352
Views
31
CrossRef citations to date
0
Altmetric
Reviews

Mechanisms of oxysterol-induced disease: insights from the biliary system

Pages 537-548 | Published online: 18 Jan 2017

References

  • Jusakul A, Yongvanit P, Loilome W, Namwat N, Kuver R. Mechanisms of oxysterol-induced carcinogenesis. Lipids Health Dis. 10, 44 (2011).
  • Otaegui-Arrazola A, Menendez-Carreno M, Ansorena D, Astiasaran I. Oxysterols: a world to explore. Food Chem. Toxicol. 48, 3289–3303 (2010).
  • Hovenkamp E, Demonty I, Plat J, Lutjohann D, Mensink RP, Trautwein EA. Biological effects of oxidized phytosterols: a review of the current knowledge. Prog. Lipid Res. 47, 37–49 (2008).
  • Guardiola F, Codony R, Addis PB, Rafecas M, Boatella J. Biological effects of oxysterols: current status. Food Chem. Toxicol. 34, 193–211 (1996).
  • Sottero B, Gamba P, Gargiulo S, Leonarduzzi G, Poli G. Cholesterol oxidation products and disease: an emerging topic of interest in medicinal chemistry. Curr. Med. Chem. 16, 685–705 (2009).
  • Vejux A, Lizard G. Cytotoxic effects of oxysterols associated with human diseases: induction of cell death (apoptosis and/or oncosis), oxidative and inflammatory activities, and phospholipidosis. Mol. Aspects Med. 30, 153–170 (2009).
  • Schroepfer GJ Jr. Oxysterols: modulators of cholesterol metabolism and other processes. Physiol. Rev. 80, 361–554 (2000).
  • Smith LL. Cholesterol Autoxidation. Plenum Press, NY, USA (1981).
  • Wollam J, Antebi A. Sterol regulation of metabolism, homeostasis, and development. Annu. Rev. Biochem. 80, 885–916 (2011).
  • Javitt NB. Oxysterols: novel biologic roles for the 21st century. Steroids 73, 149–157 (2008).
  • Crosignani A, Zuin M, Allocca M, Del Puppo M. Oxysterols in bile acid metabolism. Clin. Chim. Acta 412, 2037–2045 (2011).
  • Shibata N, Glass CK. Macrophages, oxysterols and atherosclerosis. Circulation J. 74, 2045–2051 (2010).
  • Diczfalusy U. Analysis of cholesterol oxidation products in biological samples. J. AOAC Int. 87, 467–473 (2004).
  • Wu Z, Martin KO, Javitt NB, Chiang JY. Structure and functions of human oxysterol 7a-hydroxylase cDNAs and gene CYP7B1. J. Lipid Res. 40, 2195–2203 (1999).
  • Russell DW. Oxysterol biosynthetic enzymes. Biochim. Biophys. Acta 1529, 126–135 (2000).
  • Brown AJ, Jessup W. Oxysterols: sources, cellular storage and metabolism and new insights into their roles in cholesterol homeostasis. Mol. Aspects Med. 30, 111–122 (2009).
  • Bjorkhem I, Lutjohann D, Diczfalusky U, Stahle L, Ahlborg G, Wahren J. Cholesterol homeostasis in human brain: turnover of 24(S)-hydroxycholesterol and evidence for a cerebral origin of most of this oxysterol in the circulation. J. Lipid Res. 39, 1594–1600 (1998).
  • Iuliano L. Pathways of cholesterol oxidation via non-enzymatic mechanisms. Chem. Phys. Lipids 164, 457–468 (2011).
  • McDonald JG, Smith DD, Stiles AR, Russell DW. A comprehensive method for extraction and quantitative analysis of sterols and secosteroids from human plasma. J. Lipid Res. 53, 1399–1409 (2012).
  • Xu L, Sheflin LG, Poter NA, Fliesler SJ. 7-dehydrocholesterol-derived oxysterols and retinal degeneration in a rat model of Smith–Lemli–Opitz syndrome. Biochim. Biophys. Acta 1821(6), 877–883 (2012).
  • Haigh WG, Lee SP. Identification of oxysterols in human bile and pigment gallstones. Gastroenterology 121, 118–123 (2001). ▪▪ First description of oxysterol species in human bile and pigment gallstones.
  • Yoshida T, Matsuzaki Y, Haigh WG et al. Origin of oxysterols in hepatic bile of patients with biliary infection. Am. J. Gastroenterol. 98, 2275–2280 (2003). ▪▪ Comprehensive quantitative analysis of oxysterol species in hepatic bile obtained from patients with biliary tract diseases, showing an association with infected bile.
  • Iuliano L, Michelette F, Natoli S, Ginanni Corradini S et al. Measurement of oxysterols and alpha-tocopherol in plasma and tissue samples as indices of oxidant stress status. Anal. Biochem. 312, 217–223 (2003).
  • Kidambi S, Patel SB. Cholesterol and non-cholesterol sterol transporters: ABCG5, ABCG8 and NPC1L1: a review. Xenobiotica 38, 1119–1139 (2008).
  • Staprans I, Pan XM, Rapp JH, Feingold KR. Oxidized cholesterol in the diet is a source of oxidized lipoproteins in human serum. J. Lipid Res. 44, 705–715 (2003).
  • Liseisen J, Wolfram G. Absorption of cholesterol oxidation products from ordinary foodstuff in humans. Ann. Nutr. Metab. 42, 221–230 (1998).
  • Brown MS, Goldstein JL. Cholesterol feedback: from Schoenheimer’s bottle to Scap’s MELADL. J. Lipid Res. 50(Suppl.), S15–S27 (2009).
  • Gill S, Chow R, Brown AJ. Sterol regulators of cholesterol homeostasis and beyond: the oxysterol hypothesis revisited and revised. Prog. Lipid Res. 47, 391–404 (2008).
  • Lange Y, Ory DS, Ye J, Lanier MH, Hsu FF, Seck TL. Effectors of rapid homeostatic responses of endoplasmic reticulum cholesterol and HMG-CoA reductase. J. Biol Chem. 283, 1445–1455 (2008).
  • Porter JA. Cholesterol modification of hedgehog signaling proteins in animal development. Science 274, 255–259 (1996).
  • Mann RK, Beachy PA. Novel lipid modifications of secreted protein signals. Annu. Rev. Biochem. 73, 891–923 (2004).
  • Stottmann RW, Turbe-Doan A, Tran P et al. Cholesterol metabolism is required for intracellular hedgehog signal transduction in vivo. PLoS Genet. 7, e1002224 (2011).
  • Kha HT, Basseri B, Shouhed D et al. Oxysterols regulate differentiation of mesenchymal stem cells: pro-bone and anti-fat. J. Bone Miner. Res. 19, 830–840 (2004).
  • Dwyer JR, Sever N, Carlson M, Nelson SF, Beachy PA, Parhami F. Oxysterols are novel activators of the hedgehog signaling pathway in pluripotent mesenchymal cells. J. Biol Chem. 282, 8959–8968 (2007).
  • Corcoran RB, Scott MP. Oxysterols stimulate Sonic hedgehog signal transduction and proliferation of medulloblastoma cells. Proc. Natl Acad. Sci. USA 103, 8408–8413 (2006).
  • Nachtergaele S, Mydock LK, Krishnan K et al. Oxysterols are allosteric activators of the oncoprotein Smoothened. Nat. Chem. Biol. 8, 211–220 (2012).
  • Hannedouche S, Zhang J, Yi T et al. Oxysterols direct B cell and T cell migration via EBI2. Nature 475, 524–527 (2011).
  • Liu C, Yang XV, Wu J et al. Oxysterols direct B-cell migration through EBI2. Nature 475, 519–523 (2011).
  • Ngo MH, Colbourne TR, Ridgway ND. Functional implications of sterol transport by the oxysterol-binding protein gene family. Biochem. J. 429, 13–24 (2010).
  • Lehto M, Olkkonen VM. The OSBP-related proteins: a novel protein family involved in vesicle transport, cellular lipid metabolism and cell signaling. Biochim. Biophys. Acta 1631, 1–11 (2003).
  • Raychaudhuri S, Prinz WA. The diverse functions of oxysterol-binding proteins. Annu. Rev. Cell Dev. Biol. 26, 157–177 (2010).
  • Ryan E, Chopra J, McCarthy FO, Maguire AR, O’Brien NM. Qualitative and quantitative comparison of the cytotoxic and apoptotic potential of phytosterol and oxidation products with their corresponding cholesterol oxidation products. Br. J. Nutr. 94, 443–451 (2005).
  • Bjorkhem I, Diczfalusy U. Oxysterols: friends, foes, or just fellow passengers? Arterioscler. Thromb. Vasc. Biol. 22. 734–742 (2002).
  • Kudo K, Emmons GT, Casserly EW et al. Inhibitors of sterol synthesis. Chromatography of acetate derivatives of oxygenated sterols. J. Lipid Res. 30, 1097–1111 (1989).
  • Fuda H, Javitt NB, Mitamura K, Ikegawa S, Strott CA. Oxysterols are substrates for cholesterol sulfotransferase. J. Lipid Res. 48, 1343–1352 (2007).
  • Xu L, Bai Q, Rodriguez-Agudo D, Hylemon P et al. Regulation of hepatocyte lipid metabolism and inflammatory response by 25-hydroxycholeserol and 25-hydroxycholesterol-3-sulfate. Lipids 45, 821–832 (2010).
  • Bai Q, Xu L, Kakiyama G, Runge-Morris MA, Hylemon P et al. Sulfation of 25-hydroxycholesterol by SULF2B1b decreases cellular lipids via the LXR/ SREBP-1c signaling pathway in human aortic endothelial cells. Atherosclerosis 214, 350–356 (2011).
  • Xu L, Shen S, Ma Y et al. 25-hydroxycholesterol-3-sulfate attenuates inflammatory response via PPARg signaling in human THP-1 macrophages. Am. J. Physiol. Endocrinol. Metab. 302, e788–e799 (2012).
  • Bai Q, Zhang X, Xu L et al. Oxysterol sulfation by cytosolic sulfotransferase suppresses liver X receptor/sterol regulatory element binding protein-1c signaling pathway and reduces serum and hepatic lipids in mouse models of nonalcoholic fatty liver disease. Metabolism 61, 836–845 (2012).
  • Ko CW, Lee SP. Gallstone formation. Local factors. Gastroenterol. Clin. North Am. 28, 99–115 (1999).
  • Stokes CS, Krawczyk M, Lammert F. Gallstones: environment, lifestyle and genes. Dig. Dis. 29, 191–201 (2011).
  • Lee DK, Tarr PI, Haigh WG, Lee SP. Bacterial DNA in mixed cholesterol gallstones. Am. J. Gastroenterol. 94, 3502–3506 (1999).
  • Haigh WG, Wong T, Lee SP. The production of oxysterols in bile by activated human leukocytes. Biochem. Biophys. Res. Comm. 34, 467–469 (2006).
  • Oda D, Lee SP, Hayashi A. Long-term culture and partial characterization of dog gallbladder epithelial cells. Lab. Invest. 64, 682–692 (1991).
  • Seo DW, Choi H-S, Lee SP, Kuver R. Oxysterols from human bile induce apoptosis of canine gallbladder epithelial cells in monolayer culture. Am. J. Physiol. Gastrointest. Liver Physiol. 287, G1247–G1256 (2004). ▪ Cultured dog gallbladder epithelial cells were found to undergo apoptosis via a mitochondrial pathway when exposed to model bile solutions containing cholest-4-en-3-one, cholesta-4,6-dien-3-one, and 5b-cholestan-3-one, three oxysterol species identified in bile.
  • Yoshida T, Klinkspoor JH, Kuver R et al. Effects of bile salts on cholestan-3b, 5a, 6b-triol-induced apoptosis in dog gallbladder epithelial cells. Biochim. Biophys. Acta 1530, 199–208 (2001).
  • Kuver R. The expanding universe of bile acid physiology: delving into the mysteries of dark (green) matter. J. Surg. Res. doi:10.1016/j.jss.2012.03.041 (2012) (Epub ahead of print).
  • Wang YD, Chen WD, Yu D, Forman BM, Huang W. The G-protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor kappa light-chain enhancer of activated B cells (NF–kB) in mice. Hepatology 54, 1421–1432 (2011).
  • Fiorucci S, Cipriani S, Mencarelli A, Renga B, Distrutti E, Baldelli F. Counter-regulatory role of bile acid activated receptors in immunity and inflammation. Curr. Mol. Med. 10, 579–595 (2010).
  • Cipriani S, Mencarelli A, Chini MG, Distrutti E et al. The bile acid receptor Gpbar-1 (TGR5) modulates integrity of intestinal barrier and immune response to experimental colitis. PLoS ONE 6(10), e25637 (2011).
  • Yoshida T, Klinkspoor JH, Kuver R, Wrenn SP, Kaler EW, Lee SP. Cholestan-3b, 5a, 6b-triol, but not 7-ketocholesterol, suppresses taurocholate-induced mucin secretion by cultured dog gallbladder epithelial cells. FEBS Lett. 478, 113–118 (2000).
  • Patel T. Increasing incidence and mortality of primary intrahepatic cholangiocarcinoma in the United States. Hepatology 33, 1353–1357 (2001).
  • Patel T. Cholangiocarcinoma. Nat. Clin. Pract. Gastroenterol. Hepatol. 3, 33–42 (2006).
  • Smout MJ, Sripa B, Laha T et al. Infection with the carcinogenic human liver fluke, Opisthorchis viverrini. Mol. Biosyst. 7, 1367–1375 (2011).
  • Yongvanit P, Pinlaor S, Bartsch H. Oxidative and nitrative DNA damage: key events in opisthorchiasis-induced carcinogenesis. Parasitol. Int. 61, 130–135 (2012).
  • Pinlaor S, Sripa B, Ma N et al. Nitrative and oxidative DNA damage in intrahepatic cholangiocarcinoma patients in relation to tumor invasion. World J. Gastroenterol. 11, 4644–4649 (2005).
  • Thamavit W, Bhamarapravati N, Saphaphong S, Vajrasthira S, Angsubhakorn S. Effects of dimethylnitrosamine on induction of cholangiocarcinoma in Opisthorchis viverrini-infected Syrian golden hamsters. Cancer Res. 38, 4634–4639 (1978).
  • Loilome W, Yongvanit P, Wongkham C et al. Altered gene expression in Opisthorchis viverrini-associated cholangiocarcinoma in hamster model. Mol. Carcinog. 45, 279–287 (2006). ▪ Differential display PCR on liver tissues from hamsters induced to develop cholangiocarcinoma identified an oxysterol-binding protein, OSBPL-8, among transcripts that were upregulated compared with control tissue.
  • Dechakhamphu S, Pinlaor S, Sitthithaworn P, Bartsch H, Yongvanit P. Accumulation of miscoding etheno-DNA adducts and highly expressed DNA repair during liver fluke-induced cholangiocarcinogenesis in hamsters. Mutat. Res. 691, 9–16 (2010).
  • Dechakhamphu S, Pinlaor S, Sitthithaworn P, Nair J, Bartsch H, Yongvanit P. Lipid peroxidation and etheno DNA adducts in white blood cells of liver fluke-infected patients: protection by plasma alpha-tocopherol and praziquantel. Cancer Epidemiol. Biomarkers Prev. 19, 310–318 (2010).
  • Loilome W, Wechagama P, Namwat N et al. Expression of oxysterol binding protein isoforms in opisthorchiasis-associated cholangiocarcinoma: a potential molecular marker for tumor metastasis. Parasitol. Int. 61, 136–139 (2012). ▪ In human cholangiocarcinoma tissue, increased levels of OSBPL-7 mRNA and protein were found compared with non-tumor liver tissues. Higher levels of OSBP2 and OSBPL-7 mRNA were found in blood samples from cholangiocarcinoma patients compared with normal controls.
  • Jusakul A, Loilome W, Namwat N et al. Liver fluke-induced hepatic oxysterols stimulate DNA damage and apoptosis in cultured human cholangiocytes. Mutat. Res. 731, 48–57 (2012). ▪▪ In the hamster model of cholangiocarcinoma induced by Opisthorchis viverrini and NMDA, two oxysterols (cholestan-3b, 5a, 6b-triol and 3-keto-cholesta-4, 6-diene) were found at higher levels in tumor tissues compared with livers from control animals. These two oxysterols were found to induce DNA adduct formation and induce apoptosis in cultured cholangiocytes.
  • Wehbe H, Henson R, Meng F, Mize-Berge J, Patel T. Interleukin-6 contributes to growth in cholangiocarcinoma cells by aberrant promoter methylation and gene expression. Cancer Res. 66, 10517–10524 (2006).
  • Yoon JH, Canbay AE, Werneburg NW, Lee SP, Gores GJ. Oxysterols induce cyclooxygenase-2 expression in cholangiocytes: implications for biliary tract carcinogenesis. Hepatology 39, 732–738 (2004).
  • Jaiswal M, LaRusso NF, Shapiro RA, Billiar TR, Gores GJ. Nitric oxide-mediated inhibition of DNA repair potentiates oxidative DNA damage in cholangiocytes. Gastroenterology 120, 190–199 (2001).
  • Jaiswal M, LaRusso NF, Burgart LJ, Gores GJ. Inflammatory cytokines induce DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric-oxidedependent mechanism. Cancer Res. 60, 184–190 (2000).
  • Braconi C, Huang N, Patel T. MicroRNAdependent regulation of DNA methyltransferase-1 and tumor suppressor gene expression by interleukin-6 in human malignant cholangiocytes. Hepatology 51. 881–890 (2010).
  • Gallus GN, Dotti MT, Federico A. Clinical and molecular diagnosis of cerebrotendinous xanthomatosis with a review of the mutations in the CYP27A1 gene. Neurol. Sci. 27, 143–149 (2006).
  • Campbell DJ, Dumur CI, Lamour NF, Dewitt JL, Sirica AE. Novel organotypic culture model of cholangiocarcinoma progression. Hepatol. Res. doi:10.1111/j.1872-034X.01026.x (2012) (Epub ahead of print).

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.