Cloning and characterization of the promoter of the cancer associated gene, FAT10

119 317 0
Cloning and characterization of the promoter of the cancer  associated gene, FAT10

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

CLONING AND CHARACTERIZATION OF THE PROMOTER OF THE CANCER-ASSOCIATED GENE, FAT10 ZHANG DONGWEI NATIONAL UNIVERSITY OF SINGAPORE 2007 CLONING AND CHARACTERIZATION OF THE PROMOTER OF THE CANCER-ASSOCIATED GENE, FAT10 ZHANG DONGWEI (M. Sc.) Wuhan Institute of Virology Chinese Academy of Science A THESIS SUBMITTED FOR THE DEGREE OF MASTER OF SCIENCE DEPARTMENT OF BIOCHEMISTRY NATIONAL UNIVERSITY OF SINGAPORE 2007 ACKNOWLEDGEMENTS I would like to express my greatest gratitude to my supervisor Dr. Caroline Lee for her excellent supervision, encouragement, patience and unfailing support throughout the course of this work, and for her invaluable amendments to this thesis. I would like to thank the past and present members in CFG laboratory: Ren Jianwei, Wang Baoshuang, Tan Kun, Wang Jingbo, Wang Zihua, Alvin Lee, Xiao Peiyun, Wang Yu, Wang Lipeng and Alison Kan for their kind concern, helpful discussion, technical assistance, cooperation, and friendship. Specially thank Dr. Grace Pang for English correcting. My heartfelt and deepest appreciation goes to my wife, Tian Jing, for her love, patience, understanding, and support over these years. I also would like to thank my beloved daughter Esther, for the joy and happiness she brings me. Last, but certainly not the least, this thesis is dedicated to my parents, for their unwavering support, encouragement and belief in me always. Zhang Dongwei February 2007 Table of Contents ii TABLE OF CONTENTS ACKNOWLEDGEMENTS i TABLE OF CONTENTS ii LIST OF FIGURES vi LIST OF TABLES xiii LIST OF ABREVIATIONS ix LIST OF PUBLICATIONS xi SUMMARY xii CHAPTER I 1.1 INTRODUCTION Role of Ubiquitin in posttranslational modification 1 2 1.1.1 Ubiquitin and ubiquitylation 1.1.2 Ubiquitin-like protein family 1.1.3 The function of ubiquitin-like 2 3 4 1.2 6 General Information about FAT10 1.2.1 FAT10 as a new member of UBL family 1.2.2 FAT10 and immune response 1.2.3 FAT10 and tumorigenesis 1.2.3.1 FAT10 is overexpressed in tumour tissue 1.2.3.2 FAT10 and the chromosomal stability 1.3 Research objectives Specific Aim 1: Isolated and Characterize the Promoter of the FAT10 Gene Specific Aim 2: Determine if p53 plays a role in the regulation of FAT10 at the transcript level. Specific Aim 3: Evaluate if mutations/polymorphisms or differential methylation at the FAT10 promoter can account for the aberrant over-expression of FAT10 in the tumors of HCC patients. 6 8 10 10 11 13 13 15 16 Table of Contents CHAPTER II ISOLATION AND CHARACTERIZATION OF THE PROMOTER OF FAT10 GENE iii 18 2.1 Background 19 2.2 Materials and Methods 20 2.2.1 2.2.2 2.2.3 2.2.4 2.2.5 2.2.6 Cell lines and Transfection Reverse Transcript PCR DNA sequencing reaction Determination of FAT10, p53 and β-actin protein levels Identification of promoter region in the FAT10 gene Identification of IFN-g and TNF-a responsive domain in FAT10 promoter region 20 20 22 23 23 27 Results 28 2.3 2.3.1 FAT10 promoter resides at the 5’UTR 2.3.2 The responsive domain of the FAT10 promoter to TNF-α and IFN-γ resides in the region upstream of FAT10 promoter 2.3.3 FAT10 promoter activity is different in different cell lines 28 2.4 37 Discussion CHAPTER III EVALUATION OF THE ROLE OF P53 IN THE REGULATION OF FAT10 AT THE TRANSCRIPT LEVEL 32 35 40 3.1 Background 41 3.2 3.2.1 3.2.2 3.2.3 3.2.4 3.2.5 Materials and Methods Cell lines and Transfection Quantitation of FAT10, p53, p21 and MDM2 and β-actin transcript Generation of siRNA constructs against p53 Generation of construct pFATgal-CMVp53 Determination of the binding of p53 to FAT10 promoter region using DNA and chromatin immuno-precipitation (DIP and ChIP) 43 43 43 44 46 48 Table of Contents 3.3 Results iv 50 3.3.1 p53 negatively regulates FAT10 promoter activity. 1.1.1 p53 binds to the 5’ half consensus sequence of p53 binding site of the FAT10 promoter and plays a role in the responsiveness of FAT10 promoter to p53 58 3.4 66 Discussion 50 CHAPTER IV EVALUATION OF THE ROLE OF MUTATIONS/POLYMORPHISM OR DIFFERENIAL METHYLATION AT THE FAT10 PROMOTER IN ACCOUNTING FOR THE ABERRANT OVER-EXPRESSION OF FAT10 IN THE TUMORS OF HCC PATIENTS 69 4.1 Background 70 4.2 Materials and Methods 72 4.2.1 4.2.2 4.2.3 4.2.4 Patient samples Isolation of genomic DNA and RNA from tissue samples Identification of Mutations/Polymorphisms through DNA sequencing Determination of the Methylation Status at the FAT10 promoter using Methylation-Specific Sequencing 72 72 72 4.3 Results 75 4.3.1 The expression levels of FAT10 in tumour tissue are much higher than adjacent non-tumorous liver tissues Only polymorphisms, but not mutations were identified in the ~1.3kb region of FAT10 promoter Differential Methylation at the FAT10 promoter was observed between tumour and adjacent normal liver tissues of HCC patients 4.3.2 4.3.3 4.4 Discussion 4.4.1 No mutations at the FAT10 Promoter were observed. 4.4.2 Differential methylation may account for the differences in FAT10 gene expression between HCC tumor and adjacent non-tumorous tissues 73 75 77 84 89 89 91 Table of Contents v CONCLUSION 93 REFERENCES 94 PUBLICATION Table of Contents vi LIST OF FIGURES Figure 1 Genomic structure of FAT10 gene 14 Figure 2 The strategy for TA-cloning of FAT10 promoter 25 Figure 3 Strategy for mutating the TATA box in FAT10 promoter region 26 Figure 4 Characterization of FAT10 promoter region 29 Figure 5 The diagram of FAT10 promoter that confers the highest promoter activity with important putative binding sites 30 Induction of different truncated FAT10 promoter by IFN-γ and TNF-α. 33 FAT10 promoter can not be induced by IFN-γ and TNF-α Synergistically 34 Figure 8 FAT10 promoter is more active in some cell-lines than others 36 Figure 9 The strategy to construct the SiRNA specific to p53 45 Figure 10 Strategy for making construct pFAT/β-gal-CMV/p53 47 Figure 11 p53 repression of FAT10 promoter activity 51 Figure 12 Repression of FAT10 transcription level in Hep3B cells by p53 52 Figure 13 Release the repression of p53 to FAT10 promoter by pH1-Sip53 in Hep3B cells 54 FAT10 promoter activity is enhanced by the addition of siRNA against p53 in p53+/+ KB3-1 cells and 293 cells 56 Endogenous FAT10 transcriptional level is also related to endogenous p53, along with other p53 regulated genes 57 Delineation of regions in FAT10 promoter that is responsible for the responsiveness of the promoter to p53 61 FAT10 promoter region emcompassing the p53 binding half site binds p53 in vivo 64 p53 binding the chromatin of the FAT10 promoter region emcompassing the p53 binding half site 65 Figure 6 Figure 7 Figure 14 Figure 15 Figure 16 Figure 17 Figure 18 Table of Contents Figure 19 vii Comparison of FAT10 transcript level in tumor tissues and adjacent non-tumorous tissues 76 Figure 20 SNPs in FAT10 promoter 79 Figure 21 FAT10 Promoter activity with different haplotype structures 82 Figure 22 The CG dinucleotides in FAT10 promoter region 86 Figure 23 The strategy for identifying the methylation status of CG dinucloetides in FAT10 promoter 87 Table of Contents viii LIST OF TABLES Table 1 Table 2 Table 3 Oligonucleotides for amplification of different length of fragments upstream of the transcription start site (TSS) or translation start site (TLSS) of FAT10 gene 24 Single nucleotide polymorphisms identified in the 5’-flanking region of FAT10 80 The methylation status of CG dinucleotides in FAT10 promoter in HCC patient tissues 88 Table of Contents LIST OF ABREVIATIONS aa amino acid Ab antibody β-gal β-galactosidase bp base pair ChIP Chromatin Immunoprecipitation CIN Chromosome Instability CMV Cytomegalovirus CPRG Chlorphenol red- β-D-galactopyranoside DEPC Diethyl pyrocarbonate DIP DNA immunoprecipitation EBV Epstein-Barr virus EGFP Enhanced Green Fluorescent Protein DMEM Dulbecco’s modified Eagle’s medium dNTP deoxyribonucleotide triphosphate HCC Hepatocellular Carcinoma hr hour IFN-γ Interferon-γ Kb kilo base pair KDa kilo Dalton MHC Major histocompatibility complex mins minutes ng nanogram O/N overnight ix Table of Contents ORFs Open reading frames PBS phosphate-buffered saline PCR polymerase chain reaction RT room temperature RT-PCR reverse transcription polymerase chain reaction Sec Second SiRNA Small interfering RNA SNP Single Nucleotide Polymorphism TNF-α Tumour Necrosis Factor- α TLSS Translation Start Site TSS Transcription Start site UBL Ubiquitin-like modifiers UTR Untranslated Region x Table of Contents xi LIST OF PUBLICATION S Zhang DW, Jeang KT, Lee CG. p53 negatively regulates the expression of FAT10, a gene upregulated in various cancers. Oncogene. 2006 Apr 13;25(16):2318-27. Lim CB, Zhang D, Lee CG. FAT10, a gene up-regulated in various cancers, is cellcycle regulated. Cell Div. 2006 Sep 8;1:20 Table of Contents xii SUMMARY FAT10 is a member of the ubiquitin-like modifier (UBL) family of proteins and has been implicated to play important roles such as antigen presentation, cytokine response, apoptosis and mitosis. Recently, our laboratory reported that the FAT10 gene is up regulated in 90% of hepatocellular carcinomas and over-expression of FAT10 gene may lead to chromosomal stability. As part of the studies to elucidate the mechanism behind FAT10 gene regulation, we identified and characterized the promoter of the FAT10 gene. We found that the 5’UTR, from the transcription start site to 15 bases before the start codon, displayed significant promoter activity. Regions upstream of the 5’UTR (from +26 to -1997) did not confer any promoter activity. As FAT10 expression was reported to be induced by cytokines, we also explored the role of the FAT10 promoter in cytokine responsiveness. We found that the distal promoter region, -1716 to -975, was highly responsive to interferon- γ and tumour necrosis factor-α with 4~5 times higher expression upon treatment with cytokines. FAT10 promoter activity and expression is significantly repressed in KB3-1 and HepG2 cells, which have wild-type p53, but not in p53-negative Hep3B cells. The role of p53 in regulating FAT10 expression was evident by the significant downregulation (PG), SNP5 (e1 –143 A>G) and SNP3 (5’UR –616 T>C), have high major allele frequency of greater than 10%, while the others including the two novel SNPs (SNP4 5’UR -169 C>T and SNP2 5’UR –876 G>A) were of low allele frequency (T) were observed in both the DNA of HCC samples and normal samples (Table 2). However, the other two low frequency SNPs, SNP1 (5’UR –914 Chapter IV Evaluation of the role of mutations/polymorphisms and differential methylation Figure 20. SNPs in FAT10 promoter. (A) The location of SNPs in FAT10 promoter. There are six SNPs identified in ours and previous studies in FAT10 promoter, three common SNPs which are SNP3 (5’UR -616 T>C), SNP5 (e1 -143 A>G) and SNP6 (e1 -121 A>G) and three low frequency SNPs which are SNP1 (5’UR -914 G>C), SNP2 (5’UR –876 G>A) and SNP4 (5’UR – 169 C>T). Both SNP2 and SNP4 are two new SNPs (not reported before). (B) SNPs haplotype frequency of FAT10 promoter in HCC patient samples (tumor tissue and adjacent non-tumor tissue) and normal samples from healthy population. Haplotype frequencies were derived from genotype data using Arlequin. Tumor tissue and adjacent nontumor tissue show exactly the same SNPs and haplotype profiles. Haplotype 5, 6, 7 specifically belong to HCC patient group, while haplotype 8 was only fond in normal population. No significant difference was observed in the haplotype distribution of HCC samples and normal samples (the Fisher’s exact Test, p>0.05) 78 Chapter IV Evaluation of the role of mutations/polymorphisms and differential methylation 79 Chapter IV Evaluation of the role of mutations/polymorphisms and differential methylation 80 Chapter IV Evaluation of the role of mutations/polymorphisms and differential methylation 81 G>C) and SNP2 (5’UR –876 G>A) in the FAT10 promoter were observed only in the HCC patient samples (Table 2). From the genotype data of these six SNPs, a total of seven haplotypes were inferred from HCC patients using an expectation maximisation (EM) algorithm in the ArlequinTM software program (Fig. 20B). As there was no difference in the sequence between tumor and adjacent non-tumorous tissue, no difference was observed in the haplotype distribution of tumor versus adjacent normal tissues from HCC patients. Although no significant difference in the haplotype distribution was observed between HCC patients and normal individuals (Fisher’s Exact Test, P>0.05) (Fig. 20B), there were three low frequency haplotypes that were only found in HCC patients (CGTCAG, GATCAG and GGTTGG) and one (GGCTAA) that is only observed in normal individuals (Fig. 20B). More samples may need to be examined before any conclusions can be drawn. An in silico strategy using the web-based program, MatInspector, was employed to evaluate whether the SNPs at the FAT10 promoter can potentially affect transcription factor binding. As shown in Table 2, the putative transcription factor binding sites were abolished in 2 SNPs identified in the 5’ untranslated region of FAT10 gene. The major G allele of SNP2 (5'UR-876G>A) was predicted to be bound by GATA-binding factor 2 and hepatic nuclear factor 1, whereas the alternative minor A allele was predicted not to bind any factor. Similarly, the major T allele of SNP3 (5'UR -616T>C) predicts a myocyte enhancer factor-binding site, whereas the alternative minor C allele does not. The creation of putative binding sites was predicted for two SNPs (SNPs 1 and 6) (Table 2). Finally, for the last 2 SNPs (SNPs 4 and 5), the putative binding sites predicted for the major allele not longer exist when Chapter IV Evaluation of the role of mutations/polymorphisms and differential methylation 0.5 ** ** 0.4 0.35 ** 0.3 0.25 ** 0.2 ** ** 0.15 ** 0.1 0.05 GGCTAA GGTTGG GATCAG CGTCAG GGTCGG GGTCAG GGCCAA SNP6 SNP5 SNP4 SNP3 SNP2 SNP1 GGTCAA 0 Haplotypes (OD.sec-1)βgal / RFUegfp Normalized βgal Activity 0.45 Figure 21. FAT10 Promoter activity with different haplotype structures. Promoter activity of FAT10 promoter with different haplotype structures were determined as β-gal activity (normalized against EGFP). Experiment was carried out in hep3B cells and performed in four independent occasions. The order of SNPs in a haplotype structure is from 5’ to 3’ (Figure20). The hapolotypes with high frequency were boxed. ** denotes significant difference (P0.05). 4.4.2 Differential methylation may account for the differences in FAT10 gene expression between HCC tumor and adjacent non-tumorous tissues CG dinucleotides are present in the regulatory regions of many genes (Bird, 1996). In normal cells, the cytosines in the CG dinucleotides generally remain unmethylated (Bird, 1996). However, in the promoter sequences of genes associated with certain cancers or inherited diseases, more cytosines at the promoter region were found to be methylated (Herman et al., 1996). The methylation status in the DNA of humans and other mammals play an important role in determining whether some genes are or are not expressed. Abnormal DNA methylation plays an important role in other developmental diseases as well. Abnormal increases or decreases in DNA Chapter IV Evaluation of the role of mutations/polymorphisms and differential methylation 92 methylation are often observed in human cancers and may contribute to their development (Baylin et al., 1998; Herman et al., 1996). Seven CG sites were observed to reside in FAT10 promoter region. In order to study the correlation between the methylation status of these CG dinucleotides and the aberrant expression of FAT10 in HCC patients (Lee et al., 2003), we performed methylation-specific sequencing to screen the methylation status of these CG dinucleotides at the FAT10 promoter in 11 HCC patients. We found that generally, higher FAT10 expression is correlated significantly with reduced methylation of the CGs at the FAT10 promoter (Kappa value=0.468) (Table 3). The exceptions are patients 6, 10 and 11 whereby although the HCC tumor tissues has higher FAT10 expression, the methylation status of the tumor is either more in the tumor (P6) or no different from the adjacent normal tissues (P10 and 11). It is possible that differential methylation of other unexamined sites correlates with the FAT10 expression. A previous report demonstrated that methylation status in the coding region may also play a role in the regulation of gene expression (Irvine et al., 2002). Hence, it would be worthwhile to determine the methylation status of the coding region as well as regions further upstream the 1.5 kb promoter that we examined. In summary, although no mutations were identified at the FAT10 promoter in the tumor of HCC patients, polymorphisms at this promoter was identified which facilitated differential FAT10 promoter activities. Importantly, the methylation status at this promoter was found to correlate significantly with FAT10 expression levels. Conclusion 93 Conclusion In order to elucidate the mechanism of the aberrant expression of FAT10 in liver tissues of HCC patients, we initially cloned and characterized FAT10 promoter. We found that the core promoter of FAT10 gene resides at the 5’ UTR and regions upstream of the 5’UTR did not confer any promoter activity. Interestingly, we found that FAT10 may be a downstream gene of p53 as it can be significantly repressed by p53. Results from Chromatin immunoprecipitation suggests that p53 represses FAT10 by directly binding to its consensus site which resides upstream of the transcriptional start site in FAT10 promoter. We also found that the region between -975 and -1997 bp upstream of the transcription start site may play a role in the response of FAT10 promoter to TNF-α and IFN-γ cytokines. To elucidate the mechanism behind the aberrant expression of FAT10 in the tumor tissues of HCC patients, we sequenced the ~1.3 kb of the FAT10 promoter to screen for mutations and determined the methylation status of this promoter in HCC patients. No mutations can be found in the tumor tissue of the FAT10 promoter that can account for the aberrant FAT10 expression. However, 6 polymorphisms were identified in this promoter. Haplotypes of these polymorphisms were found to mediate different FAT10 promoter activity. Significantly we found that the methylation status at the FAT10 promoter inversely correlated significantly with FAT10 expression. Reference 94 Reference Aguiar, J., Santurlidis, S., Nowok, J., Alexander, C., Rudnicki, D., Gispert, S., Schulz, W. and Auburger, G. (1999). Identification of the physiological promoter for spinocerebellar ataxia 2 gene reveals a CpG island for promoter activity situated into the exon 1 of this gene and provides data about the origin of the nonmethylated state of these types of islands. Biochem Biophys Res Commun 254, 315-8. Aklillu, E., Carrillo, J. A., Makonnen, E., Hellman, K., Pitarque, M., Bertilsson, L. and Ingelman-Sundberg, M. (2003). Genetic polymorphism of CYP1A2 in Ethiopians affecting induction and expression: characterization of novel haplotypes with single-nucleotide polymorphisms in intron 1. Mol Pharmacol 64, 659-69. Bates, E. E., Ravel, O., Dieu, M. C., Ho, S., Guret, C., Bridon, J. M., Ait-Yahia, S., Briere, F., Caux, C., Banchereau, J. et al. (1997). Identification and analysis of a novel member of the ubiquitin family expressed in dendritic cells and mature B cells. Eur J Immunol 27, 2471-7. Baylin, S. B., Herman, J. G., Graff, J. R., Vertino, P. M. and Issa, J. P. (1998). Alterations in DNA methylation: a fundamental aspect of neoplasia. Adv Cancer Res 72, 141-96. Berg, G. (1965). [Virus spread through the waterway]. Arch Hyg Bakteriol 149, 31035. Beutler, B. and Cerami, A. (1988). The history, properties, and biological effects of cachectin. Biochemistry 27, 7575-82. Beyne-Rauzy, O., Recher, C., Dastugue, N., Demur, C., Pottier, G., Laurent, G., Sabatier, L. and Mansat-De Mas, V. (2004). Tumor necrosis factor alpha induces senescence and chromosomal instability in human leukemic cells. Oncogene 23, 7507-16. Biggins, S., Ivanovska, I. and Rose, M. D. (1996). Yeast ubiquitin-like genes are involved in duplication of the microtubule organizing center. J Cell Biol 133, 133146. Bird, A. P. (1996). The relationship of DNA methylation to cancer. Cancer Surv 28, 87-101. Black, R. A., Rauch, C. T., Kozlosky, C. J., Peschon, J. J., Slack, J. L., Wolfson, M. F., Castner, B. J., Stocking, K. L., Reddy, P., Srinivasan, S. et al. (1997). A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 385, 729-33. Boddy, M. N., Howe, K., Etkin, L. D., Solomon, E. and Freemont, P. S. (1996). PIC 1, a novel ubiquitin-like protein which interacts with the PML component of a multiprotein complex that is disrupted in acute promyelocytic leukaemia. Oncogene 13, 971-82. Reference 95 Bond, G. L., Hu, W., Bond, E. E., Robins, H., Lutzker, S. G., Arva, N. C., Bargonetti, J., Bartel, F., Taubert, H., Wuerl, P. et al. (2004). A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 119, 591-602. Bressac, B., Galvin, K. M., Liang, T. J., Isselbacher, K. J., Wands, J. R. and Ozturk, M. (1990). Abnormal structure and expression of p53 gene in human hepatocellular carcinoma. Proc Natl Acad Sci U S A 87, 1973-7. Brummelkamp, T. R., Bernards, R. and Agami, R. (2002). A system for stable expression of short interfering RNAs in mammalian cells. Science 296, 550-3. Cahill, D. P., Lengauer, C., Yu, J., Riggins, G. J., Willson, J. K., Markowitz, S. D., Kinzler, K. W. and Vogelstein, B. (1998). Mutations of mitotic checkpoint genes in human cancers. Nature 392, 300-3. Canaan, A., Yu, X., Booth, C. J., Lian, J., Lazar, I., Gamfi, S. L., Castille, K., Kohya, N., Nakayama, Y., Liu, Y. C. et al. (2006). FAT10/diubiquitin-like proteindeficient mice exhibit minimal phenotypic differences. Mol Cell Biol 26, 5180-9. Chen, C. and Okayama, H. (1987). High-efficiency transformation of mammalian cells by plasmid DNA. Mol Cell Biol 7, 2745-52. Chun, A. C. and Jin, D. Y. (2003). Transcriptional regulation of mitotic checkpoint gene MAD1 by p53. J Biol Chem 278, 37439-50. Ciechanover, A., Elias, S., Heller, H., Ferber, S. and Hershko, A. (1980). Characterization of the heat-stable polypeptide of the ATP-dependent proteolytic system from reticulocytes. J Biol Chem 255, 7525-8. Clark, I. M., Rowan, A. D., Edwards, D. R., Bech-Hansen, T., Mann, D. A., Bahr, M. J. and Cawston, T. E. (1997). Transcriptional activity of the human tissue inhibitor of metalloproteinases 1 (TIMP-1) gene in fibroblasts involves elements in the promoter, exon 1 and intron 1. Biochem J 324 ( Pt 2), 611-7. Costello, J. F. and Plass, C. (2001). Methylation matters. J Med Genet 38, 285-303. Davey, C., Pennings, S. and Allan, J. (1997). CpG methylation remodels chromatin structure in vitro. J Mol Biol 267, 276-88. Deiss, L. P., Feinstein, E., Berissi, H., Cohen, O. and Kimchi, A. (1995). Identification of a novel serine/threonine kinase and a novel 15-kD protein as potential mediators of the gamma interferon-induced cell death. Genes Dev 9, 15-30. Derynck, R., Leung, D. W., Gray, P. W. and Goeddel, D. V. (1982). Human interferon gamma is encoded by a single class of mRNA. Nucleic Acids Res 10, 360515. Reference 96 Dokmanovic, M., Chang, B. D., Fang, J. and Roninson, I. B. (2002). Retinoidinduced growth arrest of breast carcinoma cells involves co-activation of multiple growth-inhibitory genes. Cancer Biol Ther 1, 24-7. Drew, P. D., Lonergan, M., Goldstein, M. E., Lampson, L. A., Ozato, K. and McFarlin, D. E. (1993). Regulation of MHC class I and beta 2-microglobulin gene expression in human neuronal cells. Factor binding to conserved cis-acting regulatory sequences correlates with expression of the genes. J Immunol 150, 3300-10. el-Deiry, W. S. (1998). Regulation of p53 downstream genes. Semin Cancer Biol 8, 345-57. Esteller, M., Fraga, M. F., Guo, M., Garcia-Foncillas, J., Hedenfalk, I., Godwin, A. K., Trojan, J., Vaurs-Barriere, C., Bignon, Y. J., Ramus, S. et al. (2001). DNA methylation patterns in hereditary human cancers mimic sporadic tumorigenesis. Hum Mol Genet 10, 3001-7. Excoffier, L. and Slatkin, M. (1995). Maximum-likelihood estimation of molecular haplotype frequencies in a diploid population. Mol Biol Evol 12, 921-7. Fan, W., Cai, W., Parimoo, S., Schwarz, D. C., Lennon, G. G. and Weissman, S. M. (1996). Identification of seven new human MHC class I region genes around the HLA-F locus. Immunogenetics 44, 97-103. Fang, G., Yu, H. and Kirschner, M. W. (1998). The checkpoint protein MAD2 and the mitotic regulator CDC20 form a ternary complex with the anaphase-promoting complex to control anaphase initiation. Genes Dev 12, 1871-83. Farmer, G., Colgan, J., Nakatani, Y., Manley, J. L. and Prives, C. (1996). Functional interaction between p53, the TATA-binding protein (TBP), andTBPassociated factors in vivo. Mol Cell Biol 16, 4295-304. Galan, J. M. and Haguenauer-Tsapis, R. (1997). Ubiquitin lys63 is involved in ubiquitination of a yeast plasma membrane protein. Embo J 16, 5847-54. Gorbsky, G. J., Chen, R. H. and Murray, A. W. (1998). Microinjection of antibody to Mad2 protein into mammalian cells in mitosis induces premature anaphase. J Cell Biol 141, 1193-205. Gruen, J. R., Nalabolu, S. R., Chu, T. W., Bowlus, C., Fan, W. F., Goei, V. L., Wei, H., Sivakamasundari, R., Liu, Y., Xu, H. X. et al. (1996). A transcription map of the major histocompatibility complex (MHC) class I region. Genomics 36, 70-85. Hardaker, E. L., Bacon, A. M., Carlson, K., Roshak, A. K., Foley, J. J., Schmidt, D. B., Buckley, P. T., Comegys, M., Panettieri, R. A., Jr., Sarau, H. M. et al. (2004). Regulation of TNF-alpha- and IFN-gamma-induced CXCL10 expression: participation of the airway smooth muscle in the pulmonary inflammatory response in chronic obstructive pulmonary disease. Faseb J 18, 191-3. Reference 97 Haupt, Y., Robles, A. I., Prives, C. and Rotter, V. (2002). Deconstruction of p53 functions and regulation. Oncogene 21, 8223-31. Hay, R. T. (2005). SUMO: a history of modification. Mol Cell 18, 1-12. Herman, J. G., Jen, J., Merlo, A. and Baylin, S. B. (1996). Hypermethylationassociated inactivation indicates a tumor suppressor role for p15INK4B. Cancer Res 56, 722-7. Hernando, E., Nahle, Z., Juan, G., Diaz-Rodriguez, E., Alaminos, M., Hemann, M., Michel, L., Mittal, V., Gerald, W., Benezra, R. et al. (2004). Rb inactivation promotes genomic instability by uncoupling cell cycle progression from mitotic control. Nature 430, 797-802. Hershko, A., Ciechanover, A., Heller, H., Haas, A. L. and Rose, I. A. (1980). Proposed role of ATP in protein breakdown: conjugation of protein with multiple chains of the polypeptide of ATP-dependent proteolysis. Proc Natl Acad Sci U S A 77, 1783-6. Hicke, L. (1997). Ubiquitin-dependent internalization and down-regulation of plasma membrane proteins. Faseb J 11, 1215-26. Hipp, M. S., Kalveram, B., Raasi, S., Groettrup, M. and Schmidtke, G. (2005). FAT10, a ubiquitin-independent signal for proteasomal degradation. Mol Cell Biol 25, 3483-91. Hipp, M. S., Raasi, S., Groettrup, M. and Schmidtke, G. (2004). NEDD8 ultimate buster-1L interacts with the ubiquitin-like protein FAT10 and accelerates its degradation. J Biol Chem 279, 16503-10. Ho, J. and Benchimol, S. (2003). Transcriptional repression mediated by the p53 tumour suppressor. Cell Death Differ 10, 404-8. Hochstrasser, M. (1996a). Protein degradation or regulation: Ub the judge. Cell 84, 813-5. Hochstrasser, M. (1996b). Ubiquitin-dependent protein degradation. Annu Rev Genet 30, 405-39. Hochstrasser, M. (2000). Evolution and function of ubiquitin-like proteinconjugation systems. Nat Cell Biol 2, E153-7. Hoffman, W. H., Biade, S., Zilfou, J. T., Chen, J. and Murphy, M. (2002). Transcriptional repression of the anti-apoptotic survivin gene by wild type p53. J Biol Chem 277, 3247-57. Reference 98 Hofmann, R. M. and Pickart, C. M. (1999). Noncanonical MMS2-encoded ubiquitin-conjugating enzyme functions in assembly of novel polyubiquitin chains for DNA repair. Cell 96, 645-53. Innocente, S. A., Abrahamson, J. L., Cogswell, J. P. and Lee, J. M. (1999). p53 regulates a G2 checkpoint through cyclin B1. Proc Natl Acad Sci U S A 96, 2147-52. Irvine, R. A., Lin, I. G. and Hsieh, C. L. (2002). DNA methylation has a local effect on transcription and histone acetylation. Mol Cell Biol 22, 6689-96. Iwai, K., Yamanaka, K., Kamura, T., Minato, N., Conaway, R. C., Conaway, J. W., Klausner, R. D. and Pause, A. (1999). Identification of the von Hippel-lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proc Natl Acad Sci U S A 96, 12436-41. Jackson P. K, Eldridge A. G, Freed E, Furstenthal L, Hsu J. Y, Kaiser B. K, Reimann J. D. 2000). The lore of the RINGs: substrate recognition and catalysis by ubiquitin ligases. Trends Cell Biol 10, 429-439. Jentsch, S. and Pyrowolakis, G. (2000). Ubiquitin and its kin: how close are the family ties? Trends Cell Biol 10, 335-42. Jesenberger, V. and Jentsch, S. (2002). Deadly encounter: ubiquitin meets apoptosis. Nat Rev Mol Cell Biol 3, 112-21. Joazeiro C. A, Weissman A. M. (2000). RING finger proteins: mediators of ubiquitin ligase activity. Cell. 102, 549-552. Johnson, D. R. and Pober, J. S. (1994). HLA class I heavy-chain gene promoter elements mediating synergy between tumor necrosis factor and interferons. Mol Cell Biol 14, 1322-32. Jones, P. A. and Baylin, S. B. (2002). The fundamental role of epigenetic events in cancer. Nat Rev Genet 3, 415-28. Jones, P. A. and Laird, P. W. (1999). Cancer epigenetics comes of age. Nat Genet 21, 163-7. Jones, P. L., Veenstra, G. J., Wade, P. A., Vermaak, D., Kass, S. U., Landsberger, N., Strouboulis, J. and Wolffe, A. P. (1998). Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat Genet 19, 187-91. Joosten, P. H., Toepoel, M., Mariman, E. C. and Van Zoelen, E. J. (2001). Promoter haplotype combinations of the platelet-derived growth factor alpha-receptor gene predispose to human neural tube defects. Nat Genet 27, 215-7. Reference 99 Kamura, T., Conrad, M. N., Yan, Q., Conaway, R. C. & Conaway, J. W. (1999). The Rbx1 subunit of SCF and VHL E3 ubiquitin ligase activates Rub1 modification of cullins Cdc53 and Cul2. Genes Dev. 13, 2928–2933. Kanakis, D., Kirches, E., Mawrin, C. and Dietzmann, K. (2003). Promoter mutations are no major cause of PTTG overexpression in pituitary adenomas. Clin Endocrinol (Oxf) 58, 151-5. Korant, B. D., Blomstrom, D. C., Jonak, G. J. and Knight, E., Jr. (1984). Interferon-induced proteins. Purification and characterization of a 15,000-dalton protein from human and bovine cells induced by interferon. J Biol Chem 259, 148359. Kurz T, Pintard L, Willis J. H, Hamill D. R, Gonczy P, Peter M, Bowerman B. (2002). Cytoskeletal regulation by the Nedd8 ubiquitin-like protein modification pathway. Science 295, 1294-1298 Laird, P. W. and Jaenisch, R. (1996). The role of DNA methylation in cancer genetic and epigenetics. Annu Rev Genet 30, 441-64. Lammer D, Mathias N, Laplaza J. M, Jiang W, Liu Y, Callis J, Goebl M, Estelle M. (1998). Modification of yeast Cdc53p by the ubiquitin-related protein rub1p affects function of the SCFCdc4 complex. Genes Dev. 12, 914-926. Lane, D. P. (1992). Cancer. p53, guardian of the genome. Nature 358, 15-6. Lapierre, L. A., Fiers, W. and Pober, J. S. (1988). Three distinct classes of regulatory cytokines control endothelial cell MHC antigen expression. Interactions with immune gamma interferon differentiate the effects of tumor necrosis factor and lymphotoxin from those of leukocyte alpha and fibroblast beta interferons. J Exp Med 167, 794-804. Lee, C. G., Ren, J., Cheong, I. S., Ban, K. H., Ooi, L. L., Yong Tan, S., Kan, A., Nuchprayoon, I., Jin, R., Lee, K. H. et al. (2003). Expression of the FAT10 gene is highly upregulated in hepatocellular carcinoma and other gastrointestinal and gynecological cancers. Oncogene 22, 2592-603. Lee, K. C., Crowe, A. J. and Barton, M. C. (1999). p53-mediated repression of alpha-fetoprotein gene expression by specific DNA binding. Mol Cell Biol 19, 127988. Lengauer, C., Kinzler, K. W. and Vogelstein, B. (1998). Genetic instabilities in human cancers. Nature 396, 643-9. Levine, A. J. (1997). p53, the cellular gatekeeper for growth and division. Cell 88, 323-31. Reference 100 Li, G. Q., Li, H. and Zhang, H. F. (2003). Mad2 and p53 expression profiles in colorectal cancer and its clinical significance. World J Gastroenterol 9, 1972-5. Li, Y. and Benezra, R. (1996). Identification of a human mitotic checkpoint gene: hsMAD2. Science 274, 246-8. Li, Y., Gorbea, C., Mahaffey, D., Rechsteiner, M. and Benezra, R. (1997). MAD2 associates with the cyclosome/anaphase-promoting complex and inhibits its activity. Proc Natl Acad Sci U S A 94, 12431-6. Liakopoulos D, Doenges G, Matuschewski K, Jentsch S. (1998). A novel protein modification pathway related to the ubiquitin system. EMBO J. 17, 2208-2214. Liu, Y., Li, J. Z., Yuan, X. H., Adler-Storthz, K. and Che, Z. (2002). An AP-1 binding site mutation in HPV-16 LCR enhances E6/E7 promoter activity in human oral epithelial cells. Virus Genes 24, 29-37. Liu, Y. C., Pan, J., Zhang, C., Fan, W., Collinge, M., Bender, J. R. and Weissman, S. M. (1999). A MHC-encoded ubiquitin-like protein (FAT10) binds noncovalently to the spindle assembly checkpoint protein MAD2. Proc Natl Acad Sci U S A 96, 4313-8. Loeb K. R, Haas A. L. (1994). Conjugates of ubiquitin cross-reactive protein distribute in a cytoskeletal pattern. Mol Cell Biol 14, 8408-8419. Luo, X., Tang, Z., Rizo, J. and Yu, H. (2002). The Mad2 spindle checkpoint protein undergoes similar major conformational changes upon binding to either Mad1 or Cdc20. Mol Cell 9, 59-71. Maciejewski, J. P., Selleri, C., Sato, T., Cho, H. J., Keefer, L. K., Nathan, C. F. and Young, N. S. (1995). Nitric oxide suppression of human hematopoiesis in vitro. Contribution to inhibitory action of interferon-gamma and tumor necrosis factoralpha. J Clin Invest 96, 1085-92. Maeda, Y., Seidel, S. D., Wei, G., Liu, X. and Sladek, F. M. (2002). Repression of hepatocyte nuclear factor 4alpha tumor suppressor p53: involvement of the ligandbinding domain and histone deacetylase activity. Mol Endocrinol 16, 402-10. Matunis, M. J., Coutavas, E. and Blobel, G. (1996). A novel ubiquitin-like modification modulates the partitioning of the Ran-GTPase-activating protein RanGAP1 between the cytosol and the nuclear pore complex. J Cell Biol 135, 145770. Michel, L. S., Liberal, V., Chatterjee, A., Kirchwegger, R., Pasche, B., Gerald, W., Dobles, M., Sorger, P. K., Murty, V. V. and Benezra, R. (2001). MAD2 haploinsufficiency causes premature anaphase and chromosome instability in mammalian cells. Nature 409, 355-9. Reference 101 Moll, U. M., Riou, G. and Levine, A. J. (1992). Two distinct mechanisms alter p53 in breast cancer: mutation and nuclear exclusion. Proc Natl Acad Sci U S A 89, 72626. Moss, M. L., Jin, S. L., Milla, M. E., Bickett, D. M., Burkhart, W., Carter, H. L., Chen, W. J., Clay, W. C., Didsbury, J. R., Hassler, D. et al. (1997). Cloning of a disintegrin metalloproteinase that processes precursor tumour-necrosis factor-alpha. Nature 385, 733-6. Muller, S., Matunis, M. J. and Dejean, A. (1998). Conjugation with the ubiquitinrelated modifier SUMO-1 regulates the partitioning of PML within the nucleus. Embo J 17, 61-70. Nan, X., Campoy, F. J. and Bird, A. (1997). MeCP2 is a transcriptional repressor with abundant binding sites in genomic chromatin. Cell 88, 471-81. Nowak, M. A., Komarova, N. L., Sengupta, A., Jallepalli, P. V., Shih Ie, M., Vogelstein, B. and Lengauer, C. (2002). The role of chromosomal instability in tumor initiation. Proc Natl Acad Sci U S A 99, 16226-31. Olek, A., Oswald, J. and Walter, J. (1996). A modified and improved method for bisulphite based cytosine methylation analysis. Nucleic Acids Res 24, 5064-6. Osaka F, Kawasaki H, Aida N, Saeki M, Chiba T, Kawashima S, Tanaka K, Kato S. (1998). A new NEDD8-ligating system for cullin-4A. Genes Dev. 12, 22632268. Pagano, M. (1997). Cell cycle regulation by the ubiquitin pathway. Faseb J 11, 106775. Patel, S. A., Graunke, D. M. and Pieper, R. O. (1997). Aberrant silencing of the CpG island-containing human O6-methylguanine DNA methyltransferase gene is associated with the loss of nucleosome-like positioning. Mol Cell Biol 17, 5813-22. Pickart, C. M. (2001). Mechanisms underlying ubiquitination. Annu Rev Biochem 70, 503-33. Pieper, R. O., Patel, S., Ting, S. A., Futscher, B. W. and Costello, J. F. (1996). Methylation of CpG island transcription factor binding sites is unnecessary for aberrant silencing of the human MGMT gene. J Biol Chem 271, 13916-24. Pines, J. (1994). Cell cycle. Ubiquitin with everything. Nature 371, 742-3. Pogribny, I. P., Pogribna, M., Christman, J. K. and James, S. J. (2000). Singlesite methylation within the p53 promoter region reduces gene expression in a reporter gene construct: possible in vivo relevance during tumorigenesis. Cancer Res 60, 58894. Reference 102 Raasi, S., Schmidtke, G., de Giuli, R. and Groettrup, M. (1999). A ubiquitin-like protein which is synergistically inducible by interferon-gamma and tumor necrosis factor-alpha. Eur J Immunol 29, 4030-6. Raasi, S., Schmidtke, G. and Groettrup, M. (2001). The ubiquitin-like protein FAT10 forms covalent conjugates and induces apoptosis. J Biol Chem 276, 35334-43. Ray, J. S., Harbison, M. L., McClain, R. M. and Goodman, J. I. (1994). Alterations in the methylation status and expression of the raf oncogene in phenobarbital-induced and spontaneous B6C3F1 mouse live tumors. Mol Carcinog 9, 155-66. Read MA, Brownell JE, Gladysheva TB, Hottelet M, Parent LA, Coggins MB, Pierce J. W, Podust V. N, Luo R. S, Chau V, Palombella V. J. (2000). Nedd8 modification of cul-1 activates SCF(beta(TrCP))-dependent ubiquitination of IkappaBalpha. Mol. Cell Biol. 20, 2326–2333. Ren, J., Kan, A., Leong, S. H., Ooi, L. L., Jeang, K. T., Chong, S. S., Kon, O. L. and Lee, C. G. (2006). FAT10 plays a role in the regulation of chromosomal stability. J Biol Chem 281, 11413-21. Ritchie, K. J., Hahn, C. S., Kim, K. I., Yan, M., Rosario, D., Li, L., de la Torre, J. C. and Zhang, D. E. (2004). Role of ISG15 protease UBP43 (USP18) in innate immunity to viral infection. Nat Med 10, 1374-8. Robzyk, K., Recht, J. and Osley, M. A. (2000). Rad6-dependent ubiquitination of histone H2B in yeast. Science 287, 501-4. Ross, M. J., Wosnitzer, M. S., Ross, M. D., Granelli, B., Gusella, G. L., Husain, M., Kaufman, L., Vasievich, M., D'Agati, V. D., Wilson, P. D. et al. (2006). Role of ubiquitin-like protein FAT10 in epithelial apoptosis in renal disease. J Am Soc Nephrol 17, 996-1004. Rossi, M., De Simone, M., Pollice, A., Santoro, R., La Mantia, G., Guerrini, L. and Calabro, V. (2006). Itch/AIP4 associates with and promotes p63 protein degradation. Cell Cycle 5, 1816-22. Rotin, D., Staub, O. and Haguenauer-Tsapis, R. (2000). Ubiquitination and endocytosis of plasma membrane proteins: role of Nedd4/Rsp5p family of ubiquitinprotein ligases. J Membr Biol 176, 1-17. Ruffner H, Joazeiro C. A, Hemmati D, Hunter T, Verma I. M. (2001). Cancerpredisposing mutations within the RING domain of BRCA1: loss of ubiquitin protein ligase activity and protection from radiation hypersensitivity. Proc Natl Acad Sci U S A. 98, 5134-5139. Scheurich, P., Kronke, M., Schluter, C., Ucer, U. and Pfizenmaier, K. (1986). Noncytocidal mechanisms of action of tumor necrosis factor-alpha on human tumor Reference 103 cells: enhancement of HLA gene expression synergistic with interferon-gamma. Immunobiology 172, 291-300. Sampson, D. A., Wang, M. and Matunis, M. J. (2001). The small ubiquitin-like modifier-1 (SUMO-1) consensus sequence mediates Ubc9 binding and is essential for SUMO-1 modification. J Biol Chem 276, 21664-9. Schmidtke, G., Kalveram, B., Weber, E., Bochtler, P., Lukasiak, S., Hipp, M. S. and Groettrup, M. (2006). The UBA domains of NUB1L are required for binding but not for accelerated degradation of the ubiquitin-like modifier FAT10. J Biol Chem 281, 20045-54. Shah, J. V. and Cleveland, D. W. (2000). Waiting for anaphase: Mad2 and the spindle assembly checkpoint. Cell 103, 997-1000. Shin, K. H., Shin, J. H., Kim, J. H. and Park, J. G. (2002). Mutational analysis of promoters of mismatch repair genes hMSH2 and hMLH1 in hereditary nonpolyposis colorectal cancer and early onset colorectal cancer patients: identification of three novel germ-line mutations in promoter of the hMSH2 gene. Cancer Res 62, 38-42. Sibley, K., Rollinson, S., Allan, J. M., Smith, A. G., Law, G. R., Roddam, P. L., Skibola, C. F., Smith, M. T. and Morgan, G. J. (2003). Functional FAS promoter polymorphisms are associated with increased risk of acute myeloid leukemia. Cancer Res 63, 4327-30. Spence, J., Gali, R. R., Dittmar, G., Sherman, F., Karin, M. and Finley, D. (2000). Cell cycle-regulated modification of the ribosome by a variant multiubiquitin chain. Cell 102, 67-76. Stahler, F. and Roemer, K. (1998). Mutant p53 can provoke apoptosis in p53deficient Hep3B cells with delayed kinetics relative to wild-type p53. Oncogene 17, 3507-12. Tateishi, K., Omata, M., Tanaka, K. and Chiba, T. (2001). The NEDD8 system is essential for cell cycle progression and morphogenetic pathway in mice. J Cell Biol 155, 571-9. Taylor, W. R., Schonthal, A. H., Galante, J. and Stark, G. R. (2001). p130/E2F4 binds to and represses the cdc2 promoter in response to p53. J Biol Chem 276, 19982006. Taylor, W. R. and Stark, G. R. (2001). Regulation of the G2/M transition by p53. Oncogene 20, 1803-15. van den Elsen, P. J., Gobin, S. J., van Eggermond, M. C. and Peijnenburg, A. (1998). Regulation of MHC class I and II gene transcription: differences and similarities. Immunogenetics 48, 208-21. Reference 104 Vilcek, J. and Lee, T. H. (1991). Tumor necrosis factor. New insights into the molecular mechanisms of its multiple actions. J Biol Chem 266, 7313-6. Vogelstein, B., Lane, D. and Levine, A. J. (2000). Surfing the p53 network. Nature 408, 307-10. Vorce, R. L. and Goodman, J. I. (1989). Hypomethylation of ras oncogenes in chemically induced and spontaneous B6C3F1 mouse liver tumors. Mol Toxicol 2, 99116. Vousden, K. H. and Lu, X. (2002). Live or let die: the cell's response to p53. Nat Rev Cancer 2, 594-604. Wang, X., Jin, D. Y., Ng, R. W., Feng, H., Wong, Y. C., Cheung, A. L. and Tsao, S. W. (2002). Significance of MAD2 expression to mitotic checkpoint control in ovarian cancer cells. Cancer Res 62, 1662-8. Wang, X., Jin, D. Y., Wong, Y. C., Cheung, A. L., Chun, A. C., Lo, A. K., Liu, Y. and Tsao, S. W. (2000). Correlation of defective mitotic checkpoint with aberrantly reduced expression of MAD2 protein in nasopharyngeal carcinoma cells. Carcinogenesis 21, 2293-7. Watson, I. R. and Irwin, M. S. (2006). Ubiquitin and ubiquitin-like modifications of the p53 family. Neoplasia 8, 655-66. Weissman A. M. (2001). Themes and variations on ubiquitylation. Nat Rev Mol Cell Biol. 2, 169-78. Xu, Y., Fang, F., Ludewig, G., Jones, G. and Jones, D. (2004). A mutation found in the promoter region of the human survivin gene is correlated to overexpression of survivin in cancer cells. DNA Cell Biol 23, 419-29. Yeh, E. T., Gong, L. and Kamitani, T. (2000). Ubiquitin-like proteins: new wines in new bottles. Gene 248, 1-14. Yoshimura, A. (2006). Signal transduction of inflammatory cytokines and tumor development. Cancer Sci 97, 439-47. Yu, H. (2002). Regulation of APC-Cdc20 by the spindle checkpoint. Curr Opin Cell Biol 14, 706-14. Zhao, R., Gish, K., Murphy, M., Yin, Y., Notterman, D., Hoffman, W. H., Tom, E., Mack, D. H. and Levine, A. J. (2000). Analysis of p53-regulated gene expression patterns using oligonucleotide arrays. Genes Dev 14, 981-93. [...]... Isolation and Characterization of the Promoter of the FAT10 Gene CHAPTER II ISOLATION AND CHARACTERIZATION OF THE PROMOTER OF FAT10 GENE Adapted from Zhang et al (2006 Apr 13;25(16):2318-27, Oncogene) 18 Chapter II 2.1 Isolation and Characterization of the Promoter of the FAT10 Gene 19 Background FAT10, a new member of UBL family, contains two ubiquitin-like domains joined by a short linker and is 29%... methylation at the FAT10 promoter can account for the aberrant over-expression of FAT10 in the tumors of HCC patients Our lab has previously reported that the expression of the FAT10 gene is upregulated in greater than 90% of HCC patients (Lee et al., 2003) To determine whether there are mutations within the FAT10 promoter that can account for the aberrant over-expression of the FAT10 gene in the tumors of HCC... account for the aberrant over-expression of FAT10 in the tumors of HCC patients The following are the specific aims that help address the primary objective of this study Specific Aim 1: Isolate and Characterize the Promoter of the FAT10 Gene FAT10 gene comprises only two exons with an intervening 3.6kb intron and an upstream putative promoter region (Fig 1) The 5’UTR resides in exon 1 To isolate and characterize... cytokine response, apoptosis and mitosis Recently, our laboratory reported that the FAT10 gene is up regulated in 90% of hepatocellular carcinomas and over-expression of FAT10 gene may lead to chromosomal stability As part of the studies to elucidate the mechanism behind FAT10 gene regulation, we identified and characterized the promoter of the FAT10 gene We found that the 5’UTR, from the transcription start... tissues of these HCC patients Two of these polymorphisms were novel We computationally inferred the haplotype of these six polymorphisms and recapitulated these SNP haplotypes in vitro in the promoter- reporter (β-gal) system to determine the effects of the different SNP haplotypes on FAT10 promoter activity We proceeded to explore if the distribution of these SNPs in HCC was different from that of non-HCC... wt p53 was introduced To demonstrate the reversal of the effect of p53 on FAT10 promoter activity, siRNA against p53 or siRNA against a random sequence with no known homology to human, mouse or rat was also introduced and FAT10 promoter activity was determined We also attempted to delineate the region of the FAT10 promoter that confers responsiveness of the FAT10 promoter to p53 In silico analyses were... in the 5’ flanking region (-616(T/C)) and two at the 5’UTR (+82(A/G) and +104(A/G)), occurred at high frequency in both the normal and HCC patients With the current data, we did not find obvious correlation between the polymorphisms at the FAT10 promoter region and the relative FAT10 expression levels in HCC patients Nonetheless, we recapitulated various combinations of these three polymorphisms and. .. enhanced FAT10 expression and promoter activity P53 was found to bind in vivo to the 5’ half-consensus sequence of Table of Contents xiii the p53 binding site located in the FAT10 promoter Hence, we propose that FAT10 is a downstream target of p53 We proceeded to investigate if the up-regulation of FAT10 expression in the tumors of HCC patients can be accounted for by mutations or aberrant methylation at the. .. methylation at the FAT10 promoter region Through sequencing of approximately 37 HCC individuals and 39 normal individuals, we did not find any mutations at the FAT10 promoter region in the tumor of the patients that could account for the differential expression of the tumor and adjacent normal liver tissues in HCC patients Nonetheless, we identified six polymorphisms, two of which were novel Three of these six... role of FAT10 in immune response To elucidate the mechanism of FAT10 gene regulation, we isolated and characterized the promoter of FAT10 Interestingly, we found significant promoter activity in the 5’ untranslated region (UTR) (+1 bp to +209 bp) of the FAT10 gene but no promoter activity in regions upstream of the 5’UTR alone (from +26 bp to -1997 bp) Region -975 to +209 conferred maximum promoter ... II Isolation and Characterization of the Promoter of the FAT10 Gene 32 2.3.2 The responsive domain of the FAT10 promoter to TNF-α and IFN-γ resides in the region upstream of FAT10 promoter To.. .CLONING AND CHARACTERIZATION OF THE PROMOTER OF THE CANCER -ASSOCIATED GENE, FAT10 ZHANG DONGWEI (M Sc.) Wuhan Institute of Virology Chinese Academy of Science A THESIS SUBMITTED FOR THE DEGREE... Chapter II 2.3 Isolation and Characterization of the Promoter of the FAT10 Gene 28 Results 2.3.1 FAT10 promoter resides at the 5’UTR To better understand the regulation of FAT10 gene expression,

Ngày đăng: 03/10/2015, 20:57

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan