1,537
Views
174
CrossRef citations to date
0
Altmetric
Review Article

Mode of action framework analysis for receptor-mediated toxicity: The peroxisome proliferator-activated receptor alpha (PPARα) as a case study

, , , , , , , , & show all
Pages 1-49 | Received 07 Jan 2013, Accepted 14 Aug 2013, Published online: 04 Nov 2013

References

  • Abdellatif AG, Preat V, Taper HS, Roberfroid M. (1991). The modulation of rat liver carcinogenesis by perfluorooctanoic acid, a peroxisome proliferator. Toxicol Appl Pharmacol, 111, 530–7
  • Abdellatif AG, Preat V, Vamecq J, et al. (1990). Peroxisome proliferation and modulation of rat liver carcinogenesis by 2,4-dichlorophenoxyacetic acid, 2,4,5-trichlorophenoxyacetic acid, perfluorooctanoic acid and nafenopindichlorophenoxyacetic acid, 2,4,5-trichlorophenoxyacetic acid, perfluorooctanoic acid and nafenopin. Carcinogenesis, 11, 1899–1902
  • Akiyama TE, Nicol CJ, Fievet C, et al. (2001). Peroxisome proliferator-activated receptor-alpha regulates lipid homeostasis, but is not associated with obesity: studies with congenic mouse lines. J Biol Chem, 276, 39088–93
  • Alsarra IA, Brockmann WG, Cunningham ML, Badr MZ. (2006). Hepatocellular proliferation in response to agonists of peroxisome proliferator-activated receptor alpha: a role for Kupffer cells? J Carcinog, 5, 26
  • Amacher DE, Beck R, Schomaker SJ, Kenny CV. (1997). Hepatic microsomal enzyme induction, beta-oxidation, and cell proliferation following administration of clofibrate, gemfibrozil, or bezafibrate in the CD rat. Toxicol Appl Pharmacol, 142, 143--50
  • Andersen ME, Preston RJ, Maier A, et al. (in press). Dose-response approaches for nuclear receptor-mediated modes of action for liver carcinogenicity: results of a workshop. Crit Rev Toxicol
  • Anderson SP, Dunn C, Laughter A, et al. (2004a). Overlapping transcriptional programs regulated by the nuclear receptors peroxisome proliferator-activated receptor alpha, retinoid X receptor, and liver X receptor in mouse liver. Mol Pharmacol, 66, 1440–52
  • Anderson SP, Howroyd P, Liu J, et al. (2004b). The transcriptional response to a peroxisome proliferator-activated receptor alpha agonist includes increased expression of proteome maintenance genes. J Biol Chem, 279, 52390–8
  • Anderson SP, Dunn CS, Cattley RC, Corton JC. (2001). Hepatocellular proliferation in response to a peroxisome proliferator does not require TNFalpha signaling. Carcinogenesis, 22, 1843–51
  • Anderson SP, Yoon L, Richard EB, et al. (2002). Delayed liver regeneration in peroxisome proliferator-activated receptor-alpha-null mice. Hepatology, 36, 544–54
  • Aoyama T, Peters J, Iritani N, et al. (1998). Altered constitutive expression of fatty acid-metabolizing enzymes in mice lacking the peroxisome proliferator-activated receptor α (PPARα). J Biol Chem, 273, 5678–84
  • Arsura M, Cavin LG. (2005). Nuclear factor-kappaB and liver carcinogenesis. Cancer Lett, 229, 157–69
  • Ashby J, Brady A, Elcombe CR, et al. (1994). Mechanistically-based human hazard assessment of peroxisome proliferator-induced hepatocarcinogenesis. Hum Exp Toxicol, 13, S1–117
  • Austin EW, Okita JR, Okita RT, et al. (1995). Modification of lipoperoxidative effects of dichloroacetate and trichloroacetate is associated with peroxisome proliferation. Toxicology, 97, 59–69
  • Auwerx J, Schoonjans K, Fruchart JC, Staels B. (1996a). Transcriptional control of triglyceride metabolism: fibrates and fatty acids change the expression of the LPL and apo C-III genes by activating the nuclear receptor PPAR. Atherosclerosis, 124, S29–37
  • Auwerx J, Schoonjans K, Fruchart JC, Staels B. (1996b). Regulation of triglyceride metabolism by PPARs: fibrates and thiazolidinediones have distinct effects. J Atheroscler Thromb, 3, 81–9
  • Barber ED, Astill BD, Moran EJ, et al. (1987). Peroxisome induction studies on seven phthalate esters. Toxicol Ind Health, 3, 7–24
  • Barrass NC, Price RJ, Lake BG, Orton TC. (1993). Comparison of the acute and chronic mitogenic effects of the peroxisome proliferators methylclofenapate and clofibric acid in rat liver. Carcinogenesis, 14, 1451–6
  • Bayly AC, Roberts RA, Dive C. (1994). Suppression of liver cell apoptosis in vitro by the non-genotoxic hepatocarcinogen and peroxisome proliferator nafenopin. J Cell Biol, 125, 197–203
  • Becuwe P, Dauça M. (2005). Comparison of cytotoxicity induced by hypolipidemic drugs via reactive oxygen species in human and rodent liver cells. Int J Mol Med, 16, 483–92
  • Bell AR, Savory R, Horley NJ, et al. (1998). Molecular basis of non-responsiveness to peroxisome proliferators: the guinea-pig PPARα is functional and mediates peroxisome proliferator-induced hypolipidaemia. Biochem J, 332, 689–93
  • Bell DR, Plant NJ, Rider CG, et al. (1993). Species-specific induction of cytochrome P-450 4A RNAs: PCR cloning of partial guinea-pig, human and mouse CYP4A cDNAs. Biochem J, 294, 173–80
  • Bentley P, Calder I, Elcombe C, et al. (1993). Hepatic peroxisome proliferation in rodents and its significance for humans. Food Chem Toxicol, 31, 857–907
  • Bichet N, Chard D, Fabre G, et al. (1990). Toxicological studies on a benzofuran derivative. III. Comparison of peroxisome proliferation in rat and human hepatocytes in primary culture. Toxicol Appl Pharmacol, 106, 509–17
  • Biddie SC, Hager GL. (2009). Glucocorticoid receptor dynamics and gene regulation. Stress, 12, 193–205
  • Biddie SC, John S, Hager GL. (2010). Genome-wide mechanisms of nuclear receptor action. Trends Endocrinol Metab, 21, 3–9
  • Biegel LB, Hurtt ME, Frame SR, et al. (2001). Mechanisms of extrahepatic tumor induction by peroxisome proliferators in male CD rats. Toxicol Sci, 60, 44–55
  • Bility MT, Thompson JT, McKee RH, et al. (2004). Activation of mouse and human peroxisome proliferator-activated receptors (PPARs) by phthalate monoesters. Toxicol Sci, 82, 170–82
  • Blumcke S, Schwartzkopff W, Lobeck H, et al. (1983). Influence of fenofibrate on cellular and subcellular liver structure in hyperlipidemic patients. Atherosclerosis, 46, 105–16
  • Bojes HK, Germolec DR, Simeonova P, et al. (1997). Antibodies to tumor necrosis factor alpha prevent increases in cell replication in liver due to the potent peroxisome proliferator WY14 643. Carcinogenesis, 18, 669–74
  • Boobis AR, Cohen SM, Dellarco V, et al. (2006). IPCS framework for analyzing the relevance of a cancer mode of action for humans. Crit Rev Toxicol, 36, 781–92
  • Boobis AR, Datson GP, Preston RJ, Olin SS. (2009). Application of key events analysis to chemical carcinogens and noncarcinogens. Crit Rev Food Sci Nutr, 49, 690–707
  • Boobis AR, Doe JE, Heinrich-Hirsch B, et al. (2008). IPCS framework for analyzing the relevance of a noncancer mode of action for humans. Crit Rev Toxicol, 38, 87–96
  • Bull RJ, Orner GA, Cheng RS, et al. (2002). Contribution of dichloroacetate and trichloroacetate to liver tumor induction in mice by trichloroethylene. Toxicol Appl Pharmacol, 182, 55–65
  • Bull RJ, Sanchez IM, Nelson MA, et al. (1990). Liver tumor induction in B6C3F1 mice by dichloroacetate and trichloroacetate. Toxicology, 63, 341–59
  • Burkhardt S, Mellert W, Reinacher M, Bahnenmann R. (2001). Zonal evaluation of proliferation and apoptosis in mice reveals new mechanistic data for PB, WY 14 643 and CH. Toxicologist, 60, 286
  • Bursch W, Lauer B, Timmermann-Trosiener I, et al. (1984). Controlled death (apoptosis) of normal and putative preneoplastic cells in rat liver following withdrawal of tumor promoters. Carcinogenesis, 5, 453–8
  • Busser MT, Lutz WK. (1987). Stimulation of DNA synthesis in rat and mouse liver by various tumor promoters. Carcinogenesis, 8, 1433–7
  • Cai Y, Appelkvist EL, DePierre JW. (1995). Hepatic oxidative stress and related defenses during treatment of mice with acetylsalicylic acid and other peroxisome proliferators. J Biochem Toxicol, 10, 87–94
  • Caira F, Clémencet MC, Cherkaoui-Malki M, et al. (1998). Differential regulation by a peroxisome proliferator of the different multifunctional proteins in guinea pig: cDNA cloning of the guinea pig D-specific multifunctional protein 2. Biochem J, 330, 1361–8
  • Calfee-Mason KG, Lee EY, Spear BT, Glauert HP. (2008). Role of the p50 subunit of NF-kappaB in vitamin E-induced changes in mice treated with the peroxisome proliferator, ciprofibrate. Food Chem Toxicol, 46, 2062–73
  • Calfee-Mason KG, Spear BT, Glauert HP. (2004). Effects of vitamin E on the NF-kappaB pathway in rats treated with the peroxisome proliferator, ciprofibrate. Toxicol Appl Pharmacol, 199, 1–9
  • Cariello NF, Romach EH, Colton HM, et al. (2005). Gene expression profiling of the PPARα agonist ciprofibrate in the cynomolgus monkey liver. Toxicol Sci, 88, 250–64
  • Cattley RC, Conway JG, Popp JA. (1987). Association of persistent peroxisome proliferation and oxidative injury with hepatocarcinogenicity in female F-344 rats fed di(2-ethylhexyl)phthalate for 2 years. Cancer Lett, 38, 15–22
  • Cattley RC, DeLuca J, Elcombe C, et al. (1998). Do peroxisome proliferating compounds pose a hepatocarcinogenic hazard to humans? Regul Toxicol Pharmacol, 27, 47–60
  • Cattley RC, Glover SE. (1993). Elevated 8-hydroxydeoxyguanosine in hepatic DNA of rats following exposure to peroxisome proliferators: relationship to carcinogenesis and nuclear localization. Carcinogenesis, 14, 2495–9
  • Cattley RC, Marsman DS, Popp JA. (1991). Age-related susceptibility to the carcinogenic effect of the peroxisome proliferator WY14 643 in rat liver. Carcinogenesis, 12, 469–73
  • Cattley RC, Popp JA. (1989). Differences between the promoting activities of the peroxisome proliferator WY-14, 643 and phenobarbital in rat liver. Cancer Res, 49, 3246–51
  • Chan E, Tan CS, Deurenberg-Yap M, et al. (2006). The V227A polymorphism at the PPARΑ locus is associated with serum lipid concentrations and modulates the association between dietary polyunsaturated fatty acid intake and serum high density lipoprotein concentrations in Chinese women. Atherosclerosis, 187, 309–15
  • Chen H, Huang CY, Wilson MW, et al. (1994). Effect of the peroxisome proliferators ciprofibrate and perfluorodecanoic acid on hepatic cell proliferation and toxicity in Sprague Dawley rats. Carcinogenesis, 15, 2847–50
  • Cheung C, Akiyama TE, Ward JM, et al. (2004). Diminished hepatocellular proliferation in mice humanized for the nuclear receptor peroxisome proliferator-activated receptor alpha. Cancer Res, 64, 3849–54
  • Chinje E, Kentish P, Jarnot B, et al. (1994). Induction of the CYP4A subfamily by perfluorodecanoic acid: the rat and the guinea pig as susceptible and non-susceptible species. Toxicol Lett, 71, 69–75
  • Choudhury AI, Chahal S, Bell AR, et al. (2000). Species differences in peroxisome proliferation; mechanisms and relevance. Mutat Res, 448, 201–12
  • Choudhury AI, Sims HM, Horley NJ, et al. (2004). Molecular analysis of peroxisome proliferation in the hamster. Toxicol Appl Pharmacol, 197, 9–18
  • Christensen JG, Gonzales AJ, Cattley RC, Goldsworthy TL. (1998). Regulation of apoptosis in mouse hepatocytes and alteration of apoptosis by nongenotoxic carcinogens. Cell Growth Differ, 9, 815–25
  • Cohen SM, Klaunig J, Meek ME, et al. (2004). Evaluating the human relevance of chemically induced animal tumors. Toxicol Sci, 78, 181–6
  • Cohen SM, Meek ME, Klaunig JE, et al. (2003). The human relevance of information on carcinogenic modes of action: overview. Crit Rev Toxicol, 33, 581–9
  • Columbano A, Ledda-Columbano GM, Pibiri M, et al. (2001). Peroxisome proliferator-activated receptor-alpha mice show enhanced hepatocyte proliferation in response to the hepatomitogen 1,4-bis [2-(3,5-dichloropyridyloxy)] benzene, a ligand of constitutive androstane receptor. Hepatology, 34, 262–6
  • Committee of Principal Investigators. (1978). A cooperative trial in the primary prevention of ischemic heart disease using clofibrate. Br Heart J, 40, 1069–118
  • Conway JG, Tomaszewski KE, Olson MJ, et al. (1989). Relationship of oxidative damage to the hepatocarcinogenicity of the peroxisome proliferators di(2-ethylhexyl)phthalate and Wy-14 643. Carcinogenesis, 10, 513–9
  • Cornu-Chagnon MC, Dupont H, Edgar A. (1995). Fenofibrate: metabolism and species differences for peroxisome proliferation in cultured hepatocytes. Fundam Appl Toxicol, 26, 63–74
  • Corton JC. (2008). Evaluation of the role of peroxisome proliferator-activated receptor alpha (PPARα in mouse liver tumor induction by trichloroethylene and metabolites. Crit Rev Toxicol, 38, 857–75
  • Corton JC. (2010). Mode of action analysis and human relevance of liver tumors induced by PPARα activation. In: Hsu C-H, Stedeford T, eds. Cancer risk assessment: chemical carcinogenesis, hazard evaluation, and risk quantification. Hoboken (NJ): John Wiley & Sons, Inc, 438--81
  • Corton JC, Anderson SP, Stauber A. (2000). Central role of peroxisome proliferator-activated receptors in the actions of peroxisome proliferators. Annu Rev Pharmacol Toxicol, 40, 491–518
  • Corton JC, Apte U, Anderson SP, et al. (2004). Mimetics of caloric restriction include agonists of lipid-activated nuclear receptors. J Biol Chem, 279, 46204–12
  • Corton JC, Bocos C, Moreno ES, et al. (1996). Rat 17 beta-hydroxysteroid dehydrogenase type IV is a novel peroxisome proliferator-inducible gene. Mol Pharmacol, 50, 1157–66
  • Corton JC, Brown-Borg HM. (2005). Peroxisome proliferator-activated receptor gamma coactivator 1 in caloric restriction and other models of longevity. J Gerontol A Biol Sci Med Sci, 60, 1494–509
  • Corton JC, Lapinskas PJ. (2005). Peroxisome proliferator-activated receptors: mediators of phthalate ester-induced effects in the male reproductive tract? Toxicol Sci, 83, 4–17
  • Costet P, Legendre C, More J, et al. (1998). Peroxisome proliferator-activated receptor alpha-isoform deficiency leads to progressive dyslipidemia with sexually dimorphic obesity and steatosis. J Biol Chem, 273, 29577–85
  • Cruciani V, Rast C, Durand MJ, et al. (1997). Comparative effects of clofibrate and methyl clofenapate on morphological transformation and intercellular communication of Syrian hamster embryo cells. Carcinogenesis, 18, 701–6
  • Cunningham ML, Collins BJ, Hejtmancik MR, et al. (2010). Effects of the PPARα agonist and widely used antihyperlipidemic drug gemfibrozil on hepatic toxicity and lipid metabolism. PPAR Res, 2010, Article ID, 681963
  • Currie RA, Bombail V, Oliver JD, et al. (2005). 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, 86, 453–69
  • Czaja MJ. (2007). Cell signaling in oxidative stress-induced liver injury. Semin Liver Dis, 27, 378–89
  • David RM, Moore MR, Cifone MA, et al. (1999). Chronic peroxisome proliferation and hepatomegaly associated with the hepatocellular tumorigenesis of di(2-ethylhexyl)phthalate and the effects of recovery. Toxicol Sci, 50, 195–205
  • David RM, Moore MR, Finney DC, Guest D. (2000a). Chronic toxicity of di(2-ethylhexyl)phthalate in mice. Toxicol Sci, 58, 377–85
  • David RM, Moore MR, Finney DC, Guest D. (2000b). Chronic toxicity of di(2-ethylhexyl)phthalate in rats. Toxicol Sci, 55, 433–43
  • de Duve C. (1996). The peroxisome in retrospect. Ann NY Acad Sci, 804, 1–10
  • de la Iglesia FA, Lewis JE, Buchanan RA, et al. (1982). Light and electron microscopy of liver in hyperlipoproteinemic patients under long-term gemfibrozil treatment. Atherosclerosis, 43, 19–37
  • De Minicis S, Bataller R, Brenner DA. (2006). NADPH oxidase in the liver: defensive, offensive, or fibrogenic? Gastroenterology, 131, 272–5
  • DeAngelo AB, Daniel FB, McMillan L, et al. (1989). Species and strain sensitivity to the induction of peroxisome proliferation by chloroacetic acids. Toxicol Appl Pharmacol, 101, 285–98
  • DeAngelo AB, Daniel FB, Most BM, Olson GR. (1997). Failure of monochloroacetic acid and trichloroacetic acid administered in the drinking water to produce liver cancer in male F344/N rats. J Toxicol Environ Health, 52, 425–45
  • Dees C, Travis C. (1994). Trichloroacetate stimulation of liver DNA synthesis in male and female mice. Toxicol Lett, 70, 343–55
  • Delerive P, Gervois P, Fruchart JC, Staels B. (2000). Induction of IkappaBalpha expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferator-activated receptor-alpha activators. J Biol Chem, 275, 36703–7
  • Dellarco VL, Baetcke K. (2005). A risk assessment perspective: application of mode of action and human relevance frameworks to the analysis of rodent tumor data. Toxicol Sci, 86, 1–3
  • DeLuca JG, Doebber TW, Kelly LJ, et al. (2000). Evidence for peroxisome proliferator-activated receptor (PPAR)alpha-independent peroxisome proliferation: effects of PPARgamma/delta-specific agonists in PPARα-null mice. Mol Pharmacol, 58, 470–6
  • Desvergne B, Ijpenberg A, Devchand PR, Wahli W. (1998). The peroxisome proliferator-activated receptors at the cross-road of diet and hormonal signaling. J Steroid Biochem Mol Biol, 65, 65–74
  • Desvergne B, Wahli W. (1999). Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev, 20, 649–88
  • Dostalek M, Hardy KD, Milne GL, et al. (2008). Development of oxidative stress by cytochrome P450 induction in rodents is selective for barbiturates and related to loss of pyridine nucleotide-dependent protective systems. J Biol Chem, 283, 17147–57
  • Doull J, Cattley R, Elcombe C, et al. (1999). A cancer risk assessment of di(2-ethylhexyl)phthalate: application of the new U.S. EPA Risk Assessment Guidelines. Regul Toxicol Pharmacol, 29, 327–57
  • Duclos S, Bride J, Ramirez LC, Bournot P. (1997). Peroxisome proliferation and beta-oxidation in Fao and MH1C1 rat hepatoma cells, HepG2 human hepatoblastoma cells and cultured human hepatocytes: effect of ciprofibrate. Eur J Cell Biol, 72, 314–23
  • Dwivedi RS, Alvares K, Nemali MR, et al. (1989). Comparison of the peroxisome proliferator-induced pleiotropic response in the liver of nine strains of mice. Toxicol Pathol, 17, 16–26
  • Elcock FJ, Chipman JK, Roberts RA. (1998). The rodent nongenotoxic hepatocarcinogen and peroxisome proliferator nafenopin inhibits intercellular communication in rat but not guinea-pig hepatocytes, perturbing S-phase but not apoptosis. Arch Toxicol, 72, 439–44
  • Elcombe CR. (1985). Species differences in carcinogenicity and peroxisome proliferation due to trichloroethylene: a biochemical human hazard assessment. Arch Toxicol, 8, S6–17
  • Elcombe CR, Bell DR, Elias E, et al. (1996). Peroxisome proliferators: species differences in response of primary hepatocytes cultures. Ann NY Acad Sci, 804, 628–35
  • Elcombe CR, Peffer RC, Wolf DC, et al. (in press). Mode of action and human relevance analysis for nuclear receptormediated liver toxicity: a case study with phenobarbital as a model constitutive androstane receptor (CAR) activator. Crit Rev Toxicol
  • Elliott BM, Elcombe CR. (1987). Lack of DNA damage or lipid peroxidation measured in vivo in the rat liver following treatment with peroxisomal proliferators. Carcinogenesis, 8, 1213–18
  • Escher P, Wahli W. (2000). Peroxisome proliferator-activated receptors: insight into multiple cellular functions. Mutat Res, 448, 121–38
  • Eveillard A, Lasserre F, de Tayrac M, et al. (2009a). Identification of potential mechanisms of toxicity after di-(2-ethylhexyl)-phthalate (DEHP) adult exposure in the liver using a systems biology approach. Toxicol Appl Pharmacol, 236, 282–92
  • Eveillard A, Mselli-Lakhal L, Mogha A, et al. (2009b). Di-(2-ethylhexyl)-phthalate (DEHP) activates the constitutive androstane receptor (CAR): a novel signalling pathway sensitive to phthalates. Biochem Pharmacol, 77, 1735–46
  • Fan CY, Pan J, Usuda N, et al. (1998). Steatohepatitis, spontaneous peroxisome proliferation and liver tumors in mice lacking peroxisomal fatty acyl-CoA oxidase. Implications for peroxisome proliferator-activated receptor alpha natural ligand metabolism. J Biol Chem, 273, 15639–45
  • Feige JN, Gerber A, Casals-Casas C, et al. (2010). The pollutant diethylhexyl phthalate regulates hepatic energy metabolism via species-specific PPARalpha-dependent mechanisms. Environ Health Perspect, 118, 234–41
  • Fischer JG, Glauert HP, Yin T, et al. (2002). Moderate iron overload enhances lipid peroxidation in livers of rats, but does not affect NF-kappaB activation induced by the peroxisome proliferator, Wy-14 643. J Nutr, 132, 2525–31
  • Fitzgerald JE, Sanyer JL, Schardein JL, et al. (1981). Carcinogen bioassay and mutagenicity studies with the hypolipidemic agent gemfibrozil. J Natl Cancer Inst, 67, 1105–16
  • Flavell DM, Pineda Torra I, Jamshidi Y, et al. (2000). Variation in the PPARα gene is associated with altered function in vitro and plasma lipid concentrations in Type II diabetic subjects. Diabetologia, 43, 673–80
  • Forman BM, Chen J, Evans RM. (1997). Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc Natl Acad Sci USA, 94, 4312–7
  • Frick MH, Elo O, Haapa K, et al. (1987). Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med, 317, 1237–45
  • Ganning AE, Olsson MJ, Brunk U, Dallner G. (1991). Effects of prolonged treatment with phthalate ester on rat liver. Pharmacol Toxicol, 67, 392–401
  • Gariot P, Barrat E, Drouin P, et al. (1987). Morphometric study of human hepatic cell modifications induced by fenofibrate. Metabolism, 36, 203–10
  • Gariot P, Barrat E, Mejean L, et al. (1983). Fenofibrate and human liver. Lack of proliferation of peroxisomes. Arch Toxicol, 53, 151–63
  • Gervois P, Torra IP, Chinetti G, et al. (1999). A truncated human peroxisome proliferator-activated receptor alpha splice variant with dominant negative activity. Mol Endocrinol, 13, 1535–49
  • Gill JH, Roberts RA, Dive C. (1998). The nongenotoxic hepatocarcinogen nafenopin suppresses rodent hepatocyte apoptosis induced by TGFb1, DNA damage and FAS. Carcinogenesis, 19, 299–304
  • Givler B, Alberts D, Wollenberg G, et al. (2000). TNF[alpha] is not required for Wy-14, 643-induced proliferation in mice. Toxicologist, 54, A1975
  • Glauert HP, Beaty MM, Clark TD, et al. (1990). Effect of dietary vitamin E on the development of altered hepatic foci and hepatic tumors induced by the peroxisome proliferator ciprofibrate. J Cancer Res Clin Oncol, 116, 351–6
  • Glauert HP, Eyigor A, Tharappel JC, et al. (2006). Inhibition of hepatocarcinogenesis by the deletion of the p50 subunit of NF-kappaB in mice administered the peroxisome proliferator Wy-14 643. Toxicol Sci, 90, 331–6
  • Glauert HP, Srinivasan S, Tatum VL, et al. (1992). Effects of the peroxisome proliferators ciprofibrate and perfluorodecanoic acid on hepatic cellular antioxidants and lipid peroxidation in rats. Biochem Pharmacol, 43, 1353–9
  • Gloire G, Legrand-Poels S, Piette J. (2006). NF-kappaB activation by reactive oxygen species: fifteen years later. Biochem Pharmacol, 72, 1493–505
  • Goel SK, Lalwani ND, Reddy JK. (1986). Peroxisome proliferation and lipid peroxidation in rat liver. Cancer Res, 46, 1324–30
  • Goetz ME, Luch A. (2008). Reactive species: a cell damaging rout assisting to chemical carcinogens. Cancer Lett, 266, 73–83
  • Goll V, Alexandre E, Viollon-Abadie C, et al. (1999). Comparison of the effects of various peroxisome proliferators on peroxisomal enzyme activities, DNA synthesis, and apoptosis in rat and human hepatocyte cultures. Toxicol Appl Pharmacol, 160, 21–32
  • Gottlicher M, Widmark E, Li Q, Gustafsson JA. (1992). Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. Proc Natl Acad Sci USA, 89, 4653–7
  • Grasl-Kraupp B, Huber W, Just W, et al. (1993a). Enhancement of peroxisomal enzymes, cytochrome P-452 and DNA synthesis in putative preneoplastic foci of rat liver treated with the peroxisome proliferator nafenopin. Carcinogenesis, 14, 1007–12
  • Grasl-Kraupp B, Huber W, Timmermann-Trosiener I, Schulte-Hermann R. (1993b). Peroxisomal enzyme induction uncoupled from enhanced DNA synthesis in putative preneoplastic liver foci of rats treated with a single dose of the peroxisome proliferator nafenopin. Carcinogenesis, 14, 2435–7
  • Grasl-Kraupp B, Waldhor T, Huber W, Schulte-Hermann R. (1993c). Glutathione S-transferase isoenzyme patterns in different subtypes of enzyme-altered rat liver foci treated with the peroxisome proliferator nafenopin or with phenobarbital. Carcinogenesis, 14, 2407–12
  • Grasl-Kraupp B, Ruttkay-Nedecky B, Mullauer L, et al. (1997). Inherent increase of apoptosis in liver tumors: implications for carcinogenesis and tumor regression. Hepatology, 25, 906–12
  • Gray RH, de la Iglesia FA. (1984). Quantitative microscopy comparison of peroxisome proliferation by the lipid-regulating agent gemfibrozil in several species. Hepatology, 4, 520–30
  • Grosse Y, Baan R, Secretan-Lauby B, et al. (2011). Carcinogenicity of chemicals in industrial and consumer products, food contaminants and flavourings, and water chlorination byproducts. WHO International Agency for Research on Cancer Monograph Working Group. Lancet Oncol, 12, 328–9
  • Guo D, Sarkar J, Suino-Powell K, et al. (2007). Induction of nuclear translocation of constitutive androstane receptor by peroxisome proliferator-activated receptor alpha synthetic ligands in mouse liver. J Biol Chem, 282, 36766–76
  • Guyton KZ, Chiu WA, Bateson TF, et al. (2009). A reexamination of the PPAR-α activation mode of action as a basis for assessing human cancer risks of environmental contaminants. Environ Health Perspect, 117, 1664–72
  • Hager GL, Varticovski L. (2012). Chromatin in time and space. Biochim Biophys Acta, 1819, 631
  • Hagmann M, Georgiev O, Schaffner W. (1997). The VP16 paradox: herpes simplex virus VP16 contains a long-range activation domain but within the natural multiprotein complex activates only from promoter-proximal positions. J Virol, 71, 5952–62
  • Hanahan D, Weinberg RA. (2011). Hallmarks of cancer: the next generation. Cell, 144, 646–74
  • Handler JA, Seed CB, Bradford BU, Thurman RG. (1992). Induction of peroxisomes by treatment with perfluorooctanoate does not increase rates of H2O2 production in intact liver. Toxicol Lett, 60, 61–8
  • Hanefeld M, Kemmer C, Kadner E. (1983). Relationship between morphological changes and lipid lowering action of p-chlorphenoxyisobutyric acid (CPIB) on hepatic mitochondria and peroxisomes in man. Atherosclerosis, 46, 239–46
  • Hanefeld M, Kemmer C, Leonhardt W, et al. (1980). Effects of p-chlorophenoxyisobutyric acid (CPIB) on the human liver. Atherosclerosis, 36, 159–72
  • Hanselman JC, Vartanian MA, Koester BP, et al. (2001). Expression of the mRNA encoding truncated PPAR alpha does not correlate with hepatic insensitivity to peroxisome proliferators. Mol Cell Biochem, 217, 91–7
  • Hashimoto T, Fujita T, Usuda N, et al. (1999). Peroxisomal and mitochondrial fatty acid beta-oxidation in mice nullizygous for both peroxisome proliferator-activated receptor alpha and peroxisomal fatty acyl-CoA oxidase. Genotype correlation with fatty liver phenotype. J Biol Chem, 274, 19228–36
  • Hasmall SC, James NH, MacDonald N, et al. (2000a). Suppression of mouse hepatocyte apoptosis by peroxisome proliferators: role of PPARα and TNFalpha. Mutat Res, 448, 193–200
  • Hasmall SC, James NH, MacDonald N, et al. (2000b). Species differences in response to diethylhexylphthalate: suppression of apoptosis, induction of DNA synthesis and peroxisome proliferator activated receptor alpha-mediated gene expression. Arch Toxicol, 74, 85–91
  • Hasmall SC, West DA, Olsen K, Roberts RA. (2000c). Role of hepatic non-parenchymal cells in the response of rat hepatocytes to the peroxisome proliferator nafenopin in vitro. Carcinogenesis, 21, 2159–65
  • Hasmall SC, James NH, MacDonald N, et al. (1999). Suppression of apoptosis and induction of DNA synthesis in vitro by the phthalate plasticizers monoethylhexylphthalate (MEHP) and diisononylphthalate (DINP): a comparison of rat and human hepatocytes in vitro. Arch Toxicol, 73, 451–6
  • Hasmall SC, James NH, Soames AR, Roberts RA. (1998). The peroxisome proliferator nafenopin does not suppress hepatocyte apoptosis in guinea-pig liver in vivo nor in human hepatocytes in vitro. Arch Toxicol, 72, 777–83
  • Hasmall SC, Roberts RA. (2000). The nongenotoxic hepatocarcinogens diethylhexylphthalate and methylclofenapate induce DNA synthesis preferentially in octoploid rat hepatocytes. Toxicol Pathol, 28, 503–9
  • Hays T, Rusyn I, Burns AM, et al. (2005). Role of peroxisome proliferator-activated receptor-alpha (PPARα) in bezafibrate-induced hepatocarcinogenesis and cholestasis. Carcinogenesis, 26, 219–27
  • Herren-Freund SL, Pereira MA, Khoury MD, Olson G. (1987). The carcinogenicity of trichloroethylene and its metabolites, trichloroacetic acid and dichloroacetic acid, in mouse liver. Toxicol Appl Pharmacol, 90, 183–9
  • Hertz R, Bishara-Shieban J, Bar-Tana J. (1995). Mode of action of peroxisome proliferators as hypolipidemic drugs. Suppression of apolipoprotein C-III. J Biol Chem, 270, 13470–5
  • Hinton RH, Mitchell FE, Mann A, et al. (1986). Effects of phthalic acid esters on the liver and thyroid. Environ Health Perspect, 70, 195–210
  • Hoivik DJ, Qualls CW Jr, Mirabile RC, et al. (2004). Fibrates induce hepatic peroxisome and mitochondrial proliferation without overt evidence of cellular proliferation and oxidative stress in cynomolgus monkeys. Carcinogenesis, 25, 1757–69
  • Holden P, Hasmall S, James N, et al. (2000). Tumour necrosis factor a (TNFa): role in suppression of apoptosis by peroxisome proliferators. Cell Mol Biol, 46, 29–39
  • Holsapple MP, Pitot HC, Cohen SM, et al. (2006). Mode of action in relevance of rodent liver tumors to human cancer risk. Toxicol Sci, 89, 51–6
  • Howroyd P, Swanson C, Dunn C, et al. (2004). Decreased longevity and enhancement of age-dependent lesions in mice lacking the nuclear receptor peroxisome proliferator-activated receptor alpha (PPARα). Toxicol Pathol, 32, 591–9
  • Hsieh LL, Shinozuka H, Weinstein IB. (1991). Changes in expression of cellular oncogenes and endogenous retrovirus-like sequences during hepatocarcinogenesis induced by a peroxisome proliferator. Br J Cancer, 64, 815–20
  • Hsu MH, Savas U, Griffin KJ, Johnson EF. (2001). Identification of peroxisome proliferator responsive human genes by elevated expression of the peroxisome proliferator-activated receptor alpha in HepG2 cells. J Biol Chem, 276, 27950–8
  • Huber W, Kraupp-Grasl B, Esterbauer H, Schulte-Hermann R. (1991). Role of oxidative stress in age dependent hepatocarcinogenesis by the peroxisome proliferator nafenopin in the rat. Cancer Res, 51, 1789–92
  • Huber WW, Grasl-Kraupp B, Stekel H, et al. (1997). Inhibition instead of enhancement of lipid peroxidation by pretreatment with the carcinogenic peroxisome proliferator nafenopin in rat liver exposed to a high single dose of corn oil. Arch Toxicol, 71, 575–81
  • Huttunen JK, Heinonen OP, Manninen V, et al. (1994). The Helsinki Heart Study: an 8.5-year safety and mortality follow-up. J Intern Med, 235, 31–9
  • IARC. (1996). Some pharmaceutical drugs. IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans. Lyon, France: IARC Press, 391–426
  • IARC. (2012). Some chemicals present in industrial and consumer products, food and drinking-water. IARC Technical Report No. 101. Lyon, France: IARC Press
  • Isenberg JS, Kamendulis LM, Ackley DC, et al. (2001). Reversibility and persistence of di-2-ethylhexyl phthalate (DEHP)- and phenobarbital-induced hepatocellular changes in rodents. Toxicol Sci, 64, 192–9
  • Isenberg JS, Kamendulis LM, Smith JH, et al. (2000). Effects of Di-2-ethylhexyl phthalate (DEHP) on gap-junctional intercellular communication (GJIC), DNA synthesis, and peroxisomal beta oxidation (PBOX) in rat, mouse, and hamster liver. Toxicol Sci, 56, 73–85
  • Isenberg JS, Kolaja KL, Ayoubi SA, et al. (1997). Inhibition of WY 14 643-induced hepatic lesion growth in mice by rotenone. Carcinogenesis, 18, 1511–9
  • Issemann I, Green S. (1990). Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature, 347, 645–50
  • Ito Y, Yamanoshita O, Asaeda N, et al. (2007). Di(2-ethylhexyl)phthalate induces hepatic tumorigenesis through a peroxisome proliferator-activated receptor alpha-independent pathway. J Occup Health, 49, 172–82
  • James NH, Gill JH, Brindle R, et al. (1998a). Peroxisome proliferator-activated receptor (PPAR) alpha-regulated growth responses and their importance to hepatocarcinogenesis. Toxicol Lett, 102–103, 91–6
  • James NH, Soames AR, Roberts RA. (1998b). Suppression of hepatocyte apoptosis and induction of DNA synthesis by the rat and mouse hepatocarcinogen diethylhexylphthalate (DEHP) and the mouse hepatocarcinogen 1,4-dichlorobenzene (DCB). Arch Toxicol, 72, 784–90
  • James NH, Roberts RA. (1996). Species differences in response to peroxisome proliferators correlate in vitro with induction of DNA synthesis rather than suppression of apoptosis. Carcinogenesis, 17, 1623–32
  • Janssen-Timmen U, Traub O, Dermietzel R, et al. (1986). Reduced number of gap junctions in rat hepatocarcinomas detected by monoclonal antibody. Carcinogenesis, 7, 1475–82
  • Jia Y, Guo GL, Surapureddi S, et al. (2005). Transcription coactivator peroxisome proliferator-activated receptor-binding protein/mediator 1 deficiency abrogates acetaminophen hepatotoxicity. Proc Natl Acad Sci, 102, 12531–6
  • Jia Y, Qi C, Zhang Z, et al. (2003). Overexpression of peroxisome proliferator-activated receptor-alpha (PPARα)-regulated genes in liver in the absence of peroxisome proliferation in mice deficient in both L- and D-forms of enoyl-CoA hydratase/dehydrogenase enzymes of peroxisomal beta-oxidation system. J Biol Chem, 278, 47232–9
  • Julien E, Boobis AR, Olin SS. (2009). The key events dose-response framework: a cross-disciplinary mode-of-action based approach to examining dose-response and thresholds. Crit Rev Food Sci Nutr, 49, 682–9
  • Kamendulis LM, Isenberg JS, Prow TW, Klaunig JE. (1997). Rotenone reversal of Wy-14 643-induced inhibition of hepatic gap junctional intercellular communication. Toxicologist, 36, 58
  • Kamendulis LM, Isenberg JS, Smith JH, et al. (2002). Comparative effects of phthalate monoesters on gap junctional intercellular communication and peroxisome proliferation in rodent and primate hepatocytes. J Toxicol Environ Health A, 65, 569–88
  • Karin M. (2006). Nuclear factor-kappaB in cancer development and progression. Nature, 441, 431–6
  • Kasai H. (1997). Analysis of a form of oxidative DNA damage, 8-hydroxy-2-deoxyguanosine, as a marker of cellular oxidative stress during carcinogenesis. Mutat Res, 387, 147–63
  • Kawashima Y, Suzuki S, Kozuka H, et al. (1994). Effects of prolonged administration of perfluorooctanoic acid on hepatic activities of enzymes which detoxify peroxide and xenobiotic in the rat. Toxicology, 93, 85–97
  • Keller H, Devchand PR, Perroud M, Wahli W. (1997). PPAR alpha structure-function relationships derived from species-specific differences in responsiveness to hypolipidemic agents. Biol Chem, 378, 651–5
  • Keller H, Dreyer C, Medin J, et al. (1993). Fatty acids and retinoids control lipid metabolism through activation of peroxisome proliferator-activated receptor-retinoid X receptor heterodimers. Proc Natl Acad Sci, 90, 2160–4
  • Kersten S, Seydoux J, Peters JM, et al. (1999). Peroxisome proliferator-activated receptor α mediates the adaptive response to fasting. J Clin Inv, 103, 1489–98
  • Ketcham CA, Klaunig JE. (1997). The inhibition of gap junctional intercellular communication by WY-14 643 through the activation of protein kinase c and oxidative stress. Toxicologist, 36, 223
  • Khetchoumian K, Teletin M, Tisserand J, et al. (2008). Trim24 (Tif1 alpha): an essential ‘brake’ for retinoic acid-induced transcription to prevent liver cancer. Cell Cycle, 7, 3647–52
  • Kim MY, Song KS, Park GH, et al. (2004). B6C3F1 mice exposed to ozone with 4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone and/or dibutyl phthalate showed toxicities through alterations of NF-kappaB, AP-1, Nrf2, and osteopontin. J Vet Sci, 5, 131–7
  • Klaunig JE, Babich MA, Baetcke KP, et al. (2003). PPARα agonist-induced rodent tumors: modes of action and human relevance. Crit Rev Toxicol, 33, 655–780
  • Klaunig JE, Hocevar BA, Kamendulis LM. (2012). Mode of action analysis of perfluorooctanoic acid (PFOA) tumorigenicity and human relevance. Reprod Toxicol, 33, 410--18
  • Klaunig JE, Ruch RJ. (1990). Role of inhibition of intercellular communication in carcinogenesis. Lab Invest, 62, 135–46
  • Klaunig JE, Ruch RJ, Weghorst CM. (1990). Comparative effects of phenobarbital, DDT, and lindane on mouse hepatocyte gap-junctional intercellular communication. Toxicol Appl Pharmacol, 102, 553–63
  • Klaunig JE, Ruch RJ, DeAngelo AB, Kaylor WH. (1988). Inhibition of mouse hepatocyte intercellular communication by phthalate monoesters. Cancer Lett, 43, 65–71
  • Klaunig JE, Ruch RJ, Lin ELC. (1989). Effects of trichloroethylene and its metabolites on rodent hepatocyte intercellular communication. Toxicol Appl Pharmacol, 99, 454–65
  • Klaunig JE, Xu Y, Isenberg JS, et al. (1998). The role of oxidative stress in chemical carcinogenesis. Environ Health Perspect, 106, 289–95
  • Kluwe WM, Huff JE, Matthews HB, et al. (1985). Comparative chronic toxicities and carcinogenic potentials of 2-ethylhexyl-containing compounds in rats and mice. Carcinogenesis, 6, 1577–83
  • Kluwe WM, McConnell EE, Huff JE, et al. (1982). Carcinogenicity testing of phthalate esters and related compounds. Environ Health Perspect, 45, 129–33, 1982
  • Krause BR, Bousley R, Kieft K, et al. (1994). Comparison of lifibrol to other lipid-regulating agents in experimental animals. Pharmacol Res, 29, 345–57
  • Lacquemant C, Lepretre F, Pineda Torra I, et al. (2000). Mutation screening of the PPARα gene in type 2 diabetes associated with coronary heart disease. Diabetes Metab, 26, 393–401
  • Lake BG, Evans JG, Cunninghame ME, Price RJ. (1993). Comparison of the hepatic effects of nafenopin and WY-14 643 on peroxisome proliferation and cell replication in the rat and Syrian hamster. Environ Health Perspect, 101, 241–7
  • Lake BG, Evans JG, Gray TJ, et al. (1989a). Comparative studies on nafenopin-induced hepatic peroxisome proliferation in the rat, Syrian hamster, guinea pig, and marmoset. Toxicol Appl Pharmacol, 99, 148–60
  • Lake BG, Gray TJ, Körösi SA, Walters DG. (1989b). Nafenopin, a peroxisome proliferator, depletes hepatic vitamin E content and elevates plasma oxidised glutathione levels in rats. Toxicol Lett., 45, 221–9
  • Lake BG, Kozlen SL, Evans JG, et al. (1987). Effect of prolonged administration of clofibric acid and di-(2-ethylhexyl)phthalate on hepatic enzyme activities and lipid peroxidation in the rat. Toxicology, 44, 213–28
  • Lake BG, Rumsby PC, Price RJ, Cunninghame ME. (2000). Species differences in hepatic peroxisome proliferation, cell replication and transforming growth factor-beta1 gene expression in the rat, Syrian hamster and guinea pig. Mutat Res, 448, 213–25
  • Lalwani ND, Reddy MK, Qureshi SA, et al. (1983). Evaluation of selected hypolipidemic agents for the induction of peroxisomal enzymes and peroxisome proliferation in the rat liver. Hum Toxicol, 2, 27–48
  • Lapinskas PJ, Brown S, Leesnitzer LM, et al. (2005). Role of PPARalpha in mediating the effects of phthalates and metabolites in the liver. Toxicology, 207, 149–63
  • Laughter AR, Dunn CS, Swanson CL, et al. (2004). Role of the peroxisome proliferator-activated receptor alpha (PPARα) in responses to trichloroethylene and metabolites, trichloroacetate and dichloroacetate in mouse liver. Toxicology, 203, 83–98
  • Lawrence JW, Li Y, Chen S, et al. (2001a). Differential gene regulation in human versus rodent hepatocytes by peroxisome proliferator-activated receptor (PPAR) alpha. PPAR alpha fails to induce peroxisome proliferation-associated genes in human cells independently of the level of receptor expression. J Biol Chem, 276, 31521–7
  • Lawrence JW, Wollenberg GK, DeLuca JG. (2001b). Tumor necrosis factor alpha is not required for WY14 643-induced cell proliferation. Carcinogenesis, 22, 381–6
  • Lee SS, Pineau T, Drago J, et al. (1995). Targeted disruption of the alpha isoform of the peroxisome proliferator-activated receptor gene in mice results in abolishment of the pleiotropic effects of peroxisome proliferators. Mol Cell Biol, 15, 3012–22
  • Li Y, Glauert HP, Spear BT. (2000a). Activation of nuclear factor-kappaB by the peroxisome proliferator ciprofibrate in H4IIEC3 rat hepatoma cells and its inhibition by the antioxidants N-acetylcysteine and vitamin E. Biochem Pharmacol, 59, 427–34
  • Li Y, Tharappel JC, Cooper S, et al. (2000b). Expression of the hydrogen peroxide-generating enzyme fatty acyl CoA oxidase activates NF-kappaB. DNA Cell Biol, 19, 113–20
  • Li Y, Kovach A, Suino-Powell K, et al. (2008). Structural and biochemical basis for the binding selectivity of peroxisome proliferator-activated receptor gamma to PGC-1alpha. J Biol Chem, 283, 19132–9
  • Li Y, Leung LK, Glauert HP, Spear BT. (1996). Treatment of rats with the peroxisome proliferator ciprofibrate results in increased liver NF-kappa B activity. Carcinogenesis, 17, 2305–9
  • Lington AW, Khalimi GH, Klaunig JE, Nikiforov AI. (1994). Effects of di-2-ethylhexyl phthalate and four metabolites on rodent hepatocyte gap-junctional intercellular communication. Toxicologist, 14, 57
  • Liu MH, Li J, Shen P, et al. (2008). A natural polymorphism in peroxisome proliferator-activated receptor-alpha hinge region attenuates transcription due to defective release of nuclear receptor corepressor from chromatin. Mol Endocrinol, 22, 1078–92
  • Loewenstein WR, Kanno Y. (1966). Intercellular communication and the control of tissue growth: lack of communication between cancer cells. Nature, 209, 1248–9
  • Lu Y, Boekschoten MV, Wopereis S, et al. (2011). Comparative transcriptomic and metabolomic analysis of fenofibrate and fish oil treatments in mice. Physiol Genomics, 43, 1307–18
  • Luebker DJ, Hansen KJ, Bass NM, et al. (2002). Interactions of fluorochemicals with rat liver fatty acid-binding protein. Toxicology, 176, 175–85
  • Ma WL, Hsu CL, Wu MH, et al. (2008). Androgen receptor is a new potential therapeutic target for the treatment of hepatocellular carcinoma. Gastroenterology, 135, 947–55, 955
  • MacDonald N, Holden PR, Roberts RA. (1999). Addition of peroxisome proliferator-activated receptor alpha to guinea pig hepatocytes confers increased responsiveness to peroxisome proliferators. Cancer Res, 59, 4776–80
  • Maeda S, Kamata H, Luo JL, et al. (2005). IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell, 121, 977–90
  • Maglich JM, Lobe DC, Moore JT. (2009). The nuclear receptor CAR (NR1I3) regulates serum triglyceride levels under conditions of metabolic stress. J Lipid Res, 50, 439–45
  • Mäkinen J, Frank C, Jyrkkärinne J, et al. (2002). Modulation of mouse and human phenobarbital-responsive enhancer module by nuclear receptors. Mol Pharmacol, 62, 366–78
  • Makowska JM, Gibson GG, Bonner FW. (1992). Species differences in ciprofibrate induction of hepatic cytochrome P450 4A1 and peroxisome proliferation. J Biochem Toxicol, 7, 183–91
  • Mally A, Chipman JK. (2002). Non-genotoxic carcinogens: early effects on gap junctions, cell proliferation and apoptosis in the rat. Toxicology, 180, 233–24
  • Maloney EK, Waxman DJ. (1999). Trans-activation of PPARα and PPARgamma by structurally diverse environmental chemicals. Toxicol Appl Pharmacol, 161, 209–18
  • Marsman D. (1995). NTP technical report on the toxicity studies of dibutyl phthalate (CAS No. 84-74-2) administered in feed to F344/N rats and B6C3F1 mice. Toxic Rep Ser, 30, 1–G5
  • Marsman DS, Cattley RC, Conway JG, Popp JA. (1988). Relationship of hepatic peroxisome proliferation and replicative DNA synthesis to the hepatocarcinogenicity of the peroxisome proliferators di(2-ethylhexyl)phthalate and [4-chloro-6-(2,3-xylidino)-2-pyrimidinylthio]acetic acid (Wy-14 643) in rats. Cancer Res, 48, 6739–44
  • Marsman DS, Goldsworthy TL, Popp JA. (1992). Contrasting hepatocytic peroxisome proliferation, lipofuscin accumulation and cell turnover for the hepatocarcinogens Wy-14 643 and clofibric acid. Carcinogenesis, 13, 1011–7
  • Marsman DS, Popp JA. (1994). Biological potential of basophilic hepatocellular foci and hepatic adenoma induced by the peroxisome proliferator, Wy-14 643. Carcinogenesis, 15, 111–7
  • Martin PG, Guillou H, Lasserre F, et al. (2007). Novel aspects of PPARα-mediated regulation of lipid and xenobiotic metabolism revealed through a nutrigenomic study. Hepatology, 45, 767–77
  • Maruyama H, Amanuma T, Tsutsumi M, et al. (1994). Inhibition by catechol and di(2-ethylhexyl)phthalate of pancreatic carcinogenesis after initiation with N-nitrosobis(2-hydroxypropyl)amine in Syrian hamsters. Carcinogenesis, 15, 1193–6
  • Mascaro C, Acosta E, Ortiz JA, et al. (1998). Control of human muscle-type carnitine palmitoyltransferase I gene transcription by peroxisome proliferator activated receptor. J Biol Chem, 273, 8560–3
  • McGuire EJ, Lucas JA, Gray RH, de la Iglesia FA. (1991). Peroxisome induction potential and lipid-regulating activity in rats. Quantitative microscopy and chemical structure-activity relationships. Am J Pathol, 139, 217–29
  • McNally JG, Muller WG, Walker D, et al. (2000). The glucocorticoid receptor: rapid exchange with regulatory sites in living cells. Science, 287, 1262–5
  • Meek ME. (2008). Recent developments in frameworks to consider human relevance of hypothesized modes of action for tumours in animals. Environ Mol Mutagen, 49, 110–6
  • Meek ME, Bucher JR, Cohen SM, et al. (2003). A framework for human relevance analysis of information on carcinogenic modes of action. Crit Rev Toxicol, 33, 591–653
  • Melnick RL, Brody C, DiGangi J, Huff J. (2003). The IARC evaluation of DEHP excludes key papers demonstrating carcinogenic effects. Int J Occup Environ Health, 9, 400–2
  • Menegazzi M, Carcereri-De Prati A, Suzuki H, et al. (1997). Liver cell proliferation induced by nafenopin and cyproterone acetate is not associated with increases in activation of transcription factors NF-kappaB and AP-1 or with expression of tumor necrosis factor alpha. Hepatology, 25, 585–92
  • Meyer K, Lee JS, Dyck PA, et al. (2003). Molecular profiling of hepatocellular carcinomas developing spontaneously in acyl-CoA oxidase deficient mice: comparison with liver tumors induced in wild-type mice by a peroxisome proliferator and a genotoxic carcinogen. Carcinogenesis, 24, 975–84
  • Mitchell FE, Price SC, Hinton RH, et al. (1985). Time and dose-response study of the effects on rats of the plasticizer di(2-ethylhexyl) phthalate. Toxicol Appl Pharmacol, 81, 371–92
  • Moennikes O, Stahl S, Bannasch P, et al. (2003). WY-14 643-mediated promotion of hepatocarcinogenesis in connexin32-wild-type and connexin32-null mice. CarcinogenesisSep, 24, 1561–5
  • Morimura K, Cheung C, Ward JM, et al. (2006). Differential susceptibility of mice humanized for peroxisome proliferator-activated receptor alpha to Wy-14 643-induced liver tumorigenesis. Carcinogenesis, 27, 1074–80
  • Mukherjee R, Jow L, Noonan D, McDonnell DP. (1994). Human and rat peroxisome proliferator activated receptors (PPARs) demonstrate similar tissue distribution but different responsiveness to PPAR activators. J Steroid Biochem Molec Biol, 51, 157–66
  • National Toxicology Program (NTP). (1982). Carcinogenesis bioassay of di(2-ethylhexyl)phthalate (CAS No. 117-81-7) in F344 rats and B6C3F1 mice (feed studies). Natl Toxicol Program Tech Rep Ser, 217, 1–127
  • Nemali MR, Usuda N, Reddy MK, et al. (1988). Comparison of constitutive and inducible levels of expression of peroxisomal beta-oxidation and catalase genes in liver and extrahepatic tissues of rat. Cancer Res, 48, 5316–24
  • Nesfield SR, Clarke CJ, Hoivik DJ, et al. (2005a). Evaluation of the carcinogenic potential of clofibrate in the rasH2 mouse. Int J Toxicol, 24, 301–11
  • Nesfield SR, Williams TC, Hoivik DJ, et al. (2005b). Evaluation of the carcinogenic potential of clofibrate in the neonatal mouse. Int J Toxicol, 24, 341–8
  • Neveu MJ, Hully JR, Paul DL, Pitot HC. (1990). Reversible alteration in the expression of the gap-junctional protein connexin 32 during tumor promotion in rat liver and its role during cell proliferation. Cancer Commun, 2, 21–31
  • Nicholls-Grzemski FA, Belling GB, Priestly BG, et al. (2000). Clofibrate pretreatment in mice confers resistance against hepatic lipid peroxidation. J Biochem Mol Toxicol, 14, 335–45
  • Nilakantan V, Spear BT, Glauert HP. (1998). Liver-specific catalase expression in transgenic mice inhibits NF-kappaB activation and DNA synthesis induced by the peroxisome proliferator ciprofibrate. Carcinogenesis, 19, 631–7
  • O’Brien ML, Cunningham ML, Spear BT, Glauert HP. (2001a). Effects of peroxisome proliferators on glutathione and glutathione-related enzymes in rats and hamsters. Toxicol Appl Pharmacol, 171, 27–37
  • O’Brien ML, Twaroski TP, Cunningham ML, et al. (2001b). Effects of peroxisome proliferators on antioxidant enzymes and antioxidant vitamins in rats and hamsters. Toxicol Sci, 60, 271–8
  • Oberhammer FA, Qin HM. (1995). Effect of three tumour promoters on the stability of hepatocytes cultures and apoptosis after transforming growth factor-beta1. Carcinogenesis, 16, 1363–71
  • Ohmura T, Ledda-Columbano GM, Piga R, et al. (1996). Hepatocyte proliferation induced by a single dose of a peroxisome proliferator. Am J Pathol, 148, 815–24
  • Pacot C, Petit M, Rollin M, et al. (1996). Difference between guinea pig and rat in the liver peroxisomal response to equivalent plasmatic level of ciprofibrate. Arch Biochem Biophys, 327, 181–8
  • Palmer CN, Hsu MH, Griffin KJ, et al. (1998). Peroxisome proliferator activated receptor-alpha expression in human liver. Mol Pharmacol, 53, 14–22
  • Parzefall W, Berger W, Kainzbauer E, et al. (2001). Peroxisome proliferators do not increase DNA synthesis in purified rat hepatocytes. Carcinogenesis, 22, 519–23
  • Patsouris D, Reddy JK, Müller M, Kersten S. (2006). Peroxisome proliferator-activated receptor alpha mediates the effects of high-fat diet on hepatic gene expression. Endocrinology, 147, 1508–16
  • Permadi H, Lundgren B, Andersson K, et al. (1993). Effects of perfluoro fatty acids on peroxisome proliferation and mitochondrial size in mouse liver: dose and time factors and effect of chain length. Xenobiotica, 23, 761–70
  • Perrone CE, Shao L, Williams GM. (1998). Effect of rodent hepatocarcinogenic peroxisome proliferators on fatty acyl-CoA oxidase, DNA synthesis, and apoptosis in cultured human and rat hepatocytes. Toxicol Appl Pharmacol, 150, 277–86
  • Peters JM, Aoyama T, Cattley RC, et al. (1998). Role of peroxisome proliferator-activated receptor alpha in altered cell cycle regulation in mouse liver. Carcinogenesis, 19, 1989–94
  • Peters JM, Cattley RC, Gonzalez FJ. (1997). Role of PPAR alpha in the mechanism of action of the nongenotoxic carcinogen and peroxisome proliferator Wy-14 643. Carcinogenesis, 18, 2029–33
  • Peters JM, Cheung C, Gonzalez FJ. (2005). Peroxisome proliferator-activated receptor-α and liver cancer: where do we stand? J Mol Med, 83, 774–85
  • Peters JM, Rusyn I, Rose ML, et al. (2000). Peroxisome proliferator-activated receptor alpha is restricted to hepatic parenchymal cells, not Kupffer cells: implications for the mechanism of action of peroxisome proliferators in hepatocarcinogenesis. Carcinogenesis, 21, 823–6
  • Plant NJ, Horley NJ, Dickins M, et al. (1998). The coordinate regulation of DNA synthesis and suppression of apoptosis is differentially regulated by the liver growth agents, phenobarbital and methylclofenapate. Carcinogenesis, 19, 1521–7
  • Price RJ, Evans JG, Lake BG. (1992). Comparison of the effects of nafenopin on hepatic peroxisome proliferation and replicative DNA synthesis in the rat and Syrian hamster. Food Chem Toxicol, 30, 937–44
  • Pugh G Jr, Isenberg JS, Kamendulis LM, et al. (2000). Effects of diisononyl phthalate, di-2-ethylhexyl phthalate, and clofibrate in cynomolgus monkeys. Toxicol Sci, 56, 181–8
  • Qu A, Shah YM, Matsubara T, et al. (2010). PPARalpha-dependent activation of cell cycle control and DNA repair genes in hepatic nonparenchymal cells. Toxicol Sci, 118, 404–10
  • Qu B, Halliwell B, Ong CN, et al. (2000). Caloric restriction prevents oxidative damage induced by the carcinogen clofibrate in mouse liver. FEBS Lett, 473, 85–8
  • Qu B, Li Q, Wong KP, et al. (2001). Mechanism of clofibrate hepatotoxicity: mitochondrial damage and oxidative stress in hepatocytes. Free Radical Biol Med, 31, 659–69
  • Rakhshandehroo M, Hooiveld G, Müller M, Kersten S. (2009). Comparative analysis of gene regulation by the transcription factor PPARα between mouse and human. PLoS One, 4, e6796
  • Rao MS, Dwivedi RS, Subbarao V, Reddy JK. (1988). Induction of peroxisome proliferation and hepatic tumours in C57BL/6N mice by ciprofibrate, a hypolipidaemic compound. Br J Cancer, 58, 46–51
  • Rao MS, Lalwani ND, Scarpelli DG, Reddy JK. (1982). The absence of gamma-glutamyl transpeptidase activity in putative preneoplastic lesions and in hepatocellular carcinomas induced in rats by the hypolipidemic peroxisome proliferator Wy-14 643. Carcinogenesis, 3, 1231–3
  • Rao MS, Lalwani ND, Watanabe TK, Reddy JK. (1984). Inhibitory effect of antioxidants ethoxyquin and 2-tert-butyl-4-hydroxyanisole on hepatic tumorigenesis in rats fed ciprofibrate, a peroxisome proliferator. Cancer Res, 44, 1072–6
  • Rao MS, Reddy JK. (1996). Hepatocarcinogenesis of peroxisome proliferators. Ann N Y Acad Sci, 804, 573–87
  • Rao MS, Subbarao V. (1997). The effect of deferoxamine on ciprofibrate-induced hepatocarcinogenesis in the rat. In Vivo, 11, 495–8
  • Rao MS, Subbarao V. (1999). Inhibition of ciprofibrate-induced hepatocarcinogenesis in the rat by dimethylthiourea, a scavenger of hydroxyl radical. Oncol Rep, 6, 1285–8
  • Rao MS, Tatematsu M, Subbarao V, et al. (1986). Analysis of peroxisome proliferator-induced preneoplastic and neoplastic lesions of rat liver for placental form of glutathione S-transferase and gamma-glutamyltranspeptidase. Cancer Res, 46, 5287–90
  • Rao MS, Thangada S, Subbarao V. (1991). Peroxisome proliferation in neoplastic nodules and hepatocellular carcinomas induced by ciprofibrate in the rat. Exp Pathol, 41, 44–9
  • Rao MS, Usuda N, Subbarao V, Reddy JK. (1987). Absence of gamma-glutamyl transpeptidase activity in neoplastic lesions induced in the liver of male F-344 rats by di-(2-ethylhexyl)phthalate, a peroxisome proliferator. Carcinogenesis, 8, 1347–50
  • Reddy JK, Lalwani ND, Reddy MK, Qureshi SA. (1982). Excessive accumulation of autofluorescent lipofuscin in the liver during hepatocarcinogenesis by methyl clofenapate and other hypolipidemic peroxisome proliferators. Cancer Res, 42, 259–66
  • Reddy JK, Qureshi SA. (1979). Tumorigenicity of the hypolipidaemic peroxisome proliferator ethyl-alpha-p-chlorophenoxyisobutyrate (clofibrate) in rats. Br J Cancer, 40, 476–82
  • Reddy JK, Rao MS, Azarnoff DL, Sell S. (1979). Mitogenic and carcinogenic effects of a hypolipidemic peroxisome proliferator, [4-chloro-6-(2,3-xylidino)-2-pyrimidinylthio]acetic acid (Wy-14, 643), in rat and mouse liver. Cancer Res, 39, 152–61
  • Reddy JK, Rao MS. (1977). Malignant tumors in rats fed nafenopin, a hepatic peroxisome proliferator. J Natl Cancer Inst, 59, 1645–50
  • Reddy JK, Rao MS. (1989). Oxidative DNA damage caused by persistent peroxisome proliferation: its role in hepatocarcinogenesis. Mutat Res, 214, 63–8
  • Ren H, Aleksunes LM, Wood C, et al. (2010). Characterization of peroxisome proliferator-activated receptor alpha-independent effects of PPARalpha activators in the rodent liver: di-(2-ethylhexyl) phthalate also activates the constitutive-activated receptor. Toxicol Sci, 113, 45–59
  • Ren H, Vallanat B, Nelson DM, et al. (2009). Evidence for the involvement of xenobiotic-responsive nuclear receptors in transcriptional effects upon perfluoroalkyl acid exposure in diverse species. Reprod Toxicol, 27, 266–77
  • Richert L, Lamboley C, Viollon-Abadie C, et al. (2003). Effects of clofibric acid on mRNA expression profiles in primary cultures of rat, mouse and human hepatocytes. Toxicol Appl Pharmacol, 191, 130–46
  • Roberts RA, Ganey PE, Ju C, et al. (2007). Role of the Kupffer cell in mediating hepatic toxicity and carcinogenesis. Toxicol Sci, 96, 2–15
  • Roberts RA, James NH, Hasmall SC, et al. (2000). Apoptosis and proliferation in nongenotoxic carcinogenesis: species differences and role of PPARα. Toxicol Lett, 112–113, 49–57
  • Roberts RA. (1999). Peroxisome proliferators: mechanisms of adverse effects in rodents and molecular basis for species differences. Arch Toxicol, 73, 413–8
  • Roglans N, Bellido A, Rodriguez C, et al. (2002). Fibrate treatment does not modify the expression of acyl coenzyme A oxidase in human liver. Clin Pharmacol Ther, 72, 692–701
  • Rolfe M, James NH, Roberts RA. (1997). Tumour necrosis factor alpha (TNF-alpha) suppresses apoptosis and induces S-phase in rodent hepatocytes: a mediator of the hepatocarcinogenicity of peroxisome proliferators? Carcinogenesis, 18, 2277–80
  • Rose ML, Cattley RC, Dunn C, et al. (1999a). Dietary glycine prevents the development of liver tumors caused by the peroxisome proliferator WY-14 643. Carcinogenesis, 20, 2075–81
  • Rose ML, Rivera CA, Bradford BU, et al. (1999b). Kupffer cell oxidant production is central to the mechanism of peroxisome proliferators. Carcinogenesis, 20, 27–33
  • Rose ML, Germolec D, Arteel GE, et al. (1997a). Dietary glycine prevents increases in hepatocyte proliferation caused by the peroxisome proliferator WY-14 643. Chem Res Toxicol, 10, 1198–204
  • Rose ML, Germolec DR, Schoonhoven R, Thurman RG. (1997b). Kupffer cells are causally responsible for the mitogenic effect of peroxisome proliferators. Carcinogenesis, 18, 1453–6
  • Rose ML, Rusyn I, Bojes HK, et al. (2000). Role of Kupffer cells and oxidants in signaling peroxisome proliferator-induced hepatocyte proliferation. Mutat Res, 448, 179–92
  • Rosen MB, Abbott BD, Wolf DC, et al. (2008a). Gene profiling in the livers of wild-type and PPARα-null mice exposed to perfluorooctanoic acid. Toxicol Pathol, 36, 592–607
  • Rosen MB, Lee JS, Ren H, et al. (2008b). Toxicogenomic dissection of the perfluorooctanoic acid transcript profile in mouse liver: evidence for the involvement of nuclear receptors PPAR alpha and CAR. Toxicol Sci, 103, 46–56
  • Rosen MB, Schmid JR, Corton JC, et al. (2010). Gene expression profiling in wild-type and PPARα-null mice exposed to perfluorooctane sulfonate reveals PPARα-independent effects. PPAR Res, 2010, Article ID, 794739. 1--23
  • Roy D, Cai Q, Felty Q, Narayan S. (2007). Estrogen-induced generation of reactive oxygen and nitrogen species, gene damage, and estrogen-dependent cancers. J Toxicol Environ Health B Crit Rev, 10, 235–57
  • Rubins HB, Robins SJ, Collins D, et al. (1999). Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med, 341, 410--18
  • Ruch RJ, Klaunig JE. (1988). Kinetics of phenobarbital inhibition of intercellular communication in mouse hepatocytes. Cancer Res, 48, 2519–23
  • Rusyn I, Asakura S, Pachkowski B, et al. (2004). Expression of base excision DNA repair genes is a sensitive biomarker for in vivo detection of chemical-induced chronic oxidative stress: identification of the molecular source of radicals responsible for DNA damage by peroxisome proliferators. Cancer Res, 64, 1050–7
  • Rusyn I, Corton JC. (2012). Mechanistic considerations for human relevance of cancer hazard of di(2-ethylhexyl) phthalate. Mutat Res, 750, 141--58
  • Rusyn I, Kadiiska MB, Dikalova A, et al. (2001). Phthalates rapidly increase production of reactive oxygen species in vivo: role of Kupffer cells. Mol Pharmacol, 59, 744–50
  • Rusyn I, Peters JM, Cunningham ML. (2006). Modes of action and species-specific effects of di-(2-ethylhexyl)phthalate in the liver. Crit Rev Toxicol, 36, 459–79
  • Rusyn I, Tsukamoto H, Thurman RG. (1998). WY-14 643 rapidly activates nuclear factor kappaB in Kupffer cells before hepatocytes. Carcinogenesis, 19, 1217–22
  • Rusyn I, Yamashina S, Segal BH, et al. (2000). Oxidants from nicotinamide adenine dinucleotide phosphate oxidase are involved in triggering cell proliferation in the liver due to peroxisome proliferators. Cancer Res, 60, 4798–803
  • Sanderson LM, de Groot PJ, Hooiveld GJ, et al. (2008). Effect of synthetic dietary triglycerides: a novel research paradigm for nutrigenomics. PLoS One, 3, e1681
  • Sapone A, Peters JM, Sakai S, et al. (2000). The human peroxisome proliferator-activated receptor alpha gene: identification and functional characterization of two natural allelic variants. Pharmacogenetics, 10, 321–33
  • Sausen PJ, Lee DC, Rose ML, Cattley RC. (1995). Elevated 8-hydroxydeoxyguanosine in hepatic DNA of rats following exposure to peroxisome proliferators: relationship to mitochondrial alterations. Carcinogenesis, 16, 1795–801
  • Schmezer P, Pool BL, Klein RG, et al. (1988). Various short-term assays and two long-term studies with the plasticizer di(2-ethylhexyl)phthalate in the Syrian golden hamster. Carcinogenesis, 9, 37–43
  • Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, et al. (1996a). PPARα and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene. EMBO J, 15, 5336–48
  • Schoonjans K, Staels B, Auwerx J. (1996b). The peroxisome proliferator activated receptors (PPARs) and their effects on lipid metabolism and adipocyte differentiation. Biochim Biophys Acta, 1302, 93–109
  • Schulte-Hermann R, Ohde G, Schuppler J, Timmermann-Trosiener I. (1981). Enhanced proliferation of putative preneoplastic cells in rat liver following treatment with the tumor promoters phenobarbital, hexachlorocyclohexane, steroid compounds, and nafenopin. Cancer Res, 41, 2556–62
  • Schwarz JJ, Chakraborty T, Martin J, et al. (1992). The basic region of myogenin cooperates with two transcription activation domains to induce muscle-specific transcription. Mol Cell Biol, 12, 266–75
  • Seed J, Carney EW, Corley RA, et al. (2005). Overview: using mode of action and life stage information to evaluate the human relevance of animal toxicity data. Crit Rev Toxicol, 35, 664–72
  • Seo KW, Kim KB, Kim YJ, et al. (2004). Comparison of oxidative stress and changes of xenobiotic metabolizing enzymes induced by phthalates in rats. Food Chem Toxicol, 42, 107–14
  • Shah YM, Morimura K, Yang Q, et al. (2007). Peroxisome proliferator-activated receptor alpha regulates a microRNA-mediated signaling cascade responsible for hepatocellular proliferation. Mol Cell Biol, 27, 4238–47
  • Shaw D, Lee R, Roberts RA. (2002). Species differences in response to the phthalate plasticizer monoisononylphthalate (MINP) in vitro: a comparison of rat and human hepatocytes. Arch Toxicol, 76, 344–50
  • Sher T, Yi HF, McBride OW, Gonzalez FJ. (1993). cDNA cloning, chromosomal mapping, and functional characterization of the human peroxisome proliferator activated receptor. Biochemistry, 32, 5598–604
  • Shipley JM, Hurst CH, Tanaka SS, et al. (2004). trans-activation of PPARα and induction of PPARα target genes by perfluorooctane-based chemicals. Toxicol Sci, 80, 151–60
  • Smith-Oliver T, Butterworth BE. (1987). Correlation of the carcinogenic potential of di(2-ethylhexyl)phthalate (DEHP) with induced hyperplasia rather than with genotoxic activity. Mutat Res, 188, 21–8
  • Soames AR, Cliffe S, Pate I, Foster JR. (1999). Quantitative analysis of the lobular distribution of S-phase in rat liver following dietary administration of di(2-ethylhexyl)phthalate. Toxicol Pathol, 27, 436–40
  • Soliman MS, Cunningham ML, Morrow JD, et al. (1997). Evidence against peroxisome proliferation-induced hepatic oxidative damage. Biochem Pharmacol, 53, 1369–74
  • Sonich-Mullin C, Fielder R, Wiltse J, et al. (2001). IPCS conceptual framework for evaluating a mode of action for chemical carcinogenesis. Regul Toxicol Pharmacol, 34, 146–52
  • Staels B, Schoonjans K, Fruchart JC, Auwerx J. (1997). The effects of fibrates and thiazolidinediones on plasma triglyceride metabolism are mediated by distinct peroxisome proliferator activated receptors (PPARs). Biochimie, 79, 95–9
  • Stanko RT, Sekas G, Isaacson IA, et al. (1995). Pyruvate inhibits clofibrate-induced hepatic peroxisomal proliferation and free radical production in rats. Metabolism, 44, 166–71
  • Stauber AJ, Brown-Borg H, Liu J, et al. (2005). Constitutive expression of peroxisome proliferator-activated receptor alpha-regulated genes in dwarf mice. Mol Pharmacol, 67, 681–94
  • Stauber AJ, Bull RJ. (1997). Differences in phenotype and cell replicative behavior of hepatic tumors induced by dichloroacetate (DCA) and trichloroacetate (TCA). Toxicol Appl Pharmacol, 144, 235–46
  • Stoner MA, Auerbach SS, Zamule SM, et al. (2007). Transactivation of a DR-1 PPRE by a human constitutive androstane receptor variant expressed from internal protein translation start sites. Nucleic Acids Res, 35, 2177–90
  • Styles JA, Kelly M, Pritchard NR, Elcombe CR. (1988). A species comparison of acute hyperplasia induced by the peroxisome proliferator methylclofenapate: involvement of the binucleated hepatocyte. Carcinogenesis, 9, 1647–55
  • Styles JA, Kelly MD, Pritchard NR, Elcombe CR. (1990). Acute hyperplasia and peroxisome proliferation induced by methylclofenapate: a species comparison and implications for liver carcinogenesis. Prog Clin Biol Res, 331, 385–93
  • Surapureddi S, Viswakarma N, Yu S, et al. (2006). PRIC320, a transcription coactivator, isolated from peroxisome proliferator-binding protein complex. Biochem Biophys Res Commun, 343, 535–43
  • Svoboda DJ, Azarnoff DL. (1979). Tumors in male rats fed ethyl chlorophenoxyisobutyrate, a hypolipidemic drug. Cancer Res, 39, 3419–28
  • Tachibana K, Kobayashi Y, Tanaka T, et al. (2005). Gene expression profiling of potential peroxisome proliferator-activated receptor (PPAR) target genes in human hepatoblastoma cell lines inducibly expressing different PPAR isoforms. Nucl Recept, 3, 1--17
  • Tai ES, Demissie S, Cupples LA, et al. (2002). Association between the PPARΑ L162V polymorphism and plasma lipid levels: the Framingham Offspring Study. Arterioscler Thromb Vasc Biol, 22, 805–10
  • Takacs ML, Abbott BD. (2007). Activation of mouse and human peroxisome proliferator-activated receptors (alpha, beta/delta, gamma) by perfluorooctanoic acid and perfluorooctane sulfonate. Toxicol Sci, 95, 108–17
  • Takagi AK, Sai T, Umemura R, et al. (1990). Significant increase of 8-hydroxydeoxyguanosine in liver DNA of rats following short-term exposure to the peroxisome proliferators di(2-ethylhexyl)phthalate and di(2-ethylhexyl)adipate. Jpn J Cancer Res, 81, 213–5
  • Takashima K, Ito Y, Gonzalez FJ, Nakajima T. (2008). Different mechanisms of DEHP-induced hepatocellular adenoma tumorigenesis in wild-type and PPAR alpha-null mice. J Occup Health, 50, 169–80
  • Tamura K, Ono A, Miyagishima T, et al. (2006). Profiling of gene expression in rat liver and rat primary cultured hepatocytes treated with peroxisome proliferators. J Toxicol Sci, 31, 471–90
  • Tanaka K, Smith PF, Stromberg PC, et al. (1992). Studies of early hepatocellular proliferation and peroxisomal proliferation in Sprague Dawley rats treated with tumorigenic doses of clofibrate. Toxicol Appl Pharmacol, 116, 71–7
  • Tanaka N, Moriya K, Kiyosawa K, et al. (2008a). Hepatitis C virus core protein induces spontaneous and persistent activation of peroxisome proliferator-activated receptor alpha in transgenic mice: implications for HCV-associated hepatocarcinogenesis. Int J Cancer, 122, 124–31
  • Tanaka N, Moriya K, Kiyosawa K, et al. (2008b). PPARα activation is essential for HCV core protein-induced hepatic steatosis and hepatocellular carcinoma in mice. J Clin Invest, 118, 683–94
  • Tharappel JC, Cunningham ML, Spear BT, Glauert HP. (2001). Differential activation of hepatic NF-kappaB in rats and hamsters by the peroxisome proliferators Wy-14 643, gemfibrozil, and dibutyl phthalate. Toxicol Sci, 62, 20–7
  • Tharappel JC, Nalca A, Owens AB, et al. (2003). Cell proliferation and apoptosis are altered in mice deficient in the NF-kappaB p50 subunit after treatment with the peroxisome proliferator ciprofibrate. Toxicol Sci, 75, 300–8
  • Thoolen B, Maronpot RR, Harada T, et al. (2010). Proliferative and nonproliferative lesions of the rat and mouse hepatobiliary system. Toxicol Pathol, 38, 5S--81S
  • Thottassery J, Winberg L, Youssef J, et al. (1992). Regulation of perfluorooctanoic acid-induced peroxisomal enzyme activities and hepatocellular growth by adrenal hormones. Hepatology, 15, 316–22
  • Tomaszewski KE, Heindel SW, Jenkins WL, Melnick RL. (1990). Induction of peroxisomal acyl CoA oxidase activity and lipid peroxidation in primary rat hepatocyte cultures. Toxicology, 65, 49–60
  • Traub O, Drüge PM, Willecke K. (1983). Degradation and resynthesis of gap junction protein in plasma membranes of regenerating liver after partial hepatectomy or cholestasis. Proc Natl Acad Sci U S A, 80, 755--9
  • Trosko JE, Chang CC, Madhukar BV. (1990). Modulation of intercellular communication during radiation and chemical carcinogenesis. Radiat Res, 123, 241–51
  • Tucker MJ, Orton TC. (1995). Comparative toxicology of hypolipidaemic fibrates. Bristol (PA): Taylor and Francis
  • Tugwood JD, Aldridge TC, Lambe KG, et al. (1996). Peroxisome proliferator-activated receptors: structures and function. Ann N Y Acad Sci, 804, 252–65
  • Tugwood JD, Holden PR, James NH, et al. (1998). A PPAR alpha cDNA cloned from guinea pig liver encodes a protein with similar properties to the mouse PPAR alpha: implications for species differences in response to peroxisome proliferators. Arch Toxicol, 72, 169–77
  • U.S. Environmental Protection Agency. (2005). Guidelines for carcinogen risk assessment and supplemental guidance for assessing susceptibility from early-life exposure to carcinogens. Fed Reg, 70, 66, 17765–817
  • Upham BL, Deocampo ND, Wurl B, Trosko JE. (1998). Inhibition of gap junctional intercellular communication by perfluorinated fatty acids is dependent on the chain length of the fluorinated tail. Int J Cancer, 78, 491–5
  • Valles EG, Laughter AR, Dunn CS, et al. (2003). Role of the peroxisome proliferator-activated receptor alpha in responses to diisononyl phthalate. Toxicology, 191, 211–25
  • Van Rafelghem MJ, Mattie DR, Bruner RH, Andersen ME. (1987). Pathological and hepatic ultrastructural effects of a single dose of perfluoro-n-decanoic acid in the rat, hamster, mouse, and guinea pig. Fundam Appl Toxicol, 9, 522–40
  • Vanden Heuvel JP, Kreder D, Belda B, et al. (2003). Comprehensive analysis of gene expression in rat and human hepatoma cells exposed to the peroxisome proliferator WY14 643. Toxicol Appl Pharmacol, 188, 185–98
  • Vanden Heuvel JP, Thompson JT, Frame SR, Gillies PJ. (2006). Differential activation of nuclear receptors by perfluorinated fatty acid analogs and natural fatty acids: a comparison of human, mouse, and rat peroxisome proliferator-activated receptor-alpha, -beta, and -gamma, liver X receptor-beta, and retinoid X receptor-alpha. Toxicol Sci, 92, 476–89
  • Varanasi U, Chu R, Huang Q, et al. (1996). Identification of a peroxisome proliferator-responsive element upstream of the human peroxisomal fatty acyl coenzyme A oxidase gene. J Biol Chem, 271, 2147–55
  • Vu-Dac N, Schoonjans K, Kosykh V, et al. (1995). Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor. J Clin Invest, 96, 741–50
  • Vu-Dac N, Schoonjans K, Laine B, et al. (1994). Negative regulation of the human apolipoprotein A-I promoter by fibrates can be attenuated by the interaction of the peroxisome proliferator-activated receptor with its response element. J Biol Chem, 269, 31012–8
  • Wada N, Marsman DS, Popp JA. (1992). Dose-related effects of the hepatocarcinogen, Wy-14 643, on peroxisomes and cell replication. Fundam Appl Toxicol, 18, 149–54
  • Wahli W, Braissant O, Desvergne B. (1995). Peroxisome proliferator activated receptors: transcriptional regulators of adipogenesis, lipid metabolism and more. Chem Biol, 2, 261–6
  • Ward JM, Hagiwara A, Anderson LM, et al. (1988). The chronic hepatic or renal toxicity of di(2-ethylhexyl) phthalate, acetaminophen, sodium barbital, and phenobarbital in male B6C3F1 mice: autoradiographic, immunohistochemical, and biochemical evidence for levels of DNA synthesis not associated with carcinogenesis or tumor promotion. Toxicol Appl Pharmacol, 96, 494–506
  • Watanabe T, Horie S, Yamada J, et al. (1989). Species differences in the effects of bezafibrate, a hypolipidemic agent, on hepatic peroxisome-associated enzymes. Biochem Pharmacol, 38, 367–71
  • Weglarz TC, Sandgren EP. (2004). Cell cross-talk mediates PPARα null hepatocyte proliferation after peroxisome proliferator exposure. Carcinogenesis, 25, 107–12
  • West D, James N, Holden P, et al. (1999). Role for tumour necrosis factor a (TNFa) receptor 1 (TNFR1) and interleukin 1 receptor (IL1R) in the suppression of apoptosis by peroxisome proliferators. Hepatology, 30, 1417–24
  • Wheeler MD, Smutney OM, Check JF, et al. (2003). Impaired Ras membrane association and activation in PPARα knockout mice after partial hepatectomy. Am J Physiol Gastrointest Liver Physiol, 284, G302–12
  • Williams GM, Perrone C. (1995). Mechanism based risk assessment of peroxisome proliferating rodent hepatocarcinogens. In: Peroxisomes: Biology and Role in Toxicology and Disease, eds. J.K. Reddy, T. Suga, and G.P. Mannaerts. Ann NY Acad Sci, 804, 554–72
  • Wolf DC, Moore T, Abbott BD, et al. (2008). Comparative hepatic effects of perfluorooctanoic acid and WY 14 643 in PPARα knockout and wild-type mice. Toxicol Pathol, 36, 632–9
  • Woods CG, Burns AM, Bradford BU, et al. (2007a). WY-14 643 induced cell proliferation and oxidative stress in mouse liver are independent of NADPH oxidase. Toxicol Sci, 98, 366–74
  • Woods CG, Burns AM, Maki A, et al. (2007b). Sustained formation of alpha-(4-pyridyl-1-oxide)-N-tert-butylnitrone radical adducts in mouse liver by peroxisome proliferators is dependent upon peroxisome proliferator-activated receptor-alpha, but not NADPH oxidase. Free Radic Biol Med, 42, 335–42
  • Woods CG, Kosyk O, Bradford BU, et al. (2007c). Time course investigation of PPARα- and Kupffer cell-dependent effects of WY-14 643 in mouse liver using microarray gene expression. Toxicol Appl Pharmacol, 225, 267–77
  • Woodyatt NJ, Lambe KG, Myers KA, et al. (1999). The peroxisome proliferator (PP) response element upstream of the human acyl CoA oxidase gene is inactive among a sample human population: significance for species differences in response to PPs. Carcinogenesis, 20, 369–72
  • Xiao S, Anderson SP, Swanson C, et al. (2006). Activation of peroxisome proliferator-activated receptor alpha enhances apoptosis in the mouse liver. Toxicol Sci, 92, 368–77
  • Xu HE, Li Y. (2008). Ligand-Dependent and -Independent Regulation of PPAR{gamma} and Orphan Nuclear Receptors. Sci Signal, 1, pe52
  • Yamakawa-Kobayashi K, Ishiguro H, Arinami T, et al. (2002). A Val227Ala polymorphism in the peroxisome proliferator activated receptor alpha (PPARα) gene is associated with variations in serum lipid levels. J Med Genet, 39, 189–91
  • Yamasaki H. (1990). Gap-junctional intercellular communication and carcinogenesis. Carcinogenesis, 11, 1051–8
  • Yang Q, Ito S, Gonzalez FJ. (2007). Hepatocyte-restricted constitutive activation of PPAR alpha induces hepatoproliferation but not hepatocarcinogenesis. Carcinogenesis, 28, 1171–7
  • Yang Q, Nagano T, Shah Y, et al. (2008). The PPAR alpha-humanized mouse: a model to investigate species differences in liver toxicity mediated by PPAR alpha. Toxicol Sci, 101, 132–9
  • Yeldandi AV, Milano M, Subbarao V, et al. (1989). Evaluation of liver cell proliferation during ciprofibrate-induced hepatocarcinogenesis. Cancer Lett, 47, 21–7
  • Yeldandi AV, Rao MS, Reddy JK. (2000). Hydrogen peroxide generation in peroxisome proliferator-induced oncogenesis. Mutat Res, 448, 159–77
  • Youssef JA, Bouziane M, Badr MZ. (2003). Age-dependent effects of nongenotoxic hepatocarcinogens on liver apoptosis in vivo. Mech Ageing Dev, 124, 333–40

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.