2D DIGE analysis of butyrate treated hepg2 cells

129 229 0
2D DIGE analysis of butyrate treated hepg2 cells

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

2D-DIGE ANALYSIS OF BUTYRATE TREATED HCC CELL LINE, HEPG2 VINCENT, LAU SIANG LIN B.Sc. (Hons.), NUS A THESIS SUBMITTED FOR THE MASTERS OF SCIENCE DEPARTMENT OF BIOCHEMISTRY NATIONAL UNIVERSITY OF SINGAPORE 2011 ACKNOWLEDGEMENTS I would like to express sincere gratitude to my principal investigator, Associate Professor Maxey Chung Ching Ming, for his guidance and support throughout the span of this project. He has given me opportunities and exposure to the different facets of scientific research. In addition, acknowledgement specifically goes to Dr. Sandra Tan and Dr. Tan Hwee Tong for their insights and assistance on the direction of the project as well as advises during times of experimental troubleshooting. I owe a large part of the rich experiences in my post-graduate studies to the members of my laboratory especially to research assistants Cynthia Liang Mui Yee, Lim Teck Kwang and Tan Gek San. Last but not least, I thank my fellow post-graduate students Hendrick Loei, Lin Qifeng and Zubaidah Ramdzan for their constant encouragement and camaraderie. TABLE OF CONTENTS PAGE ACKNOWLEDGEMENTS ABSTRACT . INDEX OF TABLES . INDEX OF FIGURES . INDEX OF ABBREVIATIONS . 12 1. LITERATURE REVIEW . 15 1.1 Hepatocellular carcinoma . 15 1.1.1 Etiological factors 15 1.1.2 Disease pathology 16 1.1.3 Disease diagnosis and classification 19 1.1.4 Current treatments of HCC 21 1.1.5 Molecular pathogenesis 23 1.2 Butyrate 25 1.2.1 Butyrate and cancer 26 1.2.2 Molecular effects of butyrate . 27 1.4 Proteomics and current platforms . 32 1.4.1 Tools in expressional proteomics . 34 1.4.2 2D-DIGE 35 1.4.3 Sample prefractionation with heparin affinity chromatography 36 2. OBJECTIVES OF THE STUDY . 39 3. MATERIALS AND METHODS 41 3.1 Materials 41 3.1.1 Cell lines . 41 3.1.2 Instruments and Equipment 42 3.1.4 General Chemicals and Reagents . 43 3.1.5 Western Blot antibodies and reagents 45 3.1.6 Softwares and Databases 46 3.2 Methods 48 3.2.1 Cell line extract preparation . 48 3.2.2 Heparin Affinity Liquid Chromatography (HPLC) . 50 3.2.3 Sodium dodecyl sulphate poly-acrylamide gel electrophoresis (SDS PAGE) 52 3.2.4 2-Dimensional gel electrophoresis (2-DE) . 53 3.2.5 2-Dimension Difference Gel Electrophoresis (2D-DIGE) . 54 3.2.6 Decyder image analysis 57 3.2.7 In-gel tryptic digestion for Mass spectrometry analysis 58 3.2.8 Matrix-assisted laser desorption/ionization tandem Mass spectrometry 59 3.2.9 Bioinformatics annotation tools . 60 3.2.10 Immunobloting . 60 3.2.11 MTT cell viability assay . 62 3.2.12 Transwell migration assay 63 3.2.13 Basement membrane matrix invasion assay . 63 3.2.14 siRNA knockdown of GSN 64 4. RESULTS . 65 4.1 HCC cell lines exhibit butyrate induced cell-growth inhibition . 65 4.3 Butyrate increases expression of p21 and p53, and decreases c-Myc levels 69 4.4 2D-DIGE analysis after heparin affinity chromatography . 69 4.5 Differential protein expression in HepG2 cells after butyrate stimulation 75 4.5.1 Localization and biological functions of deregulated proteome 81 4.5.2 Butyrate-affected pathways and cellular processes 81 4.6 Immunoblotting validation of selected regulated proteins 85 4.7 Over-expression of Gelsolin (GSN) by butyrate . 86 4.8 Functional study on GSN 87 5. DISCUSSION 91 5.1 Butyrate inhibits growth but promotes migration in HCC lines 92 5.2 Using HepG2 as a cell model for proteomic analysis of drug treatment 93 5.3 Identification of novel proteins involved in butyrate . 94 5.4 Proteins differentially-expressed in HepG2 after butyrate treatment . 96 5.4.1 Changes in Metabolic Program 97 5.4.2 Tumor suppressing effects of butyrate in HepG2 cells 102 5.5 Butyrate-inhibited metastasis . 106 5.5.1 Proteins known in metastasis . 108 5.5.2 Increased expression of gelsolin after butyrate stimulation . 110 6. CONCLUSION 112 7. REFERENCE 114 ABSTRACT Hepatocellular carcinoma (HCC) is one of the top causes of cancer deaths in Asia Pacific, and its incidence is predicted to increase in the next decade. The lethality of this disease can be seen from the high mortality rates and poor prognosis with a 5-year survival rate of only 5%. For patients with extrahepatic metastasis, the life expectancy decreases to a mere months. This is due to lack of reliable biomarkers, poor understanding of HCC tumorigenesis and limited clinical therapies. Butyrate, a physiological saccharolytic fermentative by-product of the colonic microflora, is an attractive chemotherapeutic agent with curative potentials due to its cancer-specific cytostatic and apoptotic effects. As the liver is the second site of butyrate delivery via hepatic portal transport and butyrate is well tolerated by hepatocytes, butyrate treatment could be an avenue to lengthen the lifespan of HCC patients awaiting liver transplants. Despite treatment feasibility, butyrate’s mode of actions against neoplasm, such as cell arrest induction and metastasis abrogation, remains oblique in liver cancer cells. In this study, we employed a novel proteomics workflow using heparin affinity chromatography and 2-Dimension Difference Gel Electrophoresis (2D-DIGE) to identify butyrate-induced changes in proteins in the HepG2 liver cancer cell line. We focused on cytoskeletal-related proteins in cell migration and invasion pathways which are enriched via heparin interaction. Our results revealed that the three HCC cell lines HepG2, HCC-M and Hep3B exhibited reduced cell viability significantly after butyrate treatment. The treatment however resulted in significant increased cell motility in all the cell lines. From this proteomics screening of butyrate-treated HepG2 cells, a total of 52 proteins’ expressions were detected to be dysregulated. These butyrate-induced proteins are mainly involved in glucose catabolism, urea cycle and nucleic acid synthesis. This study also yielded a list of cancer-associated proteins that were regulated after treatment thus suggesting the reversal of the malignant phenotype of HCC cells by butyrate. A group of proteins involved in cell migration and cytoskeletal reorganization was identified in this study which included LRRC15, NME1, and GSN. These proteins may help us to understand the mechanisms behind the changes in cell mobility after butyrate treatment. Gelsolin (GSN) is one of the targets up-regulated by butyrate treatment in HepG2 cells indicating its possible involvement in butyrate’s effect on cell motility or invasion. HepG2 with siRNA-mediated GSN knockdown resulted in increased basement membrane invasion. This suggested that up-regulation of GSN by butyrate is crucial for butyrate-mediated suppression of metastasis. INDEX OF TABLES Table No. Description Page No. Table 3.1: Labeling strategy for 2D-DIGE of heparin-bound and unbound 56 protein fractions. control-treated paired biological replicate were analyzed. Table 3.2: Concentration of primary and secondary antibodies used for western 62 blot Table 4.1: 76 Protein spots detected to be regulated with more than 1.5-fold from 2D-DIGE of heparin-bound and unbound samples. Spot numbers are numbered by DeCyderTM. The ‘Appearance’ column depicts the number of replicate gels that each individual spot was detected in. Table 4.2: Regulated protein spots identified by MALDI TOF/TOF mass 78 spectrometry after in-gel tryptic digestion, with total ion score C.I.% and best ion score C.I.%. Table 5.1: Cellular movement-related proteins identified in butyrate-treated 108 HepG2 dataset as categorized by IPA. INDEX OF FIGURES Figure No. Description Page No. Figure 4.1: Cell viability of (A) HepG2, (B) HCC-M, and (C) Hep3B HCC cell 66 lines were quantified using MTT assay. Cells were treated with mM butyrate treatment for 120 hrs at 12-, 24-, 48-, 72-, 96-, and 120-hr time-points. Respective non-treated controls were done in parallel. Figure 4.2: Migration potential of HepG2, HCC-M and Hep3B cell lines before 68 and after 22 hrs of mM butyrate treatment was assessed via boyden transwell assay. HCT116 colon cancer cell line was performed as an experimental control. The number of migrated cells treated with butyrate was expressed as a ratio of migrated control cells. Figure 4.3: 1-D western blot of p21, c-Myc, and p53 on mM, 24 hr butyrate- 69 treated (Trt) and control (Crtl) HepG2 whole cell extracts with GAPDH as loading control. Figure 4.4: 71 HPLC elution profile, at 280 nM absorbance, of mg (A) control and (B) butyrate-treated HepG2 cell lysates with protein fractions collected in a two-step program: (I) The larger peak contains unbound proteins and (II) a smaller peak contains heparin-bound proteins eluted at 0.8 M NaCl. Figure 4.5: 72 Representative images of 2D-DIGE analysis of (A) unbound and (B) heparin-bound protein fractions. 120 µg of proteins from Cydyelabeled internal standard, control and treatment samples were combined and electrophoresed on the same IPG strip of pH 3–10 nonlinear. Gel images were acquired with Typhoon fluorescent scanner. Figure 4.6: Representative silver-stained gel images of (A) unbound and (B) 73 heparin-bound protein fraction proteins. Figure 4.7: Distribution of identified butyrate-induced differentially expressed 82 proteins categorized by (A) cellular localization and (B) biological functions based on GO consortium. Figure 4.8: 83 Top biological functions and canonical pathways identified from butyrate-regulated proteins using IPA. (A) Top molecular and cellular functions (B) Selected study-relevant canonical pathways. All function and pathway analysis represented here are significant at p < 0.05. Figure 4.9: Top network ‘cancer’ from IPA. Schematic illustration of the 84 interaction behavior of differentially expressed proteins. Arrows described the direct associations between the proteins. Solid lines and dotted lines represent direct and indirect interactions. Proteins that are down-regulated and up-regulated are denoted in red and green respectively Figure 4.10: 2-D immunoblot validation of selected proteins regulated by mM 85 butyrate treatment for 24 hr. Figure 4.11: 2-D western detection of control and 24-hr butyrate-treated whole 86 cell extracts using anti-GSN antibodies. The spots observed at approximately 90 kDa correspond to spots 303 and 304 detected in the 2D-DIGE analysis. Figure 4.12: Western detection of GSN in control and mM butyrate-treated cell 87 extracts across 120 hours. Actin is the loading control. Figure 4.13: Western blot of siRNA knockdown GSN knockdown in control and 88 butyrate-treated cells. (A) HepG2 cells with various siRNA transfections are subjected to mM butyrate treatment for 24 hr. Lanes and are control cell extracts with no siRNA; Lanes and 10 7. REFERENCE Abe S., Ito Y., Davies E., (1995) Isolation of a heparin sensitive, ribosome sedimenting factor from the cytoskeleton fractions of peas and corn. Plant Physiology and Biochemistry. 33:463-70. Alcarraz-Vizán G., Boren J., Lee W.N., Cascante M., (2010) Histone deacetylase inhibition results in a common metabolic profile associated with HT29 differentiation. Metabolomics. 6:229-237. Anderson L. and Seihammer J., (1997) A comparison of selected mRNA and protein abundances in human liver. Electrophoresis. 18:533-37 Anthony P.P., Vogel C.L., Barker L.F., (1973) Liver cell dysplasia: a premalignant condition. J Clin Pathol. 26:217-23. Bhalla K.N., (2005) Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies. J Clin Oncol. 23:3971-93. Bingham S.A., Day N.E., Luben R., Ferrari P., Slimani N., Norat T., Clavel-Chapelon F., Kesse E., Nieters A., Boeing H., Tjønneland A., Overvad K., Martinez C., Dorronsoro M., Gonzalez C.A., Key T.J., Trichopoulou A., Naska A., Vineis P., Tumino R., Krogh V., Bueno-de-Mesquita H.B., Peeters P.H., Berglund G., Hallmans G., Lund E., Skeie G., Kaaks R., Riboli E.; European Prospective Investigation into Cancer and Nutrition, (2003) Dietary fibre in food and protection against colorectal cancer in the European 114 Prospective Investigation into Cancer and Nutrition (EPIC): an observational study. Lancet. 361:1496-501. Blouin J.M., Penot G., Collinet M., Nacfer M., Forest C., Laurent-Puig P., Coumoul X., Barouki R., Benelli C., Bortoli S., (2010) Butyrate elicits a metabolic switch in human colon cancer cells by targeting the pyruvate dehydrogenase complex. Int J Cancer. Epub ahead of print. Bolden J.E., Peart M.J., Johnstone R.W., (2006) Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 5:769-84. Boren J., Lee W.N., Bassilian S., Centelles J.J., Lim S., Ahmed S., Boros L.G., Cascante M., (2003) The stable isotope-based dynamic metabolic profile of butyrate-induced HT29 cell differentiation. J Biol Chem. 278:28395-402. Brenner C., Deplus R., Didelot C., Loriot A., Viré E., De Smet C., Gutierrez A., Danovi D., Bernard D., Boon T., Pelicci P.G., Amati B., Kouzarides T., de Launoit Y., Di Croce L., Fuks F., (2005) Myc represses transcription through recruitment of DNA methyltransferase corepressor. EMBO J. 24:336-346. Bruix J., Sherman M., Llovet J.M., Beaugrand M., Lencioni R., Burroughs A.K., Christensen E., Pagliaro L., Colombo M., Rodés J.; EASL Panel of Experts on HCC, (2001) Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona2000 EASL conference. European Association for the Study of the Liver. J Hepatol. 35:421-30. Burgstaler E.A., Pineda A.A., Ellefson R.D., (1980) Removal of plasma lipoproteins 115 from circulating blood with a heparin-agarose column. Mayo Clin Proc. 55:180-4. Bréchot C., (1998) Molecular mechanisms of hepatitis B and C related to liver carcinogenesis. Hepatogastroenterology. 45:1189-96. Cao X.X., Mohuiddin I., Ece F., McConkey D.J., Smythe W.R., (2001) Histone deacetylase inhibitor downregulation of bcl-xl gene expression leads to apoptotic cell death in mesothelioma. Am J Respir Cell Mol Biol. 25:562-8. Caso G., McNurlan M.A., McMillan N.D., Eremin O., Garlick P.J., (2004) Tumour cell growth in culture: dependence on arginine. Clin Sci (Lond). 107:371-9. Chen Y.X., Fang J.Y., Zhu H.Y., Lu R., Cheng Z.H., Qiu D.K., (2004) Histone acetylation regulates p21WAF1 expression in human colon cancer cell lines. World J Gastroenterol. 10:2643-6. Cheng P.N., Leung Y.C., Lo W.H., Tsui S.M., Lam K.C., (2005) Remission of hepatocellular carcinoma with arginine depletion induced by systemic release of endogenous hepatic arginase due to transhepatic arterial embolisation, augmented by high-dose insulin: arginase as a potential drug candidate for hepatocellular carcinoma. Cancer Lett. 224:67-80. Choi J.S., Paek A.R., Kim S.Y., You H.J., (2010) GIPC mediates the generation of reactive oxygen species and the regulation of cancer cell proliferation by insulin-like growth factor-1/IGF-1R signaling. Cancer Lett. 28:254-63. 116 Chopin V., Toillon R.A., Jouy N., Le Bourhis X., (2002) Sodium butyrate induces P53independent, Fas-mediated apoptosis in MCF-7 human breast cancer cells. Br J Pharmacol. 135:79-86. Coradini D. and Speranza A., (2005) Histone deacetylase inhibitors for treatment of hepatocellular carcinoma. Acta Pharmacol Sin. 26:1025-33. Couchie D., Holic N., Chobert M.N., Corlu A., Laperche Y., (2002) In vitro differentiation of WB-F344 rat liver epithelial cells into the biliary lineage. Differentiation. 69:209-15. Cousens L.S., Gallwitz D., Alberts B.M., (1979) Different accessibilities in chromatin to histone acetylase. J Biol Chem. 254:1716-23. Dang CV. c-Myc target genes involved in cell growth, apoptosis, and metabolism. Mol Cell Biol. 1999;19:1–11. Fantin V.R. and Richon V.M., (2007) Mechanisms of resistance to histone deactylase inhibitors and their therapeutic implications. Clin Cancer Res. 13:7237-42. Farazi P.A. and DePinho R.A., (2006) Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer. 6:674-87. Feitelson M.A., (2004) c-myc overexpression in hepatocarcinogenesis. Hum Pathol. 35:1299-302. Filmus J. and Capurro M., (2004) Glypican-3 and alphafetoprotein as diagnostic tests for hepatocellular carcinoma. Mol Diagn. 4:207-12. 117 Gadgil H., Jarrett H.W., (1999) Heparin elution of transcription factors from DNASepharose columns. J Chromatogr A. 848:131-8. Gentry P.W. and Alexander B., (1973) Specific coagulation factor adsorption to insoluble heparin. Biochem Biophys Res Commun. 50:500-9. Giardina C., Boulares H., Inan M.S., (1999) NSAIDs and butyrate sensitize a human colorectal cancer cell line to TNF-alpha and Fas ligation: the role of reactive oxygen species. Biochim Biophys Acta. 1448:425-38. Glozak M.A., Sengupta N., Zhang X., Seto E., (2005) Acetylation and deacetylation of non-histone proteins. Gene. 363:15-23. Grando-Lemaire V, Guettier C, Chevret S, Beaugrand M, Trinchet JC. (1999) Hepatocellular carcinoma without cirrhosis in the West: epidemiological factors and histopathology of the non-tumorous liver. J Hepatol. 31:508-13. Grisham J.W., (2001) Molecular genetic alterations in primary hepatocellular neoplasms: hepatocellular adenoma, hepatocellular carcinoma, and hepatoblastoma. The molecular basis of human cancer. Eds Coleman W.B. and Tsongalis G.J., 269-346. Gregoretti I.V., Lee Y.M., Goodson H.V., (2004) Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J Mol Biol. 338:1731. Guo Q.M., Malek R.L., Kim S., Chiao C., He M., Ruffy M., Sanka K., Lee N.H., Dang C.V., Liu E.T., (2000) Identification of c-myc responsive genes using rat cDNA microarray. Cancer Res. 60: 5922-5928. 118 Gygi S.P., Corthals G.L., Zhang Y., Rochon Y., Aebersold R., (2000) Evaluation of twodimensional gel electrophoresis-based proteome analysis technology. Proc Natl Acad Sci U S A. 97:9390-5. Haga K, Fujita H, Nomoto M, Sazawa A, Nakagawa K, Harabayashi T, Shinohara N, Takimoto M, Nonomura K, Kuzumaki N. (2004) Gelsolin gene silencing involving unusual hypersensitivities to dimethylsulfate and KMnO4 in vivo footprinting on its promoter region. Int J Cancer. 111:873-80. Hall M.P. and Schneider L.V., (2004) Isotope-differentiated binding energy shift tags (IDBEST) for improved targeted biomarker discovery and validation. Expert Rev Proteomics. 1:421-31. Hamer H.M., Jonkers D., Venema K., Vanhoutvin S., Troost F.J., Brummer R.J., (2008) Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther. 27:104-19. Hernandez A., Thomas R., Smith F., Sandberg J., Kim S., Chung D.H., Evers B.M., (2001) Butyrate sensitizes human colon cancer cells to TRAIL-mediated apoptosis. Surgery. 130:265-72. Hoshikawa Y., Kwon H.J., Yoshida M., Horinouchi S., Beppu T., (1994) Trichostatin A induces morphological changes and gelsolin expression by inhibiting histone deacetylase in human carcinoma cell lines. Exp Cell Res. 214:189-97. Hradec J., Dusek Z., (1978) All factors required for protein synthesis are retained on heparin bound to Sepharose. Biochem J. 172:1-7. 119 Hrebicek T., Dürrschmid K., Auer N., Bayer K., Rizzi A., (2007) Effect of CyDye minimum labeling in differential gel electrophoresis on the reliability of protein identification. Electrophoresis. 28:1161-9. Huang Y.Q., Wang L.W., Yan S.N., Gong Z.J., (2004) Effects of cell cycle on telomerase activity and on hepatitis B virus replication in HepG2 2.2.15 cells. Hepatobilary Prancreat Dis Int 3:543-7. Humprey-Smith I., Cordwell S.J., Blackstock W.P., (1997) Proteome research: complementarity and limitations with respect to the RNA and DNA worlds. Electrophoresis. 18:1217-42. Hussain S.M., Zondervan P.E., IJzermans J.N., Schalm S.W., de Man R.A., Krestin G.P., (2002) Benign versus malignant hepatic nodules: MR imaging findings with pathologic correlation. Radiographics 22:1023–36. Illi B., Dello Russo C., Colussi C., Rosati J., Pallaoro M., Spallotta F., Rotili D., Valente S., Ragone G., Martelli F., Biglioli P., Steinkuhler C., Gallinari P., Mai A., Capogrossi M.C., Gaetano C., (2008) Nitric oxide modulates chromatin folding in human endothelial cells via protein phosphatase 2A activation and class II histone deacetylases nuclear shuttling. Circ Res. 102:51-8. Ishii H, Furuse J, Kinoshita T, Konishi M, Nakagohri T, Takahashi S, Gotohda N, Nakachi K, Yoshino M. (2004) Extrahepatic spread from hepatocellular carcinoma: who are candidates for aggressive anti-cancer treatment? Jpn J Clin Oncol. 34:733-739. 120 Javitt N.B., (1990) Hep-G2 cells as a resource for metabolic studies - lipoprotein, cholesterol, and bile-acids. FASEB J. 4:161–168. Kaibori M., Ishizaki M., Matsui K., Kitade H., Matsui Y., Kwon A.H., (2011) Evaluation of metabolic factors on the prognosis of patients undergoing resection of hepatocellular carcinoma. J Gastroenterol Hepatol. 26:536-43. Kamitani H., Taniura S., Ikawa H., Watanabe T., Kelavkar U.P., Eling T.E., (2001) Expression of 15-lipoxygenase-1 is regulated by histone acetylation in human colorectal carcinoma. Carcinogenesis. 22:187-91. Katoh M., (2002) GIPC gene family (Review). Int J Mol Med. 9:585-9. Khan K.N., Tsutsumi T., Nakata K., Kato Y., (1999) Sodium butyrate induces alkaline phosphatase gene expression in human hepatoma cells. J Gastroenterol Hepatol. 14:15662. Kim Y.H., Park J.W., Lee J.Y., Kwon T.K., (2004) Sodium butyrate sensitizes TRAILmediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis. 25:1813-20. Kirikoshi H., Katoh M., (2002) Expression of human GIPC1 in normal tissues, cancer cell lines, and primary tumors. Int J Mol Med. 9:509-13. Klein A., Olendrowitz C., Schmutzler R., Hampl J., Schlag P.M., Maass N., Arnold N., Wessel R., Ramser J., Meindl A., Scherneck S., Seitz S., (2009) Identification of brainand bone-specific breast cancer metastasis genes. Cancer Lett. 276:212-20. 121 Knowles M.R., Cervino S., Skynner H.A., Hunt S.P., de Felipe C., Salim K., MenesesLorente G., McAllister G., Guest P.C., (2003) Multiplex proteomic analysis by twodimensional differential in-gel electrophoresis. Proteomics. 3:1162-71. Kobayashi T., Nishii M., Takagi Y., Titani K., Matsuzawa T., (1989) Molecular cloning and nucleotide sequence analysis of mRNA for human kidney ornithine aminotransferase. An examination of ornithine aminotransferase isozymes between liver and kidney. FEBS Lett. 255:300-4. Kwiatkowski D.J., (1999) Functions of gelsolin: motility, signaling, apoptosis, cancer. Curr Opin Cell Biol. 11:103-8. Larsson L.G., Pettersson M., Oberg F., Nilsson K., Lüscher B., (1994) Expression of mad, mxi1, max and c-myc during induced differentiation of hematopoietic cells: opposite regulation of mad and c-myc. Oncogene. 9:1247-52. Leschelle X., Delpal S., Goubern M., Blottière H.M., Blachier F., (2000) Butyrate metabolism upstream and downstream acetyl-CoA synthesis and growth control of human colon carcinoma cells. Eur J Biochem. 267:6435-42. Litwin M., Mazur A.J., Nowak D., Mannherz H.G., Malicka-Błaszkiewicz M., (2009) Gelsolin in human colon adenocarcinoma cells with different metastatic potential. Acta Biochim Pol. 56:739-43. Liu Y.B., Gao S.L., Chen X.P., Peng S.Y., Fang H.Q., Wu Y.L., Peng C.H., Tang Z., Xu B., Wang J.W., Deng G.L., Li H.J., Feng X.D., Qian H.R., (2005) Expression and 122 significance of heparanase and nm23-H1 in hepatocellular carcinoma. World J Gastroenterol. 11:1378-81. Llovet J.M., (2005) Updated treatment approach to hepatocellular carcinoma. J Gastroenterol. 40:225-35. Llovet J.M., Bruix J., (2003) Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology. 37:42942. Llovet J.M., Di Bisceglie A.M., Bruix J., Kramer B.S., Lencioni R., Zhu A.X., Sherman M., Schwartz M., Lotze M., Talwalkar J., Gores G.J., (2008) Design and endpoints of clinical trials in hepatocellular carcinoma. J Natl Cancer Inst. 100:698-711. Lopez P.M., Villanueva A., Llovet J.M., (2006) Systematic review: evidence-based management of hepatocellular carcinoma--an updated analysis of randomized controlled trials. Aliment Pharmacol Ther. 23:1535-47. Margalit H., Fischer N., Ben-Sasson S.A., (1993) Comparative analysis of structurally defined heparin binding sequences reveals a distinct spatial distribution of basic residues. J Biol Chem. 268:19228-31. Mariani M.R., Carpaneto E.M., Ulivi M., Allfrey V.G., Boffa L.C., (2003) Correlation between butyrate-induced histone hyperacetylation turn-over and c-myc expression. J Steroid Biochem Mol Biol. 86:167-71. Marks P., Rifkind R.A., Richon V.M., Breslow R., Miller T., Kelly W.K., (2001) Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer. 1:194-202. 123 Marks P.A., Dokmanovic M., (2005) Histone deacetylase inhibitors: discovery and development as anticancer agents. Expert Opin Investig Drugs. 14:1497-511. Marouga R., David S., Hawkins E., (2005) The development of the DIGE system: 2D fluorescence difference gel analysis technology. Anal Bioanal Chem. 382:669-78. Marshall J.C., Collins J., Marino N., Steeg P., (2010) The Nm23-H1 metastasis suppressor as a translational target. Eur J Cancer. 46:1278-82. Marui Y., McCall J., Gane E., Holden A., Duncan D., Yeong M.L., Chow K., Munn S., (2005) Liver transplantation for hepatocellular carcinoma in New Zealand: a prospective intent-to-treat analysis. N Z Med J. 118:U1532. Masahiro M., Masakiyo S., Ichiro A., Yoshihiko S., Seishi N., Ham-ho H., (2004) Involvement of Interferon Regulatory Factor and S100C/A11 in growth inhibition by Transforming Growth Factor in human hepatocellular carcinoma cells. Cancer Research. 64:4155-4161. Mathur A.K., Osborne N.H., Lynch R.J., Ghaferi A.A., Dimick J.B., Sonnenday C.J., (2010) Racial/ethnic disparities in access to care and survival for patients with early-stage hepatocellular carcinoma. Arch Surg. 145:1158-63. Mohiuddin I., Cao X., Fang B., Nishizaki M., Smythe W.R., (2001) Significant augmentation of pro-apoptotic gene therapy by pharmacologic bcl-xl down-regulation in mesothelioma. Cancer Gene Ther. 8:547-54. 124 Natsuizaka M., Omura T., Akaike T., Kuwata Y., Yamazaki K., Sato T., Karino Y., Toyota J., Suga T., Asaka M., (2005) Clinical features of hepatocellular carcinoma with extrahepatic metastases. J Gastroenterol Hepatol. 20:1781-178 Okusaka T, Okada S, Ishii H, Nose H, Nagahama H, Nakasuka H, Ikeda K, Yoshimori M. (1997) Prognosis of hepatocellular carcinoma patients with extrahepatic metastases. Hepatogastroenterology 44:251-257. Pajak B., Orzechowski A., Gajkowska B., (2007) Molecular basis of sodium butyratedependent proapoptotic activity in cancer cells. Adv Med Sci. 52:83-8. Pienta K.J., Partin A.W., Coffey D.S., (1989) Cancer as a disease of DNA organization and dynamic cell structure. Cancer Res. 49:2525-32. Puisieux, A., Galvin, K., Troalen, F., Bressac, B., Margais, C., Galun, E., Ponchel, F., Yakicier, C., Ji, J. & Ozturk, M. (1993) FASEB J. 7:1407-1413. Rada-Iglesias A., Enroth S., Ameur A., Koch C.M., Clelland G.K., Respuela-Alonso P., Wilcox S., Dovey O.M., Ellis P.D., Langford C.F., Dunham I., Komorowski J., Wadelius C., (2007) Butyrate mediates decrease of histone acetylation centered on transcription start sites and down-regulation of associated genes. Genome Res. 17:708-19. Reynolds P.A., Smolen G.A., Palmer R.E., Sgroi D., Yajnik V., Gerald W.L., Haber D.A., (2003) Identification of a DNA-binding site and transcriptional target for the EWSWT1(+KTS) oncoprotein. Genes Dev. 17:2094-107. Rosato R.R. and Grant S., (2005) Histone deacetylase inhibitors: insights into mechanisms of lethality. Expert Opin Ther Targets. 9:809-24. 125 Ruemmele F.M., Dionne S., Qureshi I., Sarma D.S., Levy E., Seidman E.G., (1999) Butyrate mediates Caco-2 cell apoptosis via up-regulation of pro-apoptotic BAK and inducing caspase-3 mediated cleavage of poly-(ADP-ribose) polymerase (PARP). Cell Death Differ. 6:729-35. Sakaguchi M., Miyazaki M., Takaishi M., Sakaguchi Y., Makino E., Kataoka N., Yamada H., Namba M., Huh N.H., (2003) S100C/A11 is a key mediator of Ca(2+)induced growth inhibition of human epidermal keratinocytes. Journal of Cell Biology. 163:825-35. Salama I., Malone P.S., Mihaimeed F., Jones J.L., (2008) A review of the S100 proteins in cancer. European Journal of Surgical Oncology. 34:357-364. Scatena R., Bottoni P., Pontoglio A., Giardina B., (2010) Revisiting the Warburg effect in cancer cells with proteomics. The emergence of new approaches to diagnosis, prognosis and therapy. Proteomics Clin Appl. 4:143-58. Schuetz C.S., Bonin M., Clare S.E., Nieselt K., Sotlar K., Walter M., Fehm T., Solomayer E., Riess O., Wallwiener D., Kurek R., Neubauer H.J., (2006) Progressionspecific genes identified by expression profiling of matched ductal carcinomas in situ and invasive breast tumors, combining laser capture microdissection and oligonucleotide microarray analysis. Cancer Res. 66:5278-86. Sell S. and Leffert H.L. (2008) Liver cancer stem cells. J Clin Oncol. 26:2800-5. Seow T.K., Liang R.C., Leow C.K., Chung M.C., (2001) Hepatocellular carcinoma: from bedside to proteomics. Proteomics. 1:1249-63. 126 Severi T., van Malenstein H., Verslype C., van Pelt J.F., (2010) Tumor initiation and progression in hepatocellular carcinoma: risk factors, classification, and therapeutic targets. Acta Pharmacol Sin. 31:1409-20. Sherman M., (2005) Hepatocellular carcinoma: epidemiology, risk factors, and screening. Semin Liver Dis. 25:143-54. Skolnick J., Fetrow J.S., Kolinski A., (2000) Structural genomics and its importance for gene function analysis. Nat Biotechnol. 18:283-7. Smela M.E., Currier S.S., Bailey E.A., Essigmann J.M., (2001) The chemistry and biology of aflatoxin B(1): from mutational spectrometry to carcinogenesis. Carcinogenesis. 22:535-45. Soldatenkov V.A., Prasad S., Voloshin Y., Dritschilo A., (1998) Sodium butyrate induces apoptosis and accumulation of ubiquitinated proteins in human breast carcinoma cells. Cell Death Differ. 5:307-12. Suzuki N.N., Koizumi K., Fukushima M., Matsuda A., Inagaki F., (2003) Crystallization and preliminary X-ray analysis of human uridine-cytidine kinase 2. Acta Crystallogr D Biol Crystallogr. 59:1477-8. Tabuchi Y., Arai Y., Kondo T., Takeguchi N., Asano S., (2002) Identification of genes responsive to sodium butyrate in colonic epithelial cells. Biochem Biophys Res Commun. 293:1287-94. 127 Tan H.T., Tan S., Lin Q., Lim T.K., Hew C.L., Chung M.C., (2008) Quantitative and temporal proteome analysis of butyrate-treated colorectal cancer cells. Mol Cell Proteomics. 7:1174-85. Thorgeirsson S.S. and Grisham J.W., (2002) Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet. 31:339-46. Tong A., Zhang H., Li Z., Gou L., Wang Z., Wei H., Tang M., Liang S., Chen L., Huang C., Wei Y., (2008) Proteomic analysis of liver cancer cells treated with suberonylanilide hydroxamic acid. Cancer Chemother Pharmacol. 61:791-802. Tsukamoto A. and Kaneko Y., (1991) Novobiocin and butyrate synergistically enhanced cytokeratin assembly and acetate uptake of human liver cells. Cell Biol Int Rep. 15:9834. Uka K., Aikata H., Takaki S., Shirakawa H., Jeong S.C., Yamashina K., Hiramatsu A., Kodama H., Takahashi S., Chayama K., (2007) Clinical features and prognosis of patients with extrahepatic metastases from hepatocellular carcinoma. World J Gastroenterol. 13:414-20. Unlü M., Morgan M.E., Minden J.S., (1997) Difference gel electrophoresis: a single gel method for detecting changes in protein extracts. Electrophoresis. 18:2071-7. Velázquez O.C., Jabbar A., DeMatteo R.P., Rombeau J.L., (1996) Butyrate inhibits seeding and growth of colorectal metastases to the liver in mice. Surgery. 120:440-7. Wong C.M. and Ng I.O., (2008) Molecular pathogenesis of hepatocellular carcinoma. Liver Int 28:160-74. 128 Van Lint C., Emiliani S., Verdin E., (1996) The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr. 5:245-53. Yan M.L., Wang Y.D., Lai Z.D., Tian Y.F., Chen H.B., Qiu F.N., Zhou S.Q., (2009) Pedunculated hepatocellular carcinoma and splenic metastasis. World J Gastroenterol. 15:5239-41. Zhang Q., McCorkle J.R., Novak M., Yang M., Kaetzel D.M., (2010) Metastasis suppressor function of NM23-H1 requires its 3'-5' exonuclease activity. Int J Cancer. 128:40-50. Zhu Y., Chung M.C., (2005) Proteomic analysis of HCC-M and HepG2 cells after treatment by butyrate, Unpublished thesis (MSc.), National University of Singapore. Zuo X., Echan L., Hembach P., Tang H.Y., Speicher K.D., Santoli D., Speicher D.W., (2001) Towards global analysis of mammalian proteomes using sample prefractionation prior to narrow pH range two-dimensional gels and using one-dimensional gels for insoluble and large proteins. Electrophoresis. 22:1603-15. Zubaidah R.M., Tan G.S., Tan S.B., Lim S.G., Lin Q., Chung M.C., (2008) 2D-DIGE profiling of hepatocellular carcinoma tissues identified isoforms of far upstream binding protein (FUBP) as novel candidates in liver carcinogenesis. Proteomics. 8:5086-96. 129 [...]... transcriptional activities of genes affected by butyrate treatment Changes in gene expression are ascribed to direct effects of HDACi on gene promoters as well as assorted downstream secondary effects Promoters of butyrateresponsive genes consist of the butyrate response elements,’ and one of the direct action of butyrate is mediated through transcription factors Sp1 and Sp3, such as the promoter of p21/WAF-1 For... cell surface, butyrate can modulate TNFR1, -R2, FADD and Fas-R/CD95, clearly showing its stimulation and restoration of susceptibility of tumor cells to death ligand-induced cell death The ease to commit malignant cells to apoptosize is furthered by butyrate- mediated elimination of antiapoptotic proteins such as XIAP Hitherto, one of the key characteristics of cancer cells is its loss of differentiated... treatment The immense advantages of proteomics offered by 2DE have not been utilized in this field In this study, we employed 2D- DIGE to uncover proteome changes that occur in the hepatoma cell line HepG2 In a cellular milieu, butyrate has been known to inhibit HDACs, resulting in a change of global acetylation profile Besides histones, the acetylation status of a variety of HDAC-regulated proteins is... human saliva Most of the absorbed butyrate is catabolized by the colonic epithelium, with remaining amounts of butyrate 25 transported in the portal blood The liver absorbs the rest of the micronutrient with little left for venous systemic circulation In addition of being an energy alternative for the epithelial cells, a plethora of colonic health-related processes are also influenced by butyrate These... stem cells to differentiation Butyrate s short systemic half-life has been overcome by 26 stabilization using carrier groups such as arginine butyrate rendering it an effective yet safe chemotherapeutic alternative The exploitation of butyrate however is done without a complete understanding of its molecular mechanisms Our previous work had focused on the actions of butyrate on colorectal cancer cells. .. small molecule in the liver even in cases of decompensation 1.2.2 Molecular effects of butyrate One of the main mechanism of actions through which butyrate acts is via inhibition of HDACs in the cell, resulting in the hyperacetylation of histones, chromatin relaxation and targeted changes in gene expression It is crucial to note that though similar alterations of gene expression can be observed in distinct... labelling methods, shotgun proteomics and array technologies 2-DE and 2D- DIGE of butyrate- treated HCC cells followed by tandem MS identification of differentially expressed protein spots are carried out in this study Stable-isotope labelling involves labelling of sample proteins prior to separation via liquid chromatography before tandem MS analysis Common labelling techniques in proteomics include cleavable... GSN (GSN-KD) and HCT116 as reference (B) MatrigelTM invasion assay for No siRNA, NTC and GSN-KD All migrated treated cells are expressed as a ratio of corresponding control cells without butyrate treatment 11 INDEX OF ABBREVIATIONS 1-D One-dimensional 2-DE Two-dimensional electrophoresis 2D- DIGE Two-dimensional difference gel electrophoresis Acc No Accession number ACN Acetonitrile AFB1 Aflatoxin B1... is chronic interstitial inflammation of the liver, demarcated by the replacement of normal tissue with fibrous tissue and loss of functional liver cells (Farazi, 2006) The detriment process often entails repetitive cycles of liver damage and tissue repair, preceding fibrosis, where persistent injuries are permanently filled by tough scar tissues instead of liver cells It is an irreversible process leading... cell viability assay measured at 550 nm absorbance of GSN- 89 knockdown HepG2 cells and respective non-targeting controls after 5 mM butyrate treatment at 24-, 48-, and 72-hr timepoints Figure 4.15: 90 Motility functional assay of GSN-knowndown cells (A) Transwell migration assay for HepG2 cells without siRNA treatment (No siRNA), treated with non-targeting siRNA (NTC), siRNA against GSN (GSN-KD) and . inflammation of the liver, demarcated by the replacement of normal tissue with fibrous tissue and loss of functional liver cells (Farazi, 2006). The detriment process often entails repetitive cycles of. increased cell motility in all the 3 cell lines. From this proteomics screening of butyrate-treated HepG2 cells, a total of 52 proteins’ expressions were detected to be dysregulated. These butyrate-induced. also yielded a list of cancer-associated proteins that were regulated after treatment thus suggesting the reversal of the malignant phenotype of HCC cells by butyrate. A group of proteins involved

Ngày đăng: 15/09/2015, 22:42

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan