全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Genetic and Epigenetic Events Generate Multiple Pathways in Colorectal Cancer Progression

DOI: 10.1155/2012/509348

Full-Text   Cite this paper   Add to My Lib

Abstract:

Colorectal cancer (CRC) is one of the most common causes of death, despite decades of research. Initially considered as a disease due to genetic mutations, it is now viewed as a complex malignancy because of the involvement of epigenetic abnormalities. A functional equivalence between genetic and epigenetic mechanisms has been suggested in CRC initiation and progression. A hallmark of CRC is its pathogenetic heterogeneity attained through at least three distinct pathways: a traditional (adenoma-carcinoma sequence), an alternative, and more recently the so-called serrated pathway. While the alternative pathway is more heterogeneous and less characterized, the traditional and serrated pathways appear to be more homogeneous and clearly distinct. One unsolved question in colon cancer biology concerns the cells of origin and from which crypt compartment the different pathways originate. Based on molecular and pathological evidences, we propose that the traditional and serrated pathways originate from different crypt compartments explaining their genetic/epigenetic and clinicopathological differences. In this paper, we will discuss the current knowledge of CRC pathogenesis and, specifically, summarize the role of genetic/epigenetic changes in the origin and progression of the multiple CRC pathways. Elucidation of the link between the molecular and clinico-pathological aspects of CRC would improve our understanding of its etiology and impact both prevention and treatment. 1. Background: The Molecular Basis of Colon Carcinogenesis Colorectal cancers (CRCs) arise through a multistep process in which genetic and epigenetic alterations accumulate in a sequential order. Three different pathogenetic pathways have been implicated in the development of these tumors: (1) chromosomal instability (CIN); (2) microsatellite instability (MSI); (3) CpG island methylator phenotype (CIMP). The CIN pathway is associated with the sequential deregulation of tumor suppressor genes (TSGs) and oncogenes such as, APC, KRAS, DCC/SMAD4, and TP53. It generally occurs within inherited tumors, such as familial adenomatous polyposis (FAP), but it has also been associated with the majority of sporadic CRCs. Microsatellite instability is responsible for the Lynch syndrome and sporadic tumors and is mainly caused by inactivation of the DNA mismatch repair genes (hMLH1, hMSH2, hMSH6, and hPMS2). CRCs displaying MSI tend to be rightsided, generally have high histological grades, a mucinous phenotype, and diagnosed at lower pathological stages than CIN cancers. The CpG island methylator

References

[1]  D. Cunningham, W. Atkin, H. J. Lenz et al., “Colorectal cancer,” The Lancet, vol. 375, no. 9719, pp. 1030–1047, 2010.
[2]  A. Tenesa and M. G. Dunlop, “New insights into the aetiology of colorectal cancer from genome-wide association studies,” Nature Reviews Genetics, vol. 10, no. 6, pp. 353–358, 2009.
[3]  A. Jemal, M. M. Center, E. Ward, and M. J. Thun, “Cancer occurrence,” Methods in Molecular Biology, vol. 471, pp. 3–29, 2009.
[4]  A. Jemal, R. Siegel, E. Ward et al., “Cancer statistics, 2008,” CA Cancer Journal for Clinicians, vol. 58, no. 2, pp. 71–96, 2008.
[5]  J. J. Y. Sung, J. Y. W. Lau, K. L. Goh et al., “Increasing incidence of colorectal cancer in Asia: implications for screening,” The Lancet Oncology, vol. 6, no. 11, pp. 871–876, 2005.
[6]  A. K. Rustgi, “The genetics of hereditary colon cancer,” Genes and Development, vol. 21, no. 20, pp. 2525–2538, 2007.
[7]  E. R. Fearon and B. Vogelstein, “A genetic model for colorectal tumorigenesis,” Cell, vol. 61, no. 5, pp. 759–767, 1990.
[8]  G. Smith, F. A. Carey, J. Beattie et al., “Mutations in APC, Kirsten-ras, and p53—alternative genetic pathways to colorectal cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 14, pp. 9433–9438, 2002.
[9]  S. Ogino and A. Goel, “Molecular classification and correlates in colorectal cancer,” Journal of Molecular Diagnostics, vol. 10, no. 1, pp. 13–27, 2008.
[10]  J. R. Jass, “Classification of colorectal cancer based on correlation of clinical, morphological and molecular features,” Histopathology, vol. 50, no. 1, pp. 113–130, 2007.
[11]  S. D. Markowitz and M. M. Bertagnolli, “Molecular basis of colorectal cancer,” The New England Journal of Medicine, vol. 361, no. 25, pp. 2404–2460, 2009.
[12]  J. Young, M. Jenkins, S. Parry et al., “Serrated pathway colorectal cancer in the population: genetic consideration,” Gut, vol. 56, no. 10, pp. 1453–1459, 2007.
[13]  S. Sharma, T. K. Kelly, and P. A. Jones, “Epigenetics in cancer,” Carcinogenesis, vol. 31, no. 1, pp. 27–36, 2009.
[14]  M. Esteller, “Molecular origins of cancer: epigenetics in cancer,” The New England Journal of Medicine, vol. 358, no. 11, pp. 1148–1096, 2008.
[15]  R. Bonasio, S. Tu, and D. Reinberg, “Molecular signals of epigenetic states,” Science, vol. 330, no. 6004, pp. 612–616, 2010.
[16]  D. Hanahan and R. A. Weinberg, “Hallmarks of cancer: the next generation,” Cell, vol. 144, no. 5, pp. 646–674, 2011.
[17]  R. A. Beckman and L. A. Loeb, “Genetic instability in cancer: theory and experiment,” Seminars in Cancer Biology, vol. 15, no. 6, pp. 423–435, 2005.
[18]  M. S. Pino and D. C. Chung, “The chromosomal instability pathway in colon cancer,” Gastroenterology, vol. 138, no. 6, pp. 2059–2072, 2010.
[19]  C. R. Boland and A. Goel, “Microsatellite instability in colorectal cancer,” Gastroenterology, vol. 138, no. 6, pp. 2073–2087, 2010.
[20]  L. A. Loeb, C. F. Springgate, and N. Battula, “Errors in DNA replication as a basis of malignant changes,” Cancer Research, vol. 34, no. 9, pp. 2311–2321, 1974.
[21]  C. A. Klein, “Random mutations, selected mutations: a PIN opens the door to new genetic landscapes,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 48, pp. 18033–18034, 2006.
[22]  M. Toyota, N. Ahuja, M. Ohe-Toyota, J. G. Herman, S. B. Baylin, and J. P. J. Issa, “CpG island methylator phenotype in colorectal cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 15, pp. 8681–8686, 1999.
[23]  C. R. Boland, N. L. Komarova, and A. Goel, “Chromosomal instability and cancer: not just one CINgle mechanism,” Gut, vol. 58, no. 2, pp. 163–164, 2009.
[24]  W. S. Samowitz, H. Albertsen, J. Herrick et al., “Evaluation of a large, population-based sample supports a CpG island methylator phenotype in colon cancer,” Gastroenterology, vol. 129, no. 3, pp. 837–845, 2005.
[25]  J. P. J. Issa, L. Shen, and M. Toyota, “CIMP, at last,” Gastroenterology, vol. 129, no. 3, pp. 1121–1124, 2005.
[26]  K. Curtin, M. L. Slattery, and W. S. Samowitz, “CpG island methylation in colorectal cancer: past, present and future,” Pathology Research International, vol. 2011, Article ID 902674, 8 pages, 2011.
[27]  Y. Xu, B. Hu, A. J. Choi, et al., “Unique DNA methylome profiles in CpG island methylator phenotype colon cancers,” Genome Research, vol. 22, no. 2, pp. 283–291, 2012.
[28]  T. Hinoue, D. J. Weisenberger, C. P. Lange, et al., “Genome-scale analysis of aberrant DNA methylation in colorectal cancer,” Genome Research, vol. 22, no. 2, pp. 271–282, 2012.
[29]  B. P. Berman, D. J. Weisenberger, J. F. Aman, et al., “Regions of focal DNA hypermethylation and long-range hypomethylation in colorectal cancer coincide with nuclear lamina-associated domains,” Nature Genetics, vol. 44, no. 1, pp. 40–46, 2012.
[30]  J. P. Issa, “Colon cancer: it’s CIN or CIMP,” Clinical Cancer Research, vol. 14, no. 19, pp. 5939–5940, 2008.
[31]  I. M. Shih, T. L. Wang, G. Traverso et al., “Top-down morphogenesis of colorectal tumors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 5, pp. 2640–2645, 2001.
[32]  A. P. Feinberg, R. Ohlsson, and S. Henikoff, “The epigenetic progenitor origin of human cancer,” Nature Reviews Genetics, vol. 7, no. 1, pp. 21–33, 2006.
[33]  R. A. Irizarry, C. Ladd-Acosta, B. Wen et al., “The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores,” Nature Genetics, vol. 41, no. 2, pp. 178–186, 2009.
[34]  J. E. Ohm, K. M. McGarvey, X. Yu et al., “A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing,” Nature Genetics, vol. 39, no. 2, pp. 237–242, 2007.
[35]  H. P. Mohammad, Y. Cai, K. M. McGarvey et al., “Polycomb CBX7 promotes initiation of heritable repression of genes frequently silenced with cancer-specific DNA hypermethylation,” Cancer Research, vol. 69, no. 15, pp. 6322–6330, 2009.
[36]  D. C. Snover, “Update on the serrated pathway to colorectal carcinoma,” Human Pathology, vol. 42, no. 1, pp. 1–10, 2011.
[37]  B. Leggett and V. Whitehall, “Role of the serrated pathway in colorectal cancer pathogenesis,” Gastroenterology, vol. 138, no. 6, pp. 2088–2100, 2010.
[38]  S. M. Dieter, C. R. Ball, C. M. Hoffmann, et al., “Distinct types of tumor-initiating cells form human colon cancer tumors and metastases,” Cell Stem Cell, vol. 9, no. 4, pp. 375–365, 2011.
[39]  J. Terzic, S. Grivennicov, E. Karin, and M. Karin, “Inflammation and colon cancer,” Gastroenterology, vol. 138, no. 6, pp. 2101–2114, 2010.
[40]  A. P. Feinberg and B. Vogelstein, “Hypomethylation distinguishes genes of some human cancers from their normal counterparts,” Nature, vol. 301, no. 5895, pp. 89–92, 1983.
[41]  R. J. Taft, K. C. Pang, T. R. Mercer, M. Dinger, and J. S. Mattick, “Non-coding RNAs: regulators of disease,” Journal of Pathology, vol. 220, no. 2, pp. 126–139, 2010.
[42]  A. Portela and M. Esteller, “Epigenetic modifications and human disease,” Nature Biotechnology, vol. 28, no. 10, pp. 1057–1068, 2010.
[43]  M. Pancione, L. Sabatino, A. Fucci et al., “Epigenetic silencing of peroxisome proliferator- activated receptor γ is a biomarker for colorectal cancer progression and adverse patients' outcome,” PLoS ONE, vol. 5, no. 12, article e14229, 2010.
[44]  R. K. Chodavarapu, S. Feng, Y. V. Bernatavichute et al., “Relationship between nucleosome positioning and DNA methylation,” Nature, vol. 466, no. 7304, pp. 388–392, 2010.
[45]  K. N. Harikrishnan, M. Z. Chow, E. K. Baker et al., “Brahma links the SWI/SNF chromatin-remodeling complex with MeCP2-dependent transcriptional silencing,” Nature Genetics, vol. 37, no. 3, pp. 254–264, 2005.
[46]  D. Reisman, S. Glaros, and E. A. Thompson, “The SWI/SNF complex and cancer,” Oncogene, vol. 28, no. 14, pp. 1653–1668, 2009.
[47]  L. Ho, J. L. Ronan, J. Wu et al., “An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self-renewal and pluripotency,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 13, pp. 5181–5186, 2009.
[48]  P. P. Medina and M. Sanchez Cespedes, “Involvement of the chromatin-remodeling factor BRG1/SMARCA4 in human cancer,” Epigenetics, vol. 3, no. 2, pp. 64–68, 2008.
[49]  D. N. Reisman, M. W. Strobeck, B. L. Betz et al., “Concomitant down-regulation of BRM and BRG1 in human tumor cell lines: differential effects on RB-mediated growth arrest vs CD44 expression,” Oncogene, vol. 21, no. 8, pp. 1196–1207, 2002.
[50]  A. K. C. Wong, F. Shanahan, Y. Chen et al., “BRG1, a component of the SWI-SNF complex, is mutated in multiple human tumor cell lines,” Cancer Research, vol. 60, no. 21, pp. 6171–6177, 2000.
[51]  S. Bultman, T. Gebuhr, D. Yee et al., “A Brg1 null mutation in the mouse reveals functional differences among mammalian SWI/SNF complexes,” Molecular Cell, vol. 6, no. 6, pp. 1287–1295, 2000.
[52]  T. Watanabe, S. Semba, and H. Yokozaki, “Regulation of PTEN expression by the SWI/SNF chromatin-remodelling protein BRG1 in human colorectal carcinoma cells,” British Journal of Cancer, vol. 104, no. 1, pp. 146–154, 2011.
[53]  E. Sánchez-Tilló, A. Lázaro, R. Torrent et al., “ZEB1 represses E-cadherin and induces an EMT by recruiting the SWI/SNF chromatin-remodeling protein BRG1,” Oncogene, vol. 29, no. 24, pp. 3490–3500, 2010.
[54]  N. Yamamichi, K. I. Inada, M. Ichinose et al., “Frequent loss of Brm expression in gastric cancer correlates with histologic features and differentiation state,” Cancer Research, vol. 67, no. 22, pp. 10727–10735, 2007.
[55]  C. G. Sansam and C. W. M. Roberts, “Epigenetics and cancer: altered chromatin remodeling via Snf5 loss leads to aberrant cell cycle regulation,” Cell Cycle, vol. 5, no. 6, pp. 621–624, 2006.
[56]  N. Sévenet, A. Lellouch-Tubiana, D. Schofield et al., “Spectrum of hSNF5/INI1 somatic mutations in human cancer and genotype-phenotype correlations,” Human Molecular Genetics, vol. 8, no. 13, pp. 2359–2368, 1999.
[57]  J. A. Biegel, J. Y. Zhou, L. B. Rorke, C. Stenstrom, L. M. Wainwright, and B. Fogelgren, “Germ-line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors,” Cancer Research, vol. 59, no. 1, pp. 74–79, 1999.
[58]  C. W. M. Roberts and J. A. Biegel, “The role of SMARCB1/INI1 in development of rhabdoid tumor,” Cancer Biology and Therapy, vol. 8, no. 5, pp. 412–416, 2009.
[59]  A. Remo, C. Zanella, E. Molinari, et al., “Rhabdoid carcinoma of the colon, a distinct entity with a very aggressive behaviour. A case report, associated with a polyposis coli and review of the Literature,” International Journal of Surgical Pathology, vol. 2, no. 20, pp. 185–190, 2012.
[60]  M. Pancione, A. Di Blasi, L. Sabatino et al., “A novel case of rhabdoid colon carcinoma associated with a positive CpG island methylator phenotype and BRAF mutation,” Human Pathology, vol. 42, no. 7, pp. 1047–1052, 2011.
[61]  H. R. Moinova, W. D. Chen, L. Shen et al., “HLTF gene silencing in human colon cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 7, pp. 4562–4567, 2002.
[62]  S. Popat, R. Hubner, and R. S. Houlston, “Systematic review of microsatellite instability and colorectal cancer prognosis,” Journal of Clinical Oncology, vol. 23, no. 3, pp. 609–618, 2005.
[63]  A. Lugli, I. Zlobec, P. Minoo et al., “Prognostic significance of the wnt signalling pathway molecules APC, β-catenin and E-cadherin in colorectal cancer—a tissue microarray-based analysis,” Histopathology, vol. 50, no. 4, pp. 453–464, 2007.
[64]  R. Kalluri and R. A. Weinberg, “The basics of epithelial-mesenchymal transition,” Journal of Clinical Investigation, vol. 119, no. 6, pp. 1420–1428, 2009.
[65]  J. G. Clohessy and P. P. Pandolfi, “β-tting on p63 as a Metastatic Suppressor,” Cell, vol. 137, no. 1, pp. 28–30, 2009.
[66]  G. Moreno-Bueno, H. Peinado, P. Molina et al., “The morphological and molecular features of the epithelial-to-mesenchymal transition,” Nature Protocols, vol. 4, no. 11, pp. 1591–1613, 2009.
[67]  M. Guarino, B. Rubino, and G. Ballabio, “The role of epithelial-mesenchymal transition in cancer pathology,” Pathology, vol. 39, no. 3, pp. 305–318, 2007.
[68]  M. J. Larriba, J. Casado-Vela, N. Pendás-Franco et al., “Novel snail1 target proteins in human colon cancer identified by proteomic analysis,” PLoS ONE, vol. 5, no. 4, article e10221, 2010.
[69]  G. Moreno-Bueno, E. Cubillo, D. Sarrió et al., “Genetic profiling of epithelial cells expressing E-cadherin repressors reveals a distinct role for snail, Slug, and E47 Factors in epithelial- mesenchymal transition,” Cancer Research, vol. 66, no. 19, pp. 9543–9556, 2006.
[70]  M. Amano, M. Nakayama, and K. Kaibuchi, “Rho-kinase/ROCK: a key regulator of the cytoskeleton and cell polarity,” Cytoskeleton, vol. 67, no. 9, pp. 545–554, 2010.
[71]  P. Ortega, A. Morán, C. De Juan et al., “Differential Wnt pathway gene expression and E-cadherin truncation in sporadic colorectal cancers with and without microsatellite instability,” Clinical Cancer Research, vol. 14, no. 4, pp. 995–1001, 2008.
[72]  H. Suzuki, N. Masuda, T. Shimura et al., “Nuclear β-catenin expression at the invasive front and in the vessels predicts liver metastasis in colorectal carcinoma,” Anticancer Research, vol. 28, no. 3, pp. 1821–1830, 2008.
[73]  D. Horst, S. Reu, L. Kriegl, J. Engel, T. Kirchner, and A. Jung, “The intratumoral distribution of nuclear β-catenin is a prognostic marker in colon cancer,” Cancer, vol. 115, no. 10, pp. 2063–2070, 2009.
[74]  M. Pancione, N. Forte, A. Fucci et al., “Prognostic role of β-catenin and p53 expression in the metastatic progression of sporadic colorectal cancer,” Human Pathology, vol. 41, no. 6, pp. 867–876, 2010.
[75]  J. García-Solano, P. Conesa-Zamora, J. Trujillo-Santos, et al., “Immunohistochemical expression profile of β-catenin, E-cadherin, P-cadherin, laminin-5γ2 chain, and SMAD4 in colorectal serrated adenocarcinoma,” Human Pathology, vol. 43, no. 7, pp. 1094–1102, 2012.
[76]  M. S. Pino, H. Kikuchi, M. Zeng et al., “Epithelial to mesenchymal transition is impaired in colon cancer cells with microsatellite instability,” Gastroenterology, vol. 138, no. 4, pp. 1406–1417, 2010.
[77]  G. H. Kang, “Four molecular subtypes of colorectal cancer and their precursor lesions,” Archives of Pathology and Laboratory Medicine, vol. 135, no. 6, pp. 698–703, 2011.
[78]  J. M. Bae, M. J. Kim, J. H. Kim et al., “Differential clinicopathological features in microsatellite instability-positive colorectal cancers depending on CIMP status,” Virchows Archiv, vol. 459, no. 1, pp. 55–63, 2011.
[79]  R. N. Jorissen, P. Gibbs, M. Christie et al., “Metastasis-associated gene expression changes predict poor outcomes in patients with Dukes stage B and C colorectal cancer,” Clinical Cancer Research, vol. 15, no. 24, pp. 7642–7651, 2009.
[80]  J. J. Smith, N. G. Deane, F. Wu et al., “Experimentally derived metastasis gene expression profile predicts recurrence and death in patients with colon cancer,” Gastroenterology, vol. 138, no. 3, pp. 958–968, 2010.
[81]  A. Loboda, M. V. Nebozhyn, J. W. Watters et al., “EMT is the dominant program in human colon cancer,” BMC Medical Genomics, vol. 4, article 9, 2011.
[82]  F. Prall, “Tumour budding in colorectal carcinoma,” Histopathology, vol. 50, no. 1, pp. 151–162, 2007.
[83]  E. Karamitopoulou, A. Lugli, I. Panayiotides et al., “Systematic assessment of protein phenotypes characterizing high-grade tumour budding in mismatch repair-proficient colorectal cancer,” Histopathology, vol. 57, no. 2, pp. 233–243, 2010.
[84]  I. Zlobec, F. Molinari, V. Martin et al., “Tumor budding predicts response to anti-EGFR therapies in metastatic colorectal cancer patients,” World Journal of Gastroenterology, vol. 16, no. 38, pp. 4823–4831, 2010.
[85]  I. Zlobec and A. Lugli, “Epithelial mesenchymal transition and tumor budding in aggressive colorectal cancer: tumor budding as oncotarget,” Oncotarget, vol. 1, no. 7, pp. 651–661, 2010.
[86]  W. S. Samowitz, C. Sweeney, J. Herrick et al., “Poor survival associated with the BRAF V600E mutation in microsatellite-stable colon cancers,” Cancer Research, vol. 65, no. 14, pp. 6063–6070, 2005.
[87]  M. S. Kim, J. Lee, and D. Sidransky, “DNA methylation markers in colorectal cancer,” Cancer and Metastasis Reviews, vol. 29, no. 1, pp. 181–206, 2010.
[88]  W. M. Grady and J. M. Carethers, “Genomic and epigenetic instability in colorectal cancer pathogenesis,” Gastroenterology, vol. 135, no. 4, pp. 1079–1099, 2008.
[89]  J. J. L. Wong, N. J. Hawkins, R. L. Ward, and M. P. Hitchins, “Methylation of the 3p22 region encompassing MLH1 is representative of the CpG island methylator phenotype in colorectal cancer,” Modern Pathology, vol. 24, no. 3, pp. 396–411, 2011.
[90]  I. B. Georgiades, L. J. Curtis, R. M. Morris, C. C. Bird, and A. H. Wyllie, “Heterogeneity studies identify a subset of sporadic colorectal cancers without evidence for chromosomal or microsatellite instability,” Oncogene, vol. 18, no. 56, pp. 7933–7940, 1999.
[91]  D. R. English, J. P. Young, J. A. Simpson et al., “Ethnicity and risk for colorectal cancers showing somatic BRAF V600E mutation or CpG island methylator phenotype,” Cancer Epidemiology Biomarkers and Prevention, vol. 17, no. 7, pp. 1774–1780, 2008.
[92]  L. S. Rozek, C. M. Herron, J. K. Greenson et al., “Smoking, gender, and ethnicity predict somatic BRAF mutations in colorectal cancer,” Cancer Epidemiology Biomarkers and Prevention, vol. 19, no. 3, pp. 838–843, 2010.
[93]  S. Ogino, K. Nosho, G. J. Kirkner et al., “CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer,” Gut, vol. 58, no. 1, pp. 90–96, 2009.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413