Skip to main content
Top
Published in: Cancer Causes & Control 11/2012

01-11-2012 | Review Article

Estrogen in obesity-associated colon cancer: friend or foe? Protecting postmenopausal women but promoting late-stage colon cancer

Authors: Jiezhong Chen, Don Iverson

Published in: Cancer Causes & Control | Issue 11/2012

Login to get access

Abstract

Obesity is associated with the increased incidence of colon cancer. Many cancer risk factors have been identified including increased blood levels of insulin, leptin, interleukin-6, interleukin-17, tumor necrosis factor-alpha, and decreased blood levels of adiponectin. However, the role of blood levels of estrogen in obesity-associated colon cancer is controversial. Evidence showed that obesity affected men more strongly than women in the carcinogenesis of colon cancer, indicating protective effect of estrogen which is increased in obesity. However, an epidemiological study has also shown that endogenous estradiol level is an independent risk factor for colon cancer, positively associated with colon cancer after normalizing insulin, IGF-1. The controversial opinions may be caused by different effects of ER-alpha and ER-beta. ER-alpha can increase colon cancer cell proliferation and increase cancer incidence. ER-beta has the opposite effect to ER-alpha, and it causes apoptosis of colon cancer cells. The normal colonocytes mainly express ER-beta. Therefore, increased estrogen in obesity may have protective effect via ER-beta in obesity-associated colon cancer. However, with the development of colon cancer, ER-alpha is increased and ER-beta is decreased. In the late stage of colon cancer, estrogen may promote cancer development via ER-alpha. The different effects and expression of ER-alpha and ER-beta may explain the different results observed in several epidemiological studies as well as several animal experiments. Therefore, manipulation of estrogen-caused signal pathways to inhibit ER-alpha and stimulate ER-beta may have preventive and therapeutic effect for obesity-associated colon cancer.
Literature
1.
go back to reference Renehan AG et al (2008) Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet 371(9612):569–578PubMedCrossRef Renehan AG et al (2008) Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet 371(9612):569–578PubMedCrossRef
2.
go back to reference Calle EE, Kaaks R (2004) Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4(8):579–591PubMedCrossRef Calle EE, Kaaks R (2004) Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4(8):579–591PubMedCrossRef
3.
go back to reference Gislette T, Chen J (2010) The possible role of IL-17 in obesity-associated cancer. Sci World J 10:2265–2271CrossRef Gislette T, Chen J (2010) The possible role of IL-17 in obesity-associated cancer. Sci World J 10:2265–2271CrossRef
4.
go back to reference Renehan AG et al (2004) Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: systematic review and meta-regression analysis. Lancet 363(9418):1346–1353PubMedCrossRef Renehan AG et al (2004) Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: systematic review and meta-regression analysis. Lancet 363(9418):1346–1353PubMedCrossRef
5.
go back to reference Chen J et al (2011) Insulin decreases therapeutic efficacy in colon cancer cell line HT29 via the activation of the PI3 K/Akt pathway. Curr Drug Discov Technol 8(2):119–125PubMedCrossRef Chen J et al (2011) Insulin decreases therapeutic efficacy in colon cancer cell line HT29 via the activation of the PI3 K/Akt pathway. Curr Drug Discov Technol 8(2):119–125PubMedCrossRef
6.
go back to reference Paz-Filho G et al (2011) Associations between adipokines and obesity-related cancer. Front Biosci 16:1634–1650PubMedCrossRef Paz-Filho G et al (2011) Associations between adipokines and obesity-related cancer. Front Biosci 16:1634–1650PubMedCrossRef
7.
go back to reference Renehan A, Dive C (2011) Obesity, insulin and chemoresistance in colon cancer. J Gastrointest Oncol 2(1):8–10PubMed Renehan A, Dive C (2011) Obesity, insulin and chemoresistance in colon cancer. J Gastrointest Oncol 2(1):8–10PubMed
8.
go back to reference Chen J et al (2011) Insulin caused drug resistance to oxaliplatin in colon cancer cell HT29. J Gastrointest Oncol 2:27–33PubMed Chen J et al (2011) Insulin caused drug resistance to oxaliplatin in colon cancer cell HT29. J Gastrointest Oncol 2:27–33PubMed
9.
10.
go back to reference Huang XF, Chen J (2009) Obesity, the PI3 K/Akt signal pathway and colon cancer. Obes Rev 10(6):610–616PubMedCrossRef Huang XF, Chen J (2009) Obesity, the PI3 K/Akt signal pathway and colon cancer. Obes Rev 10(6):610–616PubMedCrossRef
11.
go back to reference Chen J (2012) Targeted therapy of obesity-associated colon cancer. Transl Gastrointest Cancer 1(1):44–57 Chen J (2012) Targeted therapy of obesity-associated colon cancer. Transl Gastrointest Cancer 1(1):44–57
12.
go back to reference Murphy TK et al (2000) Body mass index and colon cancer mortality in a large prospective study. Am J Epidemiol 152(9):847–854PubMedCrossRef Murphy TK et al (2000) Body mass index and colon cancer mortality in a large prospective study. Am J Epidemiol 152(9):847–854PubMedCrossRef
13.
go back to reference Ben Q et al (2012) Body mass index increases risk for colorectal adenomas based on meta-analysis. Gastroenterology 142(4):762–772PubMedCrossRef Ben Q et al (2012) Body mass index increases risk for colorectal adenomas based on meta-analysis. Gastroenterology 142(4):762–772PubMedCrossRef
14.
go back to reference Pischon T et al (2006) Body size and risk of colon and rectal cancer in the European Prospective Investigation Into Cancer and Nutrition (EPIC). J Natl Cancer Inst 98(13):920–931PubMedCrossRef Pischon T et al (2006) Body size and risk of colon and rectal cancer in the European Prospective Investigation Into Cancer and Nutrition (EPIC). J Natl Cancer Inst 98(13):920–931PubMedCrossRef
15.
go back to reference Kim BC et al (2012) Association of colorectal adenoma with components of metabolic syndrome. Cancer Causes Control 23(5):727–735PubMedCrossRef Kim BC et al (2012) Association of colorectal adenoma with components of metabolic syndrome. Cancer Causes Control 23(5):727–735PubMedCrossRef
16.
go back to reference Terry PD, Miller AB, Rohan TE (2002) Obesity and colorectal cancer risk in women. Gut 51(2):191–194PubMedCrossRef Terry PD, Miller AB, Rohan TE (2002) Obesity and colorectal cancer risk in women. Gut 51(2):191–194PubMedCrossRef
17.
go back to reference Hou L et al (2006) Body mass index and colon cancer risk in Chinese people: menopause as an effect modifier. Eur J Cancer 42(1):84–90PubMedCrossRef Hou L et al (2006) Body mass index and colon cancer risk in Chinese people: menopause as an effect modifier. Eur J Cancer 42(1):84–90PubMedCrossRef
18.
go back to reference Slattery ML et al (2001) Estrogens reduce and withdrawal of estrogens increase risk of microsatellite instability-positive colon cancer. Cancer Res 61(1):126–130PubMed Slattery ML et al (2001) Estrogens reduce and withdrawal of estrogens increase risk of microsatellite instability-positive colon cancer. Cancer Res 61(1):126–130PubMed
19.
go back to reference Sikalidis AK, Varamini B (2011) Roles of hormones and signalling molecules in describing the relationship between obesity and colon cancer. Pathol Oncol Res 17(4):785–790PubMedCrossRef Sikalidis AK, Varamini B (2011) Roles of hormones and signalling molecules in describing the relationship between obesity and colon cancer. Pathol Oncol Res 17(4):785–790PubMedCrossRef
20.
go back to reference Gunter MJ et al (2008) Insulin, insulin-like growth factor-I, endogenous estradiol, and risk of colorectal cancer in postmenopausal women. Cancer Res 68(1):329–337PubMedCrossRef Gunter MJ et al (2008) Insulin, insulin-like growth factor-I, endogenous estradiol, and risk of colorectal cancer in postmenopausal women. Cancer Res 68(1):329–337PubMedCrossRef
21.
go back to reference English MA et al (1999) Loss of estrogen inactivation in colonic cancer. J Clin Endocrinol Metab 84(6):2080–2085PubMedCrossRef English MA et al (1999) Loss of estrogen inactivation in colonic cancer. J Clin Endocrinol Metab 84(6):2080–2085PubMedCrossRef
22.
go back to reference Lin J et al (2004) Body mass index and risk of colorectal cancer in women (United States). Cancer Causes Control 15(6):581–589PubMedCrossRef Lin J et al (2004) Body mass index and risk of colorectal cancer in women (United States). Cancer Causes Control 15(6):581–589PubMedCrossRef
23.
go back to reference Di Domenico M et al (1996) Estradiol activation of human colon carcinoma-derived Caco-2 cell growth. Cancer Res 56(19):4516–4521PubMed Di Domenico M et al (1996) Estradiol activation of human colon carcinoma-derived Caco-2 cell growth. Cancer Res 56(19):4516–4521PubMed
24.
go back to reference Narayan S et al (1992) Estradiol is trophic for colon cancer in mice: effect on ornithine decarboxylase and c-myc messenger RNA. Gastroenterology 103(6):1823–1832PubMed Narayan S et al (1992) Estradiol is trophic for colon cancer in mice: effect on ornithine decarboxylase and c-myc messenger RNA. Gastroenterology 103(6):1823–1832PubMed
25.
go back to reference Rondini EA et al (2011) Energy balance modulates colon tumour growth: interactive roles of insulin and estrogen. Mol Carcinog 50(5):370–382PubMedCrossRef Rondini EA et al (2011) Energy balance modulates colon tumour growth: interactive roles of insulin and estrogen. Mol Carcinog 50(5):370–382PubMedCrossRef
26.
go back to reference Enmark E et al (1997) Human estrogen receptor beta-gene structure, chromosomal localization, and expression pattern. J Clin Endocrinol Metab 82(12):4258–4265PubMedCrossRef Enmark E et al (1997) Human estrogen receptor beta-gene structure, chromosomal localization, and expression pattern. J Clin Endocrinol Metab 82(12):4258–4265PubMedCrossRef
27.
go back to reference Menasce LP et al (1993) Localization of the estrogen receptor locus (ESR) to chromosome 6q25.1 by FISH and a simple post-FISH banding technique. Genomics 17(1):263–265PubMedCrossRef Menasce LP et al (1993) Localization of the estrogen receptor locus (ESR) to chromosome 6q25.1 by FISH and a simple post-FISH banding technique. Genomics 17(1):263–265PubMedCrossRef
28.
go back to reference Motylewska E, Stasikowska O, Melen-Mucha G (2009) The inhibitory effect of diarylpropionitrile, a selective agonist of estrogen receptor beta, on the growth of MC38 colon cancer line. Cancer Lett 276(1):68–73PubMedCrossRef Motylewska E, Stasikowska O, Melen-Mucha G (2009) The inhibitory effect of diarylpropionitrile, a selective agonist of estrogen receptor beta, on the growth of MC38 colon cancer line. Cancer Lett 276(1):68–73PubMedCrossRef
29.
go back to reference Greene GL et al (1986) Sequence and expression of human estrogen receptor complementary DNA. Science 231(4742):1150–1154PubMedCrossRef Greene GL et al (1986) Sequence and expression of human estrogen receptor complementary DNA. Science 231(4742):1150–1154PubMedCrossRef
30.
go back to reference Cho MA et al (2007) Expression and role of estrogen receptor alpha and beta in medullary thyroid carcinoma: different roles in cancer growth and apoptosis. J Endocrinol 195(2):255–263PubMedCrossRef Cho MA et al (2007) Expression and role of estrogen receptor alpha and beta in medullary thyroid carcinoma: different roles in cancer growth and apoptosis. J Endocrinol 195(2):255–263PubMedCrossRef
31.
go back to reference Chow SK, Chan JY, Fung KP (2004) Suppression of cell proliferation and regulation of estrogen receptor alpha signalling pathway by arsenic trioxide on human breast cancer MCF-7 cells. J Endocrinol 182(2):325–337PubMedCrossRef Chow SK, Chan JY, Fung KP (2004) Suppression of cell proliferation and regulation of estrogen receptor alpha signalling pathway by arsenic trioxide on human breast cancer MCF-7 cells. J Endocrinol 182(2):325–337PubMedCrossRef
32.
go back to reference Nilsson S et al (2001) Mechanisms of estrogen action. Physiol Rev 81(4):1535–1565PubMed Nilsson S et al (2001) Mechanisms of estrogen action. Physiol Rev 81(4):1535–1565PubMed
33.
go back to reference de Batistuzzo Medeiros SR (1997) Functional interactions between the estrogen receptor and the transcription activator Sp1 regulate the estrogen-dependent transcriptional activity of the vitellogenin A1 io promoter. J Biol Chem 272(29):18250–18260CrossRef de Batistuzzo Medeiros SR (1997) Functional interactions between the estrogen receptor and the transcription activator Sp1 regulate the estrogen-dependent transcriptional activity of the vitellogenin A1 io promoter. J Biol Chem 272(29):18250–18260CrossRef
34.
go back to reference Porter W et al (1997) Functional synergy between the transcription factor Sp1 and the estrogen receptor. Mol Endocrinol 11(11):1569–1580PubMedCrossRef Porter W et al (1997) Functional synergy between the transcription factor Sp1 and the estrogen receptor. Mol Endocrinol 11(11):1569–1580PubMedCrossRef
35.
go back to reference Qin C, Singh P, Safe S (1999) Transcriptional activation of insulin-like growth factor-binding protein-4 by 17beta-estradiol in MCF-7 cells: role of estrogen receptor-Sp1 complexes. Endocrinology 140(6):2501–2508PubMedCrossRef Qin C, Singh P, Safe S (1999) Transcriptional activation of insulin-like growth factor-binding protein-4 by 17beta-estradiol in MCF-7 cells: role of estrogen receptor-Sp1 complexes. Endocrinology 140(6):2501–2508PubMedCrossRef
36.
go back to reference Webb P et al (1999) The estrogen receptor enhances AP-1 activity by two distinct mechanisms with different requirements for receptor transactivation functions. Mol Endocrinol 13(10):1672–1685PubMedCrossRef Webb P et al (1999) The estrogen receptor enhances AP-1 activity by two distinct mechanisms with different requirements for receptor transactivation functions. Mol Endocrinol 13(10):1672–1685PubMedCrossRef
37.
go back to reference Wang C et al (2011) Estrogen induces c-myc gene expression via an upstream enhancer activated by the estrogen receptor and the AP-1 transcription factor. Mol Endocrinol 25(9):1527–1538PubMedCrossRef Wang C et al (2011) Estrogen induces c-myc gene expression via an upstream enhancer activated by the estrogen receptor and the AP-1 transcription factor. Mol Endocrinol 25(9):1527–1538PubMedCrossRef
38.
go back to reference Bjornstrom L, Sjoberg M (2005) Mechanisms of estrogen receptor signalling: convergence of genomic and nongenomic actions on target genes. Mol Endocrinol 19(4):833–842PubMedCrossRef Bjornstrom L, Sjoberg M (2005) Mechanisms of estrogen receptor signalling: convergence of genomic and nongenomic actions on target genes. Mol Endocrinol 19(4):833–842PubMedCrossRef
39.
go back to reference Hewitt SC et al (2005) Global uterine genomics in vivo: microarray evaluation of the estrogen receptor alpha-growth factor cross-talk mechanism. Mol Endocrinol 19(3):657–668PubMedCrossRef Hewitt SC et al (2005) Global uterine genomics in vivo: microarray evaluation of the estrogen receptor alpha-growth factor cross-talk mechanism. Mol Endocrinol 19(3):657–668PubMedCrossRef
40.
go back to reference Simoncini T et al (2000) Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature 407(6803):538–541PubMedCrossRef Simoncini T et al (2000) Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature 407(6803):538–541PubMedCrossRef
41.
go back to reference Le Romancer M et al (2008) Regulation of estrogen rapid signalling through arginine methylation by PRMT1. Mol Cell 31(2):212–221PubMedCrossRef Le Romancer M et al (2008) Regulation of estrogen rapid signalling through arginine methylation by PRMT1. Mol Cell 31(2):212–221PubMedCrossRef
42.
go back to reference Yang Z, Barnes CJ, Kumar R (2004) Human epidermal growth factor receptor 2 status modulates subcellular localization of and interaction with estrogen receptor alpha in breast cancer cells. Clin Cancer Res 10(11):3621–3628PubMedCrossRef Yang Z, Barnes CJ, Kumar R (2004) Human epidermal growth factor receptor 2 status modulates subcellular localization of and interaction with estrogen receptor alpha in breast cancer cells. Clin Cancer Res 10(11):3621–3628PubMedCrossRef
43.
go back to reference Duan R et al (2001) Estrogen receptor-mediated activation of the serum response element in MCF-7 cells through MAPK-dependent phosphorylation of Elk-1. J Biol Chem 276(15):11590–11598PubMedCrossRef Duan R et al (2001) Estrogen receptor-mediated activation of the serum response element in MCF-7 cells through MAPK-dependent phosphorylation of Elk-1. J Biol Chem 276(15):11590–11598PubMedCrossRef
44.
go back to reference Acconcia F et al (2005) Survival versus apoptotic 17beta-estradiol effect: role of ER alpha and ER beta activated non-genomic signalling. J Cell Physiol 203(1):193–201PubMedCrossRef Acconcia F et al (2005) Survival versus apoptotic 17beta-estradiol effect: role of ER alpha and ER beta activated non-genomic signalling. J Cell Physiol 203(1):193–201PubMedCrossRef
45.
go back to reference La Rosa P et al (2011) 17beta-Estradiol-induced cell proliferation requires estrogen receptor (ER) alpha monoubiquitination. Cell Signal 23(7):1128–1135PubMedCrossRef La Rosa P et al (2011) 17beta-Estradiol-induced cell proliferation requires estrogen receptor (ER) alpha monoubiquitination. Cell Signal 23(7):1128–1135PubMedCrossRef
46.
go back to reference Marino M, Acconcia F, Trentalance A (2003) Biphasic estradiol-induced AKT phosphorylation is modulated by PTEN via MAP kinase in HepG2 cells. Mol Biol Cell 14(6):2583–2591PubMedCrossRef Marino M, Acconcia F, Trentalance A (2003) Biphasic estradiol-induced AKT phosphorylation is modulated by PTEN via MAP kinase in HepG2 cells. Mol Biol Cell 14(6):2583–2591PubMedCrossRef
47.
go back to reference Song RX et al (2004) The role of Shc and insulin-like growth factor 1 receptor in mediating the translocation of estrogen receptor alpha to the plasma membrane. Proc Natl Acad Sci USA 101(7):2076–2081PubMedCrossRef Song RX et al (2004) The role of Shc and insulin-like growth factor 1 receptor in mediating the translocation of estrogen receptor alpha to the plasma membrane. Proc Natl Acad Sci USA 101(7):2076–2081PubMedCrossRef
48.
go back to reference Zhang Z et al (2004) The role of adapter protein Shc in estrogen non-genomic action. Steroids 69(8–9):523–529PubMedCrossRef Zhang Z et al (2004) The role of adapter protein Shc in estrogen non-genomic action. Steroids 69(8–9):523–529PubMedCrossRef
49.
go back to reference Fernando RI, Wimalasena J (2004) Estradiol abrogates apoptosis in breast cancer cells through inactivation of BAD: Ras-dependent nongenomic pathways requiring signalling through ERK and Akt. Mol Biol Cell 15(7):3266–3284PubMedCrossRef Fernando RI, Wimalasena J (2004) Estradiol abrogates apoptosis in breast cancer cells through inactivation of BAD: Ras-dependent nongenomic pathways requiring signalling through ERK and Akt. Mol Biol Cell 15(7):3266–3284PubMedCrossRef
50.
go back to reference Marino M et al (2002) Distinct nongenomic signal transduction pathways controlled by 17beta-estradiol regulate DNA synthesis and cyclin D(1) gene transcription in HepG2 cells. Mol Biol Cell 13(10):3720–3729PubMedCrossRef Marino M et al (2002) Distinct nongenomic signal transduction pathways controlled by 17beta-estradiol regulate DNA synthesis and cyclin D(1) gene transcription in HepG2 cells. Mol Biol Cell 13(10):3720–3729PubMedCrossRef
51.
go back to reference Bredfeldt TG et al (2010) Xenoestrogen-induced regulation of EZH2 and histone methylation via estrogen receptor signalling to PI3 K/AKT. Mol Endocrinol 24(5):993–1006PubMedCrossRef Bredfeldt TG et al (2010) Xenoestrogen-induced regulation of EZH2 and histone methylation via estrogen receptor signalling to PI3 K/AKT. Mol Endocrinol 24(5):993–1006PubMedCrossRef
52.
go back to reference Gustafsson JA (1999) Estrogen receptor beta—a new dimension in estrogen mechanism of action. J Endocrinol 163(3):379–383PubMedCrossRef Gustafsson JA (1999) Estrogen receptor beta—a new dimension in estrogen mechanism of action. J Endocrinol 163(3):379–383PubMedCrossRef
53.
go back to reference Kuiper GG et al (1996) Cloning of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci USA 93(12):5925–5930PubMedCrossRef Kuiper GG et al (1996) Cloning of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci USA 93(12):5925–5930PubMedCrossRef
54.
go back to reference Zhao C, Dahlman-Wright K, Gustafsson JA (2010) Estrogen signalling via estrogen receptor {beta}. J Biol Chem 285(51):39575–39579PubMedCrossRef Zhao C, Dahlman-Wright K, Gustafsson JA (2010) Estrogen signalling via estrogen receptor {beta}. J Biol Chem 285(51):39575–39579PubMedCrossRef
55.
go back to reference Galluzzo P et al (2007) Role of ERbeta palmitoylation in the inhibition of human colon cancer cell proliferation. Endocr Relat Cancer 14(1):153–167PubMedCrossRef Galluzzo P et al (2007) Role of ERbeta palmitoylation in the inhibition of human colon cancer cell proliferation. Endocr Relat Cancer 14(1):153–167PubMedCrossRef
56.
go back to reference Weige CC, Allred KF, Allred CD (2009) Estradiol alters cell growth in nonmalignant colonocytes and reduces the formation of preneoplastic lesions in the colon. Cancer Res 69(23):9118–9124PubMedCrossRef Weige CC, Allred KF, Allred CD (2009) Estradiol alters cell growth in nonmalignant colonocytes and reduces the formation of preneoplastic lesions in the colon. Cancer Res 69(23):9118–9124PubMedCrossRef
57.
go back to reference Edvardsson K et al (2011) Estrogen receptor beta induces antiinflammatory and antitumorigenic networks in colon cancer cells. Mol Endocrinol 25(6):969–979PubMedCrossRef Edvardsson K et al (2011) Estrogen receptor beta induces antiinflammatory and antitumorigenic networks in colon cancer cells. Mol Endocrinol 25(6):969–979PubMedCrossRef
58.
59.
go back to reference Dhanasekaran DN, Johnson GL (2007) MAPKs: function, regulation, role in cancer and therapeutic targeting. Oncogene 26(22):3097–3099PubMedCrossRef Dhanasekaran DN, Johnson GL (2007) MAPKs: function, regulation, role in cancer and therapeutic targeting. Oncogene 26(22):3097–3099PubMedCrossRef
60.
go back to reference Cuenda A, Rousseau S (2007) p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim Biophys Acta 1773(8):1358–1375PubMedCrossRef Cuenda A, Rousseau S (2007) p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim Biophys Acta 1773(8):1358–1375PubMedCrossRef
61.
go back to reference Porras A et al (2004) P38 alpha mitogen-activated protein kinase sensitizes cells to apoptosis induced by different stimuli. Mol Biol Cell 15(2):922–933PubMedCrossRef Porras A et al (2004) P38 alpha mitogen-activated protein kinase sensitizes cells to apoptosis induced by different stimuli. Mol Biol Cell 15(2):922–933PubMedCrossRef
62.
go back to reference Hui L et al (2007) p38alpha: a suppressor of cell proliferation and tumorigenesis. Cell Cycle 6(20):2429–2433PubMedCrossRef Hui L et al (2007) p38alpha: a suppressor of cell proliferation and tumorigenesis. Cell Cycle 6(20):2429–2433PubMedCrossRef
63.
go back to reference Shimada K et al (2003) Roles of p38- and c-jun NH2-terminal kinase-mediated pathways in 2-methoxyestradiol-induced p53 induction and apoptosis. Carcinogenesis 24(6):1067–1075PubMedCrossRef Shimada K et al (2003) Roles of p38- and c-jun NH2-terminal kinase-mediated pathways in 2-methoxyestradiol-induced p53 induction and apoptosis. Carcinogenesis 24(6):1067–1075PubMedCrossRef
64.
go back to reference Hui L et al (2007) p38alpha suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway. Nat Genet 39(6):741–749PubMedCrossRef Hui L et al (2007) p38alpha suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway. Nat Genet 39(6):741–749PubMedCrossRef
65.
go back to reference Iwasa H, Han J, Ishikawa F (2003) Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells 8(2):131–144PubMedCrossRef Iwasa H, Han J, Ishikawa F (2003) Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells 8(2):131–144PubMedCrossRef
66.
go back to reference Aguirre-Ghiso JA, Ossowski L, Rosenbaum SK (2004) Green fluorescent protein tagging of extracellular signal-regulated kinase and p38 pathways reveals novel dynamics of pathway activation during primary and metastatic growth. Cancer Res 64(20):7336–7345PubMedCrossRef Aguirre-Ghiso JA, Ossowski L, Rosenbaum SK (2004) Green fluorescent protein tagging of extracellular signal-regulated kinase and p38 pathways reveals novel dynamics of pathway activation during primary and metastatic growth. Cancer Res 64(20):7336–7345PubMedCrossRef
67.
go back to reference Feng Y, Wen J, Chang CC (2009) p38 Mitogen-activated protein kinase and hematologic malignancies. Arch Pathol Lab Med 133(11):1850–1856PubMed Feng Y, Wen J, Chang CC (2009) p38 Mitogen-activated protein kinase and hematologic malignancies. Arch Pathol Lab Med 133(11):1850–1856PubMed
68.
go back to reference Gompel A et al (2004) Steroidal hormones and proliferation, differentiation and apoptosis in breast cells. Maturitas 49(1):16–24PubMedCrossRef Gompel A et al (2004) Steroidal hormones and proliferation, differentiation and apoptosis in breast cells. Maturitas 49(1):16–24PubMedCrossRef
69.
go back to reference Lin JH et al (2012) Postmenopausal hormone therapy is associated with a reduced risk of colorectal cancer lacking CDKN1A expression. Cancer Res 72(12):3020–3028PubMedCrossRef Lin JH et al (2012) Postmenopausal hormone therapy is associated with a reduced risk of colorectal cancer lacking CDKN1A expression. Cancer Res 72(12):3020–3028PubMedCrossRef
70.
go back to reference Tanaka Y et al (2008) Medroxyprogesterone acetate inhibits proliferation of colon cancer cell lines by modulating cell cycle-related protein expression. Menopause 15(3):442–453PubMedCrossRef Tanaka Y et al (2008) Medroxyprogesterone acetate inhibits proliferation of colon cancer cell lines by modulating cell cycle-related protein expression. Menopause 15(3):442–453PubMedCrossRef
71.
go back to reference Gizard F et al (2005) Progesterone inhibits human breast cancer cell growth through transcriptional upregulation of the cyclin-dependent kinase inhibitor p27Kip1 gene. FEBS Lett 579(25):5535–5541PubMedCrossRef Gizard F et al (2005) Progesterone inhibits human breast cancer cell growth through transcriptional upregulation of the cyclin-dependent kinase inhibitor p27Kip1 gene. FEBS Lett 579(25):5535–5541PubMedCrossRef
72.
go back to reference Konstantinopoulos PA et al (2003) Oestrogen receptor beta (ERbeta) is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer 39(9):1251–1258PubMedCrossRef Konstantinopoulos PA et al (2003) Oestrogen receptor beta (ERbeta) is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer 39(9):1251–1258PubMedCrossRef
73.
go back to reference Qiu Y et al (2002) Oestrogen-induced apoptosis in colonocytes expressing oestrogen receptor beta. J Endocrinol 174(3):369–377PubMedCrossRef Qiu Y et al (2002) Oestrogen-induced apoptosis in colonocytes expressing oestrogen receptor beta. J Endocrinol 174(3):369–377PubMedCrossRef
74.
go back to reference Jassam N et al (2005) Loss of expression of oestrogen receptor beta in colon cancer and its association with Dukes’ staging. Oncol Rep 14(1):17–21PubMed Jassam N et al (2005) Loss of expression of oestrogen receptor beta in colon cancer and its association with Dukes’ staging. Oncol Rep 14(1):17–21PubMed
75.
go back to reference Fang YJ et al (2010) Prognostic impact of ERbeta and MMP7 expression on overall survival in colon cancer. Tumour Biol 31(6):651–658PubMedCrossRef Fang YJ et al (2010) Prognostic impact of ERbeta and MMP7 expression on overall survival in colon cancer. Tumour Biol 31(6):651–658PubMedCrossRef
76.
go back to reference Wong NA et al (2005) ERbeta isoform expression in colorectal carcinoma: an in vivo and in vitro study of clinicopathological and molecular correlates. J Pathol 207(1):53–60PubMedCrossRef Wong NA et al (2005) ERbeta isoform expression in colorectal carcinoma: an in vivo and in vitro study of clinicopathological and molecular correlates. J Pathol 207(1):53–60PubMedCrossRef
77.
go back to reference Wu AH et al (2010) Hormone therapy, DNA methylation and colon cancer. Carcinogenesis 31(6):1060–1067PubMedCrossRef Wu AH et al (2010) Hormone therapy, DNA methylation and colon cancer. Carcinogenesis 31(6):1060–1067PubMedCrossRef
78.
go back to reference Lin KJ et al (2012) The effect of estrogen vs. combined estrogen-progestogen therapy on the risk of colorectal cancer. Int J Cancer 130(2):419–430PubMedCrossRef Lin KJ et al (2012) The effect of estrogen vs. combined estrogen-progestogen therapy on the risk of colorectal cancer. Int J Cancer 130(2):419–430PubMedCrossRef
79.
go back to reference Hiraoka S et al (2010) Methylation status of normal background mucosa is correlated with occurrence and development of neoplasia in the distal colon. Hum Pathol 41(1):38–47PubMedCrossRef Hiraoka S et al (2010) Methylation status of normal background mucosa is correlated with occurrence and development of neoplasia in the distal colon. Hum Pathol 41(1):38–47PubMedCrossRef
80.
go back to reference Anghel A et al (2010) Estrogen receptor alpha polymorphisms and the risk of malignancies. Pathol Oncol Res 16(4):485–496PubMedCrossRef Anghel A et al (2010) Estrogen receptor alpha polymorphisms and the risk of malignancies. Pathol Oncol Res 16(4):485–496PubMedCrossRef
81.
go back to reference Worthley DL et al (2010) DNA methylation within the normal colorectal mucosa is associated with pathway-specific predisposition to cancer. Oncogene 29(11):1653–1662PubMedCrossRef Worthley DL et al (2010) DNA methylation within the normal colorectal mucosa is associated with pathway-specific predisposition to cancer. Oncogene 29(11):1653–1662PubMedCrossRef
82.
go back to reference Honma et al (2012) Estrogen receptor-beta gene polymorphism and colorectal cancer risk: effect modified by body mass index and isoflavone intake. Int J Cancer Honma et al (2012) Estrogen receptor-beta gene polymorphism and colorectal cancer risk: effect modified by body mass index and isoflavone intake. Int J Cancer
83.
go back to reference Saleiro et al (2012) Estrogen receptor-beta protects against colitis-associated neoplasia in mice. Int J Cancer Saleiro et al (2012) Estrogen receptor-beta protects against colitis-associated neoplasia in mice. Int J Cancer
84.
go back to reference Gallo D et al (2012) Estrogen receptor beta in cancer: an attractive target for therapy. Curr Pharm Des 18(19):2734–2757PubMedCrossRef Gallo D et al (2012) Estrogen receptor beta in cancer: an attractive target for therapy. Curr Pharm Des 18(19):2734–2757PubMedCrossRef
85.
go back to reference Yi KW et al (2008) Role of estrogen receptor-alpha and -beta in regulating leptin expression in 3T3-L1 adipocytes. Obes (Silver Spring) 16(11):2393–2399CrossRef Yi KW et al (2008) Role of estrogen receptor-alpha and -beta in regulating leptin expression in 3T3-L1 adipocytes. Obes (Silver Spring) 16(11):2393–2399CrossRef
86.
go back to reference Nejati-Koshki et al (2012) Inhibition of leptin gene expression and secretion by silibinin: possible role of estrogen receptors. Cytotechnology Nejati-Koshki et al (2012) Inhibition of leptin gene expression and secretion by silibinin: possible role of estrogen receptors. Cytotechnology
87.
go back to reference Mendoza RA et al (2011) Interactions between IGF-I, estrogen receptor-alpha (ERalpha), and ERbeta in regulating growth/apoptosis of MCF-7 human breast cancer cells. J Endocrinol 208(1):1–9PubMedCrossRef Mendoza RA et al (2011) Interactions between IGF-I, estrogen receptor-alpha (ERalpha), and ERbeta in regulating growth/apoptosis of MCF-7 human breast cancer cells. J Endocrinol 208(1):1–9PubMedCrossRef
88.
go back to reference Harris DM et al (2005) Phytoestrogens induce differential estrogen receptor alpha- or Beta-mediated responses in transfected breast cancer cells. Exp Biol Med (Maywood) 230(8):558–568 Harris DM et al (2005) Phytoestrogens induce differential estrogen receptor alpha- or Beta-mediated responses in transfected breast cancer cells. Exp Biol Med (Maywood) 230(8):558–568
89.
go back to reference Schleipen B et al (2011) ERbeta-specific agonists and genistein inhibit proliferation and induce apoptosis in the large and small intestine. Carcinogenesis 32(11):1675–1683PubMedCrossRef Schleipen B et al (2011) ERbeta-specific agonists and genistein inhibit proliferation and induce apoptosis in the large and small intestine. Carcinogenesis 32(11):1675–1683PubMedCrossRef
90.
go back to reference Bielecki A et al (2011) Estrogen receptor-beta mediates the inhibition of DLD-1 human colon adenocarcinoma cells by soy isoflavones. Nutr Cancer 63(1):139–150PubMed Bielecki A et al (2011) Estrogen receptor-beta mediates the inhibition of DLD-1 human colon adenocarcinoma cells by soy isoflavones. Nutr Cancer 63(1):139–150PubMed
91.
go back to reference Cotterchio M et al (2006) Dietary phytoestrogen intake is associated with reduced colorectal cancer risk. J Nutr 136(12):3046–3053PubMed Cotterchio M et al (2006) Dietary phytoestrogen intake is associated with reduced colorectal cancer risk. J Nutr 136(12):3046–3053PubMed
92.
go back to reference Yang G et al (2009) Prospective cohort study of soy food intake and colorectal cancer risk in women. Am J Clin Nutr 89(2):577–583PubMedCrossRef Yang G et al (2009) Prospective cohort study of soy food intake and colorectal cancer risk in women. Am J Clin Nutr 89(2):577–583PubMedCrossRef
93.
go back to reference Tu Z et al (2012) Estrogen receptor beta potentiates the antiproliferative effect of raloxifene and affects the cell migration and invasion in HCT-116 colon cancer cells. J Cancer Res Clin Oncol 138(7):1091–1103PubMedCrossRef Tu Z et al (2012) Estrogen receptor beta potentiates the antiproliferative effect of raloxifene and affects the cell migration and invasion in HCT-116 colon cancer cells. J Cancer Res Clin Oncol 138(7):1091–1103PubMedCrossRef
Metadata
Title
Estrogen in obesity-associated colon cancer: friend or foe? Protecting postmenopausal women but promoting late-stage colon cancer
Authors
Jiezhong Chen
Don Iverson
Publication date
01-11-2012
Publisher
Springer Netherlands
Published in
Cancer Causes & Control / Issue 11/2012
Print ISSN: 0957-5243
Electronic ISSN: 1573-7225
DOI
https://doi.org/10.1007/s10552-012-0066-z

Other articles of this Issue 11/2012

Cancer Causes & Control 11/2012 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine