugs that can combat this disease. In this study, we show that targeting the formation of proangiogenic epoxyeicosatrienoic acids (EET) by the cytochrome P450 arachidonic acid epoxygenases (Cyp2c) represents a new and safe mechanism to treat NSCLC growth and progression. In the transgenic murine K-Ras model and human orthotopic models of NSCLC, we found that Cyp2c44 could be downregulated by activating the transcription factor PPARα with the ligands bezafibrate and Wyeth-14,643. Notably, both treatments reduced primary and metastatic NSCLC growth, tumor angiogenesis, endothelial Cyp2c44 expression, and circulating EET levels. These beneficial effects were independent of the time of administration, whether before or after the onset of primary NSCLC, and they persisted after drug withdrawal, suggesting the benefits were durable. Our findings suggest that strategies to downregulate Cyp2c expression and/or its enzymatic activity may provide a safer and effective strategy to treat NSCLC. Moreover, as bezafibrate is a well-tolerated clinically approved drug used for managing lipidemia, our findings provide an immediate cue for clinical studies to evaluate the utility of PPARα ligands as safe agents for the treatment of lung cancer in humans.
Collapse
Affiliation(s)
- Nataliya Skrypnyk
- Authors' Affiliations: Departments of Medicine, Division of Nephrology, Cancer Biology, and Biochemistry, Vanderbilt University; Department of Medicine, Veterans Affairs Hospital, Nashville, Tennessee; and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Benigni R, Bossa C, Tcheremenskaia O. Nongenotoxic carcinogenicity of chemicals: mechanisms of action and early recognition through a new set of structural alerts. Chem Rev 2013; 113:2940-57. [PMID: 23469814 DOI: 10.1021/cr300206t] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Romualdo Benigni
- Istituto Superiore di Sanita' Environment and Health Department, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | |
Collapse
|
18
|
Rusyn I, Corton JC. Mechanistic considerations for human relevance of cancer hazard of di(2-ethylhexyl) phthalate. Mutat Res 2011; 750:141-158. [PMID: 22198209 DOI: 10.1016/j.mrrev.2011.12.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 12/06/2011] [Accepted: 12/12/2011] [Indexed: 12/28/2022]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a peroxisome proliferator agent that is widely used as a plasticizer to soften polyvinylchloride plastics and non-polymers. Both occupational (e.g., by inhalation during its manufacture and use as a plasticizer of polyvinylchloride) and environmental (medical devices, contamination of food, or intake from air, water and soil) routes of exposure to DEHP are of concern for human health. There is sufficient evidence for carcinogenicity of DEHP in the liver in both rats and mice; however, there is little epidemiological evidence on possible associations between exposure to DEHP and liver cancer in humans. Data are available to suggest that liver is not the only target tissue for DEHP-associated toxicity and carcinogenicity in both humans and rodents. The debate regarding human relevance of the findings in rats or mice has been informed by studies on the mechanisms of carcinogenesis of the peroxisome proliferator class of chemicals, including DEHP. Important additional mechanistic information became available in the past decade, including, but not limited to, sub-acute, sub-chronic and chronic studies with DEHP in peroxisome proliferator-activated receptor (PPAR) α-null mice, as well as experiments utilizing several transgenic mouse lines. Activation of PPARα and the subsequent downstream events mediated by this transcription factor represent an important mechanism of action for DEHP in rats and mice. However, additional data from animal models and studies in humans exposed to DEHP from the environment suggest that multiple molecular signals and pathways in several cell types in the liver, rather than a single molecular event, contribute to the cancer in rats and mice. In addition, the toxic and carcinogenic effects of DEHP are not limited to liver. The International Agency for Research on Cancer working group concluded that the human relevance of the molecular events leading to cancer elicited by DEHP in several target tissues (e.g., liver and testis) in rats and mice can not be ruled out and DEHP was classified as possibly carcinogenic to humans (Group 2B).
Collapse
Affiliation(s)
- Ivan Rusyn
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, NC 27599-7431, USA.
| | - J Christopher Corton
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| |
Collapse
|
19
|
Klaunig JE, Hocevar BA, Kamendulis LM. Mode of Action analysis of perfluorooctanoic acid (PFOA) tumorigenicity and Human Relevance. Reprod Toxicol 2011; 33:410-418. [PMID: 22120428 DOI: 10.1016/j.reprotox.2011.10.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 10/01/2011] [Accepted: 10/24/2011] [Indexed: 11/19/2022]
Abstract
Perfluorooctanoic acid (PFOA) is an environmentally persistent chemical used in the manufacturing of a wide array of industrial and commercial products. PFOA has been shown to induce tumors of the liver, testis and pancreas (tumor triad) in rats following chronic dietary administration. PFOA belongs to a group of compounds that are known to activate the PPARα receptor. The PPARα activation Mode of Action was initially addressed in 2003 [9] and further refined in subsequent reviews [92-94]. In the intervening time, additional information on PFOA effects as well as a further refinement of the Mode of Action framework warrants a re-examination of this compound for its cancer induction Mode of Action. This review will address the rodent (rat) cancer data and cancer Mode of Action of PFOA for tumors of the liver, testes and pancreas.
Collapse
Affiliation(s)
- James E Klaunig
- Indiana University, Department of Environmental Health, Bloomington, IN 47405, United States.
| | - Barbara A Hocevar
- Indiana University, Department of Environmental Health, Bloomington, IN 47405, United States
| | - Lisa M Kamendulis
- Indiana University, Department of Environmental Health, Bloomington, IN 47405, United States
| |
Collapse
|
20
|
Yamana D, Shimizu T, Fan Y, Miura T, Nanashima N, Yamada T, Hakamada K, Tsuchida S. Decrease of hepatic stellate cells in rats with enhanced sensitivity to clofibrate-induced hepatocarcinogenesis. Cancer Sci 2011; 102:735-41. [PMID: 21214675 PMCID: PMC11158309 DOI: 10.1111/j.1349-7006.2011.01856.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Revised: 12/27/2010] [Accepted: 12/31/2010] [Indexed: 12/28/2022] Open
Abstract
To examine the possible involvement of nonparenchymal cells in the development of preneoplastic hepatic lesions induced by clofibrate (CF), alterations of these cells were investigated immunohistochemically in glutathione S-transferase M1 gene polymorphic rats (KS and NC types) with different cancer susceptibilities. After CF administration for 8 weeks, α-smooth muscle actin (α-SMA)-positive hepatic stellate cells (HSC) were markedly decreased in sensitive KS-type rats, but not in the NC-type rats. Kupffer cells were decreased with similar extents between them. The sinusoidal endothelial cells were not changed in either type. The other markers for HSC, vimentin and CRBP1, also confirmed the decrease of HSC in the KS type. The decrease of HSC was not observed at 4 weeks of CF administration. Preneoplastic peroxisomal bifunctional enzyme-negative foci were detected in the KS-type rats at 8 weeks of CF administration, but not at 4 weeks. Human HSC were cultured in the presence of clofibric acid and expression of most HSC marker genes, such as vimentin and α-SMA (ACTA2), evaluated by a microarray, was not altered by the treatment, suggesting that HSC loss in the KS-type rats was not due to the direct toxic effect of CF. The expression levels of most HSC marker genes were low in both control and CF-treated rat livers. A possible link between HSC loss and the development of preneoplastic hepatic foci is discussed.
Collapse
Affiliation(s)
- Daisuke Yamana
- Department of Biochemistry and Genome Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Benigni R, Bossa C. Mechanisms of Chemical Carcinogenicity and Mutagenicity: A Review with Implications for Predictive Toxicology. Chem Rev 2011; 111:2507-36. [PMID: 21265518 DOI: 10.1021/cr100222q] [Citation(s) in RCA: 239] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Romualdo Benigni
- Istituto Superiore di Sanita’, Environment and Health Department, Viale Regina Elena, 299 00161 Rome, Italy
| | - Cecilia Bossa
- Istituto Superiore di Sanita’, Environment and Health Department, Viale Regina Elena, 299 00161 Rome, Italy
| |
Collapse
|
22
|
Yuan X, Jonker MJ, de Wilde J, Verhoef A, Wittink FR, van Benthem J, Bessems JG, Hakkert BC, Kuiper RV, van Steeg H, Breit TM, Luijten M. Finding maximal transcriptome differences between reprotoxic and non-reprotoxic phthalate responses in rat testis. J Appl Toxicol 2010; 31:421-30. [DOI: 10.1002/jat.1601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 08/30/2010] [Accepted: 08/31/2010] [Indexed: 11/07/2022]
|
23
|
Shimizu T, Fan Y, Yamana D, Miura T, Nanashima N, Yamada T, Tsuchida S. Glutathione S-transferase A4 is a positive marker for rat hepatic foci induced by clofibrate and genotoxic carcinogens. Cancer Sci 2010; 101:1093-8. [PMID: 20180811 PMCID: PMC11158787 DOI: 10.1111/j.1349-7006.2010.01508.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Peroxisome proliferators (PP), including clofibrate (CF), are non-genotoxic rodent carcinogens, and oxidative DNA damages are suggested as a causative event for carcinogenesis. Gene expression profiles differ between hepatic lesions induced by PP and genotoxic carcinogens. Our previous study revealed that expression of L-bifunctional enzyme (enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase, BE) was repressed in preneoplastic lesions induced by PP, whereas it was enhanced in the surrounding tissues. In the present study, we immunohistochemically examined expression of the specific glutathione S-transferase (GST) form, GST-A4, which detoxifies 4-hydroxy-alkenal, the end-product of lipid peroxides, and nuclear factor-erythroid 2-related factor 2 (Nrf2), a transcription factor for many genes encoding drug-metabolizing enzymes and defending enzymes against oxidative stress, during rat hepatocarcinogenesis induced by CF and genotoxic carcinogens. GST-A4 and Nrf2 were not expressed in BE-negative foci at 8 weeks of CF administration, but were expressed in the foci at 60 weeks. GST-A4-positive foci appeared at later stages than BE-negative foci, but its localization was coincidental with that of the latter foci. The areas of GST-A4-positive foci were larger than those of BE-negative foci without GST-A4 expression. Most GST-A4-positive foci were also positive for Nrf2. In rat livers induced by genotoxic carcinogens, GST-P-negative foci as well as GST-P-positive foci were demonstrated. GST-A4 and Nrf2 were expressed in GST-P-negative foci, whereas they were not expressed in most GST-P-positive foci. Thus, GST-A4-positive foci developed in rat livers by CF and genotoxic carcinogen administration, indicating that the enzyme is a positive marker for hepatic foci induced by these different carcinogens.
Collapse
Affiliation(s)
- Takeshi Shimizu
- Department of Biochemistry and Genome Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Therapeutic Implications of PPARgamma in Human Osteosarcoma. PPAR Res 2010; 2010:956427. [PMID: 20182546 PMCID: PMC2825651 DOI: 10.1155/2010/956427] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 11/21/2009] [Accepted: 11/24/2009] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma (OS) is the most common nonhematologic malignancy of bone in children and adults. Although dysregulation of tumor suppressor genes and oncogenes, such as Rb, p53, and the genes critical to cell cycle control, genetic stability, and apoptosis have been identified in OS, consensus genetic changes that lead to OS development are poorly understood. Disruption of the osteogenic differentiation pathway may be at least in part responsible for OS tumorigenesis. Current OS management involves chemotherapy and surgery. Peroxisome proliferator-activated receptor (PPAR) agonists and/or retinoids can inhibit OS proliferation and induce apoptosis and may inhibit OS growth by promoting osteoblastic terminal differentiation. Thus, safe and effective PPAR agonists and/or retinoid derivatives can be then used as adjuvant therapeutic drugs for OS therapy. Furthermore, these agents have the potential to be used as chemopreventive agents for the OS patients who undergo the resection of the primary bone tumors in order to prevent local recurrence and/or distal pulmonary metastasis.
Collapse
|
25
|
Jollow DJ, Bruckner JV, McMillan DC, Fisher JW, Hoel DG, Mohr LC. Trichloroethylene risk assessment: a review and commentary. Crit Rev Toxicol 2010; 39:782-97. [PMID: 19852561 DOI: 10.3109/10408440903222177] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Trichloroethylene (TCE) is a widespread environmental contaminant that is carcinogenic when given in high, chronic doses to certain strains of mice and rats. The capacity of TCE to cause cancer in humans is less clear. The current maximum contaminant level (MCL) of 5 ppb (microg/L) is based on an US Environment Protection Agency (USEPA) policy decision rather than the underlying science. In view of major advances in understanding the etiology and mechanisms of chemically induced cancer, USEPA began in the late 1990s to revise its guidelines for cancer risk assessment. TCE was chosen as the pilot chemical. The USEPA (2005) final guidelines emphasized a "weight-of-evidence" approach with consideration of dose-response relationships, modes of action, and metabolic/toxicokinetic processes. Where adequate data are available to support reversible binding of the carcinogenic moiety to biological receptors as the initiating event (i.e., a threshold exists), a nonlinear approach is to be used. Otherwise, the default assumption of a linear (i.e., nonthreshold) dose-response is utilized. When validated physiologically based pharmacokinetic (PBPK) models are available, they are to be used to predict internal dosimetry as the basis for species and dose extrapolations. The present article reviews pertinent literature and discusses areas where research may resolve some outstanding issues and facilitate the reassessment process. Key research needs are proposed, including role of dichloroacetic acid (DCA) in TCE-induced liver tumorigenesis in humans; extension of current PBPK models to predict target organ deposition of trichloroacetic acid (TCA) and DCA in humans ingesting TCE in drinking water; use of human hepatocytes to ascertain metabolic rate constants for use in PBPK models that incorporate variability in metabolism of TCE by potentially sensitive subpopulations; measurement of the efficiency of first-pass elimination of trace levels of TCE in drinking water; and assessment of exogenous factors' (e.g., alcohol, drugs) ability to alter metabolic activation and risks at such low-level exposure.
Collapse
Affiliation(s)
- David J Jollow
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
26
|
Vallanat B, Anderson SP, Brown-Borg HM, Ren H, Kersten S, Jonnalagadda S, Srinivasan R, Corton JC. Analysis of the heat shock response in mouse liver reveals transcriptional dependence on the nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha). BMC Genomics 2010; 11:16. [PMID: 20059764 PMCID: PMC2823686 DOI: 10.1186/1471-2164-11-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 01/07/2010] [Indexed: 11/22/2022] Open
Abstract
Background The nuclear receptor peroxisome proliferator-activated receptor alpha (PPARα) regulates responses to chemical or physical stress in part by altering expression of genes involved in proteome maintenance. Many of these genes are also transcriptionally regulated by heat shock (HS) through activation by HS factor-1 (HSF1). We hypothesized that there are interactions on a genetic level between PPARα and the HS response mediated by HSF1. Results Wild-type and PPARα-null mice were exposed to HS, the PPARα agonist WY-14,643 (WY), or both; gene and protein expression was examined in the livers of the mice 4 or 24 hrs after HS. Gene expression profiling identified a number of Hsp family members that were altered similarly in both mouse strains. However, most of the targets of HS did not overlap between strains. A subset of genes was shown by microarray and RT-PCR to be regulated by HS in a PPARα-dependent manner. HS also down-regulated a large set of mitochondrial genes specifically in PPARα-null mice that are known targets of PPARγ co-activator-1 (PGC-1) family members. Pretreatment of PPARα-null mice with WY increased expression of PGC-1β and target genes and prevented the down-regulation of the mitochondrial genes by HS. A comparison of HS genes regulated in our dataset with those identified in wild-type and HSF1-null mouse embryonic fibroblasts indicated that although many HS genes are regulated independently of both PPARα and HSF1, a number require both factors for HS responsiveness. Conclusions These findings demonstrate that the PPARα genotype has a dramatic effect on the transcriptional targets of HS and support an expanded role for PPARα in the regulation of proteome maintenance genes after exposure to diverse forms of environmental stress including HS.
Collapse
Affiliation(s)
- Beena Vallanat
- NHEERL Toxicogenomics Core, US EPA, Research Triangle Park, NC 27711, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Rakhshandehroo M, Hooiveld G, Müller M, Kersten S. Comparative analysis of gene regulation by the transcription factor PPARalpha between mouse and human. PLoS One 2009; 4:e6796. [PMID: 19710929 PMCID: PMC2729378 DOI: 10.1371/journal.pone.0006796] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 07/15/2009] [Indexed: 12/18/2022] Open
Abstract
Background Studies in mice have shown that PPARα is an important regulator of hepatic lipid metabolism and the acute phase response. However, little information is available on the role of PPARα in human liver. Here we set out to compare the function of PPARα in mouse and human hepatocytes via analysis of target gene regulation. Methodology/Principal Findings Primary hepatocytes from 6 human and 6 mouse donors were treated with PPARα agonist Wy14643 and gene expression profiling was performed using Affymetrix GeneChips followed by a systems biology analysis. Baseline PPARα expression was similar in human and mouse hepatocytes. Depending on species and time of exposure, Wy14643 significantly induced the expression of 362–672 genes. Surprisingly minor overlap was observed between the Wy14643-regulated genes from mouse and human, although more substantial overlap was observed at the pathway level. Xenobiotics metabolism and apolipoprotein synthesis were specifically regulated by PPARα in human hepatocytes, whereas glycolysis-gluconeogenesis was regulated specifically in mouse hepatocytes. Most of the genes commonly regulated in mouse and human were involved in lipid metabolism and many represented known PPARα targets, including CPT1A, HMGCS2, FABP1, ACSL1, and ADFP. Several genes were identified that were specifically induced by PPARα in human (MBL2, ALAS1, CYP1A1, TSKU) or mouse (Fbp2, lgals4, Cd36, Ucp2, Pxmp4). Furthermore, several putative novel PPARα targets were identified that were commonly regulated in both species, including CREB3L3, KLF10, KLF11 and MAP3K8. Conclusions/Significance Our results suggest that PPARα activation has a major impact on gene regulation in human hepatocytes. Importantly, the role of PPARα as master regulator of hepatic lipid metabolism is generally well-conserved between mouse and human. Overall, however, PPARα regulates a mostly divergent set of genes in mouse and human hepatocytes.
Collapse
Affiliation(s)
- Maryam Rakhshandehroo
- Nutrigenomics Consortium, Top Institute (TI) Food and Nutrition, Wageningen, the Netherlands
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Guido Hooiveld
- Nutrigenomics Consortium, Top Institute (TI) Food and Nutrition, Wageningen, the Netherlands
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Michael Müller
- Nutrigenomics Consortium, Top Institute (TI) Food and Nutrition, Wageningen, the Netherlands
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Sander Kersten
- Nutrigenomics Consortium, Top Institute (TI) Food and Nutrition, Wageningen, the Netherlands
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
- * E-mail:
| |
Collapse
|
28
|
Corton JC. Evaluation of the role of peroxisome proliferator-activated receptor alpha (PPARalpha) in mouse liver tumor induction by trichloroethylene and metabolites. Crit Rev Toxicol 2009; 38:857-75. [PMID: 18821149 DOI: 10.1080/10408440802209796] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Trichloroethylene (TCE) is an industrial solvent and a widespread environmental contaminant. Induction of liver cancer in mice by TCE is thought to be mediated by two metabolites, dichloroacetate (DCA) and trichloroacetate (TCA), both of which are themselves mouse liver carcinogens. TCE, TCA, and DCA are relatively weak peroxisome proliferators (PP), a group of rodent hepatocarcinogens that activate a nuclear receptor, PP-activated receptor alpha (PPARalpha. The objective of this review is to assess the weight of evidence (WOE) that PPARalpha is or is not mechanistically involved in mouse liver tumor induction by TCE and metabolites. Based on similarities of TCE and TCA to typical PP, including dose-response characteristics showing PPARalpha-dependent responses coincident with liver tumor induction and abolishment of TCE and TCA effects in PPARalpha-null mice, the WOE supports the hypothesis that PPARalpha plays a dominant role in TCE- and TCA-induced hepatocarcinogenesis. Data indicates that the MOA for DCA tumor induction is PPARalpha-independent. Uncertainties remain regarding the genesis of the TCE-induced tumors. In contrast to the TCA-induced tumors, which have molecular features similar to those induced by typical PP, there is evidence, albeit weak, that TCE tumors arise by a mode of action (MOA) different from that of TCA tumors, based largely on dissimilarities in molecular markers found in TCE versus TCA-induced tumors. In summary, the WOE indicates that TCA-induced liver tumors arise by a PPARalpha-dependent MOA. Although the TCE MOA is likely dominated by a PPARalpha-dependent contribution from TCA, the contribution of a PPARalpha-independent MOA from DCA cannot be ruled out.
Collapse
Affiliation(s)
- J Christopher Corton
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA.
| |
Collapse
|
29
|
Lubet RA, Fischer SM, Steele VE, Juliana MM, Desmond R, Grubbs CJ. Rosiglitazone, a PPAR gamma agonist: potent promoter of hydroxybutyl(butyl)nitrosamine-induced urinary bladder cancers. Int J Cancer 2008; 123:2254-9. [PMID: 18712722 DOI: 10.1002/ijc.23765] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In an initial study to determine if rosiglitazone had chemopreventive activity, Fischer-344 female rats were administered twice weekly doses of hydroxybutyl(butyl)nitrosamine (OH-BBN), a urinary bladder specific carcinogen, for 8 weeks. Two weeks following the last dose of OH-BBN, rats were administered rosiglitazone (50 mg/kg BW) daily by gavage for the remainder of the study (7 months). Only 57% of OH-BBN-treated animals developed palpable urinary bladder cancers during the course of the study, while all of the OH-BBN plus rosiglitazone treated rats developed large cancers (p < 0.01). Surprisingly, examination for PPAR gamma by immunohistochemistry in the urinary bladders of rats showed that while untreated bladder urothelium and preneoplastic lesions clearly expressed PPAR gamma, frank carcinomas exhibited significantly lower levels. This was confirmed by employing microarray studies of the same samples. In additional studies, lower doses of rosiglitazone (10, 2 and 0.4 mg/kg BW/day) were administered. The 10 mg/kg BW/day dose greatly enhanced bladder cancer incidence (p < 0.01). The dose of 2 mg/kg BW/day, which is roughly equivalent to a standard human dose, also significantly increased bladder cancer incidence (controls, 48%; rosiglitazone-treated, 84%). The lowest dose did not significantly increase tumor incidence (rosiglitazone at 0.4 mg/kg BW/day, 64%) or tumor weight in the rats, although there was a trend in that direction. Rosiglitazone alone (10 mg/kg BW/day) given in the absence of OH-BBN did not result in bladder cancer formation when given for 10 months. In summary, rosiglitazone over a wide dose range enhanced urinary bladder carcinogenesis in the OH-BBN model in rats.
Collapse
Affiliation(s)
- Ronald A Lubet
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA.
| | | | | | | | | | | |
Collapse
|
30
|
O'Brien ML, Spear BT, Glauert HP. Role of Oxidative Stress in Peroxisome Proliferator-Mediated Carcinogenesis. Crit Rev Toxicol 2008; 35:61-88. [PMID: 15742903 DOI: 10.1080/10408440590905957] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In this review, the evidence about the role of oxidative stress in the induction of hepatocellular carcinomas by peroxisome proliferators is examined. The activation of PPAR-alpha by peroxisome proliferators in rats and mice may produce oxidative stress, due to the induction of enzymes like fatty acyl coenzyme A (CoA) oxidase (AOX) and cytochrome P-450 4A1. The effect of peroxisome proliferators on the antioxidant defense system is reviewed, as is the effect on endpoints resulting from oxidative stress that may be important in carcinogenesis, such as lipid peroxidation, oxidative DNA damage, and transcription factor activation. Peroxisome proliferators clearly inhibit several enzymes in the antioxidant defense system, but studies examining effects on lipid peroxidation and oxidative DNA damage are conflicting. There is a profound species difference in the induction of hepatocellular carcinomas by peroxisome proliferators, with rats and mice being sensitive, whereas species such as nonhuman primates and guinea pigs are not susceptible to the effects of peroxisome proliferators. The possible role of oxidative stress in these species differences is also reviewed. Overall, peroxisome proliferators produce changes in oxidative stress, but whether these changes are important in the carcinogenic process is not clear at this time.
Collapse
Affiliation(s)
- Michelle L O'Brien
- Graduate Centerfor Toxicology, University of Kentucky, Lexington, Kentucky 40506-0054, USA
| | | | | |
Collapse
|
31
|
Tong Z, Chandrasekaran A, Jordan R, Markiewicz V, Li H, Xiang Q, Shen L, Scatina J. Effects of ertiprotafib on hepatic cytochrome P450 and peroxisomal enzymes in rats and dogs, and in rat and human primary hepatocytes. Xenobiotica 2008; 37:1-18. [PMID: 17178630 DOI: 10.1080/00498250600965115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Ertiprotafib (ERTI) significantly increased liver weights in male and female rats, and moderately increased liver weights in male dogs after treatment for 28 days. The present study tested the hypotheses that the organ weight increases were associated with peroxisome proliferation in rats and induction of hepatic enzymes in rats and dogs, and would have limited impacts on humans. At a dosage of 200 mg kg-1 day-1, CYP4A was induced by tenfold in male rats and 2.4-fold in female rats. In male rats, CYP2B was induced by 1.2-fold and CYP3A was induced by 1.7-fold. Palmitoyl CoA oxidase was induced by 5.1-fold in male rats and 2.9-fold in female rats; carnitine acetyltransferase was induced by 10.4-fold in male rats and 5.2-fold in female rats. CYP3A, CYP4A and peroxisomal enzymes were not induced in dogs at 150/200 mg kg-1 day-1. ERTI at 50 microM markedly induced the mRNA level of CYP4A by up to fivefold in rat hepatocytes, but not in human primary hepatocytes. In conclusion, the liver weight increases observed in rats treated with ERTI appears to be due to rodent-specific peroxisome proliferation and the substantial induction of CYP4A1. ERTI is not a potent P450 inducer in dogs or in human hepatocytes. Therefore, ERTI is not expected to exert any significant effects on hepatic drug-metabolizing enzymes in humans.
Collapse
Affiliation(s)
- Z Tong
- Biotransformation Division, Wyeth Research, 500 Arcola Road, Collegeville, PA 19426, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Calfee-Mason KG, Lee EY, Spear BT, Glauert HP. Role of the p50 subunit of NF-kappaB in vitamin E-induced changes in mice treated with the peroxisome proliferator, ciprofibrate. Food Chem Toxicol 2008; 46:2062-73. [PMID: 18336980 PMCID: PMC2600965 DOI: 10.1016/j.fct.2008.01.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 10/01/2007] [Accepted: 01/28/2008] [Indexed: 12/18/2022]
Abstract
Peroxisome proliferators (PPs) are a diverse class of chemicals, which cause a dramatic increase in the size and number of hepatic peroxisomes in rodents and eventually lead to the development of hepatic tumors. Nuclear factor-kappaB (NF-kappaB) is a transcription factor activated by reactive oxygen and is involved in cell proliferation and apoptosis. Previously we found that the peroxisome proliferator ciprofibrate (CIP) activates NF-kappaB and that dietary vitamin E decreases CIP-induced NF-kappaB DNA binding. We, therefore, hypothesized that inhibition of NF-kappaB by vitamin E is necessary for effects of vitamin E on CIP-induced cell proliferation and the inhibition of apoptosis by CIP. Sixteen B6129 female mice (p50+/+) and twenty mice deficient in the p50 subunit of NF-kappaB (p50-/-) were fed a purified diet containing 10 or 250mg/kg vitamin E (alpha-tocopherol acetate) for 28 days. At that time, half of the mice were placed on the same diet with 0.01% CIP for 10 days. CIP treatment increased the DNA binding activity of NF-kappaB and cell proliferation, but had no significant effect on apoptosis. Compared to wild-type mice, the p50-/- mice had lower NF-kappaB activation, higher basal levels of cell proliferation and apoptosis, and a lower ratio of reduced glutathione to oxidized glutathione (GSH/GSSG). There was approximately a 60% reduction in cell proliferation in the CIP-treated p50-/- mice fed higher vitamin E in comparison to the p50-/- mice fed lower vitamin E. Dietary vitamin E also inhibited the DNA binding activity of NF-kappaB, increased apoptosis, and increased the GSH/GSSG ratio. This study shows the effects of vitamin E on cell growth parameters do not appear to be solely through decreased NF-kappaB activation, suggesting that vitamin E is acting by other molecular mechanisms.
Collapse
Affiliation(s)
- Karen G. Calfee-Mason
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY 40506, USA
| | - Eun Y. Lee
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Brett T. Spear
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY 40506, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Graduate Center for Toxicology; University of Kentucky, Lexington, KY 40506, USA
| | - Howard P. Glauert
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY 40506, USA
- Graduate Center for Toxicology; University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
33
|
Nishimura J, Dewa Y, Okamura T, Muguruma M, Jin M, Saegusa Y, Umemura T, Mitsumori K. Possible involvement of oxidative stress in fenofibrate-induced hepatocarcinogenesis in rats. Arch Toxicol 2008; 82:641-54. [PMID: 18253720 DOI: 10.1007/s00204-007-0278-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Accepted: 12/20/2007] [Indexed: 12/16/2022]
Abstract
To clarify whether oxidative stress is involved in the development of hepatocellular preneoplastic foci induced by fenofibrate (FF), a peroxisome proliferator-activated receptor alpha agonist, male F344/N rats were fed a diet containing 6,000, 3,000, or 0 ppm of FF for 13 weeks after N-diethylnitrosamine initiation. Two-third partial hepatectomy was performed 1 week after the FF treatment. Histopathologically, the number of hepatocellular altered foci significantly increased in the FF-treated groups with a concomitant increase in the number of hepatocytes positive for anti-Ki-67 antibody, but the number and area of glutathione S-transferase placental form (GST-P)-positive foci decreased in these groups, as compared to those in the controls. Microarray analysis or quantitative real-time reverse transcription-polymerase chine reaction demonstrated the significant up-regulations of Aco and Cyp4a1 (genes related to lipid metabolism); Gpx2, Yc2, Cat, Cyp2b15, and Ugt1a6 (metabolic oxidative stress-related genes); Apex1, Mgmt, Xrcc5, Nbn, and Gadd45a (DNA repair-related genes); and Ccnd1 (cell cycle-related genes) in the FF-treated groups, and the significant down-regulations of Cyp1a2, Gsta2, Gstm2, and Gstm3 (phase I or II metabolism-related genes); Mlh1 and Top1 (DNA repair-related genes); and Cdkn1a, Cdkn1b, Chek2, and Gadd45b (cell cycle/apoptosis-related genes) in these rats. FF-treatment increased the activity of enzymes such as carnitine acetyltransferase, carnitine palmitoyltransferase, fatty acyl-CoA oxidizing system, and catalase in the liver, but not superoxide dismutase in the liver. In addition, 8-OHdG level in liver DNA, lipofuscin deposition in hepatocytes, and in vitro reactive oxygen species production in microsomes significantly increased due to FF treatment. These results suggest that oxidative stress is involved in the development of FF-induced hepatocellular preneoplastic foci in rats.
Collapse
Affiliation(s)
- Jihei Nishimura
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
DeAngelo AB, Daniel FB, Wong DM, George MH. The induction of hepatocellular neoplasia by trichloroacetic acid administered in the drinking water of the male B6C3F1 mouse. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2008; 71:1056-68. [PMID: 18569617 DOI: 10.1080/15287390802111952] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The prevalence (percent of animals with a tumor) and multiplicity (number of tumors per animal) of hepatocellular neoplasia in the male B6C3F1 mouse exposed to trichloroacetic acid (TCA) in the drinking water were determined. Male mice were exposed to 0.05, 0.5, and 5 g/L TCA for 60 wk (Study 1), to 4.5 g/L TCA for 104 wk (Study 2) and to 0.05 and 0.5 g/L TCA for 104 wk (Study 3). Time-weighted mean daily doses measured for the low, medium, and high dose groups were consistent over the three studies, 6-8, 58-68, and 572-602 mg/kg-d for the 0.05, 0.5, and the 4.5-5 g/L treatment groups, respectively. No significant changes in animal survival were noted across the studies. A significant increase in the prevalence and multiplicity of hepatocellular tumors was found in the 58-68 and 572-602 mg/kg/d TCA dose groups. Nonhepatoproliferative changes (cytoplasmic alterations, inflammation, and necrosis) in mice treated with TCA were mild and dose related. A TCA-induced increase in liver palmitoyl CoA oxidase activity, a marker of peroxisome proliferation, correlated with tumor induction. A linear association was found between peroxisome proliferation and tumor induction. Sporadic increases in the labeling index of nuclei outside of proliferative lesions were observed at carcinogenic doses throughout the studies. Given that there are no compelling data demonstrating genotoxic activity of either TCA or any metabolite, data are consistent with an epigenetic mode of action. The studies provide dose-response data on the development of hepatocellular neoplasia in male mice over a lifetime exposure to TCA. A no-observed-effect-level (NOEL) of 6 mg/kg/d was calculated for neoplastic and nonproliferative liver pathology.
Collapse
Affiliation(s)
- Anthony B DeAngelo
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | |
Collapse
|
35
|
Pozzi A, Capdevila JH. PPARalpha Ligands as Antitumorigenic and Antiangiogenic Agents. PPAR Res 2008; 2008:906542. [PMID: 18725983 PMCID: PMC2517125 DOI: 10.1155/2008/906542] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Accepted: 07/01/2008] [Indexed: 12/30/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor family of ligand-activated transcription factors. This subfamily is composed of three members-PPARalpha, PPARdelta, and PPARgamma-that differ in their cell and tissue distribution as well as in their target genes. PPARalpha is abundantly expressed in liver, brown adipose tissue, kidney, intestine, heart, and skeletal muscle; and its ligands have been used to treat diseases such as obesity and diabetes. The recent finding that members of the PPAR family, including the PPARalpha, are expressed by tumor and endothelial cells together with the observation that PPAR ligands regulate cell growth, survival, migration, and invasion, suggested that PPARs also play a role in cancer. In this review, we focus on the contribution of PPARalpha to tumor and endothelial cell functions and provide compelling evidence that PPARalpha can be viewed as a new class of ligand activated tumor "suppressor" gene with antiangiogenic and antitumorigenic activities. Given that PPAR ligands are currently used in medicine as hypolipidemic drugs with excellent tolerance and limited toxicity, PPARalpha activation might offer a novel and potentially low-toxic approach for the treatment of tumor-associated angiogenesis and cancer.
Collapse
Affiliation(s)
- Ambra Pozzi
- Department of Medicine, Division of Nephrology and Hypertension, S-3223 Medical Center North, Vanderbilt University, Nashville, TN 37232, USA
| | - Jorge H. Capdevila
- Department of Medicine, Division of Nephrology and Hypertension, S-3223 Medical Center North, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
36
|
Seedorf U, Aberle J. Emerging roles of PPARdelta in metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2007; 1771:1125-31. [PMID: 17588807 DOI: 10.1016/j.bbalip.2007.04.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 04/05/2007] [Accepted: 04/25/2007] [Indexed: 02/04/2023]
Abstract
PPARdelta differs from the other two PPAR isotypes (alpha and gamma) by its more wide-spread tissue-specific expression pattern, its involvement in developmental processes and its profound impact on muscle and heart fat metabolism. Activation of PPARdelta modulates inflammatory responses of macrophages and is linked to altered lipoprotein metabolism, most importantly a significant raise of HDL cholesterol. PPARdelta activation in the liver decreases hepatic glucose output, thereby contributing to improved glucose tolerance and insulin sensitivity. Several studies have shown that PPARdelta polymorphisms are associated with plasma lipid levels, body mass index and the risk for diabetes and coronary heart disease. These findings support that high affinity PPARdelta agonists may be promising drugs of the future to treat the metabolic syndrome which is an expanding overweight-related health threat characterized by insulin resistance, hyperglycemia, dyslipidemia, hypertension, and accelerated atherosclerosis.
Collapse
Affiliation(s)
- Udo Seedorf
- Leibniz-Institut für Arteriosklerosforschung an der Universität Münster, Münster, Germany.
| | | |
Collapse
|
37
|
Rusyn I, Peters JM, Cunningham ML. Modes of action and species-specific effects of di-(2-ethylhexyl)phthalate in the liver. Crit Rev Toxicol 2006; 36:459-79. [PMID: 16954067 PMCID: PMC2614359 DOI: 10.1080/10408440600779065] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The industrial plasticizer di-(2-ethylhexyl)phthalate (DEHP) is used in manufacturing of a wide variety of polyvinyl chloride (PVC)-containing medical and consumer products. DEHP belongs to a class of chemicals known as peroxisome proliferators (PPs). PPs are a structurally diverse group of compounds that share many (but perhaps not all) biological effects and are characterized as non-genotoxic rodent carcinogens. This review focuses on the effect of DEHP in liver, a primary target organ for the pleiotropic effects of DEHP and other PPs. Specifically, liver parenchymal cells, identified herein as hepatocytes, are a major cell type that are responsive to exposure to PPs, including DEHP; however, other cell types in the liver may also play a role. The PP-induced increase in the number and size of peroxisomes in hepatocytes, so called 'peroxisome proliferation' that results in elevation of fatty acid metabolism, is a hallmark response to these compounds in the liver. A link between peroxisome proliferation and tumor formation has been a predominant, albeit questioned, theory to explain the cause of a hepatocarcinogenic effect of PPs. Other molecular events, such as induction of cell proliferation, decreased apoptosis, oxidative DNA damage, and selective clonal expansion of the initiated cells have been also been proposed to be critically involved in PP-induced carcinogenesis in liver. Considerable differences in the metabolism and molecular changes induced by DEHP in the liver, most predominantly the activation of the nuclear receptor peroxisome proliferator-activated receptor (PPAR)alpha, have been identified between species. Both sexes of rats and mice develop adenomas and carcinomas after prolonged feeding with DEHP; however, limited DEHP-specific human data are available, even though exposure to DEHP and other phthalates is common in the general population. This likely constitutes the largest gap in our knowledge on the potential for DEHP to cause liver cancer in humans. Overall, it is believed that the sequence of key events that are relevant to DEHP-induced liver carcinogenesis in rodents involves the following events whereby the combination of the molecular signals and multiple pathways, rather than a single hallmark event (such as induction of PPARalpha and peroxisomal genes, or cell proliferation) contribute to the formation of tumors: (i) rapid metabolism of the parental compound to primary and secondary bioactive metabolites that are readily absorbed and distributed throughout the body; (ii) receptor-independent activation of hepatic macrophages and production of oxidants; (iii) activation of PPARalpha in hepatocytes and sustained increase in expression of peroxisomal and non-peroxisomal metabolism-related genes; (iv) enlargement of many hepatocellular organelles (peroxisomes, mitochondria, etc.); (v) rapid but transient increase in cell proliferation, and a decrease in apoptosis; (vi) sustained hepatomegaly; (vii) chronic low-level oxidative stress and accumulation of DNA damage; (viii) selective clonal expansion of the initiated cells; (ix) appearance of the pre-neoplastic nodules; (x) development of adenomas and carcinomas.
Collapse
Affiliation(s)
- Ivan Rusyn
- Department of Environmental Sciences and Engineering, School of Public Health, University of North Carolina, Chapel Hill, 27599-7431, USA.
| | | | | |
Collapse
|
38
|
Kim HS, Ishizaka M, Kazusaka A, Fujita S. Di-(2-ethylhexyl) phthalate suppresses tamoxifen-induced apoptosis in GH3 pituitary cells. Arch Toxicol 2006; 81:27-33. [PMID: 16874505 DOI: 10.1007/s00204-006-0132-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Accepted: 07/06/2006] [Indexed: 10/24/2022]
Abstract
Tamoxifen, an estrogen receptor antagonist, has been clinically used as an antitumor drug and induces apoptosis in GH3 pituitary cells. Although di-(2-ethylhexyl) phthalate (DEHP) is a well-known environmental estrogen and the exposure to this chemical is well expected, reports are limited regarding effects of DEHP on tamoxifen-induced apoptosis in pituitary cells. In the cytotoxicity assay, the reduced cell viability in tamoxifen-treated GH3 cells was reversed by DEHP (250 microM) treatment for 4 days. To characterize cell death, cells were stained using Hoechst 33258. Apoptotic morphological change such as chromatin condensation induced by tamoxifen was suppressed by treatment with DEHP. Flow cytometric analysis revealed that the number of apoptotic cells induced by tamoxifen was significantly decreased by DEHP treatment. Enhanced poly (ADP-ribose) polymerase (PARP) cleavage by tamoxifen treatment was also inhibited by DEHP. These results suggest that DEHP suppresses tamoxifen-induced apoptosis in association with its estrogenic effect in GH3 cells and might counteract the therapeutic effect of tamoxifen.
Collapse
Affiliation(s)
- H-S Kim
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, N18, W9, Sapporo 060-0818, Japan
| | | | | | | |
Collapse
|
39
|
Hastings KL. The use of hepatotoxic signals to predict the safety of peroxisome proliferator-activated receptor agonists. Expert Opin Drug Metab Toxicol 2006; 2:489-92. [PMID: 16859399 DOI: 10.1517/17425255.2.4.489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
40
|
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer related mortality worldwide. The incidence of HCC is rising worldwide, especially in the United States. The overall survival of patients with HCC is grim and currently no efficient secondary prevention or systemic treatments are available. Recent evidence suggests that COX-2 signaling is implicated in hepatocarcinogenesis and COX-2 inhibitors prevent HCC cell growth in vitro and in animal models. However, given the recently reported side effect associated with some of the COX-2 inhibitors, it is imperative to develop chemotherapeutic strategy that simultaneously targets COX-2 and other related key molecules in hepatocarcinogenesis or to utilize agents inhibiting COX-2 signaling in conjunction with other standard chemotherapy or radiation therapy. Such combinational therapeutic approaches are expected to provide synergistic anti-tumor effect with lesser side effect. In this regard, the recently delineated interplay between COX-2-derived PG signaling and other growth-regulatory pathways such as EGFR, Met, iNOS, VEGF and n-3 polyunsaturated fatty acids is expected to provide important therapeutic implications. This review summarizes the recent advances in understanding the mechanisms for COX-2-derived PG signaling in hepatocarcinogenesis and focuses on the newly unveiled interactions between PG cascade and other key signaling pathways that coordinately regulate HCC growth. Understanding these mechanisms and interplays will facilitate the development of more effective chemopreventive and therapeutic strategies.
Collapse
Affiliation(s)
- Tong Wu
- Department of Pathology, University of Pittsburgh School of Medicine, MUH E-740, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|
41
|
Corton JC, Brown-Borg HM. Peroxisome Proliferator-Activated Receptor Coactivator 1 in Caloric Restriction and Other Models of Longevity. J Gerontol A Biol Sci Med Sci 2005; 60:1494-509. [PMID: 16424281 DOI: 10.1093/gerona/60.12.1494] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dietary restriction of calories (caloric restriction [CR]) increases longevity in phylogenetically diverse species. CR retards or prevents age-dependent deterioration of tissues and an array of spontaneous and chemically induced diseases associated with obesity including cardiovascular disease, diabetes, and cancer. An understanding of the molecular mechanisms that underlie the beneficial effects of CR will help identify novel dietary, pharmacological, and lifestyle strategies for slowing the rate of aging and preventing these diseases as well as identify factors which modulate chemical toxicity. Here, we review the involvement of transcriptional coactivator proteins, peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1 (PGC-1) alpha and beta, and regulated nuclear receptors (NR) in mediating the phenotypic changes found in models of longevity which include rodent CR models and mouse mutants in which insulin and/or insulin-like growth factor-I signaling is attenuated. PGC-1alpha is transcriptionally or posttranslationally regulated in mammals by: 1) forkhead box "other" (FoxO) transcription factors through an insulin/insulin-like growth factor-I -dependent pathway, 2) glucagon-stimulated cellular AMP (cAMP) response element binding protein, 3) stress-activated kinase signaling through p38 mitogen-activated protein kinase, and 4) the deacetylase and longevity factor sirtuin 1 (SIRT1). PGC-1alpha and PGC-1beta regulate the ligand-dependent and -independent activation of a large number of NR including PPARalpha and constitutive activated receptor (CAR). These NR regulate genes involved in nutrient and xenobiotic transport and metabolism as well as resistance to stress. CR reverses age-dependent decreases in PGC-1alpha, PPARalpha, and regulated genes. Strategies that target one or multiple PGC-1-regulated NR could be used to mimic the beneficial health effects found in models of longevity.
Collapse
Affiliation(s)
- J Christopher Corton
- United States Environmental Protection Agency, Division of Environmental Carcinogenesis, Research Triangle Park, NC 27711, USA.
| | | |
Collapse
|
42
|
Tachibana K, Kobayashi Y, Tanaka T, Tagami M, Sugiyama A, Katayama T, Ueda C, Yamasaki D, Ishimoto K, Sumitomo M, Uchiyama Y, Kohro T, Sakai J, Hamakubo T, Kodama T, Doi T. Gene expression profiling of potential peroxisome proliferator-activated receptor (PPAR) target genes in human hepatoblastoma cell lines inducibly expressing different PPAR isoforms. NUCLEAR RECEPTOR 2005; 3:3. [PMID: 16197558 PMCID: PMC1262764 DOI: 10.1186/1478-1336-3-3] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Accepted: 10/03/2005] [Indexed: 11/10/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors and commonly play an important role in the regulation of lipid homeostasis. To identify human PPARs-responsive genes, we established tetracycline-regulated human hepatoblastoma cell lines that can be induced to express each human PPAR and investigated the gene expression profiles of these cells. RESULTS The expression of each introduced PPAR gene was investigated using the various concentrations of doxycycline in the culture media. We found that the expression of each PPAR subtype was tightly controlled by the concentration of doxycycline in these established cell lines. DNA microarray analyses using these cell lines were performed with or without adding each subtype ligand and provided much important information on the PPAR target genes involved in lipid metabolism, transport, storage and other activities. Interestingly, it was noted that while ligand-activated PPARdelta induced target gene expression, unliganded PPARdelta repressed these genes. The real-time RT-PCR was used to verify the altered expression of selected genes by PPARs and we found that these genes were induced to express in the same pattern as detected in the microarray analyses. Furthermore, we analysed the 5'-flanking region of the human adipose differentiation-related protein (adrp) gene that responded to all subtypes of PPARs. From the detailed analyses by reporter assays, the EMSAs, and ChIP assays, we determined the functional PPRE of the human adrp gene. CONCLUSION The results suggest that these cell lines are important tools used to identify the human PPARs-responsive genes.
Collapse
Affiliation(s)
- Keisuke Tachibana
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yumi Kobayashi
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Toshiya Tanaka
- Laboratory for System Biology and Medicine, The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo, Japan
| | - Masayuki Tagami
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akira Sugiyama
- Laboratory for System Biology and Medicine, The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo, Japan
- Perseus Proteomics Inc, Tokyo, Japan
| | - Tatsuya Katayama
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Chihiro Ueda
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Daisuke Yamasaki
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kenji Ishimoto
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Mikako Sumitomo
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yasutoshi Uchiyama
- Laboratory for System Biology and Medicine, The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo, Japan
- Perseus Proteomics Inc, Tokyo, Japan
| | - Takahide Kohro
- Laboratory for System Biology and Medicine, The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo, Japan
| | - Juro Sakai
- Laboratory for System Biology and Medicine, The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo, Japan
| | - Takao Hamakubo
- Laboratory for System Biology and Medicine, The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Laboratory for System Biology and Medicine, The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo, Japan
| | - Takefumi Doi
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
43
|
Lupp A, Karge E, Danz M, Deufel T, Oelschläger H, Klinger W. Single and chronic administration of ciprofibrate or of ciprofibrate-glycinate in male Fischer 344 rats: Comparison of the effects on morphological and biochemical parameters in liver and blood. Eur J Drug Metab Pharmacokinet 2005; 30:203-18. [PMID: 16250258 DOI: 10.1007/bf03190621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Fibrates lead to a reduction of serum triglycerides and cholesterol in hyperlipidemic patients. Their therapeutic use, however, can be associated with adverse effects like gastrointestinal disorders, myalgia, myositis and hepatotoxicity. Large doses can even cause hepatocellular carcinoma in rodents. Additionally, interactions with the biotransformation of other compounds at the cytochrome P450 (CYP) system have been observed. Thus, the discovery of new derivatives with less of these side effects is of great interest. In the present study a single (10 mg/kg body weight) or a 4-week (1 or 10 mg/kg body weight daily) oral administration of ciprofibrate or of the newly synthesized ciprofibrate-glycinate was investigated in adult male Fischer 344 rats. Serum lipid concentrations were distinctly decreased after single but only slightly after chronic administration of the two fibrates, whereas liver parameters revealed a slight concentration and time dependent hepatotoxicity. Histologically, a hypertrophy, an eosinophilia, a reduced glycogen content and also an apoptosis of the hepatocytes was observed. Effects were more pronounced after chronic treatment and after application of the higher dosage. All CYP enzymes investigated were induced in a time and concentration dependent manner. Resulting CYP mediated monooxygenase and oxidase activities showed a dependency both on enzyme induction and hepatotoxic effects. With no parameter investigated major differences were seen between ciprofibrate and ciprofibrate-glycinate. Thus, the present investigations revealed no noticeable advantages of ciprofibrate-glycinate over its parent compound ciprofibrate.
Collapse
Affiliation(s)
- Amelie Lupp
- Institute of Pharmacology and Toxicology, Friedrich Schiller University Jena, Germany
| | | | | | | | | | | |
Collapse
|
44
|
Maire MA, Rast C, Vasseur P. Di-(2-ethylhexyl)phthalate (DEHP) increases Bcl-2/Bax ratio and modifies c-myc expression in Syrian hamster embryo (SHE) cells. Toxicol Lett 2005; 158:237-45. [PMID: 15923092 DOI: 10.1016/j.toxlet.2005.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Revised: 04/04/2005] [Accepted: 04/05/2005] [Indexed: 11/18/2022]
Abstract
The objective of this work was to study the anti-apoptotic properties of the non-genotoxic rodent carcinogen, di(2-ethylhexyl)phthalate (DEHP) in Syrian hamster embryo (SHE) cells. We demonstrated that a 24 h pre-treatment of SHE cells with 50 microM DEHP inhibited apoptosis triggered by growth factors deprivation. The RNA expression levels of the regulator genes involved in the apoptotic pathway, bcl-2, bax and of c-myc were measured using Western blotting and RT-PCR. We showed that a 24 h treatment of SHE cells with 50 microM DEHP increased (P < 0.05) the bcl-2 expression, while c-myc expression was decreased. No effect on bax expression was observed in the range of 10-50 microM. The defective regulation of apoptosis caused by DEHP treatment could contribute to its carcinogenicity.
Collapse
Affiliation(s)
- M A Maire
- ESE, Université de Metz, Faculté des Sciences, Campus Bridoux, rue du Général Delestraint, 57070 Metz, France.
| | | | | |
Collapse
|
45
|
Yu J, Leung WK, Chen J, Ebert MPA, Malfertheiner P, Sung JJY. Expression of peroxisome proliferator-activated receptor delta in human gastric cancer and its response to specific COX-2 inhibitor. Cancer Lett 2005; 223:11-7. [PMID: 15890232 DOI: 10.1016/j.canlet.2004.09.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2004] [Revised: 09/25/2004] [Accepted: 09/26/2004] [Indexed: 11/26/2022]
Abstract
Peroxisome proliferator-activated receptor delta (PPARdelta) is ligand-activated transcription factor of the nuclear receptor superfamily which is recently implicated in carcinogenesis. We examined the expression profiles of PPARdelta in human gastric cancer, normal gastric mucosa and gastric cancer cell lines by RT-PCR, Western blot and immunohistochemistry. PPARdelta mRNA and protein was found to be ubiquitously expressed in all 5 gastric cancer cell lines, 40 gastric cancer samples and 10 normal gastric mucosa from non-cancer patients. Positive immunoreactivity was detected in the nuclei of normal and malignant gastric epithelium. Treatment of gastric cell line MKN45 that overexpressed cyclooxygenase-2 (COX-2) with specific COX-2 inhibitor NS398 resulted in a time- and dose-dependent suppression of PPARdelta expression. In contrast, there was no suppression of PPARdelta in MKN28 gastric cell line with low COX-2 expression. Our results demonstrated the ubiquitous expression of PPARdelta in normal and cancer gastric epithelium. Suppression of PPARdelta may be one of the mechanisms underlying the chemopreventive effects of COX-2 inhibitor.
Collapse
Affiliation(s)
- Jun Yu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
46
|
Zhang GY, Ahmed N, Riley C, Oliva K, Barker G, Quinn MA, Rice GE. Enhanced expression of peroxisome proliferator-activated receptor gamma in epithelial ovarian carcinoma. Br J Cancer 2005; 92:113-9. [PMID: 15583697 PMCID: PMC2361744 DOI: 10.1038/sj.bjc.6602244] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) belong to a subclass of nuclear hormone receptor that executes important cellular transcriptional functions. Previous studies have demonstrated the expression of PPARγ in several tumours including colon, breast, bladder, prostate, lung and stomach. This study demonstrates the relative expression of PPARγ in normal ovaries and different pathological grades of ovarian tumours of serous, mucinous, endometrioid, clear cell and mixed subtypes. A total of 56 ovarian specimens including 10 normal, eight benign, 10 borderline, seven grade 1, nine grade 2 and 12 grade 3 were analysed using immunohistochemistry. Immunoreactive PPARγ was not expressed in normal ovaries. Out of eight benign and 10 borderline tumours, only one tumour in each group showed weak cytoplasmic PPARγ expression. In contrast, 26 out of 28 carcinomas studied were positive for PPARγ expression with staining confined to cytoplasmic and nuclear regions. An altered staining pattern of PPARγ was observed in high-grade ovarian tumours with PPARγ being mostly localized in the nuclei with little cytoplasmic immunoreactivity. On the other hand, predominant cytoplasmic staining was observed in lower-grade tumours. Significantly increased PPARγ immunoreactivity was observed in malignant ovarian tumours (grade 1, 2 and 3) compared to benign and borderline tumours (χ2=48.80, P<0.001). Western blot analyses showed significant elevation in the expression of immunoreactive PPARγ in grade 3 ovarian tumours compared with that of normal ovaries and benign ovarian tumours (P<0.01). These findings suggest an involvement of PPARγ in the onset and development of ovarian carcinoma and provide an insight into the regulation of this molecule in the progression of the disease.
Collapse
Affiliation(s)
- G Y Zhang
- Department of Obstetrics and Gynaecology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, PR China
| | - N Ahmed
- Gynaecological Cancer Research Centre, The Royal Women's Hospital, 132 Grattan Street, Carlton, Victoria 3053, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia
- Gynaecological Cancer Research Centre, The Royal Women's Hospital, 132 Grattan Street, Carlton, Victoria 3053, Australia. E-mail:
| | - C Riley
- Gynaecological Cancer Research Centre, The Royal Women's Hospital, 132 Grattan Street, Carlton, Victoria 3053, Australia
| | - K Oliva
- Gynaecological Cancer Research Centre, The Royal Women's Hospital, 132 Grattan Street, Carlton, Victoria 3053, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia
| | - G Barker
- Gynaecological Cancer Research Centre, The Royal Women's Hospital, 132 Grattan Street, Carlton, Victoria 3053, Australia
| | - M A Quinn
- Gynaecological Cancer Research Centre, The Royal Women's Hospital, 132 Grattan Street, Carlton, Victoria 3053, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia
| | - G E Rice
- Gynaecological Cancer Research Centre, The Royal Women's Hospital, 132 Grattan Street, Carlton, Victoria 3053, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
47
|
Bhattacharya N, Dufour JM, Vo MN, Okita J, Okita R, Kim KH. Differential Effects of Phthalates on the Testis and the Liver1. Biol Reprod 2005; 72:745-54. [PMID: 15564602 DOI: 10.1095/biolreprod.104.031583] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Phthalates have been shown to elicit contrasting effects on the testis and the liver, causing testicular degeneration and promoting abnormal hepatocyte proliferation and carcinogenesis. In the present study, we compared the effects of phthalates on testicular and liver cells to better understand the mechanisms by which phthalates cause testicular degeneration. In vivo treatment of rats with di-(2-ethylhexyl) phthalate (DEHP) caused a threefold increase of germ cell apoptosis in the testis, whereas apoptosis was not changed significantly in livers from the same animals. Western blot analyses revealed that peroxisome proliferator-activated receptor (PPAR) alpha is equally abundant in the liver and the testis, whereas PPAR gamma and retinoic acid receptor (RAR) alpha are expressed more in the testis. To determine whether the principal metabolite of DEHP, mono-(2-ethylhexyl) phthalate (MEHP), or a strong peroxisome proliferator, 4-chloro-6(2,3-xylindino)-2-pyrimidinylthioacetic acid (Wy-14,643), have a differential effect in Sertoli and liver cells by altering the function of RAR alpha and PPARs, their nuclear trafficking patterns were compared in Sertoli and liver cells after treatment. Both MEHP and Wy-14,643 increased the nuclear localization of PPAR alpha and PPAR gamma in Sertoli cells, but they decreased the nuclear localization of RAR alpha, as previously shown. Both PPAR alpha and PPAR gamma were in the nucleus and cytoplasm of liver cells, but RAR alpha was predominant in the cytoplasm, regardless of the treatment. At the molecular level, MEHP and Wy-14,643 reduced the amount of phosphorylated mitogen-activated protein kinase (activated MAPK) in Sertoli cells. In comparison, both MEHP and Wy-14,643 increased phosphorylated MAPK in liver cells. These results suggest that phthalates may cause contrasting effects on the testis and the liver by differential activation of the MAPK pathway, RAR alpha, PPAR alpha, and PPAR gamma in these organs.
Collapse
Affiliation(s)
- Nandini Bhattacharya
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, Pullman, Washington 99164, USA
| | | | | | | | | | | |
Collapse
|
48
|
Calfee-Mason KG, Spear BT, Glauert HP. Effects of vitamin E on the NF-κB pathway in rats treated with the peroxisome proliferator, ciprofibrate. Toxicol Appl Pharmacol 2004; 199:1-9. [PMID: 15289085 DOI: 10.1016/j.taap.2004.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2003] [Accepted: 03/08/2004] [Indexed: 10/26/2022]
Abstract
Peroxisome proliferators (PPs) are a diverse group of nongenotoxic compounds, which induce hepatic tumors in rodents. The mechanisms leading to hepatic tumors have not been elucidated, but oxidative stress may play a role in the process. Previous studies in our laboratory have shown that peroxisome proliferators activate the transcription factor nuclear factor-kappa B (NF-kappaB) and that this activation is mediated at least in part by oxidative stress. We therefore hypothesized that increased dietary vitamin E would decrease NF-kappaB DNA binding in rodents treated with ciprofibrate (CIP). In this study, 36 male Sprague-Dawley rats were fed a purified diet containing varying levels of vitamin E (10, 50, 250 ppm alpha-tocopherol acetate). After 28 days on the purified diet, seven animals per vitamin E group received 0.01% CIP in the diet for 10 days. Electrophoretic mobility shift assays (EMSAs) showed that CIP treatment increased DNA binding of NF-kappaB. Increased dietary alpha-tocopherol acetate inhibited CIP-induced NF-kappaB DNA binding. Because NF-kappaB translocates to the nucleus upon the phosphorylation and degradation of inhibitor of IkappaB, we also used Western blots to measure cytosolic protein levels of IkappaBalpha and IkappaBbeta, and the IkappaB kinases, IKKalpha and IKKbeta. IkappaBalpha protein levels were decreased in all three CIP-treated groups, with the 10 ppm vitamin E diet also decreasing IkappaBalpha levels in control rats. No difference in IkappaBbeta protein levels was observed among any of the groups. The CIP-treated rats generally had lower protein levels of IKKalpha and IKKbeta. This study supports our working hypothesis that an increased antioxidant environment can inhibit CIP-mediated NF-kappaB induction.
Collapse
Affiliation(s)
- Karen G Calfee-Mason
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY 40506-0054, USA
| | | | | |
Collapse
|
49
|
Abstract
During the past several years, important advances have been made in our understanding of the mechanisms that regulate the expression of genes that determine drug clearance, including phase I and phase II drug-metabolising enzymes and drug transporters. Orphan nuclear receptors have been recognised as key mediators of drug-induced changes in both metabolism and efflux mechanisms. In this review, we summarise recent findings regarding the function of nuclear receptors in regulating drug-metabolising and transport systems, and the relevance of these receptors to clinical drug-drug interactions and the development of new drugs. Emphasis is given to two newly recognised 'orphan' receptors (the pregnane X receptor [PXR] and the constitutive androstane receptor [CAR]) and their regulation of cytochrome P450 enzymes, such as CYP3A4, CYP2Cs and CYP2B6; and transporters, such as P-glycoprotein (MDR1), multidrug resistance-associated proteins (MRPs) and organic anion transporter peptide 2 (OATP2). Although 'cross-talk' occurs between these two receptors and their target sequences, significant species differences exist between ligand-binding and activation profiles for both receptors, and PXR appears to be the predominant or 'master' regulator of hepatic drug disposition in humans. Several important physiological processes, such as cholesterol synthesis and bile acid metabolism, are also tightly controlled by certain ligand-activated orphan nuclear receptors (farnesoid X receptor [FXR] and liver X receptor [LXR]). In general, their ability to bind a broad range of ligands and regulate an extensive array of genes that are involved in drug clearance and disposition makes these orphan receptors attractive targets for drug development. Drugs have the capacity to alter nuclear receptor expression (modulators) and/or serve as ligands for the receptors (agonists or antagonists), and thus can have synergistic or antagonistic effects on the expression of drug-metabolising enzymes and transporters. Coadministration of drugs that are nuclear receptor agonists or antagonists can lead to severe toxicity, a loss of therapeutic efficacy or an imbalance in physiological substrates, providing a novel molecular mechanism for drug-drug interactions.
Collapse
Affiliation(s)
- Hongbing Wang
- Division of Drug Delivery and Disposition, School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina 27599, USA
| | | |
Collapse
|
50
|
Saez E, Rosenfeld J, Livolsi A, Olson P, Lombardo E, Nelson M, Banayo E, Cardiff RD, Izpisua-Belmonte JC, Evans RM. PPAR gamma signaling exacerbates mammary gland tumor development. Genes Dev 2004; 18:528-40. [PMID: 15037548 PMCID: PMC374235 DOI: 10.1101/gad.1167804] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer cell lines that express the nuclear peroxisome proliferator-activated receptor gamma (PPAR gamma) can be prompted to undergo growth arrest and differentiation when treated with synthetic PPAR gamma ligands. To evaluate the therapeutic potential of increased PPAR gamma signaling in vivo, we generated transgenic mice that express a constitutively active form of PPAR gamma in mammary gland. These mice are indistinguishable from their wild-type littermates. However, when bred to a transgenic strain prone to mammary gland cancer, bigenic animals develop tumors with greatly accelerated kinetics. Surprisingly, in spite of their more malignant nature, bigenic tumors are more secretory and differentiated. The molecular basis of this tumor-promoting effect may be an increase in Wnt signaling, as ligand activation of PPAR gamma potentiates Wnt function in an in vivo model of this pathway. These results suggest that once an initiating event has taken place, increased PPAR gamma signaling serves as a tumor promoter in the mammary gland.
Collapse
Affiliation(s)
- Enrique Saez
- The Salk Institute for Biological Studies, and Howard Hughes Medical Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|