Skip to main content
Top
Published in: Diagnostic Pathology 1/2018

Open Access 01-12-2018 | Review

Genetic and molecular origins of colorectal Cancer among the Iranians: an update

Authors: Mohammad Reza Abbaszadegan, Meysam Moghbeli

Published in: Diagnostic Pathology | Issue 1/2018

Login to get access

Abstract

Background

Colorectal cancer (CRC) is one the leading causes of cancer related deaths among Iranians. Despite the various progresses in new therapeutic methods, it has still a low rate of survival. This high ratio of mortality is mainly related to the late diagnosis, in which the patients refer for treatment in advanced stages of tumor.

Main body

colorectal cancer progression is largely associated with molecular and genetic bases. Although Iran has a high ratio of CRC mortality, there is not an efficient genetic panel for detection and prognosis. Therefore, it is critical to introduce new diagnostic markers with ability to detect in early stages.

Conclusion

Present review summarizes all of the genetic and epigenetic factors which are reported in CRC until now among the Iranian patients to pave the way of incorporation of new ethnic specific markers into the clinical practice and development of new targeted therapeutic methods.
Literature
1.
go back to reference Ansari R, et al. Incidence and age distribution of colorectal cancer in Iran: results of a population-based cancer registry. Cancer Lett. 2006;240(1):143–7.PubMedCrossRef Ansari R, et al. Incidence and age distribution of colorectal cancer in Iran: results of a population-based cancer registry. Cancer Lett. 2006;240(1):143–7.PubMedCrossRef
2.
go back to reference Hosseini SV, Izadpanah A, Yarmohammadi H. Epidemiological changes in colorectal cancer in shiraz, Iran: 1980-2000. ANZ J Surg. 2004;74(7):547–9.PubMedCrossRef Hosseini SV, Izadpanah A, Yarmohammadi H. Epidemiological changes in colorectal cancer in shiraz, Iran: 1980-2000. ANZ J Surg. 2004;74(7):547–9.PubMedCrossRef
3.
go back to reference Morimoto LM, et al. Risk factors for hyperplastic and adenomatous polyps: evidence for malignant potential? Cancer Epidemiol Biomark Prev. 2002;11(10 Pt 1):1012–8. Morimoto LM, et al. Risk factors for hyperplastic and adenomatous polyps: evidence for malignant potential? Cancer Epidemiol Biomark Prev. 2002;11(10 Pt 1):1012–8.
4.
6.
go back to reference Ting WC, et al. Common genetic variants in Wnt signaling pathway genes as potential prognostic biomarkers for colorectal cancer. PLoS One. 2013;8(2):e56196.PubMedPubMedCentralCrossRef Ting WC, et al. Common genetic variants in Wnt signaling pathway genes as potential prognostic biomarkers for colorectal cancer. PLoS One. 2013;8(2):e56196.PubMedPubMedCentralCrossRef
7.
go back to reference Yokota T. Are KRAS/BRAF mutations potent prognostic and/or predictive biomarkers in colorectal cancers? Anti Cancer Agents Med Chem. 2012;12(2):163–71.CrossRef Yokota T. Are KRAS/BRAF mutations potent prognostic and/or predictive biomarkers in colorectal cancers? Anti Cancer Agents Med Chem. 2012;12(2):163–71.CrossRef
8.
10.
go back to reference Kihara M, Kihara M, Noda K. Risk of smoking for squamous and small cell carcinomas of the lung modulated by combinations of CYP1A1 and GSTM1 gene polymorphisms in a Japanese population. Carcinogenesis. 1995;16(10):2331–6.PubMedCrossRef Kihara M, Kihara M, Noda K. Risk of smoking for squamous and small cell carcinomas of the lung modulated by combinations of CYP1A1 and GSTM1 gene polymorphisms in a Japanese population. Carcinogenesis. 1995;16(10):2331–6.PubMedCrossRef
11.
go back to reference Nebert DW, McKinnon RA, Puga A. Human drug-metabolizing enzyme polymorphisms: effects on risk of toxicity and cancer. DNA Cell Biol. 1996;15(4):273–80.PubMedCrossRef Nebert DW, McKinnon RA, Puga A. Human drug-metabolizing enzyme polymorphisms: effects on risk of toxicity and cancer. DNA Cell Biol. 1996;15(4):273–80.PubMedCrossRef
12.
go back to reference Singhal SS, et al. Induction of glutathione S-transferase hGST 5.8 is an early response to oxidative stress in RPE cells. Invest Ophthalmol Vis Sci. 1999;40(11):2652–9.PubMed Singhal SS, et al. Induction of glutathione S-transferase hGST 5.8 is an early response to oxidative stress in RPE cells. Invest Ophthalmol Vis Sci. 1999;40(11):2652–9.PubMed
13.
go back to reference Vanhaecke T, et al. Effect of ethanol on the expression of hepatic glutathione S-transferase: an in vivo/in vitro study. Biochem Pharmacol. 2000;60(10):1491–6.PubMedCrossRef Vanhaecke T, et al. Effect of ethanol on the expression of hepatic glutathione S-transferase: an in vivo/in vitro study. Biochem Pharmacol. 2000;60(10):1491–6.PubMedCrossRef
14.
go back to reference Nomani H, et al. Glutathione S-transferases activity in patients with colorectal cancer. Clin Biochem. 2005;38(7):621–4.PubMedCrossRef Nomani H, et al. Glutathione S-transferases activity in patients with colorectal cancer. Clin Biochem. 2005;38(7):621–4.PubMedCrossRef
15.
go back to reference Ebrahimkhani S, et al. Association of GSTM1, GSTT1, GSTP1 and CYP2E1 single nucleotide polymorphisms with colorectal cancer in Iran. Pathol Oncol Res. 2012;18(3):651–6.PubMedCrossRef Ebrahimkhani S, et al. Association of GSTM1, GSTT1, GSTP1 and CYP2E1 single nucleotide polymorphisms with colorectal cancer in Iran. Pathol Oncol Res. 2012;18(3):651–6.PubMedCrossRef
16.
go back to reference Jamhiri I, Saadat I, Omidvari S. Genetic polymorphisms of superoxide dismutase-1 A251G and catalase C-262T with the risk of colorectal cancer. Mol Biol Res Commun. 2017;6(2):85–90.PubMedPubMedCentral Jamhiri I, Saadat I, Omidvari S. Genetic polymorphisms of superoxide dismutase-1 A251G and catalase C-262T with the risk of colorectal cancer. Mol Biol Res Commun. 2017;6(2):85–90.PubMedPubMedCentral
17.
go back to reference Bassermann F, Pagano M. Dissecting the role of ubiquitylation in the DNA damage response checkpoint in G2. Cell Death Differ. 2010;17(1):78–85.PubMedCrossRef Bassermann F, Pagano M. Dissecting the role of ubiquitylation in the DNA damage response checkpoint in G2. Cell Death Differ. 2010;17(1):78–85.PubMedCrossRef
18.
go back to reference van Wijk SJ, Timmers HT. The family of ubiquitin-conjugating enzymes (E2s): deciding between life and death of proteins. FASEB J. 2010;24(4):981–93.PubMedCrossRef van Wijk SJ, Timmers HT. The family of ubiquitin-conjugating enzymes (E2s): deciding between life and death of proteins. FASEB J. 2010;24(4):981–93.PubMedCrossRef
19.
go back to reference Mokarram P, et al. Promoter methylation status of two novel human genes, UBE2Q1 and UBE2Q2, in colorectal Cancer: a new finding in Iranian patients. Asian Pac J Cancer Prev. 2015;16(18):8247–52.PubMedCrossRef Mokarram P, et al. Promoter methylation status of two novel human genes, UBE2Q1 and UBE2Q2, in colorectal Cancer: a new finding in Iranian patients. Asian Pac J Cancer Prev. 2015;16(18):8247–52.PubMedCrossRef
21.
go back to reference Derynck R, Akhurst RJ, Balmain A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet. 2001;29(2):117–29.PubMedCrossRef Derynck R, Akhurst RJ, Balmain A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet. 2001;29(2):117–29.PubMedCrossRef
22.
go back to reference Amirghofran Z, et al. Genetic polymorphism in the transforming growth factor beta1 gene (−509 C/T and −800 G/a) and colorectal cancer. Cancer Genet Cytogenet. 2009;190(1):21–5.PubMedCrossRef Amirghofran Z, et al. Genetic polymorphism in the transforming growth factor beta1 gene (−509 C/T and −800 G/a) and colorectal cancer. Cancer Genet Cytogenet. 2009;190(1):21–5.PubMedCrossRef
23.
go back to reference Akbari Z, et al. Lack of influence of the SMAD7 gene rs2337107 polymorphism on risk of colorectal cancer in an Iranian population. Asian Pac J Cancer Prev. 2014;15(11):4437–41.PubMedCrossRef Akbari Z, et al. Lack of influence of the SMAD7 gene rs2337107 polymorphism on risk of colorectal cancer in an Iranian population. Asian Pac J Cancer Prev. 2014;15(11):4437–41.PubMedCrossRef
24.
go back to reference Damavand B, et al. Intronic polymorphisms of the SMAD7 gene in association with colorectal cancer. Asian Pac J Cancer Prev. 2015;16(1):41–4.PubMedCrossRef Damavand B, et al. Intronic polymorphisms of the SMAD7 gene in association with colorectal cancer. Asian Pac J Cancer Prev. 2015;16(1):41–4.PubMedCrossRef
25.
go back to reference Moghbeli M, et al. Role of Msi1 and PYGO2 in esophageal squamous cell carcinoma depth of invasion. J Cell Commun Signal. 2016;10(1):49–53.PubMedCrossRef Moghbeli M, et al. Role of Msi1 and PYGO2 in esophageal squamous cell carcinoma depth of invasion. J Cell Commun Signal. 2016;10(1):49–53.PubMedCrossRef
26.
go back to reference Kimelman D, Xu W. Beta-catenin destruction complex: insights and questions from a structural perspective. Oncogene. 2006;25(57):7482–91.PubMedCrossRef Kimelman D, Xu W. Beta-catenin destruction complex: insights and questions from a structural perspective. Oncogene. 2006;25(57):7482–91.PubMedCrossRef
27.
go back to reference Giles RH, van Es JH, Clevers H. Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 2003;1653(1):1–24.PubMed Giles RH, van Es JH, Clevers H. Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 2003;1653(1):1–24.PubMed
28.
go back to reference Thorstensen L, et al. Genetic and epigenetic changes of components affecting the WNT pathway in colorectal carcinomas stratified by microsatellite instability. Neoplasia. 2005;7(2):99–108.PubMedPubMedCentralCrossRef Thorstensen L, et al. Genetic and epigenetic changes of components affecting the WNT pathway in colorectal carcinomas stratified by microsatellite instability. Neoplasia. 2005;7(2):99–108.PubMedPubMedCentralCrossRef
29.
go back to reference Naghibalhossaini F, et al. Epigenetic and genetic analysis of WNT signaling pathway in sporadic colorectal cancer patients from Iran. Mol Biol Rep. 2012;39(5):6171–8.PubMedCrossRef Naghibalhossaini F, et al. Epigenetic and genetic analysis of WNT signaling pathway in sporadic colorectal cancer patients from Iran. Mol Biol Rep. 2012;39(5):6171–8.PubMedCrossRef
30.
go back to reference Samaei NM, et al. Promoter methylation analysis of WNT/beta-catenin pathway regulators and its association with expression of DNMT1 enzyme in colorectal cancer. J Biomed Sci. 2014;21:73.PubMedPubMedCentralCrossRef Samaei NM, et al. Promoter methylation analysis of WNT/beta-catenin pathway regulators and its association with expression of DNMT1 enzyme in colorectal cancer. J Biomed Sci. 2014;21:73.PubMedPubMedCentralCrossRef
31.
go back to reference Naini MA, et al. Sensitive and noninvasive detection of aberrant SFRP2 and MGMT-B methylation in Iranian patients with Colon polyps. Asian Pac J Cancer Prev. 2016;17(4):2185–93.PubMedCrossRef Naini MA, et al. Sensitive and noninvasive detection of aberrant SFRP2 and MGMT-B methylation in Iranian patients with Colon polyps. Asian Pac J Cancer Prev. 2016;17(4):2185–93.PubMedCrossRef
32.
go back to reference Rakoff-Nahoum S. Why cancer and inflammation? Yale J Biol Med. 2006;79(3–4):123–30.PubMed Rakoff-Nahoum S. Why cancer and inflammation? Yale J Biol Med. 2006;79(3–4):123–30.PubMed
33.
go back to reference Shi Y, et al. The role of interleukin-17A in colorectal tumorigenesis. Cancer Biother Radiopharm. 2013;28(6):429–32.PubMedCrossRef Shi Y, et al. The role of interleukin-17A in colorectal tumorigenesis. Cancer Biother Radiopharm. 2013;28(6):429–32.PubMedCrossRef
34.
go back to reference Nemati K, et al. Interleukin-17FT7488 allele is associated with a decreased risk of colorectal cancer and tumor progression. Gene. 2015;561(1):88–94.PubMedCrossRef Nemati K, et al. Interleukin-17FT7488 allele is associated with a decreased risk of colorectal cancer and tumor progression. Gene. 2015;561(1):88–94.PubMedCrossRef
35.
go back to reference Aukrust P, et al. Inflammatory and anti-inflammatory cytokines in chronic heart failure: potential therapeutic implications. Ann Med. 2005;37(2):74–85.PubMedCrossRef Aukrust P, et al. Inflammatory and anti-inflammatory cytokines in chronic heart failure: potential therapeutic implications. Ann Med. 2005;37(2):74–85.PubMedCrossRef
36.
37.
go back to reference van Horssen R, Ten Hagen TL, Eggermont AM. TNF-alpha in cancer treatment: molecular insights, antitumor effects, and clinical utility. Oncologist. 2006;11(4):397–408.PubMedCrossRef van Horssen R, Ten Hagen TL, Eggermont AM. TNF-alpha in cancer treatment: molecular insights, antitumor effects, and clinical utility. Oncologist. 2006;11(4):397–408.PubMedCrossRef
38.
go back to reference Azimzadeh P, et al. Interleukin-16 (IL-16) gene polymorphisms in Iranian patients with colorectal cancer. J Gastrointestin Liver Dis. 2011;20(4):371–6.PubMed Azimzadeh P, et al. Interleukin-16 (IL-16) gene polymorphisms in Iranian patients with colorectal cancer. J Gastrointestin Liver Dis. 2011;20(4):371–6.PubMed
39.
go back to reference Al-Samadi A, et al. Distinctive expression pattern of interleukin-17 cytokine family members in colorectal cancer. Tumour Biol. 2016;37(2):1609–15.PubMedCrossRef Al-Samadi A, et al. Distinctive expression pattern of interleukin-17 cytokine family members in colorectal cancer. Tumour Biol. 2016;37(2):1609–15.PubMedCrossRef
40.
go back to reference Tian Y, et al. Differential effects of NOD2 polymorphisms on colorectal cancer risk: a meta-analysis. Int J Color Dis. 2010;25(2):161–8.CrossRef Tian Y, et al. Differential effects of NOD2 polymorphisms on colorectal cancer risk: a meta-analysis. Int J Color Dis. 2010;25(2):161–8.CrossRef
41.
go back to reference Landi D, et al. Polymorphisms within micro-RNA-binding sites and risk of sporadic colorectal cancer. Carcinogenesis. 2008;29(3):579–84.PubMedCrossRef Landi D, et al. Polymorphisms within micro-RNA-binding sites and risk of sporadic colorectal cancer. Carcinogenesis. 2008;29(3):579–84.PubMedCrossRef
42.
go back to reference Ahangari F, et al. A miRNA-binding site single nucleotide polymorphism in the 3’-UTR region of the NOD2 gene is associated with colorectal cancer. Med Oncol. 2014;31(9):173.PubMedCrossRef Ahangari F, et al. A miRNA-binding site single nucleotide polymorphism in the 3’-UTR region of the NOD2 gene is associated with colorectal cancer. Med Oncol. 2014;31(9):173.PubMedCrossRef
43.
go back to reference Azimzadeh P, et al. Association of co-stimulatory human B-lymphocyte antigen B7-2 (CD86) gene polymorphism with colorectal cancer risk. Gastroenterol Hepatol Bed Bench. 2013;6(2):86–91.PubMedPubMedCentral Azimzadeh P, et al. Association of co-stimulatory human B-lymphocyte antigen B7-2 (CD86) gene polymorphism with colorectal cancer risk. Gastroenterol Hepatol Bed Bench. 2013;6(2):86–91.PubMedPubMedCentral
44.
go back to reference Abtahi S, et al. Dual association of serum interleukin-10 levels with colorectal cancer. J Cancer Res Ther. 2017;13(2):252–6.PubMedCrossRef Abtahi S, et al. Dual association of serum interleukin-10 levels with colorectal cancer. J Cancer Res Ther. 2017;13(2):252–6.PubMedCrossRef
45.
go back to reference Jaiswal M, LaRusso NF, Gores GJ. Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am J Physiol Gastrointest Liver Physiol. 2001;281(3):G626–34.PubMedCrossRef Jaiswal M, LaRusso NF, Gores GJ. Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am J Physiol Gastrointest Liver Physiol. 2001;281(3):G626–34.PubMedCrossRef
46.
go back to reference Kim PK, et al. The regulatory role of nitric oxide in apoptosis. Int Immunopharmacol. 2001;1(8):1421–41.PubMedCrossRef Kim PK, et al. The regulatory role of nitric oxide in apoptosis. Int Immunopharmacol. 2001;1(8):1421–41.PubMedCrossRef
48.
go back to reference Weinberg JB. Nitric oxide synthase 2 and cyclooxygenase 2 interactions in inflammation. Immunol Res. 2000;22(2–3):319–41.PubMedCrossRef Weinberg JB. Nitric oxide synthase 2 and cyclooxygenase 2 interactions in inflammation. Immunol Res. 2000;22(2–3):319–41.PubMedCrossRef
49.
go back to reference Habibollahi P, et al. Correlation between inducible nitric oxide synthase and cyclooxygenase-2 expression in human colorectal adenocarcinoma: a cross-sectional study. Pathol Oncol Res. 2010;16(3):327–35.PubMedCrossRef Habibollahi P, et al. Correlation between inducible nitric oxide synthase and cyclooxygenase-2 expression in human colorectal adenocarcinoma: a cross-sectional study. Pathol Oncol Res. 2010;16(3):327–35.PubMedCrossRef
50.
go back to reference Tivol EA, et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3(5):541–7.PubMedCrossRef Tivol EA, et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3(5):541–7.PubMedCrossRef
51.
go back to reference Walunas TL, Bakker CY, Bluestone JA. CTLA-4 ligation blocks CD28-dependent T cell activation. J Exp Med. 1996;183(6):2541–50.PubMedCrossRef Walunas TL, Bakker CY, Bluestone JA. CTLA-4 ligation blocks CD28-dependent T cell activation. J Exp Med. 1996;183(6):2541–50.PubMedCrossRef
52.
go back to reference Hadinia A, et al. CTLA-4 gene promoter and exon 1 polymorphisms in Iranian patients with gastric and colorectal cancers. J Gastroenterol Hepatol. 2007;22(12):2283–7.PubMedCrossRef Hadinia A, et al. CTLA-4 gene promoter and exon 1 polymorphisms in Iranian patients with gastric and colorectal cancers. J Gastroenterol Hepatol. 2007;22(12):2283–7.PubMedCrossRef
53.
go back to reference Shafaei S, et al. The association between CD166 detection rate and clinicopathologic parameters of patients with colorectal cancer. Caspian J Intern Med. 2013;4(4):768–72.PubMedPubMedCentral Shafaei S, et al. The association between CD166 detection rate and clinicopathologic parameters of patients with colorectal cancer. Caspian J Intern Med. 2013;4(4):768–72.PubMedPubMedCentral
54.
go back to reference Nishida M, Funahashi T, Shimomura I. Pathophysiological significance of adiponectin. Med Mol Morphol. 2007;40(2):55–67.PubMedCrossRef Nishida M, Funahashi T, Shimomura I. Pathophysiological significance of adiponectin. Med Mol Morphol. 2007;40(2):55–67.PubMedCrossRef
55.
go back to reference Sugiyama M, et al. Adiponectin inhibits colorectal cancer cell growth through the AMPK/mTOR pathway. Int J Oncol. 2009;34(2):339–44.PubMed Sugiyama M, et al. Adiponectin inhibits colorectal cancer cell growth through the AMPK/mTOR pathway. Int J Oncol. 2009;34(2):339–44.PubMed
56.
57.
go back to reference Albanell J, et al. Activated extracellular signal-regulated kinases: association with epidermal growth factor receptor/transforming growth factor alpha expression in head and neck squamous carcinoma and inhibition by anti-epidermal growth factor receptor treatments. Cancer Res. 2001;61(17):6500–10.PubMed Albanell J, et al. Activated extracellular signal-regulated kinases: association with epidermal growth factor receptor/transforming growth factor alpha expression in head and neck squamous carcinoma and inhibition by anti-epidermal growth factor receptor treatments. Cancer Res. 2001;61(17):6500–10.PubMed
58.
go back to reference Motlagh A, et al. Expression of epidermal growth factor receptor as a predictive factor for rectal cancer. Arch Iran Med. 2007;10(3):301–8.PubMed Motlagh A, et al. Expression of epidermal growth factor receptor as a predictive factor for rectal cancer. Arch Iran Med. 2007;10(3):301–8.PubMed
59.
go back to reference Tavangar SM, Shariftabrizi A, Soroush AR. Her-2/neu over-expression correlates with more advanced disease in Iranian colorectal cancer patients. Med Sci Monit. 2005;11(3):CR123–6.PubMed Tavangar SM, Shariftabrizi A, Soroush AR. Her-2/neu over-expression correlates with more advanced disease in Iranian colorectal cancer patients. Med Sci Monit. 2005;11(3):CR123–6.PubMed
60.
go back to reference Liu L, et al. A novel protein tyrosine kinase NOK that shares homology with platelet- derived growth factor/fibroblast growth factor receptors induces tumorigenesis and metastasis in nude mice. Cancer Res. 2004;64(10):3491–9.PubMedCrossRef Liu L, et al. A novel protein tyrosine kinase NOK that shares homology with platelet- derived growth factor/fibroblast growth factor receptors induces tumorigenesis and metastasis in nude mice. Cancer Res. 2004;64(10):3491–9.PubMedCrossRef
61.
go back to reference Orang AV, et al. Diagnostic relevance of overexpressed serine threonine tyrosine kinase/novel oncogene with kinase domain (STYK1/ NOK) mRNA in colorectal cancer. Asian Pac J Cancer Prev. 2014;15(16):6685–9.PubMedCrossRef Orang AV, et al. Diagnostic relevance of overexpressed serine threonine tyrosine kinase/novel oncogene with kinase domain (STYK1/ NOK) mRNA in colorectal cancer. Asian Pac J Cancer Prev. 2014;15(16):6685–9.PubMedCrossRef
62.
63.
go back to reference Tavakoli Koudehi A, et al. AKAP4, SPAG9 and NY-ESO-1 in Iranian colorectal Cancer patients as probable diagnostic and prognostic biomarkers. Asian Pac J Cancer Prev. 2018;19(2):463–9.PubMed Tavakoli Koudehi A, et al. AKAP4, SPAG9 and NY-ESO-1 in Iranian colorectal Cancer patients as probable diagnostic and prognostic biomarkers. Asian Pac J Cancer Prev. 2018;19(2):463–9.PubMed
64.
go back to reference Lee S, et al. Clinicopathological features of CpG island methylator phenotype-positive colorectal cancer and its adverse prognosis in relation to KRAS/BRAF mutation. Pathol Int. 2008;58(2):104–13.PubMedCrossRef Lee S, et al. Clinicopathological features of CpG island methylator phenotype-positive colorectal cancer and its adverse prognosis in relation to KRAS/BRAF mutation. Pathol Int. 2008;58(2):104–13.PubMedCrossRef
65.
go back to reference Hamzehzadeh L, et al. Common KRAS and NRAS gene mutations in sporadic colorectal cancer in northeastern Iranian patients. Curr Probl Cancer. 2018;42(6):572–81.PubMedCrossRef Hamzehzadeh L, et al. Common KRAS and NRAS gene mutations in sporadic colorectal cancer in northeastern Iranian patients. Curr Probl Cancer. 2018;42(6):572–81.PubMedCrossRef
66.
go back to reference Fransen K, et al. Mutation analysis of the BRAF, ARAF and RAF-1 genes in human colorectal adenocarcinomas. Carcinogenesis. 2004;25(4):527–33.PubMedCrossRef Fransen K, et al. Mutation analysis of the BRAF, ARAF and RAF-1 genes in human colorectal adenocarcinomas. Carcinogenesis. 2004;25(4):527–33.PubMedCrossRef
67.
go back to reference Guedes JG, et al. High resolution melting analysis of KRAS, BRAF and PIK3CA in KRAS exon 2 wild-type metastatic colorectal cancer. BMC Cancer. 2013;13:169.PubMedPubMedCentralCrossRef Guedes JG, et al. High resolution melting analysis of KRAS, BRAF and PIK3CA in KRAS exon 2 wild-type metastatic colorectal cancer. BMC Cancer. 2013;13:169.PubMedPubMedCentralCrossRef
68.
go back to reference Koochak A, et al. Mutation analysis of KRAS and BRAF genes in metastatic colorectal Cancer: a first large scale study from Iran. Asian Pac J Cancer Prev. 2016;17(2):603–8.PubMedCrossRef Koochak A, et al. Mutation analysis of KRAS and BRAF genes in metastatic colorectal Cancer: a first large scale study from Iran. Asian Pac J Cancer Prev. 2016;17(2):603–8.PubMedCrossRef
69.
go back to reference Omidifar NM, Geramizadeh BM, Mirzai MM. K-ras mutation in colorectal Cancer, a report from southern Iran. Iran J Med Sci. 2015;40(5):454–60. Omidifar NM, Geramizadeh BM, Mirzai MM. K-ras mutation in colorectal Cancer, a report from southern Iran. Iran J Med Sci. 2015;40(5):454–60.
70.
go back to reference Zhao R, et al. Analysis of p53-regulated gene expression patterns using oligonucleotide arrays. Genes Dev. 2000;14(8):981–93.PubMedPubMedCentral Zhao R, et al. Analysis of p53-regulated gene expression patterns using oligonucleotide arrays. Genes Dev. 2000;14(8):981–93.PubMedPubMedCentral
71.
go back to reference Ghavam-Nasiri MR, et al. Expression of p53 in colorectal carcinoma: correlation with clinicopathologic features. Arch Iran Med. 2007;10(1):38–42.PubMed Ghavam-Nasiri MR, et al. Expression of p53 in colorectal carcinoma: correlation with clinicopathologic features. Arch Iran Med. 2007;10(1):38–42.PubMed
72.
go back to reference Sternberg MJ, et al. Progress in protein structure prediction: assessment of CASP3. Curr Opin Struct Biol. 1999;9(3):368–73.PubMedCrossRef Sternberg MJ, et al. Progress in protein structure prediction: assessment of CASP3. Curr Opin Struct Biol. 1999;9(3):368–73.PubMedCrossRef
73.
go back to reference Zheng HC, et al. Expression of Fas ligand and caspase-3 contributes to formation of immune escape in gastric cancer. World J Gastroenterol. 2003;9(7):1415–20.PubMedPubMedCentralCrossRef Zheng HC, et al. Expression of Fas ligand and caspase-3 contributes to formation of immune escape in gastric cancer. World J Gastroenterol. 2003;9(7):1415–20.PubMedPubMedCentralCrossRef
74.
75.
go back to reference Golmohammadi R, et al. Characterization and prognostic value of mutations in exons 5 and 6 of the p53 gene in patients with colorectal cancers in Central Iran. Gut Liver. 2013;7(3):295–302.PubMedPubMedCentralCrossRef Golmohammadi R, et al. Characterization and prognostic value of mutations in exons 5 and 6 of the p53 gene in patients with colorectal cancers in Central Iran. Gut Liver. 2013;7(3):295–302.PubMedPubMedCentralCrossRef
76.
go back to reference Atoum MF, Tchoporyan MN. Association between circulating vitamin D, the Taq1 vitamin D receptor gene polymorphism and colorectal cancer risk among Jordanians. Asian Pac J Cancer Prev. 2014;15(17):7337–41.PubMedCrossRef Atoum MF, Tchoporyan MN. Association between circulating vitamin D, the Taq1 vitamin D receptor gene polymorphism and colorectal cancer risk among Jordanians. Asian Pac J Cancer Prev. 2014;15(17):7337–41.PubMedCrossRef
77.
go back to reference Moossavi M, et al. Positive correlation between vitamin D receptor gene FokI polymorphism and colorectal cancer susceptibility in South-Khorasan of Iran. J Cell Biochem. 2018;119(10):8190–4.PubMedCrossRef Moossavi M, et al. Positive correlation between vitamin D receptor gene FokI polymorphism and colorectal cancer susceptibility in South-Khorasan of Iran. J Cell Biochem. 2018;119(10):8190–4.PubMedCrossRef
78.
go back to reference Mahmoudi T, et al. Vitamin D receptor gene ApaI polymorphism is associated with susceptibility to colorectal cancer. Dig Dis Sci. 2010;55(7):2008–13.PubMedCrossRef Mahmoudi T, et al. Vitamin D receptor gene ApaI polymorphism is associated with susceptibility to colorectal cancer. Dig Dis Sci. 2010;55(7):2008–13.PubMedCrossRef
79.
go back to reference Limburg PJ, et al. Insulin, glucose, insulin resistance, and incident colorectal cancer in male smokers. Clin Gastroenterol Hepatol. 2006;4(12):1514–21.PubMedPubMedCentralCrossRef Limburg PJ, et al. Insulin, glucose, insulin resistance, and incident colorectal cancer in male smokers. Clin Gastroenterol Hepatol. 2006;4(12):1514–21.PubMedPubMedCentralCrossRef
80.
go back to reference Larsson SC, Orsini N, Wolk A. Diabetes mellitus and risk of colorectal cancer: a meta-analysis. J Natl Cancer Inst. 2005;97(22):1679–87.PubMedCrossRef Larsson SC, Orsini N, Wolk A. Diabetes mellitus and risk of colorectal cancer: a meta-analysis. J Natl Cancer Inst. 2005;97(22):1679–87.PubMedCrossRef
82.
go back to reference Mahmoudi T, et al. Gly972Arg variant of insulin receptor substrate 1 gene and colorectal cancer risk in overweight/obese subjects. Int J Biol Markers. 2016;31(1):e68–72.PubMedCrossRef Mahmoudi T, et al. Gly972Arg variant of insulin receptor substrate 1 gene and colorectal cancer risk in overweight/obese subjects. Int J Biol Markers. 2016;31(1):e68–72.PubMedCrossRef
83.
go back to reference Hua FF, et al. MiRNA-338-3p regulates cervical cancer cells proliferation by targeting MACC1 through MAPK signaling pathway. Eur Rev Med Pharmacol Sci. 2017;21(23):5342–52.PubMed Hua FF, et al. MiRNA-338-3p regulates cervical cancer cells proliferation by targeting MACC1 through MAPK signaling pathway. Eur Rev Med Pharmacol Sci. 2017;21(23):5342–52.PubMed
84.
go back to reference Bian X, et al. HDAC inhibitor suppresses proliferation and invasion of breast cancer cells through regulation of miR-200c targeting CRKL. Biochem Pharmacol. 2018;147:30–7.PubMedCrossRef Bian X, et al. HDAC inhibitor suppresses proliferation and invasion of breast cancer cells through regulation of miR-200c targeting CRKL. Biochem Pharmacol. 2018;147:30–7.PubMedCrossRef
85.
go back to reference Mahdavinezhad A, et al. Evaluation of miR-141, miR-200c, miR-30b expression and Clinicopathological features of bladder Cancer. Int J Mol Cell Med. 2015;4(1):32–9.PubMedPubMedCentral Mahdavinezhad A, et al. Evaluation of miR-141, miR-200c, miR-30b expression and Clinicopathological features of bladder Cancer. Int J Mol Cell Med. 2015;4(1):32–9.PubMedPubMedCentral
86.
go back to reference Karimi Mazraehshah M, et al. Anticancer effects of miR-200c in colorectal cancer through BMI1. J Cell Biochem. 2018;119(12):10005–12.PubMedCrossRef Karimi Mazraehshah M, et al. Anticancer effects of miR-200c in colorectal cancer through BMI1. J Cell Biochem. 2018;119(12):10005–12.PubMedCrossRef
88.
89.
go back to reference Zhao BS, et al. Screening of microRNA in patients with esophageal cancer at same tumor node metastasis stage with different prognoses. Asian Pac J Cancer Prev. 2013;14(1):139–43.PubMedCrossRef Zhao BS, et al. Screening of microRNA in patients with esophageal cancer at same tumor node metastasis stage with different prognoses. Asian Pac J Cancer Prev. 2013;14(1):139–43.PubMedCrossRef
90.
go back to reference Orang AV, et al. Diagnostic and prognostic value of miR-205 in colorectal cancer. Asian Pac J Cancer Prev. 2014;15(9):4033–7.PubMedCrossRef Orang AV, et al. Diagnostic and prognostic value of miR-205 in colorectal cancer. Asian Pac J Cancer Prev. 2014;15(9):4033–7.PubMedCrossRef
91.
go back to reference Basati G, et al. Elevated level of microRNA-21 in the serum of patients with colorectal cancer. Med Oncol. 2014;31(10):205.PubMedCrossRef Basati G, et al. Elevated level of microRNA-21 in the serum of patients with colorectal cancer. Med Oncol. 2014;31(10):205.PubMedCrossRef
92.
go back to reference Bastaminejad S, et al. Investigation of MicroRNA-21 expression levels in serum and stool as a potential non-invasive biomarker for diagnosis of colorectal Cancer. Iran Biomed J. 2017;21(2):106–13.PubMedPubMedCentralCrossRef Bastaminejad S, et al. Investigation of MicroRNA-21 expression levels in serum and stool as a potential non-invasive biomarker for diagnosis of colorectal Cancer. Iran Biomed J. 2017;21(2):106–13.PubMedPubMedCentralCrossRef
93.
94.
go back to reference Xiao L, et al. miR-29b represses intestinal mucosal growth by inhibiting translation of cyclin-dependent kinase 2. Mol Biol Cell. 2013;24(19):3038–46.PubMedPubMedCentralCrossRef Xiao L, et al. miR-29b represses intestinal mucosal growth by inhibiting translation of cyclin-dependent kinase 2. Mol Biol Cell. 2013;24(19):3038–46.PubMedPubMedCentralCrossRef
95.
go back to reference Basati G, et al. Circulating levels of the miRNAs, miR-194, and miR-29b, as clinically useful biomarkers for colorectal cancer. Tumour Biol. 2016;37(2):1781–8.PubMedCrossRef Basati G, et al. Circulating levels of the miRNAs, miR-194, and miR-29b, as clinically useful biomarkers for colorectal cancer. Tumour Biol. 2016;37(2):1781–8.PubMedCrossRef
96.
go back to reference Hur K, et al. MicroRNA-200c modulates epithelial-to-mesenchymal transition (EMT) in human colorectal cancer metastasis. Gut. 2013;62(9):1315–26.PubMedCrossRef Hur K, et al. MicroRNA-200c modulates epithelial-to-mesenchymal transition (EMT) in human colorectal cancer metastasis. Gut. 2013;62(9):1315–26.PubMedCrossRef
97.
go back to reference Paterson EL, et al. Down-regulation of the miRNA-200 family at the invasive front of colorectal cancers with degraded basement membrane indicates EMT is involved in cancer progression. Neoplasia. 2013;15(2):180–91.PubMedPubMedCentralCrossRef Paterson EL, et al. Down-regulation of the miRNA-200 family at the invasive front of colorectal cancers with degraded basement membrane indicates EMT is involved in cancer progression. Neoplasia. 2013;15(2):180–91.PubMedPubMedCentralCrossRef
98.
go back to reference Khalili M, et al. Downregulation of the genes involved in reprogramming (SOX2, c-MYC, miR-302, miR-145, and P21) in gastric adenocarcinoma. J Gastrointest Cancer. 2015;46(3):251–8.PubMedCrossRef Khalili M, et al. Downregulation of the genes involved in reprogramming (SOX2, c-MYC, miR-302, miR-145, and P21) in gastric adenocarcinoma. J Gastrointest Cancer. 2015;46(3):251–8.PubMedCrossRef
99.
go back to reference Li S, et al. miR-145 suppresses colorectal cancer cell migration and invasion by targeting an ETS-related gene. Oncol Rep. 2016;36(4):1917–26.PubMedCrossRef Li S, et al. miR-145 suppresses colorectal cancer cell migration and invasion by targeting an ETS-related gene. Oncol Rep. 2016;36(4):1917–26.PubMedCrossRef
100.
go back to reference Valeri N, et al. MicroRNA-135b promotes cancer progression by acting as a downstream effector of oncogenic pathways in colon cancer. Cancer Cell. 2014;25(4):469–83.PubMedPubMedCentralCrossRef Valeri N, et al. MicroRNA-135b promotes cancer progression by acting as a downstream effector of oncogenic pathways in colon cancer. Cancer Cell. 2014;25(4):469–83.PubMedPubMedCentralCrossRef
101.
go back to reference Cottonham CL, Kaneko S, Xu L. miR-21 and miR-31 converge on TIAM1 to regulate migration and invasion of colon carcinoma cells. J Biol Chem. 2010;285(46):35293–302.PubMedPubMedCentralCrossRef Cottonham CL, Kaneko S, Xu L. miR-21 and miR-31 converge on TIAM1 to regulate migration and invasion of colon carcinoma cells. J Biol Chem. 2010;285(46):35293–302.PubMedPubMedCentralCrossRef
102.
go back to reference Sun D, et al. MicroRNA-31 activates the RAS pathway and functions as an oncogenic MicroRNA in human colorectal cancer by repressing RAS p21 GTPase activating protein 1 (RASA1). J Biol Chem. 2013;288(13):9508–18.PubMedPubMedCentralCrossRef Sun D, et al. MicroRNA-31 activates the RAS pathway and functions as an oncogenic MicroRNA in human colorectal cancer by repressing RAS p21 GTPase activating protein 1 (RASA1). J Biol Chem. 2013;288(13):9508–18.PubMedPubMedCentralCrossRef
103.
go back to reference Xu T, et al. microRNA-20a enhances the epithelial-to-mesenchymal transition of colorectal cancer cells by modulating matrix metalloproteinases. Exp Ther Med. 2015;10(2):683–8.PubMedPubMedCentralCrossRef Xu T, et al. microRNA-20a enhances the epithelial-to-mesenchymal transition of colorectal cancer cells by modulating matrix metalloproteinases. Exp Ther Med. 2015;10(2):683–8.PubMedPubMedCentralCrossRef
104.
go back to reference Zhang GJ, et al. miR20a is an independent prognostic factor in colorectal cancer and is involved in cell metastasis. Mol Med Rep. 2014;10(1):283–91.PubMedCrossRef Zhang GJ, et al. miR20a is an independent prognostic factor in colorectal cancer and is involved in cell metastasis. Mol Med Rep. 2014;10(1):283–91.PubMedCrossRef
105.
go back to reference Eslamizadeh S, et al. The role of MicroRNA signature as diagnostic biomarkers in different clinical stages of colorectal Cancer. Cell J. 2018;20(2):220–30.PubMedPubMedCentral Eslamizadeh S, et al. The role of MicroRNA signature as diagnostic biomarkers in different clinical stages of colorectal Cancer. Cell J. 2018;20(2):220–30.PubMedPubMedCentral
106.
go back to reference Soreide K, et al. Microsatellite instability in colorectal cancer. Br J Surg. 2006;93(4):395–406.PubMedCrossRef Soreide K, et al. Microsatellite instability in colorectal cancer. Br J Surg. 2006;93(4):395–406.PubMedCrossRef
107.
go back to reference Montazer Haghighi M, et al. Four novel germline mutations in the MLH1 and PMS2 mismatch repair genes in patients with hereditary nonpolyposis colorectal cancer. Int J Color Dis. 2009;24(8):885–93.CrossRef Montazer Haghighi M, et al. Four novel germline mutations in the MLH1 and PMS2 mismatch repair genes in patients with hereditary nonpolyposis colorectal cancer. Int J Color Dis. 2009;24(8):885–93.CrossRef
108.
go back to reference Shahmoradi S, et al. Two novel mutations in hMLH1 gene in Iranian hereditary non-polyposis colorectal cancer patients. Familial Cancer. 2012;11(1):13–7.PubMedCrossRef Shahmoradi S, et al. Two novel mutations in hMLH1 gene in Iranian hereditary non-polyposis colorectal cancer patients. Familial Cancer. 2012;11(1):13–7.PubMedCrossRef
109.
go back to reference Boland CR, et al. A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58(22):5248–57.PubMed Boland CR, et al. A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58(22):5248–57.PubMed
110.
go back to reference Moghbeli M, et al. High frequency of microsatellite instability in sporadic colorectal cancer patients in Iran. Genet Mol Res. 2011;10(4):3520–9.PubMedCrossRef Moghbeli M, et al. High frequency of microsatellite instability in sporadic colorectal cancer patients in Iran. Genet Mol Res. 2011;10(4):3520–9.PubMedCrossRef
111.
go back to reference Faghani M, et al. The correlation between microsatellite instability and the features of sporadic colorectal Cancer in the north part of Iran. Gastroenterol Res Pract. 2012;2012:756263.PubMedPubMedCentralCrossRef Faghani M, et al. The correlation between microsatellite instability and the features of sporadic colorectal Cancer in the north part of Iran. Gastroenterol Res Pract. 2012;2012:756263.PubMedPubMedCentralCrossRef
112.
go back to reference Nazemalhosseini Mojarad E, et al. Low level of microsatellite instability correlates with poor clinical prognosis in stage II colorectal Cancer patients. J Oncol. 2016;2016:2196703.PubMed Nazemalhosseini Mojarad E, et al. Low level of microsatellite instability correlates with poor clinical prognosis in stage II colorectal Cancer patients. J Oncol. 2016;2016:2196703.PubMed
113.
go back to reference Brim H, et al. Impact of BRAF, MLH1 on the incidence of microsatellite instability high colorectal cancer in populations based study. Mol Cancer. 2008;7:68.PubMedPubMedCentralCrossRef Brim H, et al. Impact of BRAF, MLH1 on the incidence of microsatellite instability high colorectal cancer in populations based study. Mol Cancer. 2008;7:68.PubMedPubMedCentralCrossRef
114.
go back to reference Khatami F, et al. Effects of amino acid substitution polymorphisms of two DNA methyltransferases on susceptibility to sporadic colorectal cancer. Asian Pac J Cancer Prev. 2009;10(6):1183–8.PubMed Khatami F, et al. Effects of amino acid substitution polymorphisms of two DNA methyltransferases on susceptibility to sporadic colorectal cancer. Asian Pac J Cancer Prev. 2009;10(6):1183–8.PubMed
115.
go back to reference Farzanehfar M, et al. Evaluation of methylation of MGMT (O(6)-methylguanine-DNA methyltransferase) gene promoter in sporadic colorectal cancer. DNA Cell Biol. 2013;32(7):371–7.PubMedCrossRef Farzanehfar M, et al. Evaluation of methylation of MGMT (O(6)-methylguanine-DNA methyltransferase) gene promoter in sporadic colorectal cancer. DNA Cell Biol. 2013;32(7):371–7.PubMedCrossRef
116.
117.
go back to reference Nemati M, et al. Deregulated expression of HDAC3 in colorectal cancer and its clinical significance. Adv Clin Exp Med. 2018;27(3):305–11.PubMedCrossRef Nemati M, et al. Deregulated expression of HDAC3 in colorectal cancer and its clinical significance. Adv Clin Exp Med. 2018;27(3):305–11.PubMedCrossRef
118.
go back to reference Frank D, Doenecke D, Albig W. Differential expression of human replacement and cell cycle dependent H3 histone genes. Gene. 2003;312:135–43.PubMedCrossRef Frank D, Doenecke D, Albig W. Differential expression of human replacement and cell cycle dependent H3 histone genes. Gene. 2003;312:135–43.PubMedCrossRef
120.
go back to reference Fang JY, Richardson BC. The MAPK signalling pathways and colorectal cancer. Lancet Oncol. 2005;6(5):322–7.PubMedCrossRef Fang JY, Richardson BC. The MAPK signalling pathways and colorectal cancer. Lancet Oncol. 2005;6(5):322–7.PubMedCrossRef
121.
go back to reference Ayoubi HA, Mahjoubi F, Mirzaei R. Investigation of the human H3.3B (H3F3B) gene expression as a novel marker in patients with colorectal cancer. J Gastrointest Oncol. 2017;8(1):64–9.PubMedPubMedCentralCrossRef Ayoubi HA, Mahjoubi F, Mirzaei R. Investigation of the human H3.3B (H3F3B) gene expression as a novel marker in patients with colorectal cancer. J Gastrointest Oncol. 2017;8(1):64–9.PubMedPubMedCentralCrossRef
122.
go back to reference Wei YH, Lee HC. Oxidative stress, mitochondrial DNA mutation, and impairment of antioxidant enzymes in aging. Exp Biol Med (Maywood). 2002;227(9):671–82.CrossRef Wei YH, Lee HC. Oxidative stress, mitochondrial DNA mutation, and impairment of antioxidant enzymes in aging. Exp Biol Med (Maywood). 2002;227(9):671–82.CrossRef
123.
go back to reference Akouchekian M, et al. High rate of mutation in mitochondrial DNA displacement loop region in human colorectal cancer. Dis Colon Rectum. 2009;52(3):526–30.PubMedCrossRef Akouchekian M, et al. High rate of mutation in mitochondrial DNA displacement loop region in human colorectal cancer. Dis Colon Rectum. 2009;52(3):526–30.PubMedCrossRef
124.
go back to reference Sternberg D, et al. Exhaustive scanning approach to screen all the mitochondrial tRNA genes for mutations and its application to the investigation of 35 independent patients with mitochondrial disorders. Hum Mol Genet. 1998;7(1):33–42.PubMedCrossRef Sternberg D, et al. Exhaustive scanning approach to screen all the mitochondrial tRNA genes for mutations and its application to the investigation of 35 independent patients with mitochondrial disorders. Hum Mol Genet. 1998;7(1):33–42.PubMedCrossRef
125.
go back to reference Mohammed F, et al. Mitochondrial A12308G alteration in tRNA(Leu(CUN)) in colorectal cancer samples. Diagn Pathol. 2015;10:115.PubMedCrossRef Mohammed F, et al. Mitochondrial A12308G alteration in tRNA(Leu(CUN)) in colorectal cancer samples. Diagn Pathol. 2015;10:115.PubMedCrossRef
126.
go back to reference Kim YI. Folate and carcinogenesis: evidence, mechanisms, and implications. J Nutr Biochem. 1999;10(2):66–88.PubMedCrossRef Kim YI. Folate and carcinogenesis: evidence, mechanisms, and implications. J Nutr Biochem. 1999;10(2):66–88.PubMedCrossRef
127.
go back to reference Shrubsole MJ, et al. Dietary folate intake and breast cancer risk: results from the Shanghai breast Cancer study. Cancer Res. 2001;61(19):7136–41.PubMed Shrubsole MJ, et al. Dietary folate intake and breast cancer risk: results from the Shanghai breast Cancer study. Cancer Res. 2001;61(19):7136–41.PubMed
128.
go back to reference Haghighi MM, et al. Association between the 1793G> a MTHFR polymorphism and sporadic colorectal cancer in Iran. Asian Pac J Cancer Prev. 2008;9(4):659–62.PubMed Haghighi MM, et al. Association between the 1793G> a MTHFR polymorphism and sporadic colorectal cancer in Iran. Asian Pac J Cancer Prev. 2008;9(4):659–62.PubMed
129.
go back to reference McCann SE, et al. Diet in the epidemiology of endometrial cancer in western New York (United States). Cancer Causes Control. 2000;11(10):965–74.PubMedCrossRef McCann SE, et al. Diet in the epidemiology of endometrial cancer in western New York (United States). Cancer Causes Control. 2000;11(10):965–74.PubMedCrossRef
130.
go back to reference Rampersaud GC, et al. Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr. 2000;72(4):998–1003.PubMedCrossRef Rampersaud GC, et al. Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr. 2000;72(4):998–1003.PubMedCrossRef
131.
go back to reference Naghibalhossaini F, et al. MTHFR C677T and A1298C variant genotypes and the risk of microsatellite instability among Iranian colorectal cancer patients. Cancer Genet Cytogenet. 2010;197(2):142–51.PubMedPubMedCentralCrossRef Naghibalhossaini F, et al. MTHFR C677T and A1298C variant genotypes and the risk of microsatellite instability among Iranian colorectal cancer patients. Cancer Genet Cytogenet. 2010;197(2):142–51.PubMedPubMedCentralCrossRef
132.
go back to reference Leonard GD, Fojo T, Bates SE. The role of ABC transporters in clinical practice. Oncologist. 2003;8(5):411–24.PubMedCrossRef Leonard GD, Fojo T, Bates SE. The role of ABC transporters in clinical practice. Oncologist. 2003;8(5):411–24.PubMedCrossRef
133.
go back to reference Luqmani YA. Mechanisms of drug resistance in cancer chemotherapy. Med Princ Pract. 2005;14(Suppl 1):35–48.PubMedCrossRef Luqmani YA. Mechanisms of drug resistance in cancer chemotherapy. Med Princ Pract. 2005;14(Suppl 1):35–48.PubMedCrossRef
134.
go back to reference Petrova DT, et al. No association between MDR1 (ABCB1) 2677G>T and 3435C>T polymorphism and sporadic colorectal cancer among Bulgarian patients. J Cancer Res Clin Oncol. 2008;134(3):317–22.PubMedCrossRef Petrova DT, et al. No association between MDR1 (ABCB1) 2677G>T and 3435C>T polymorphism and sporadic colorectal cancer among Bulgarian patients. J Cancer Res Clin Oncol. 2008;134(3):317–22.PubMedCrossRef
135.
go back to reference Arceci RJ. Clinical significance of P-glycoprotein in multidrug resistance malignancies. Blood. 1993;81(9):2215–22.PubMed Arceci RJ. Clinical significance of P-glycoprotein in multidrug resistance malignancies. Blood. 1993;81(9):2215–22.PubMed
136.
go back to reference Samanian S, et al. MDR1 gene polymorphisms: possible association with its expression and clinicopathology characteristics in colorectal cancer patients. Asian Pac J Cancer Prev. 2011;12(11):3141–5.PubMed Samanian S, et al. MDR1 gene polymorphisms: possible association with its expression and clinicopathology characteristics in colorectal cancer patients. Asian Pac J Cancer Prev. 2011;12(11):3141–5.PubMed
137.
go back to reference Khedri A, et al. Association of the colorectal cancer and MDR1 gene polymorphism in an Iranian population. Mol Biol Rep. 2011;38(5):2939–43.PubMedCrossRef Khedri A, et al. Association of the colorectal cancer and MDR1 gene polymorphism in an Iranian population. Mol Biol Rep. 2011;38(5):2939–43.PubMedCrossRef
138.
go back to reference Ardalan Khales S, et al. SALL4 as a new biomarker for early colorectal cancers. J Cancer Res Clin Oncol. 2015;141(2):229–35.PubMedCrossRef Ardalan Khales S, et al. SALL4 as a new biomarker for early colorectal cancers. J Cancer Res Clin Oncol. 2015;141(2):229–35.PubMedCrossRef
139.
go back to reference Monk M, Hitchins M, Hawes S. Differential expression of the embryo/cancer gene ECSA (DPPA2), the cancer/testis gene BORIS and the pluripotency structural gene OCT4, in human preimplantation development. Mol Hum Reprod. 2008;14(6):347–55.PubMedCrossRef Monk M, Hitchins M, Hawes S. Differential expression of the embryo/cancer gene ECSA (DPPA2), the cancer/testis gene BORIS and the pluripotency structural gene OCT4, in human preimplantation development. Mol Hum Reprod. 2008;14(6):347–55.PubMedCrossRef
140.
go back to reference Monk M, Holding C. Human embryonic genes re-expressed in cancer cells. Oncogene. 2001;20(56):8085–91.PubMedCrossRef Monk M, Holding C. Human embryonic genes re-expressed in cancer cells. Oncogene. 2001;20(56):8085–91.PubMedCrossRef
141.
go back to reference Hutvagner G, Simard MJ. Argonaute proteins: key players in RNA silencing. Nat Rev Mol Cell Biol. 2008;9(1):22–32.PubMedCrossRef Hutvagner G, Simard MJ. Argonaute proteins: key players in RNA silencing. Nat Rev Mol Cell Biol. 2008;9(1):22–32.PubMedCrossRef
142.
143.
go back to reference Raeisossadati R, et al. Aberrant expression of DPPA2 and HIWI genes in colorectal cancer and their impacts on poor prognosis. Tumour Biol. 2014;35(6):5299–305.PubMedCrossRef Raeisossadati R, et al. Aberrant expression of DPPA2 and HIWI genes in colorectal cancer and their impacts on poor prognosis. Tumour Biol. 2014;35(6):5299–305.PubMedCrossRef
144.
145.
146.
go back to reference Mirzaei A, et al. Upregulation of circulating cancer stem cell marker, DCLK1 but not Lgr5, in chemoradiotherapy-treated colorectal cancer patients. Tumour Biol. 2015;36(6):4801–10.PubMedCrossRef Mirzaei A, et al. Upregulation of circulating cancer stem cell marker, DCLK1 but not Lgr5, in chemoradiotherapy-treated colorectal cancer patients. Tumour Biol. 2015;36(6):4801–10.PubMedCrossRef
147.
go back to reference Micalizzi DS, Ford HL. Epithelial-mesenchymal transition in development and cancer. Future Oncol. 2009;5(8):1129–43.PubMedCrossRef Micalizzi DS, Ford HL. Epithelial-mesenchymal transition in development and cancer. Future Oncol. 2009;5(8):1129–43.PubMedCrossRef
148.
go back to reference Ogrodnik M, et al. Dynamic JUNQ inclusion bodies are asymmetrically inherited in mammalian cell lines through the asymmetric partitioning of vimentin. Proc Natl Acad Sci U S A. 2014;111(22):8049–54.PubMedPubMedCentralCrossRef Ogrodnik M, et al. Dynamic JUNQ inclusion bodies are asymmetrically inherited in mammalian cell lines through the asymmetric partitioning of vimentin. Proc Natl Acad Sci U S A. 2014;111(22):8049–54.PubMedPubMedCentralCrossRef
149.
go back to reference Niknami Z, et al. The association of vimentin and fibronectin gene expression with epithelial-mesenchymal transition and tumor malignancy in colorectal carcinoma. EXCLI J. 2017;16:1009–17.PubMedPubMedCentral Niknami Z, et al. The association of vimentin and fibronectin gene expression with epithelial-mesenchymal transition and tumor malignancy in colorectal carcinoma. EXCLI J. 2017;16:1009–17.PubMedPubMedCentral
150.
go back to reference Hosseini SM, et al. Nebulette expression is associated with lymph node metastasis in patients with colorectal Cancer. Middle East J Dig Dis. 2018;10(3):174–9.PubMedPubMedCentralCrossRef Hosseini SM, et al. Nebulette expression is associated with lymph node metastasis in patients with colorectal Cancer. Middle East J Dig Dis. 2018;10(3):174–9.PubMedPubMedCentralCrossRef
152.
go back to reference Sandrock K, et al. Characterization of human septin interactions. Biol Chem. 2011;392(8–9):751–61.PubMed Sandrock K, et al. Characterization of human septin interactions. Biol Chem. 2011;392(8–9):751–61.PubMed
153.
go back to reference Behrouz Sharif S, et al. Detection of aberrant methylated SEPT9 and NTRK3 genes in sporadic colorectal cancer patients as a potential diagnostic biomarker. Oncol Lett. 2016;12(6):5335–43.PubMedPubMedCentralCrossRef Behrouz Sharif S, et al. Detection of aberrant methylated SEPT9 and NTRK3 genes in sporadic colorectal cancer patients as a potential diagnostic biomarker. Oncol Lett. 2016;12(6):5335–43.PubMedPubMedCentralCrossRef
154.
go back to reference Raeisossadati R, et al. Quantitative analysis of TEM-8 and CEA tumor markers indicating free tumor cells in the peripheral blood of colorectal cancer patients. Int J Color Dis. 2011;26(10):1265–70.CrossRef Raeisossadati R, et al. Quantitative analysis of TEM-8 and CEA tumor markers indicating free tumor cells in the peripheral blood of colorectal cancer patients. Int J Color Dis. 2011;26(10):1265–70.CrossRef
Metadata
Title
Genetic and molecular origins of colorectal Cancer among the Iranians: an update
Authors
Mohammad Reza Abbaszadegan
Meysam Moghbeli
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Diagnostic Pathology / Issue 1/2018
Electronic ISSN: 1746-1596
DOI
https://doi.org/10.1186/s13000-018-0774-0

Other articles of this Issue 1/2018

Diagnostic Pathology 1/2018 Go to the issue