1
|
Wu S, Daston G, Rose J, Blackburn K, Fisher J, Reis A, Selman B, Naciff J. Identifying chemicals based on receptor binding/bioactivation/mechanistic explanation associated with potential to elicit hepatotoxicity and to support structure activity relationship-based read-across. Curr Res Toxicol 2023; 5:100108. [PMID: 37363741 PMCID: PMC10285556 DOI: 10.1016/j.crtox.2023.100108] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
The liver is the most common target organ in toxicology studies. The development of chemical structural alerts for identifying hepatotoxicity will play an important role in in silico model prediction and help strengthen the identification of analogs used in structure activity relationship (SAR)- based read-across. The aim of the current study is development of an SAR-based expert-system decision tree for screening of hepatotoxicants across a wide range of chemistry space and proposed modes of action for clustering of chemicals using defined core chemical categories based on receptor-binding or bioactivation. The decision tree is based on ∼ 1180 different chemicals that were reviewed for hepatotoxicity information. Knowledge of chemical receptor binding, metabolism and mechanistic information were used to group these chemicals into 16 different categories and 102 subcategories: four categories describe binders to 9 different receptors, 11 categories are associated with possible reactive metabolites (RMs) and there is one miscellaneous category. Each chemical subcategory has been associated with possible modes of action (MOAs) or similar key structural features. This decision tree can help to screen potential liver toxicants associated with core structural alerts of receptor binding and/or RMs and be used as a component of weight of evidence decisions based on SAR read-across, and to fill data gaps.
Collapse
|
2
|
Effect of Leucine-enkephalin on Lipid Deposition and GSK-3β/mTOR Signaling in the Liver of Zebrafish. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-023-10506-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
3
|
Zubrzycki A, Wrońska A, Kotulak-Chrząszcz A, Wierzbicki PM, Kmieć Z. Fenofibrate impairs liver function and structure more pronounced in old than young rats. Arch Gerontol Geriatr 2020; 91:104244. [PMID: 32927318 DOI: 10.1016/j.archger.2020.104244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/27/2020] [Accepted: 08/24/2020] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Since old animals are known to accumulate lipids in some organs, we compared effects of fenofibrate (FN) on systemic lipid metabolism, activity of liver marker enzymes and structure in young and old rats. MATERIAL AND METHODS Young and old rats were fed chow supplemented with 0.1 % or 0.5 % FN. After 30 days, intraperitoneal glucose tolerance test (IPGTT) was performed, and blood and liver samples were collected. RESULTS In young rats, 0.1 % FN, but not 0.5 % FN, decreased serum Chol by 74 %, and did not affect TG levels at either doses. In old rats, 0.5 % FN, but not 0.1 % FN, decreased Chol and TG level by 56 % and 49 %, respectively. In young rats, 0.1 % and 0.5 % FN increased serum activity of ALP by 227 % and 260 %, respectively, and did not affect AST and ALT activities. In old rats, only 0.5 % FN increased serum ALP activity by 150 %, respectively. In old rats, neither dose of FN affected serum AST activity, and only 0.5 % FN increased serum ALT activity by 200 %. The histological examination of liver structure revealed that both doses of FN impaired lobular architecture, expansion of bile canaliculi, and degeneration of parenchymal cells with the presence of cells containing fat droplets; administration of FN increased area occupied by collagen fibers. CONCLUSIONS Although 0.5 % FN decreased serum Chol concentration, it increased serum ALP activity and impaired liver structure in both in both age groups of rats. Thus, FN treatment should be under the control of liver function, especially in older patients.
Collapse
Affiliation(s)
- Adrian Zubrzycki
- Department of Histology, Medical University of Gdansk, Gdansk, Poland.
| | - Agata Wrońska
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| | | | | | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
4
|
Li IH, Shih JH, Tsai CS, Chien WC, Kao HH, Pan KT, Cheng YD, Kao LT. Inverse Association of Fibrates and Liver Cancer: A Population-Based Case-Control Study in Taiwan. J Clin Pharmacol 2019; 59:1170-1176. [PMID: 30964561 DOI: 10.1002/jcph.1419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/20/2019] [Indexed: 11/09/2022]
Abstract
This large-scale case-control study in Taiwan elucidated the potential connection between fibrate use and liver cancer by using the Longitudinal Health Insurance Database 2005 with a propensity-score-matching design. In total, 4173 patients diagnosed as having liver cancer were included as cases, and 4173 propensity-score-matched patients without liver cancer were identified as controls. The association between previous fibrate use and liver cancer occurrence was demonstrated using conditional logistic regression. Fibrate use was noted in 371 (8.89%) cases and 481 (11.53%) controls. After adjustments, the cases had significantly lower odds of previous fibrate use than did the controls (adjusted odds ratio 0.70, 95%CI 0.60-0.82); moreover, regardless of the patients' sex, age group, and comorbidities, the cases were less likely to have used fibrates than were the controls. Dose-dependent analysis revealed that 1-695 cumulative defined daily doses of fibrates may significantly induce a protective effect for liver cancer. Although other fibrate dose intervals did not reach statistical significance, the dose-response curve presented the trend of a protective effect for liver cancer among the fibrate users. In summary, fibrate use had a significant protective effect against liver cancer in this Asian population.
Collapse
Affiliation(s)
- I-Hsun Li
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan.,School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Jui-Hu Shih
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan.,School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wu-Chien Chien
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Hui-Han Kao
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ke-Ting Pan
- Institute of Environmental Design and Engineering, Bartlett School, UCL, London, United Kingdom.,Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yih-Dih Cheng
- Department of Pharmacy, China Medical University Hospital, Taichung City, Taiwan
| | - Li-Ting Kao
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan.,School of Pharmacy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,School of Public Health, National Defense Medical Center, Taipei, Taiwan
| |
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
|
Hines IN, Kremer M, Moore SM, Wheeler MD. Impaired T cell-mediated hepatitis in peroxisome proliferator activated receptor alpha (PPARα)-deficient mice. Biol Res 2018; 51:5. [PMID: 29448959 PMCID: PMC5815252 DOI: 10.1186/s40659-018-0153-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/06/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Peroxisome proliferator activated receptor alpha (PPARα), a regulator of enzymes involved in β oxidation, has been reported to influence lymphocyte activation. The purpose of this study was to determine whether PPARα plays a role in T cell-mediated hepatitis induced by Concanavalin A (ConA). METHODS Wild type (wt) or PPARα-deficient (PPARα-/-) mice were treated with ConA (15 mg/kg) by intravenous injection 0, 10 or 24 h prior to sacrifice and serum and tissue collection for analysis of tissue injury, cytokine response, T cell activation and characterization. RESULTS Ten and 24 h following ConA administration, wt mice had significant liver injury as demonstrated by serum transaminase levels, inflammatory cell infiltrate, hepatocyte apoptosis, and expression of several cytokines including interleukin 4 (IL4) and interferon gamma (IFNγ). In contrast, PPARα-/- mice were protected from ConA-induced liver injury with significant reductions in serum enzyme release, greatly reduced inflammatory cell infiltrate, hepatocellular apoptosis, and IFNγ expression, despite having similar levels of hepatic T cell activation and IL4 expression. This resistance to liver injury was correlated with reduced numbers of hepatic natural killer T (NKT) cells and their in vivo responsiveness to alpha-galactosylceramide. Interestingly, adoptive transfer of either wt or PPARα-/- splenocytes reconstituted ConA liver injury and cytokine production in lymphocyte-deficient, severe combined immunodeficient mice implicating PPARα within the liver, possibly through support of IL15 expression and/or suppression of IL12 production and not the lymphocyte as the key regulator of T cell activity and ConA-induced liver injury. CONCLUSION Taken together, these data suggest that PPARα within the liver plays an important role in ConA-mediated liver injury through regulation of NKT cell recruitment and/or survival.
Collapse
Affiliation(s)
- Ian N. Hines
- Department of Nutrition Science, College of Allied Health Sciences, East Carolina University, Health Sciences Bldg. Room 4165F, Greenville, NC 27834 USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Michael Kremer
- Department of General Surgery, University of Ulm, Ulm, Germany
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Sherri M. Moore
- Department of Nutrition Science, College of Allied Health Sciences, East Carolina University, Health Sciences Bldg. Room 4165F, Greenville, NC 27834 USA
| | - Michael D. Wheeler
- Department of Nutrition Science, College of Allied Health Sciences, East Carolina University, Health Sciences Bldg. Room 4165F, Greenville, NC 27834 USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599 USA
| |
Collapse
|
7
|
Gellrich L, Merk D. Therapeutic Potential of Peroxisome Proliferator-Activated Receptor Modulation in Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. NUCLEAR RECEPTOR RESEARCH 2017. [DOI: 10.11131/2017/101310] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
8
|
Peroxisome proliferator-activated receptor alpha deficiency impairs regulatory T cell functions: Possible application in the inhibition of melanoma tumor growth in mice. Biochimie 2016; 131:1-10. [PMID: 27613402 DOI: 10.1016/j.biochi.2016.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 09/01/2016] [Indexed: 11/21/2022]
Abstract
Regulatory T (Treg) cells are important to induce and maintain immunological self-tolerance. Although the progress accomplished in understanding the functional mechanism of Treg cells, intracellular molecules that control the mechanisms of their suppressive capacity are still on investigation. The present study showed that peroxisome proliferator-activated receptor-alpha deficiency impaired the suppressive activity of Treg cells on CD4+CD25- and CD8+ T cell proliferation. In Treg cells, PPARα gene deletion also induced a decrease of migratory abilities, and downregulated the expression of chemokine receptors (CCR-4, CCR-8 and CXCR-4) and p27KIP1 mRNA. Treg cells from PPARα-/- mice also lost their anergic property. Since low Treg activity, as observed in PPARα-/- mice, is known to be associated with the inhibition of tumor growth, we inoculated these mice with B16 melanoma cells and assessed tumor proliferation. In PPARα-/- mice, cancer growth was significantly curtailed, and it was correlated with high expression of granzyme B and perforin mRNA in tumor bed. Degranulation of cytolytic molecules by CD8+ T cells, assessed by a perforin-release marker CD107a expression, was higher in PPARα-/- mice than that in wild-type mice. Tumor-infiltrating lymphocytes (TIL) in melanoma tumors in PPARα-/- mice exhibited high pro-inflammatory Th1 phenotype. Consistently, adoptive transfer into lymphopenic RAG2-/- mice of total PPARα-/-splenic T cells inhibited more the growth rate of B16 tumor than the wild type splenic T cells. Our findings suggest that PPARα deficiency, by diminishing Treg cell functions and upregulating pro-inflammatory T cell phenotype, exerts an in vivo anti-cancer properties.
Collapse
|
9
|
Kim KI, Chung HK, Park JH, Lee YJ, Kang JH. Alpha-fetoprotein-targeted reporter gene expression imaging in hepatocellular carcinoma. World J Gastroenterol 2016; 22:6127-6134. [PMID: 27468205 PMCID: PMC4945974 DOI: 10.3748/wjg.v22.i27.6127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/02/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers in Eastern Asia, and its incidence is increasing globally. Numerous experimental models have been developed to better our understanding of the pathogenic mechanism of HCC and to evaluate novel therapeutic approaches. Molecular imaging is a convenient and up-to-date biomedical tool that enables the visualization, characterization and quantification of biologic processes in a living subject. Molecular imaging based on reporter gene expression, in particular, can elucidate tumor-specific events or processes by acquiring images of a reporter gene’s expression driven by tumor-specific enhancers/promoters. In this review, we discuss the advantages and disadvantages of various experimental HCC mouse models and we present in vivo images of tumor-specific reporter gene expression driven by an alpha-fetoprotein (AFP) enhancer/promoter system in a mouse model of HCC. The current mouse models of HCC development are established by xenograft, carcinogen induction and genetic engineering, representing the spectrum of tumor-inducing factors and tumor locations. The imaging analysis approach of reporter genes driven by AFP enhancer/promoter is presented for these different HCC mouse models. Such molecular imaging can provide longitudinal information about carcinogenesis and tumor progression. We expect that clinical application of AFP-targeted reporter gene expression imaging systems will be useful for the detection of AFP-expressing HCC tumors and screening of increased/decreased AFP levels due to disease or drug treatment.
Collapse
|
10
|
Rieswijk L, Brauers KJJ, Coonen MLJ, Jennen DGJ, van Breda SGJ, Kleinjans JCS. Exploiting microRNA and mRNA profiles generated in vitro from carcinogen-exposed primary mouse hepatocytes for predicting in vivo genotoxicity and carcinogenicity. Mutagenesis 2016; 31:603-15. [PMID: 27338304 DOI: 10.1093/mutage/gew027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The well-defined battery of in vitro systems applied within chemical cancer risk assessment is often characterised by a high false-positive rate, thus repeatedly failing to correctly predict the in vivo genotoxic and carcinogenic properties of test compounds. Toxicogenomics, i.e. mRNA-profiling, has been proven successful in improving the prediction of genotoxicity in vivo and the understanding of underlying mechanisms. Recently, microRNAs have been discovered as post-transcriptional regulators of mRNAs. It is thus hypothesised that using microRNA response-patterns may further improve current prediction methods. This study aimed at predicting genotoxicity and non-genotoxic carcinogenicity in vivo, by comparing microRNA- and mRNA-based profiles, using a frequently applied in vitro liver model and exposing this to a range of well-chosen prototypical carcinogens. Primary mouse hepatocytes (PMH) were treated for 24 and 48h with 21 chemical compounds [genotoxins (GTX) vs. non-genotoxins (NGTX) and non-genotoxic carcinogens (NGTX-C) versus non-carcinogens (NC)]. MicroRNA and mRNA expression changes were analysed by means of Exiqon and Affymetrix microarray-platforms, respectively. Classification was performed by using Prediction Analysis for Microarrays (PAM). Compounds were randomly assigned to training and validation sets (repeated 10 times). Before prediction analysis, pre-selection of microRNAs and mRNAs was performed by using a leave-one-out t-test. No microRNAs could be identified that accurately predicted genotoxicity or non-genotoxic carcinogenicity in vivo. However, mRNAs could be detected which appeared reliable in predicting genotoxicity in vivo after 24h (7 genes) and 48h (2 genes) of exposure (accuracy: 90% and 93%, sensitivity: 65% and 75%, specificity: 100% and 100%). Tributylinoxide and para-Cresidine were misclassified. Also, mRNAs were identified capable of classifying NGTX-C after 24h (5 genes) as well as after 48h (3 genes) of treatment (accuracy: 78% and 88%, sensitivity: 83% and 83%, specificity: 75% and 93%). Wy-14,643, phenobarbital and ampicillin trihydrate were misclassified. We conclude that genotoxicity and non-genotoxic carcinogenicity probably cannot be accurately predicted based on microRNA profiles. Overall, transcript-based prediction analyses appeared to clearly outperform microRNA-based analyses.
Collapse
Affiliation(s)
- Linda Rieswijk
- Department of Toxicogenomics, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, Netherlands and Netherlands Toxicogenomics Centre (NTC), Universiteitssingel 40, 6229ER Maastricht, Netherlands
| | - Karen J J Brauers
- Department of Toxicogenomics, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, Netherlands and
| | - Maarten L J Coonen
- Department of Toxicogenomics, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, Netherlands and Netherlands Toxicogenomics Centre (NTC), Universiteitssingel 40, 6229ER Maastricht, Netherlands
| | - Danyel G J Jennen
- Department of Toxicogenomics, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, Netherlands and Netherlands Toxicogenomics Centre (NTC), Universiteitssingel 40, 6229ER Maastricht, Netherlands
| | - Simone G J van Breda
- Department of Toxicogenomics, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, Netherlands and
| | - Jos C S Kleinjans
- Department of Toxicogenomics, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, Netherlands and Netherlands Toxicogenomics Centre (NTC), Universiteitssingel 40, 6229ER Maastricht, Netherlands
| |
Collapse
|
11
|
Mathew S, Abdel-Hafiz H, Raza A, Fatima K, Qadri I. Host nucleotide polymorphism in hepatitis B virus-associated hepatocellular carcinoma. World J Hepatol 2016; 8:485-498. [PMID: 27057306 PMCID: PMC4820640 DOI: 10.4254/wjh.v8.i10.485] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 12/04/2015] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is etiologically linked with hepatitis B virus (HBV) and is the leading cause of death amongst 80% of HBV patients. Among HBV affected patients, genetic factors are also involved in modifying the risk factors of HCC. However, the genetic factors that regulate progression to HCC still remain to be determined. In this review, we discuss several single nucleotide polymorphisms (SNPs) which were reportedly associated with increased or reduced risk of HCC occurrence in patients with chronic HBV infection such as cyclooxygenase (COX)-2 expression specifically at COX-2 -1195G/A in Chinese, Turkish and Egyptian populations, tumor necrosis factor α and the three most commonly studied SNPs: PAT-/+, Lys939Gln (A33512C, rs2228001) and Ala499Val (C21151T, rs2228000). In genome-wide association studies, strong associations have also been found at loci 1p36.22, 11q22.3, 6p21 (rs1419881, rs3997872, rs7453920 and rs7768538), 8p12 (rs2275959 and rs37821974) and 22q11.21. The genes implicated in these studies include HLA-DQB2, HLA-DQA1, TCF19, HLA-C, UBE2L3, LTL, FDX1, MICA, UBE4B and PG. The SNPs found to be associated with the above-mentioned genes still require validation in association studies in order to be considered good prognostic candidates for HCC. Screening of these polymorphisms is very beneficial in clinical experiments to stratify the higher or lower risk for HCC and may help in designing effective and efficient HCC surveillance programs for chronic HBV-infected patients if further genetic vulnerabilities are detected.
Collapse
|
12
|
Urbatzka R, Galante-Oliveira S, Rocha E, Lobo-da-Cunha A, Castro LFC, Cunha I. Effects of the PPARα agonist WY-14,643 on plasma lipids, enzymatic activities and mRNA expression of lipid metabolism genes in a marine flatfish, Scophthalmus maximus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 164:155-162. [PMID: 25974001 DOI: 10.1016/j.aquatox.2015.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/02/2015] [Accepted: 05/04/2015] [Indexed: 06/04/2023]
Abstract
Fibrates and other lipid regulator drugs are widespread in the aquatic environment including estuaries and coastal zones, but little is known on their chronic effects on non-target organisms as marine fish. In the present study, turbot juveniles were exposed to the PPARα model agonist WY-14,643 for 21 days by repeated injections at the concentrations of 5mg/kg (lo-WY) and 50mg/kg (hi-WY), and samples taken after 7 and 21 days. Enzyme activity and mRNA expression of palmitoyl-CoA oxidase and catalase in the liver were analyzed as first response, which validated the experiment by demonstrating interactions with the peroxisomal fatty acid oxidation and oxidative stress pathways in the hi-WY treatment. In order to get mechanistic insights, alterations of plasma lipids (free cholesterol, FC; HDL associated cholesterol, C-HDL; triglycerides, TG; non-esterified fatty acids, NEFA) and hepatic mRNA expression of 17 genes involved in fatty acid and lipid metabolism were studied. The exposure to hi-WY reduced the quantity of plasma FC, C-HDL, and NEFA. Microsomal triglyceride transfer protein and apolipoprotein E mRNA expression were higher in hi-WY, and indicated an increased formation of VLDL particles and energy mobilization from liver. It is speculated that energy depletion by PPARα agonists may contribute to a higher susceptibility to environmental stressors.
Collapse
Affiliation(s)
- R Urbatzka
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), CIMAR Associated Laboratory (CIMAR LA), University of Porto (U.Porto), Portugal.
| | - S Galante-Oliveira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), CIMAR Associated Laboratory (CIMAR LA), University of Porto (U.Porto), Portugal; Department of Biology & CESAM, University of Aveiro, Portugal
| | - E Rocha
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), CIMAR Associated Laboratory (CIMAR LA), University of Porto (U.Porto), Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (U.Porto), Portugal
| | - A Lobo-da-Cunha
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), CIMAR Associated Laboratory (CIMAR LA), University of Porto (U.Porto), Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (U.Porto), Portugal
| | - L F C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), CIMAR Associated Laboratory (CIMAR LA), University of Porto (U.Porto), Portugal
| | - I Cunha
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), CIMAR Associated Laboratory (CIMAR LA), University of Porto (U.Porto), Portugal
| |
Collapse
|
13
|
Effects of clofibric acid alone and in combination with 17β-estradiol on mRNA abundance in primary hepatocytes isolated from rainbow trout. Toxicol In Vitro 2014; 28:1106-16. [DOI: 10.1016/j.tiv.2014.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 05/11/2014] [Accepted: 05/14/2014] [Indexed: 12/08/2022]
|
14
|
KIM JEEYOUNG, AHN HUIJEONG, WOO HEUNGMYONG, LEE EUNSONG, LEE GEUNSHIK. Generation of liver-specific TGF-α and c-Myc-overexpressing fibroblasts for future creation of a liver cancer porcine model. Mol Med Rep 2014; 10:329-35. [DOI: 10.3892/mmr.2014.2217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 03/28/2014] [Indexed: 11/05/2022] Open
|
15
|
Integrated physiology and systems biology of PPARα. Mol Metab 2014; 3:354-71. [PMID: 24944896 PMCID: PMC4060217 DOI: 10.1016/j.molmet.2014.02.002] [Citation(s) in RCA: 422] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 12/23/2022] Open
Abstract
The Peroxisome Proliferator Activated Receptor alpha (PPARα) is a transcription factor that plays a major role in metabolic regulation. This review addresses the functional role of PPARα in intermediary metabolism and provides a detailed overview of metabolic genes targeted by PPARα, with a focus on liver. A distinction is made between the impact of PPARα on metabolism upon physiological, pharmacological, and nutritional activation. Low and high throughput gene expression analyses have allowed the creation of a comprehensive map illustrating the role of PPARα as master regulator of lipid metabolism via regulation of numerous genes. The map puts PPARα at the center of a regulatory hub impacting fatty acid uptake, fatty acid activation, intracellular fatty acid binding, mitochondrial and peroxisomal fatty acid oxidation, ketogenesis, triglyceride turnover, lipid droplet biology, gluconeogenesis, and bile synthesis/secretion. In addition, PPARα governs the expression of several secreted proteins that exert local and endocrine functions.
Collapse
|
16
|
Nakamura MT, Yudell BE, Loor JJ. Regulation of energy metabolism by long-chain fatty acids. Prog Lipid Res 2013; 53:124-44. [PMID: 24362249 DOI: 10.1016/j.plipres.2013.12.001] [Citation(s) in RCA: 500] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 12/03/2013] [Accepted: 12/04/2013] [Indexed: 12/12/2022]
Abstract
In mammals, excess energy is stored primarily as triglycerides, which are mobilized when energy demands arise. This review mainly focuses on the role of long chain fatty acids (LCFAs) in regulating energy metabolism as ligands of peroxisome proliferator-activated receptors (PPARs). PPAR-alpha expressed primarily in liver is essential for metabolic adaptation to starvation by inducing genes for beta-oxidation and ketogenesis and by downregulating energy expenditure through fibroblast growth factor 21. PPAR-delta is highly expressed in skeletal muscle and induces genes for LCFA oxidation during fasting and endurance exercise. PPAR-delta also regulates glucose metabolism and mitochondrial biogenesis by inducing FOXO1 and PGC1-alpha. Genes targeted by PPAR-gamma in adipocytes suggest that PPAR-gamma senses incoming non-esterified LCFAs and induces the pathways to store LCFAs as triglycerides. Adiponectin, another important target of PPAR-gamma may act as a spacer between adipocytes to maintain their metabolic activity and insulin sensitivity. Another topic of this review is effects of skin LCFAs on energy metabolism. Specific LCFAs are required for the synthesis of skin lipids, which are essential for water barrier and thermal insulation functions of the skin. Disturbance of skin lipid metabolism often causes apparent resistance to developing obesity at the expense of normal skin function.
Collapse
Affiliation(s)
- Manabu T Nakamura
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA.
| | - Barbara E Yudell
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA
| | - Juan J Loor
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA
| |
Collapse
|
17
|
Distinguishing between genotoxic and non-genotoxic hepatocarcinogens by gene expression profiling and bioinformatic pathway analysis. Sci Rep 2013; 3:2783. [PMID: 24089152 PMCID: PMC6505678 DOI: 10.1038/srep02783] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 09/06/2013] [Indexed: 01/09/2023] Open
Abstract
A rapid and sensitive method to determine the characteristics of carcinogens is needed. In this study, we used a microarray-based genomics approach, with a short-term in vivo model, in combination with insights from statistical and mechanistic analyses to determine the characteristics of carcinogens. Carcinogens were evaluated based on the different mechanisms involved in the responses to genotoxic carcinogens and non-genotoxic carcinogens. Gene profiling was performed at two time points after treatment with six training and four test carcinogens. We mapped the DEG (differentially expressed gene)-related pathways to analyze cellular processes, and we discovered significant mechanisms that involve critical cellular components. Classification results were further supported by Comet and Micronucleus assays. Mechanistic studies based on gene expression profiling enhanced our understanding of the characteristics of different carcinogens. Moreover, the efficiency of this study was demonstrated by the short-term nature of the animal experiments that were conducted.
Collapse
|
18
|
Urbatzka R, Galante-Oliveira S, Rocha E, Castro LFC, Cunha I. Tissue expression of PPAR-α isoforms in Scophthalmus maximus and transcriptional response of target genes in the heart after exposure to WY-14643. FISH PHYSIOLOGY AND BIOCHEMISTRY 2013; 39:1043-1055. [PMID: 23266856 DOI: 10.1007/s10695-012-9761-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 12/13/2012] [Indexed: 06/01/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are involved in the regulation of lipid and carbohydrate metabolism and can be activated either by natural ligands as fatty acids or by synthetic ligands including several environmental chemicals. In this study, two PPARα isoforms (α1 and α2) were analyzed in turbot (Scophthalmus maximus) for a different tissue distribution. PPARα1 was ubiquitously expressed, while the PPARα2 was predominantly expressed in the heart. Following this result, turbot juveniles were exposed by injection to a synthetic selective PPARα agonist, WY-14643, for 14 days. Suppression subtractive hybridization (SSH) was performed with pools of heart samples of control and exposed fish to get insights into PPARα-regulated genes in the heart of juvenile turbot. Four genes were positively identified in the forward-subtracted and 12 genes in the reverse-subtracted cDNA SSH library, corresponding to the down-regulated and up-regulated genes in response to the WY-14643 treatment, respectively. The confirmation of these results in individual samples of juvenile turbot exposed to WY-14643 revealed a statistically significant mRNA induction of two cardiac muscle proteins (myosin light chain 2 and tropomyosin 4), which were shown to be involved in heart contraction and heartbeat regulation in other teleost species. Herewith, we showed for the first time that PPARα2 is predominantly expressed in the heart and that a PPARα agonist can induce the mRNA expression of cardiac muscle proteins in teleosts.
Collapse
Affiliation(s)
- R Urbatzka
- Laboratory of Cellular, Molecular and Analytical Studies, Interdisciplinary Centre of Marine and Environmental Research, CIMAR Associated Laboratory, University of Porto (U.Porto), Rua dos Bragas 289, 4050-123 Porto, Portugal.
| | | | | | | | | |
Collapse
|
19
|
El-Shinnawy NA. The therapeutic applications of celery oil seed extract on the plasticizer di(2-ethylhexyl) phthalate toxicity. Toxicol Ind Health 2013; 31:355-66. [PMID: 23377116 DOI: 10.1177/0748233713475515] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The present study investigated the impact of two doses, 500 mg/kg and 1000 mg/kg, of di(2-ethylhexyl) phthalate (DEHP) and studied the possible therapeutic dose of celery oil seed extract for 6 weeks on some atheroscelerogenic, obesogenic, antioxidant and liver functions in rats. Both doses of DEHP caused over-expression of peroxisome proliferator-activated receptor alpha (PPARα) messenger RNA with significant increase in liver weights, relative liver weights, serum cholesterol (Chol), triglycerides, low-density lipoprotein Chol, liver total lipids, along with an increase in the activities of serum aspartate aminotransferase, alanine aminotransferase, serum endothelin 1 and liver tissue thiobarbituric acid reactive substances (TBARS). Additionally, DEHP administration to rats resulted in significant decrease in final body weights, serum total protein, albumin, liver total protein and serum total nitric oxide. Our study confirmed the role of oral combination of Apium graveolens (celery) oil seed extract at small cumulative doses (50 µl/kg for 6 weeks) with DEHP in ameliorating the toxicological effects of DEHP, which was revealed in reducing the expression of PPARα, lipid profile, with restoring liver functions, vascular oxidative stress and inhibition of TBARS activity.
Collapse
Affiliation(s)
- Nashwa A El-Shinnawy
- Department of Zoology, Women College for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| |
Collapse
|
20
|
Rogue A, Renaud MP, Claude N, Guillouzo A, Spire C. Comparative gene expression profiles induced by PPARγ and PPARα/γ agonists in rat hepatocytes. Toxicol Appl Pharmacol 2011; 254:18-31. [PMID: 21515302 DOI: 10.1016/j.taap.2011.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/21/2011] [Accepted: 04/07/2011] [Indexed: 02/09/2023]
Abstract
Species-differential toxic effects have been described with PPARα and PPARγ agonists between rodent and human liver. PPARα agonists (fibrates) are potent hypocholesterolemic agents in humans while they induce peroxisome proliferation and tumors in rodent liver. By contrast, PPARγ agonists (glitazones) and even dual PPARα/γ agonists (glitazars) have caused idiosyncratic hepatic and nonhepatic toxicities in human without evidence of any damage in rodent during preclinical studies. The mechanisms involved in such differences remain largely unknown. Several studies have identified the major target genes of PPARα agonists in rodent liver while no comprehensive analysis has been performed on gene expression changes induced by PPARγ and dual PPARα/γ agonists. Here, we investigated transcriptomes of rat hepatocytes after 24h treatment with two PPARγ (troglitazone and rosiglitazone) and two PPARα/γ (muraglitazar and tesaglitazar) agonists. Although, hierarchical clustering revealed a gene expression profile characteristic of each PPAR agonist class, only a limited number of genes was specifically deregulated by glitazars. Functional analyses showed that many genes known as PPARα targets were also modulated by both PPARγ and PPARα/γ agonists and quantitative differences in gene expression profiles were observed between these two classes. Moreover, most major genes modulated in rat hepatocytes were also found to be deregulated in rat liver after tesaglitazar treatment. Taken altogether, these results support the conclusion that differential toxic effects of PPARα and PPARγ agonists in rodent liver do not result from transcriptional deregulation of major PPAR target genes but rather from qualitative and/or quantitative differential responses of a small subset of genes.
Collapse
Affiliation(s)
- Alexandra Rogue
- UMR INSERM U991, Faculté des Sciences Pharmaceutiques et Biologiques, Rennes, France
| | | | | | | | | |
Collapse
|
21
|
Wanders RJA, Komen J, Kemp S. Fatty acid omega-oxidation as a rescue pathway for fatty acid oxidation disorders in humans. FEBS J 2010; 278:182-94. [PMID: 21156023 DOI: 10.1111/j.1742-4658.2010.07947.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Fatty acids (FAs) can be degraded via different mechanisms including α-, β- and ω-oxidation. In humans, a range of different genetic diseases has been identified in which either mitochondrial FA β-oxidation, peroxisomal FA β-oxidation or FA α-oxidation is impaired. Treatment options for most of these disorders are limited. This has prompted us to study FA ω-oxidation as a rescue pathway for these disorders, based on the notion that if the ω-oxidation of specific FAs could be upregulated one could reduce the accumulation of these FAs and the subsequent detrimental effects in the different groups of disorders. In this minireview, we describe our current state of knowledge in this area with special emphasis on Refsum disease and X-linked adrenoleukodystrophy.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
22
|
Fenofibrate induces effective apoptosis in mantle cell lymphoma by inhibiting the TNFalpha/NF-kappaB signaling axis. Leukemia 2010; 24:1476-86. [PMID: 20520642 DOI: 10.1038/leu.2010.117] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Mantle cell lymphoma (MCL) is a type of aggressive B-cell non-Hodgkin's lymphoma characterized by frequent resistance to conventional chemotherapy. In this study we provided evidence that fenofibrate, which is widely known as an agonist for peroxisome proliferator-activated receptor-alpha (PPARalpha), can induce effective apoptosis in treating MCL cells. Addition of fenofibrate to MCL cell lines significantly decreased the number of viable cells by 50% at approximately 20 microM at 72 h. This decrease in cell growth was due to apoptosis, as evidenced by the cleavage of caspase 3 and poly(ADP-ribose) polymerase. The fenofibrate-mediated effects were not significantly affected by GW6471, a specific PPARalpha antagonist. Using an apoptosis pathway-specific oligonucleotide array, we found that fenofibrate significantly downregulated several pro-survival genes, including tumor necrosis factor-alpha (TNFalpha). Importantly, addition of recombinant TNF-alpha conferred partial protection against fenofibrate-induced apoptosis. Fenofibrate also decreased the nuclear translocation of nuclear factor (NF)-kappaB-p65 and significantly inhibited the DNA binding of NF-kappaB in a dose-dependent manner. To conclude, fenofibrate shows efficacy against MCL, and the mechanism can be attributed to its inhibitory effects on the TNF-alpha/NF-kappaB signaling axis. In view of the documented safety of fenofibrate in humans, it may provide a valuable therapeutic option for MCL patients.
Collapse
|
23
|
Inflammatory mediators and insulin resistance in obesity: role of nuclear receptor signaling in macrophages. Mediators Inflamm 2010; 2010:219583. [PMID: 20508742 PMCID: PMC2874923 DOI: 10.1155/2010/219583] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 03/16/2010] [Indexed: 02/07/2023] Open
Abstract
Visceral obesity is coupled to a general low-grade chronic inflammatory state characterized by macrophage activation and inflammatory cytokine production, leading to insulin resistance (IR). The balance between proinflammatory M1 and antiinflammatory M2 macrophage phenotypes within visceral adipose tissue appears to be crucially involved in the development of obesity-associated IR and consequent metabolic abnormalities. The ligand-dependent transcription factors peroxisome proliferator activated receptors (PPARs) have recently been implicated in the determination of the M1/M2 phenotype. Liver X receptors (LXRs), which form another subgroup of the nuclear receptor superfamily, are also important regulators of proinflammatory cytokine production in macrophages. Disregulation of macrophage-mediated inflammation by PPARs and LXRs therefore underlies the development of IR. This review summarizes the role of PPAR and LXR signaling in macrophages and current knowledge about the impact of these actions in the manifestation of IR and obesity comorbidities such as liver steatosis and diabetic osteopenia.
Collapse
|
24
|
Islinger M, Cardoso MJR, Schrader M. Be different--the diversity of peroxisomes in the animal kingdom. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:881-97. [PMID: 20347886 DOI: 10.1016/j.bbamcr.2010.03.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 03/15/2010] [Accepted: 03/18/2010] [Indexed: 10/19/2022]
Abstract
Peroxisomes represent so-called "multipurpose organelles" as they contribute to various anabolic as well as catabolic pathways. Thus, with respect to the physiological specialization of an individual organ or animal species, peroxisomes exhibit a functional diversity, which is documented by significant variations in their proteome. These differences are usually regarded as an adaptational response to the nutritional and environmental life conditions of a specific organism. Thus, human peroxisomes can be regarded as an in part physiologically unique organellar entity fulfilling metabolic functions that differ from our animal model systems. In line with this, a profound understanding on how peroxisomes acquired functional heterogeneity in terms of an evolutionary and mechanistic background is required. This review summarizes our current knowledge on the heterogeneity of peroxisomal physiology, providing insights into the genetic and cell biological mechanisms, which lead to the differential localization or expression of peroxisomal proteins and further gives an overview on peroxisomal biochemical pathways, which are specialized in different animal species and organs. Moreover, it addresses the impact of proteome studies on our understanding of differential peroxisome function describing the utility of mass spectrometry and computer-assisted algorithms to identify peroxisomal target sequences for the detection of new organ- or species-specific peroxisomal proteins.
Collapse
Affiliation(s)
- M Islinger
- Department of Anatomy and Cell Biology, Ruprecht-Karls University, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
25
|
Makino T, Kinoshita J, Arakawa S, Ito K, Ando Y, Yamoto T, Teranishi M, Sanbuissho A, Nakayama H. Comprehensive analysis of hepatic gene and protein expression profiles on phenobarbital- or clofibrate-induced hepatic hypertrophy in dogs. J Toxicol Sci 2010; 34:647-61. [PMID: 19952500 DOI: 10.2131/jts.34.647] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In order to characterize the hepatic effects of phenobarbital (PB) and clofibrate (CPIB) in dogs, PB and CPIB were administered to male beagle dogs for 14 days, and biochemical and histopathological examinations and comprehensive genomic and proteomic analyses, including GeneChip analysis and proteomics analysis using the 2-dimension difference gel electrophoresis (2D-DIGE) technique, were performed. Both compounds caused centrilobular hepatocellular hypertrophy, which were related to smooth endoplasmic reticulum (SER) proliferation in PB-treated dogs and to mitochondrial proliferation in CPIB-treated dogs. In the PB-treated dogs, drug-metabolizing enzyme induction was observed by Western blot and genomic analyses. CYP proteins could not be detected by the 2D-DIGE analysis, but increases in several endoplasmic reticulum (ER)-related proteins were observed. In the CPIB-treated dogs, drug-metabolizing enzyme induction was not clearly observed by any of Western blot, genomic and proteomic analyses. Genomic and proteomic analyses revealed that mitochondrial genes and proteins, including carnitine palmytoiltransferase II, acyl-CoA deheydrogenase and hydroxyacyl-CoA dehydrogenase, pyruvate carboxylase and ATP synthase beta chain were induced. There is a relatively good correlation among the morphology and the genomic and proteomic data, but some differences exist between the genomic and proteomic data. Comprehensive evaluation using these techniques in addition to morphological evaluation may provide a useful tool for safety assessment of the liver.
Collapse
Affiliation(s)
- Toshihiko Makino
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., Shizuoka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hattis D, Chu M, Rahmioglu N, Goble R, Verma P, Hartman K, Kozlak M. A preliminary operational classification system for nonmutagenic modes of action for carcinogenesis. Crit Rev Toxicol 2009; 39:97-138. [PMID: 19009457 DOI: 10.1080/10408440802307467] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This article proposes a system of categories for nonmutagenic modes of action for carcinogenesis. The classification is of modes of action rather than individual carcinogens, because the same compound can affect carcinogenesis in more than one way. Basically, we categorize modes of action as: (1) co-initiation (facilitating the original mutagenic changes in stem and progenitor cells that start the cancer process) (e.g. induction of activating enzymes for other carcinogens); (2) promotion (enhancing the relative growth vs differentiation/death of initiated clones (e.g. inhibition of growth-suppressing cell-cell communication); (3) progression (enhancing the growth, malignancy, or spread of already developed tumors) (e.g. suppression of immune surveillance, hormonally mediated growth stimulation for tumors with appropriate receptors by estrogens); and (4) multiphase (e.g., "epigenetic" silencing of tumor suppressor genes). A priori, agents that act at relatively early stages in the process are expected to manifest greater relative susceptibility in early life, whereas agents that act via later stage modes will tend to show greater susceptibility for exposures later in life.
Collapse
Affiliation(s)
- D Hattis
- George Perkins Marsh Institute, Clark University, Worcester, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Gillum N, Karabekian Z, Swift LM, Brown RP, Kay MW, Sarvazyan N. Clinically relevant concentrations of di (2-ethylhexyl) phthalate (DEHP) uncouple cardiac syncytium. Toxicol Appl Pharmacol 2009; 236:25-38. [PMID: 19344669 DOI: 10.1016/j.taap.2008.12.027] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 11/24/2008] [Accepted: 12/18/2008] [Indexed: 11/18/2022]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer found in a variety of polyvinyl chloride (PVC) medical products. The results of studies in experimental animals suggest that DEHP leached from flexible PVC tubing may cause health problems in some patient populations. While the cancerogenic and reproductive effects of DEHP are well recognized, little is known about the potential adverse impact of phthalates on the heart. This study examined the effects of clinically relevant concentrations of DEHP on neonatal rat cardiomyocytes. It was found that application of DEHP to a confluent, synchronously beating cardiac cell network, leads to a marked, concentration-dependent decrease in conduction velocity and asynchronous cell beating. The mechanism behind these changes was a loss of gap junctional connexin-43, documented using Western blot analysis, dye-transfer assay and immunofluorescence. In addition to its effect on electrical coupling, DEHP treatment also affected the mechanical movement of myocyte layers. The latter was linked to the decreased stiffness of the underlying fibroblasts, as the amount of triton-insoluble vimentin was significantly decreased in DEHP-treated samples. The data indicate that DEHP, in clinically relevant concentrations, can impair the electrical and mechanical behavior of a cardiac cell network. Applicability of these findings to human patients remains to be established.
Collapse
Affiliation(s)
- Nikki Gillum
- Pharmacology and Physiology Department, The George Washington University, 2300 Eye Street, Washington, DC 20037, USA
| | | | | | | | | | | |
Collapse
|
28
|
Leenders MWH, Nijkamp MW, Rinkes IHMB. Mouse models in liver cancer research: A review of current literature. World J Gastroenterol 2008; 14:6915-23. [PMID: 19058325 PMCID: PMC2773853 DOI: 10.3748/wjg.14.6915] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer remains one of the most lethal malignancies worldwide. Due to differences in prevalence of etiological factors the incidence of primary liver cancer varies among the world, with a peak in East-Asia. As this disease is still lethal in most of the cases, research has to be done to improve our understanding of the disease, offering insights for possible treatment options. For this purpose, animal models are widely used, especially mouse models. In this review, we describe the different types of mouse models used in liver cancer research, with emphasis on genetically engineered mice used in this field. We focus on hepatocellular carcinoma (HCC), as this is by far the most common type of primary liver cancer, accounting for 70%-85% of cases.
Collapse
|
29
|
Kane CD, Stevens KA, Fischer JE, Haghpassand M, Royer LJ, Aldinger C, Landschulz KT, Zagouras P, Bagley SW, Hada W, Dullea R, Hayward CM, Francone OL. Molecular characterization of novel and selective peroxisome proliferator-activated receptor alpha agonists with robust hypolipidemic activity in vivo. Mol Pharmacol 2008; 75:296-306. [PMID: 18971326 DOI: 10.1124/mol.108.051656] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha) is recognized as the primary target of the fibrate class of hypolipidemic drugs and mediates lipid lowering in part by activating a transcriptional cascade that induces genes involved in the catabolism of lipids. We report here the characterization of three novel PPARalpha agonists with therapeutic potential for treating dyslipidemia. These structurally related compounds display potent and selective binding to human PPARalpha and support robust recruitment of coactivator peptides in vitro. These compounds markedly potentiate chimeric transcription systems in cell-based assays and strikingly lower serum triglycerides in vivo. The transcription networks induced by these selective PPARalpha agonists were assessed by transcriptional profiling of mouse liver after short- and long-term treatment. The induction of several known PPARalpha target genes involved with fatty acid metabolism were observed, reflecting the expected pharmacology associated with PPARalpha activation. We also noted the down-regulation of a number of genes related to immune cell function, the acute phase response, and glucose metabolism, suggesting that these compounds may have anti-inflammatory action in the mammalian liver. Whereas these compounds are efficacious in acute preclinical models, extended safety studies and further clinical testing will be required before the full therapeutic promise of a selective PPARalpha agonist is realized.
Collapse
Affiliation(s)
- Christopher D Kane
- Departments of Atherosclerosis Biology, Pfizer Global Research & Development, Pfizer Inc, Groton, Connecticut 06340, 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
|
Maher JM, Aleksunes LM, Dieter MZ, Tanaka Y, Peters JM, Manautou JE, Klaassen CD. Nrf2- and PPAR alpha-mediated regulation of hepatic Mrp transporters after exposure to perfluorooctanoic acid and perfluorodecanoic acid. Toxicol Sci 2008; 106:319-28. [PMID: 18757529 DOI: 10.1093/toxsci/kfn177] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Perfluorooctanoic acid and perfluorodecanoic acid (PFDA) are commonly used as emulsifiers and surfactants in fluoropolymer manufacturing and are known peroxisome proliferator-activated receptor alpha (PPAR alpha) agonists. PPAR alpha activation induces beta- and omega-oxidation enzymes such as Cyp4a14 and acyl-CoA oxidase, which are a likely cause of subsequent oxidative stress and peroxisome proliferation. Conversely, NF-E2-related factor-2 (Nrf2) is a transcription factor that protects against oxidative stress and inflammation by regulating several detoxification and xenobiotic transporter genes. Because PFDA markedly induces hepatic metabolism and oxidative stress, we hypothesized that PFDA exposure would increase expression of hepatic efflux multidrug resistance-associated protein (Mrp) transporters. A single PFDA dose (80 mg/kg) administered to mice increased hepatic Mrp3 (fourfold) and Mrp4 (31-fold) mRNA expression. Upregulation of Mrp3 and Mrp4 correlated with elevated serum-conjugated bilirubin and bile acids, respectively. To determine the mechanism of Mrp3 and Mrp4 induction, PFDA was administered to Nrf2-null mice, PPAR alpha-null mice, and mice pretreated with gadolinium chloride, a Kupffer cell-depleting chemical capable of inhibiting the inflammatory cytokine response. In both PPAR alpha- and Nrf2-null mice, maximal induction of Mrp3 and Mrp4 mRNA after PFDA administration was attenuated. Gadolinium chloride pretreatment reduced serum and hepatic tumor necrosis factor-alpha levels after PFDA treatment, as well as Mrp4 mRNA expression by 30%, suggesting that Kupffer cell-derived mediators may contribute to Mrp induction. Thus, after PFDA administration, the liver mounts a compensatory hepatoprotective response via PPAR alpha and Nrf2, markedly increasing Mrp3 and Mrp4 expression, with corresponding increases in serum of known Mrp3 and Mrp4 substrates.
Collapse
Affiliation(s)
- Jonathan M Maher
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
As the introduction of the concept on tumor-associated macrophages (TAMs), the two-way role of macrophages in tumor development has been more and more focused. Other than inhibiting the development of tumors as traditional thinking says, macrophages can also promote the proceeding of several kinds of tumors through angiogenesis and matrix remolding, etc. As a special kind of macrophages, Kupffer cells also play a two-way role in the development of liver cancer. The lucubrating of the mechanism of macrophage in the proceeding of tumors must be beneficial to the therapy of tumors.
Collapse
|
33
|
Woo HG, Park ES, Cheon JH, Kim JH, Lee JS, Park BJ, Kim W, Park SC, Chung YJ, Kim BG, Yoon JH, Lee HS, Kim CY, Yi NJ, Suh KS, Lee KU, Chu IS, Roskams T, Thorgeirsson SS, Kim YJ. Gene expression-based recurrence prediction of hepatitis B virus-related human hepatocellular carcinoma. Clin Cancer Res 2008; 14:2056-64. [PMID: 18381945 DOI: 10.1158/1078-0432.ccr-07-1473] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE The poor prognosis of hepatocellular carcinoma (HCC) is, in part, due to the high rate of recurrence even after "curative resection" of tumors. Therefore, it is axiomatic that the development of an effective prognostic prediction model for HCC recurrence after surgery would, at minimum, help to identify in advance those who would most benefit from the treatment, and at best, provide new therapeutic strategies for patients with a high risk of early recurrence. EXPERIMENTAL DESIGN For the prediction of the recurrence time in patients with HCC, gene expression profiles were generated in 65 HCC patients with hepatitis B infections. RESULT Recurrence-associated gene expression signatures successfully discriminated between patients at high-risk and low-risk of early recurrence (P=1.9 x 10(-6), log-rank test). To test the consistency and robustness of the recurrence signature, we validated its prognostic power in an independent HCC microarray data set. CD24 was identified as a putative biomarker for the prediction of early recurrence. Genetic network analysis suggested that SP1 and peroxisome proliferator-activated receptor-alpha might have regulatory roles for the early recurrence of HCC. CONCLUSION We have identified a gene expression signature that effectively predicted early recurrence of HCC independent of microarray platforms and cohorts, and provided novel biological insights into the mechanisms of tumor recurrence.
Collapse
Affiliation(s)
- Hyun Goo Woo
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Verma M, Martin HJ, Haq W, Connor TRO, Maser E, Balendiran GK. Inhibiting wild-type and C299S mutant AKR1B10; a homologue of aldose reductase upregulated in cancers. Eur J Pharmacol 2008; 584:213-21. [PMID: 18325492 PMCID: PMC6193476 DOI: 10.1016/j.ejphar.2008.01.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 12/05/2007] [Accepted: 01/24/2008] [Indexed: 11/16/2022]
Abstract
AKR1B10 is an aldose reductase (AR) homologue overexpressed in liver cancer and various forms of that enzyme in carcinomas catalyze the reduction of anticancer drugs, potential cytostatic drug, and dl-glyceraldehyde but do not catalyze the reduction of glucose. Kinetic parameters for wild-type and C299S mutant AKR1B10 indicate that substitution of serine for cysteine at position 299 reduces the affinity of this protein for dl-glyceraldehyde and enhances its catalytic activity. Fibrates suppress peroxisome proliferation and the development of liver cancer in human. Here we report the potency of fibrate-mediated inhibition of the carbonyl reduction catalyzed by wild-type and C299S mutant AKR1B10 and compare it with known AR inhibitors. Wild-type AKR1B10-catalyzed carbonyl reduction follows pure non-competitive inhibition kinetics using zopolrestat, EBPC or sorbinil, whereas fenofibrate, Wy 14,643, ciprofibrate and fenofibric acid follow mixed non-competitive inhibition kinetics. In contrast, catalysis of reaction by the C299S AKR1B10 mutant is not inhibited by sorbinil and EBPC. Despite these differences, the C299S AKR1B10 mutant still manifests kinetics similar to the wild-type protein with other fibrates including zopolrestat, fenofibrate, Wy 14,346, gemfibrozil and ciprofibrate that show mixed non-competitive inhibition kinetics. The reaction of the mutant AKR1B10 is inhibited by fenofibric acid, but manifests pure non-competitive inhibition kinetics that are different from those demonstrated for the wild-type enzyme.
Collapse
Affiliation(s)
- Malkhey Verma
- Division of Immunology, Beckman Research Institute, City of Hope National Medical Center, 1450 E. Duarte Road, Duarte, CA 91010, USA
| | - Hans-Joerg Martin
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Campus Kiel,Brunswiker Str. 10, 24105 Kiel, Germany
| | - Wahajul Haq
- Medicinal Chemistry Division, Central Drug Research Institute, Lucknow 226001, India
| | - Timothy R. O. Connor
- Division of Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California 91010, USA
| | - Edmund Maser
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Campus Kiel,Brunswiker Str. 10, 24105 Kiel, Germany
| | - Ganesaratnam K. Balendiran
- Division of Immunology, Beckman Research Institute, City of Hope National Medical Center, 1450 E. Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
35
|
He N, Park K, Zhang Y, Huang J, Lu S, Wang L. Epigenetic inhibition of nuclear receptor small heterodimer partner is associated with and regulates hepatocellular carcinoma growth. Gastroenterology 2008; 134:793-802. [PMID: 18325392 DOI: 10.1053/j.gastro.2008.01.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2007] [Accepted: 12/13/2007] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS Aberrant hypermethylation of promoter regions in cytosine-guanine dinucleotides (CpG) islands has been shown to be associated with transcriptional silencing of tumor-suppressor genes in many cancers. This study evaluated the methylation profile and the tumor-suppressive function of the small heterodimer partner (SHP, NR0B2) in the development of human hepatocellular carcinoma (HCC). METHODS Human HCC pathologic specimens and cell lines were used as model systems in this study. RESULTS The expression of SHP is diminished in HCC pathologic specimens and cell lines by epigenetic silencing owing to SHP promoter hypermethylation. In vitro methylation decreased SHP promoter transactivation and nuclear receptor LRH-1 binding, an event that was reversed by demethylation. Overexpression of SHP inhibited HCC foci formation, arrested HCC tumor growth in xenografted nude mice, and increased the sensitivity of HCC cells to apoptotic stimuli. Further analysis of a total of 19 normal liver and 57 HCC specimens showed that down-regulation of SHP gene expression may be a common denominator of HCC. CONCLUSIONS We propose that SHP functions as a novel tumor suppressor in the development of HCC. These findings provide new insight into the molecular mechanisms leading to this common cancer and may have both diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Nan He
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | | | | | | | | |
Collapse
|
36
|
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
|
37
|
Cindoruk M, Kerem M, Karakan T, Salman B, Akin O, Alper M, Erdem O, Unal S. Peroxisome proliferators-activated alpha agonist treatment ameliorates hepatic damage in rats with obstructive jaundice: an experimental study. BMC Gastroenterol 2007; 7:44. [PMID: 18045488 PMCID: PMC2219993 DOI: 10.1186/1471-230x-7-44] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Accepted: 11/28/2007] [Indexed: 11/17/2022] Open
Abstract
Background Peroxisome proliferators-activated receptor alpha (PPARα) activation modulates cholesterol metabolism and suppresses bile acid synthesis. This study aims to evaluate the effect of short-term administration of fenofibrate, a PPARα agonist, on proinflammatory cytokines, apoptosis, and hepatocellular damage in cholestasis. Methods Forty male Wistar rats were randomly divided into four groups: I = sham operated, II = bile duct ligation (BDL), III = BDL + vehicle (gum Arabic), IV = BDL + fenofibrate (100 mg/kg/day). All rats were sacrificed on 7th day after obtaining blood samples and liver tissue. Total bilirubin, aminotransferase (AST), alanine aminotransferase (ALT) and alkaline phosphatase (ALP), gamma-glutamyl transferase, (GGT), tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1 β), and total bile acid (TBA) in serum, and liver damage scores; portal inflammation, necrosis, bile duct number, in liver tissue were evaluated. Apoptosis in liver was also assessed by immunohistochemical staining. Results Fenofibrate administration significantly reduced serum total bilirubin, AST, ALT, ALP, and GGT, TNF-α, IL-1 β levels, and TBA (P < 0.01). Hepatic portal inflammation, hepatic necrosis, number of the bile ducts and apoptosis in rats with BDL were more prominent than the sham-operated animals (P < 0.01). PPARα induction improved all histopathologic parameters (P < 0.01), except for the number of the bile duct, which was markedly increased by fenofibrate therapy (P < 0.01). Conclusion Short-term administration of fenofibrate to the BDL rats exerts beneficial effects on hepatocellular damage and apoptosis.
Collapse
Affiliation(s)
- Mehmet Cindoruk
- Department of Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Balendiran GK, Verma M, Perry E. Chemistory of Fibrates. ACTA ACUST UNITED AC 2007; 1:311-316. [PMID: 34485047 DOI: 10.2174/187231307781662198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since the description of the synthetic chemical clofibrate in 1962, various derivatives of fibrates with a diversity of chemical structures have been developed. Several of these are used clinically to treat dyslipidemia because they are generally effective in lowering elevated plasma triglycerides and cholesterol. Studies suggest that several biochemical mechanisms underlie fibrate-mediated modulation of lipoprotein and related metabolites. These mechanisms are: 1) induced lipoprotein lipolysis; 2) induced hepatic fatty acid uptake and reduced hepatic triglyceride formation; 3) amplified removal of low density lipoprotein (LDL) particles; 4) reduced neutral lipid (cholesteryl ester and triglyceride) exchange between very low density lipoprotein (VLDL) and high density lipoprotein (HDL) resulting from decreased plasma levels of triglyceride-rich lipoprotein (TRL); and 5) increased HDL production and stimulation of reverse cholesterol transport. Recent studies of structure-based inhibitor design strategy revealed that an independent enzyme, aldose reductase (AR), is a target of fibrate activity, an additional biochemical mechanism. AR has been implicated as a major player in the development of diabetes and diabetic complications because of its ability to catalyze the conversion of glucose to sorbitol. This article discusses various targets of fibrate action, biochemical pathways and commonalities in potential molecular interactions.
Collapse
Affiliation(s)
- Ganesaratnam K Balendiran
- Beckman Research Institute and City of Hope National Medical Center, 1450 E. Duarte Road, Duarte, CA 91010, USA
| | - Malkhey Verma
- Beckman Research Institute and City of Hope National Medical Center, 1450 E. Duarte Road, Duarte, CA 91010, USA
| | - Elise Perry
- Beckman Research Institute and City of Hope National Medical Center, 1450 E. Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
39
|
Kaipainen A, Kieran MW, Huang S, Butterfield C, Bielenberg D, Mostoslavsky G, Mulligan R, Folkman J, Panigrahy D. PPARalpha deficiency in inflammatory cells suppresses tumor growth. PLoS One 2007; 2:e260. [PMID: 17327920 PMCID: PMC1800345 DOI: 10.1371/journal.pone.0000260] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Accepted: 02/02/2007] [Indexed: 01/09/2023] Open
Abstract
Inflammation in the tumor bed can either promote or inhibit tumor growth. Peroxisome proliferator-activated receptor (PPAR)α is a central transcriptional suppressor of inflammation, and may therefore modulate tumor growth. Here we show that PPARα deficiency in the host leads to overt inflammation that suppresses angiogenesis via excess production of the endogenous angiogenesis inhibitor thrombospondin-1 and prevents tumor growth. Bone marrow transplantation and granulocyte depletion show that PPARα expressing granulocytes are necessary for tumor growth. Neutralization of thrombospondin-1 restores tumor growth in PPARα-deficient mice. These findings suggest that the absence of PPARα activity renders inflammatory infiltrates tumor suppressive and, thus, may provide a target for inhibiting tumor growth by modulating stromal processes, such as angiogenesis.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/metabolism
- Bone Marrow Transplantation
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/pathology
- Cell Line, Transformed/transplantation
- Corneal Neovascularization/genetics
- Granulocytes/physiology
- Inflammation
- Male
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/genetics
- Melanoma, Experimental/pathology
- Mice
- Mice, Knockout
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Pathologic/prevention & control
- PPAR alpha/deficiency
- PPAR alpha/genetics
- PPAR alpha/physiology
- Radiation Chimera
- Thrombospondin 1/physiology
- Vascular Endothelial Growth Factor A/physiology
Collapse
Affiliation(s)
- Arja Kaipainen
- Vascular Biology Program, Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mark W. Kieran
- Vascular Biology Program, Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sui Huang
- Vascular Biology Program, Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Catherine Butterfield
- Vascular Biology Program, Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Diane Bielenberg
- Vascular Biology Program, Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gustavo Mostoslavsky
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard Mulligan
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Judah Folkman
- Vascular Biology Program, Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dipak Panigrahy
- Vascular Biology Program, Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
40
|
Kane CD, Francone OL, Stevens KA. Differential regulation of the cynomolgus, human, and rat acyl-CoA oxidase promoters by PPARα. Gene 2006; 380:84-94. [PMID: 16828988 DOI: 10.1016/j.gene.2006.05.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Revised: 05/11/2006] [Accepted: 05/15/2006] [Indexed: 11/21/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARalpha) is a member of the nuclear receptor family of transcription factors and is recognized as the molecular target of the hypolipidemic fibrate drugs. Fibrates promote lipid catabolism by inducing genes involved in fatty acid beta-oxidation. In rodents this is accompanied by peroxisome proliferation, and after chronic dosing hepatocarcinoma, whereas epidemiological studies suggest these adverse events are lacking in humans. Rodents such as rats and mice appear particularly sensitive to PPARalpha-induced peroxisome proliferation while humans are resistant. These findings question the utility of rodent models for safety monitoring of experimental PPARalpha agonists and highlight the need for additional preclinical models that display greater physiological relevance for human response. Thus we have focused on elucidating the molecular mechanism of species-dependent peroxisome proliferation by understanding the PPARalpha-dependent regulation of the acyl-CoA oxidase (AOX) promoter, the rate-limiting step of peroxisomal beta-oxidation. We have chosen the cynomolgus monkey as a model that is modestly responsive to peroxisome proliferation and functionally characterized it against the highly responsive rat and non-responsive human species. We report the identification of a putative peroxisome proliferator response element (PPRE) within the 2.3 kb proximal promoter of the cynomolgus monkey AOX gene. Characterization of these promoters using a series of constitutively active, PPARalpha constructs demonstrate that the PPREs within the proximal cynomolgus and human AOX promoters are non-responsive to PPARalpha whereas the rat PPRE is highly responsive. These findings were verified in vivo using a small molecule PPARalpha agonist. Taken together, we demonstrate concordant regulation of the AOX promoter by PPARalpha in cynomolgus monkeys and humans and suggest that this model is superior to rodent models with respect to preclinical evaluation of PPARalpha agonists.
Collapse
Affiliation(s)
- Christopher D Kane
- Pfizer Global Research & Development, Department of Cardiovascular, Metabolic & Endocrine Diseases, MS8220-3201, Groton, Connecticut, 06340, USA.
| | | | | |
Collapse
|
41
|
Abstract
Most xenobiotics that enter the body are subjected to metabolism that functions primarily to facilitate their elimination. Metabolism of certain xenobiotics can also result in the production of electrophilic derivatives that can cause cell toxicity and transformation. Many xenobiotics can also activate receptors that in turn induce the expression of genes encoding xenobiotic-metabolizing enzymes and xenobiotic transporters. However, there are marked species differences in the way mammals respond to xenobiotics, which are due in large part to molecular differences in receptors and xenobiotic-metabolizing enzymes. This presents a problem in extrapolating data obtained with rodent model systems to humans. There are also polymorphisms in xenobiotic-metabolizing enzymes that can impact drug therapy and cancer susceptibility. In an effort to generate more reliable in vivo systems to study and predict human response to xenobiotics, humanized mice are under development.
Collapse
Affiliation(s)
- Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
42
|
Li Y, Nara TY, Nakamura MT. Peroxisome proliferator-activated receptor α is required for feedback regulation of highly unsaturated fatty acid synthesis. J Lipid Res 2005; 46:2432-40. [PMID: 16106047 DOI: 10.1194/jlr.m500237-jlr200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Delta6 desaturase (D6D), the rate-limiting enzyme for highly unsaturated fatty acid (HUFA) synthesis, is induced by essential fatty acid-deficient diets. Sterol regulatory element-binding protein-1c (SREBP-1c) in part mediates this induction. Paradoxically, D6D is also induced by ligands of peroxisome proliferator-activated receptor alpha (PPARalpha). Here, we report a novel physiological role of PPARalpha in the induction of genes specific for HUFA synthesis by essential fatty acid-deficient diets. D6D mRNA induction by essential fatty acid-deficient diets in wild-type mice was diminished in PPARalpha-null mice. This impaired D6D induction in PPARalpha-null mice was not attributable to feedback suppression by tissue HUFAs because PPARalpha-null mice had lower HUFAs in liver phospholipids than did wild-type mice. Furthermore, PPARalpha-responsive genes were induced in wild-type mice under essential fatty acid deficiency, suggesting the generation of endogenous PPARalpha ligand(s). Contrary to genes for HUFA synthesis, the induction of other lipogenic genes under essential fatty acid deficiency was higher in PPARalpha-null mice than in wild-type mice even though mature SREBP-1c protein did not differ between the genotypes. The expression of PPARgamma was markedly increased in PPARalpha-null mice and might have contributed to the induction of genes for de novo lipogenesis. Our study suggests that PPARalpha, together with SREBP-1c, senses HUFA status and confers pathway-specific induction of HUFA synthesis by essential fatty acid-deficient diets.
Collapse
Affiliation(s)
- Yue Li
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | |
Collapse
|
43
|
Cariello NF, Romach EH, Colton HM, Ni H, Yoon L, Falls JG, Casey W, Creech D, Anderson SP, Benavides GR, Hoivik DJ, Brown R, Miller RT. Gene expression profiling of the PPAR-alpha agonist ciprofibrate in the cynomolgus monkey liver. Toxicol Sci 2005; 88:250-64. [PMID: 16081524 DOI: 10.1093/toxsci/kfi273] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Fibrates, such as ciprofibrate, fenofibrate, and clofibrate, are peroxisome proliferator-activated receptor-alpha (PPARalpha) agonists that have been in clinical use for many decades for treatment of dyslipidemia. When mice and rats are given PPARalpha agonists, these drugs cause hepatic peroxisome proliferation, hypertrophy, hyperplasia, and eventually hepatocarcinogenesis. Importantly, primates are relatively refractory to these effects; however, the mechanisms for the species differences are not clearly understood. Cynomolgus monkeys were exposed to ciprofibrate at various dose levels for either 4 or 15 days, and the liver transcriptional profiles were examined using Affymetrix human GeneChips. Strong upregulation of many genes relating to fatty acid metabolism and mitochondrial oxidative phosphorylation was observed; this reflects the known pharmacology and activity of the fibrates. In addition, (1) many genes related to ribosome and proteasome biosynthesis were upregulated, (2) a large number of genes downregulated were in the complement and coagulation cascades, (3) a number of key regulatory genes, including members of the JUN, MYC, and NFkappaB families were downregulated, which appears to be in contrast to the rodent, where JUN and MYC are reported to upregulated after PPARalpha agonist treatment, (4) no transcriptional signal for DNA damage or oxidative stress was observed, and (5) transcriptional signals consistent with an anti-proliferative and a pro-apoptotic effect were seen. We also compared the primate data to literature reports of hepatic transcriptional profiling in PPARalpha-treated rodents, which showed that the magnitude of induction in beta-oxidation pathways was substantially greater in the rodent than the primate.
Collapse
Affiliation(s)
- Neal F Cariello
- GlaxoSmithKline Inc., Safety Assessment, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cheon Y, Nara TY, Band MR, Beever JE, Wallig MA, Nakamura MT. Induction of overlapping genes by fasting and a peroxisome proliferator in pigs: evidence of functional PPARα in nonproliferating species. Am J Physiol Regul Integr Comp Physiol 2005; 288:R1525-35. [PMID: 15650118 DOI: 10.1152/ajpregu.00751.2004] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Peroxisome proliferator-activated receptor α (PPARα), a key regulator of fatty acid oxidation, is essential for adaptation to fasting in rats and mice. However, physiological functions of PPARα in other species, including humans, are controversial. A group of PPARα ligands called peroxisome proliferators (PPs) causes peroxisome proliferation and hepatocarcinogenesis only in rats and mice. To elucidate the role of PPARα in adaptation to fasting in nonproliferating species, we compared gene expressions in pig liver from fasted and clofibric acid (a PP)-fed groups against a control diet-fed group. As in rats and mice, fasting induced genes involved with mitochondrial fatty acid oxidation and ketogenesis in pigs. Those genes were also induced by clofibric acid feeding, indicating that PPARα mediates the induction of these genes. In contrast to rats and mice, little or no induction of genes for peroxisomal or microsomal fatty acid oxidation was observed in clofibric acid-fed pigs. Histology showed no significant hyperplasia or hepatomegaly in the clofibric acid-fed pigs, whereas it showed a reduction of glycogen by clofibric acid, an effect of PPs also observed in rats. Copy number of PPARα mRNA was higher in pigs than in mice and rats, suggesting that peroxisomal proliferation and hyperresponse of several genes to PPs seen only in rats and mice are unrelated to the abundance of PPARα. In conclusion, PPARα is likely to play a central role in adaptation to fasting in pig liver as in rats and mice.
Collapse
Affiliation(s)
- Yewon Cheon
- Division of Nutritional Sciences, Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 905 South Goodwin Ave., Bevier Hall #439, Urbana, IL, USA
| | | | | | | | | | | |
Collapse
|
45
|
Currie RA, Bombail V, Oliver JD, Moore DJ, Lim FL, Gwilliam V, Kimber I, Chipman K, Moggs JG, Orphanides G. Gene Ontology Mapping as an Unbiased Method for Identifying Molecular Pathways and Processes Affected by Toxicant Exposure: Application to Acute Effects Caused by the Rodent Non-Genotoxic Carcinogen Diethylhexylphthalate. Toxicol Sci 2005; 86:453-69. [DOI: 10.1093/toxsci/kfi207] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
46
|
Gloerich J, van Vlies N, Jansen GA, Denis S, Ruiter JPN, van Werkhoven MA, Duran M, Vaz FM, Wanders RJA, Ferdinandusse S. A phytol-enriched diet induces changes in fatty acid metabolism in mice both via PPARalpha-dependent and -independent pathways. J Lipid Res 2005; 46:716-26. [PMID: 15654129 DOI: 10.1194/jlr.m400337-jlr200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Branched-chain fatty acids (such as phytanic and pristanic acid) are ligands for the nuclear hormone receptor peroxisome proliferator-activated receptor alpha (PPARalpha) in vitro. To investigate the effects of these physiological compounds in vivo, wild-type and PPARalpha-deficient (PPARalpha-/-) mice were fed a phytol-enriched diet. This resulted in increased plasma and liver levels of the phytol metabolites phytanic and pristanic acid. In wild-type mice, plasma fatty acid levels decreased after phytol feeding, whereas in PPARalpha-/- mice, the already elevated fatty acid levels increased. In addition, PPARalpha-/- mice were found to be carnitine deficient in both plasma and liver. Dietary phytol increased liver free carnitine in wild-type animals but not in PPARalpha-/- mice. Investigation of carnitine biosynthesis revealed that PPARalpha is likely involved in the regulation of carnitine homeostasis. Furthermore, phytol feeding resulted in a PPARalpha-dependent induction of various peroxisomal and mitochondrial beta-oxidation enzymes. In addition, a PPARalpha-independent induction of catalase, phytanoyl-CoA hydroxylase, carnitine octanoyltransferase, peroxisomal 3-ketoacyl-CoA thiolase, and straight-chain acyl-CoA oxidase was observed. In conclusion, branched-chain fatty acids are physiologically relevant ligands of PPARalpha in mice. These findings are especially relevant for disorders in which branched-chain fatty acids accumulate, such as Refsum disease and peroxisome biogenesis disorders.
Collapse
Affiliation(s)
- J Gloerich
- University of Amsterdam, Academic Medical Center, Departments of Clinical Chemistry and Pediatrics, Laboratory for Genetic Metabolic Diseases, 1100 DE Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Barbier O, Fontaine C, Fruchart JC, Staels B. Genomic and non-genomic interactions of PPARalpha with xenobiotic-metabolizing enzymes. Trends Endocrinol Metab 2004; 15:324-30. [PMID: 15350604 DOI: 10.1016/j.tem.2004.07.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The hypolipidemic properties of fibrates, synthetic activators of the nuclear receptor, peroxisome proliferator-activated receptor alpha (PPARalpha), have been studied extensively. Recent observations indicate, however, that PPARalpha also functions as a regulator of endobiotic and xenobiotic metabolism in rodents and humans. Activators of PPARalpha affect xenobiotic-metabolizing enzymes (XMEs) at different levels. At the genomic level, the expression of numerous cytochrome P450 (CYP) and phase II conjugating genes is altered in a species-distinct manner on treatment with PPARalpha activators. As a result of such regulatory processes, PPARalpha affects the homeostasis of both its own natural ligands and other compounds including bile acids. At the non-genomic level, PPARalpha activators can act as competitive inhibitors for inactivating other molecules, leading to drug-drug interactions. These global effects of PPARalpha activators on the activity of XMEs are of physiological and pharmaceutical importance, and demonstrate that thorough studies of the actions on XMEs of each novel PPARalpha agonist are warranted.
Collapse
Affiliation(s)
- Olivier Barbier
- UR 545 INSERM, Département d'Athérosclérose, Institut Pasteur de Lille and the Faculté de Pharmacie, Université Lille II, Lille, 59019 France
| | | | | | | |
Collapse
|
48
|
Schlezinger JJ, Howard GJ, Hurst CH, Emberley JK, Waxman DJ, Webster T, Sherr DH. Environmental and Endogenous Peroxisome Proliferator-Activated Receptor γ Agonists Induce Bone Marrow B Cell Growth Arrest and Apoptosis: Interactions between Mono(2-ethylhexyl)phthalate, 9-cis-Retinoic Acid, and 15-Deoxy-Δ12,14-prostaglandin J2. THE JOURNAL OF IMMUNOLOGY 2004; 173:3165-77. [PMID: 15322177 DOI: 10.4049/jimmunol.173.5.3165] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The common commercial use of phthalate esters has resulted in significant human exposure to these bioactive compounds. The facts that phthalate ester metabolites, like endogenous PGs, are peroxisome proliferator-activated receptor (PPAR) agonists, and that PPARgamma agonists induce lymphocyte apoptosis suggest that phthalate esters are immunosuppressants that could act together with PGs to modulate early B cell development. In this study we examined the effects of a metabolite of one environmental phthalate, mono(2-ethylhexyl)phthalate (MEHP), and 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)), on developing B cells. MEHP inhibited [(3)H]thymidine incorporation by primary murine bone marrow B cells and a nontransformed murine pro/pre-B cell line (BU-11). Cotreatment with a retinoid X receptor alpha ligand, 9-cis-retinoic acid, decreased [(3)H]thymidine incorporation synergistically, thereby implicating activation of a PPARgamma-retinoid X receptor alpha complex. These results were similar to those obtained with the natural PPARgamma ligand 15d-PGJ(2). At moderate MEHP concentrations (25 or 100 microM for primary pro-B cells and a pro/pre-B cell line, respectively), inhibition of [(3)H]thymidine incorporation resulted primarily from apoptosis induction, whereas at lower concentrations, the inhibition probably reflected growth arrest without apoptosis. Cotreatment of bone marrow B cells with 15d-PGJ(2) and MEHP significantly enhanced the inhibition of [(3)H]thymidine incorporation seen with MEHP alone, potentially mimicking exposure in the bone marrow microenvironment where PG concentrations are high. Finally, MEHP- and 15d-PGJ(2)-induced death does not result from a decrease in NF-kappaB activation. These data demonstrate that environmental phthalates can cooperate with an endogenous ligand, 15d-PGJ(2), to inhibit proliferation of and induce apoptosis in developing bone marrow B cells, potentially via PPARgamma activation.
Collapse
Affiliation(s)
- Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Cornwell PD, De Souza AT, Ulrich RG. Profiling of hepatic gene expression in rats treated with fibric acid analogs. Mutat Res 2004; 549:131-45. [PMID: 15120967 DOI: 10.1016/j.mrfmmm.2003.12.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2003] [Revised: 12/09/2003] [Accepted: 12/10/2003] [Indexed: 04/29/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptors whose ligands include fatty acids, eicosanoids and the fibrate class of drugs. In humans, fibrates are used to treat dyslipidemias. In rodents, fibrates cause peroxisome proliferation, a change that might explain the observed hepatomegaly. In this study, rats were treated with multiple dose levels of six fibric acid analogs (including fenofibrate) for up to two weeks. Pathological analysis identified hepatocellular hypertrophy as the only sign of hepatotoxicity, and only one compound at the highest dose caused any significant increase in serum ALT or AST activity. RNA profiling revealed that the expression of 1288 genes was related to dose or length of treatment and correlated with hepatocellular hypertrophy. This gene list included expression changes that were consistent with increased mitochondrial and peroxisomal beta-oxidation, increased fatty acid transport, increased hepatic uptake of LDL-cholesterol, decreased hepatic uptake of glucose, decreased gluconeogenesis and decreased glycolysis. These changes are likely linked to many of the clinical benefits of fibrate drugs, including decreased serum triglycerides, decreased serum LDL-cholesterol and increased serum HDL-cholesterol. In light of the fact that all six compounds stimulated similar or identical changes in the expression of this set of 1288 genes, these results indicate that hepatomegaly is due to PPARalpha activation, although signaling through other receptors (e.g. PPARgamma, RXR) or through non-receptor pathways cannot be excluded.
Collapse
Affiliation(s)
- Paul D Cornwell
- Rosetta Inpharmatics-Merck Research Laboratories, 401 Terry Ave N, Seattle, WA 98109, USA.
| | | | | |
Collapse
|
50
|
Daniel H, tom Dieck H. Nutrient-gene interactions: a single nutrient and hundreds of target genes. Biol Chem 2004; 385:571-83. [PMID: 15318805 DOI: 10.1515/bc.2004.071] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
AbstractBased on the effects of a selective experimental zinc deficiency in a rodent model we explore the use of transcriptome profiling for assessing nutrient-gene interactions in the liver at the molecular and cellular levels. Zinc deficiency caused pleiotropic alterations in mRNA/protein levels of hundreds of genes. In the context of observed metabolic alterations in hepatic metabolism, possible mechanisms are discussed for how a low zinc status may be sensed and transmitted into changes in various metabolic pathways. However, it also becomes obvious that analysis of such complex nutrient-gene interactions beyond the descriptional level is a real challenge for systems biology.
Collapse
Affiliation(s)
- Hannelore Daniel
- Molecular Nutrition Unit, Technical University of Munich, Life and Food Science Center, Hochfeldweg 2, D-85350 Freising-Weihenstephan, Germany.
| | | |
Collapse
|