Skip to main content
Top
Published in: Discover Oncology 1/2024

Open Access 01-12-2024 | Colorectal Cancer | Review

Delineating the role of nuclear receptors in colorectal cancer, a focused review

Authors: Mukesh Kumar Manickasamy, Sujitha Jayaprakash, Sosmitha Girisa, Aviral Kumar, Hiu Yan Lam, Elena Okina, Huiyan Eng, Mohammed S. Alqahtani, Mohamed Abbas, Gautam Sethi, Alan Prem Kumar, Ajaikumar B. Kunnumakkara

Published in: Discover Oncology | Issue 1/2024

Login to get access

Abstract

Colorectal cancer (CRC) stands as one of the most prevalent form of cancer globally, causing a significant number of deaths, surpassing 0.9 million in the year 2020. According to GLOBOCAN 2020, CRC ranks third in incidence and second in mortality in both males and females. Despite extensive studies over the years, there is still a need to establish novel therapeutic targets to enhance the patients’ survival rate in CRC. Nuclear receptors (NRs) are ligand-activated transcription factors (TFs) that regulate numerous essential biological processes such as differentiation, development, physiology, reproduction, and cellular metabolism. Dysregulation and anomalous expression of different NRs has led to multiple alterations, such as impaired signaling cascades, mutations, and epigenetic changes, leading to various diseases, including cancer. It has been observed that differential expression of various NRs might lead to the initiation and progression of CRC, and are correlated with poor survival outcomes in CRC patients. Despite numerous studies on the mechanism and role of NRs in this cancer, it remains of significant scientific interest primarily due to the diverse functions that various NRs exhibit in regulating key hallmarks of this cancer. Thus, modulating the expression of NRs with their agonists and antagonists, based on their expression levels, holds an immense prospect in the diagnosis, prognosis, and therapeutical modalities of CRC. In this review, we primarily focus on the role and mechanism of NRs in the pathogenesis of CRC and emphasized the significance of targeting these NRs using a variety of agents, which may represent a novel and effective strategy for the prevention and treatment of this cancer.

Graphical Abstract

Literature
5.
go back to reference Clinton SK, Giovannucci EL, Hursting SD. The world cancer research fund/American institute for cancer research third expert report on diet, nutrition, physical activity, and cancer: impact and future directions. J Nutr. 2020;150:663–71.PubMedCrossRef Clinton SK, Giovannucci EL, Hursting SD. The world cancer research fund/American institute for cancer research third expert report on diet, nutrition, physical activity, and cancer: impact and future directions. J Nutr. 2020;150:663–71.PubMedCrossRef
7.
go back to reference Grassadonia A, Di Marino P, Ficorella C, Cortellini A, Cannita K, Parisi A, Gamucci T, Zoratto F, Vici P, Barba M, et al. Impact of primary tumor location in patients with RAS wild-type metastatic colon cancer treated with first-line chemotherapy plus anti-EGFR or anti-VEGF monoclonal antibodies: a retrospective multicenter study. J Cancer. 2019;10:5926–34. https://doi.org/10.7150/jca.34550.CrossRefPubMedPubMedCentral Grassadonia A, Di Marino P, Ficorella C, Cortellini A, Cannita K, Parisi A, Gamucci T, Zoratto F, Vici P, Barba M, et al. Impact of primary tumor location in patients with RAS wild-type metastatic colon cancer treated with first-line chemotherapy plus anti-EGFR or anti-VEGF monoclonal antibodies: a retrospective multicenter study. J Cancer. 2019;10:5926–34. https://​doi.​org/​10.​7150/​jca.​34550.CrossRefPubMedPubMedCentral
18.
go back to reference Kunnumakkara AB, Sung B, Ravindran J, Diagaradjane P, Deorukhkar A, Dey S, Koca C, Tong Z, Gelovani JG, Guha S, et al. Zyflamend suppresses growth and sensitizes human pancreatic tumors to gemcitabine in an orthotopic mouse model through modulation of multiple targets. Int J Cancer. 2012;131:E292-303. https://doi.org/10.1002/ijc.26442.CrossRefPubMed Kunnumakkara AB, Sung B, Ravindran J, Diagaradjane P, Deorukhkar A, Dey S, Koca C, Tong Z, Gelovani JG, Guha S, et al. Zyflamend suppresses growth and sensitizes human pancreatic tumors to gemcitabine in an orthotopic mouse model through modulation of multiple targets. Int J Cancer. 2012;131:E292-303. https://​doi.​org/​10.​1002/​ijc.​26442.CrossRefPubMed
19.
go back to reference Padmavathi G, Rathnakaram SR, Monisha J, Bordoloi D, Roy NK, Kunnumakkara AB. Potential of butein, a tetrahydroxychalcone to obliterate cancer. Phytomedicine. 2015;22:1163–71.PubMedCrossRef Padmavathi G, Rathnakaram SR, Monisha J, Bordoloi D, Roy NK, Kunnumakkara AB. Potential of butein, a tetrahydroxychalcone to obliterate cancer. Phytomedicine. 2015;22:1163–71.PubMedCrossRef
20.
go back to reference Henamayee S, Banik K, Sailo BL, Shabnam B, Harsha C, Srilakshmi S, Vgm N, Baek SH, Ahn KS, Kunnumakkara AB. Therapeutic emergence of rhein as a potential anticancer drug: a review of its molecular targets and anticancer properties. Molecules. 2020;25:2278.PubMedPubMedCentralCrossRef Henamayee S, Banik K, Sailo BL, Shabnam B, Harsha C, Srilakshmi S, Vgm N, Baek SH, Ahn KS, Kunnumakkara AB. Therapeutic emergence of rhein as a potential anticancer drug: a review of its molecular targets and anticancer properties. Molecules. 2020;25:2278.PubMedPubMedCentralCrossRef
21.
go back to reference Nair A, Amalraj A, Jacob J, Kunnumakkara AB, Gopi S. Non-curcuminoids from turmeric and their potential in cancer therapy and anticancer drug delivery formulations. Biomolecules. 2019;9:13.PubMedPubMedCentralCrossRef Nair A, Amalraj A, Jacob J, Kunnumakkara AB, Gopi S. Non-curcuminoids from turmeric and their potential in cancer therapy and anticancer drug delivery formulations. Biomolecules. 2019;9:13.PubMedPubMedCentralCrossRef
22.
go back to reference Singh YP, Girisa S, Banik K, Ghosh S, Swathi P, Deka M, Padmavathi G, Kotoky J, Sethi G, Fan L. Potential application of zerumbone in the prevention and therapy of chronic human diseases. J Funct Foods. 2019;53:248–58.CrossRef Singh YP, Girisa S, Banik K, Ghosh S, Swathi P, Deka M, Padmavathi G, Kotoky J, Sethi G, Fan L. Potential application of zerumbone in the prevention and therapy of chronic human diseases. J Funct Foods. 2019;53:248–58.CrossRef
23.
go back to reference Devi Daimary U, Girisa S, Parama D, Verma E, Kumar A, Kunnumakkara AB. Embelin: A novel XIAP inhibitor for the prevention and treatment of chronic diseases. J Biochem Mol Toxicol. 2022;36:e22950.PubMedCrossRef Devi Daimary U, Girisa S, Parama D, Verma E, Kumar A, Kunnumakkara AB. Embelin: A novel XIAP inhibitor for the prevention and treatment of chronic diseases. J Biochem Mol Toxicol. 2022;36:e22950.PubMedCrossRef
26.
go back to reference Cassidy J, Tabernero J, Twelves C, Brunet R, Butts C, Conroy T, Debraud F, Figer A, Grossmann J, Sawada N. XELOX (capecitabine plus oxaliplatin): active first-line therapy for patients with metastatic colorectal cancer. J Clin Oncol. 2004;22:2084–91.PubMedCrossRef Cassidy J, Tabernero J, Twelves C, Brunet R, Butts C, Conroy T, Debraud F, Figer A, Grossmann J, Sawada N. XELOX (capecitabine plus oxaliplatin): active first-line therapy for patients with metastatic colorectal cancer. J Clin Oncol. 2004;22:2084–91.PubMedCrossRef
27.
go back to reference Colucci G, Gebbia V, Paoletti G, Giuliani F, Caruso M, Gebbia N, Cartenì G, Agostara B, Pezzella G, Manzione L. Phase III randomized trial of FOLFIRI versus FOLFOX4 in the treatment of advanced colorectal cancer: a multicenter study of the Gruppo Oncologico Dell’Italia Meridionale. J Clin Oncol. 2005;23:4866–75.PubMedCrossRef Colucci G, Gebbia V, Paoletti G, Giuliani F, Caruso M, Gebbia N, Cartenì G, Agostara B, Pezzella G, Manzione L. Phase III randomized trial of FOLFIRI versus FOLFOX4 in the treatment of advanced colorectal cancer: a multicenter study of the Gruppo Oncologico Dell’Italia Meridionale. J Clin Oncol. 2005;23:4866–75.PubMedCrossRef
28.
go back to reference Goldberg RM, Sargent DJ, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK, Findlay BP, Pitot HC, Alberts SR. A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol. 2004;22:23–30.PubMedCrossRef Goldberg RM, Sargent DJ, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK, Findlay BP, Pitot HC, Alberts SR. A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol. 2004;22:23–30.PubMedCrossRef
31.
go back to reference Kunnumakkara AB, Bordoloi D, Sailo BL, Roy NK, Thakur KK, Banik K, Shakibaei M, Gupta SC, Aggarwal BB. Cancer drug development: The missing links. Exp Biol Med. 2019;244:663–89.CrossRef Kunnumakkara AB, Bordoloi D, Sailo BL, Roy NK, Thakur KK, Banik K, Shakibaei M, Gupta SC, Aggarwal BB. Cancer drug development: The missing links. Exp Biol Med. 2019;244:663–89.CrossRef
37.
go back to reference Huggins C, Hodges CV. Studies on prostatic cancer: I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA Cancer J Clin. 1972;22:232–40.PubMedCrossRef Huggins C, Hodges CV. Studies on prostatic cancer: I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA Cancer J Clin. 1972;22:232–40.PubMedCrossRef
40.
go back to reference Thomas C, Gustafsson J-Å. Estrogen receptor mutations and functional consequences for breast cancer. Trends Endocrinol Metab. 2015;26:467–76.PubMedCrossRef Thomas C, Gustafsson J-Å. Estrogen receptor mutations and functional consequences for breast cancer. Trends Endocrinol Metab. 2015;26:467–76.PubMedCrossRef
41.
go back to reference Zhang Y, Hagedorn CH, Wang L. Role of nuclear receptor SHP in metabolism and cancer. Biochim Biophys Acta. 2011;1812:893–908.PubMedCrossRef Zhang Y, Hagedorn CH, Wang L. Role of nuclear receptor SHP in metabolism and cancer. Biochim Biophys Acta. 2011;1812:893–908.PubMedCrossRef
44.
go back to reference Girisa S, Rana V, Parama D, Dutta U, Kunnumakkara AB. Differential roles of farnesoid X receptor (FXR) in modulating apoptosis in cancer cells. Adv Protein Chem Struct Biol. 2021;126:63–90.PubMedCrossRef Girisa S, Rana V, Parama D, Dutta U, Kunnumakkara AB. Differential roles of farnesoid X receptor (FXR) in modulating apoptosis in cancer cells. Adv Protein Chem Struct Biol. 2021;126:63–90.PubMedCrossRef
47.
go back to reference Alberg AJ, Gordon GB, Hoffman SC, Comstock GW, Helzlsouer KJ. Serum dehydroepiandrosterone and dehydroepiandrosterone sulfate and the subsequent risk of developing colon cancer. Cancer Epidemiol Biomark Prev. 2000;9:517–21. Alberg AJ, Gordon GB, Hoffman SC, Comstock GW, Helzlsouer KJ. Serum dehydroepiandrosterone and dehydroepiandrosterone sulfate and the subsequent risk of developing colon cancer. Cancer Epidemiol Biomark Prev. 2000;9:517–21.
48.
go back to reference Chlebowski RT, Wactawski-Wende J, Ritenbaugh C, Hubbell FA, Ascensao J, Rodabough RJ, Rosenberg CA, Taylor VM, Harris R, Chen C. Estrogen plus progestin and colorectal cancer in postmenopausal women. N Engl J Med. 2004;350:991–1004.PubMedCrossRef Chlebowski RT, Wactawski-Wende J, Ritenbaugh C, Hubbell FA, Ascensao J, Rodabough RJ, Rosenberg CA, Taylor VM, Harris R, Chen C. Estrogen plus progestin and colorectal cancer in postmenopausal women. N Engl J Med. 2004;350:991–1004.PubMedCrossRef
49.
go back to reference Slattery ML, Sweeney C, Murtaugh M, Ma KN, Wolff RK, Potter JD, Caan BJ, Samowitz W. Associations between ERα, ERβ, and AR genotypes and colon and rectal cancer. Cancer Epidemiol Biomark Prev. 2005;14:2936–42.CrossRef Slattery ML, Sweeney C, Murtaugh M, Ma KN, Wolff RK, Potter JD, Caan BJ, Samowitz W. Associations between ERα, ERβ, and AR genotypes and colon and rectal cancer. Cancer Epidemiol Biomark Prev. 2005;14:2936–42.CrossRef
50.
go back to reference Anagnostopoulou V, Pediaditakis I, Alkahtani S, Alarifi SA, Schmidt EM, Lang F, Gravanis A, Charalampopoulos I, Stournaras C. Differential effects of dehydroepiandrosterone and testosterone in prostate and colon cancer cell apoptosis: the role of nerve growth factor (NGF) receptors. Endocrinology. 2013;154:2446–56. https://doi.org/10.1210/en.2012-2249.CrossRefPubMed Anagnostopoulou V, Pediaditakis I, Alkahtani S, Alarifi SA, Schmidt EM, Lang F, Gravanis A, Charalampopoulos I, Stournaras C. Differential effects of dehydroepiandrosterone and testosterone in prostate and colon cancer cell apoptosis: the role of nerve growth factor (NGF) receptors. Endocrinology. 2013;154:2446–56. https://​doi.​org/​10.​1210/​en.​2012-2249.CrossRefPubMed
51.
go back to reference Amos-Landgraf JM, Heijmans J, Wielenga MC, Dunkin E, Krentz KJ, Clipson L, Ederveen AG, Groothuis PG, Mosselman S, Muncan V. Sex disparity in colonic adenomagenesis involves promotion by male hormones, not protection by female hormones. Proc Natl Acad Sci. 2014;111:16514–9.ADSPubMedPubMedCentralCrossRef Amos-Landgraf JM, Heijmans J, Wielenga MC, Dunkin E, Krentz KJ, Clipson L, Ederveen AG, Groothuis PG, Mosselman S, Muncan V. Sex disparity in colonic adenomagenesis involves promotion by male hormones, not protection by female hormones. Proc Natl Acad Sci. 2014;111:16514–9.ADSPubMedPubMedCentralCrossRef
59.
go back to reference Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835.PubMedPubMedCentralCrossRef Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835.PubMedPubMedCentralCrossRef
62.
go back to reference Jayaprakash S, Hegde M, Girisa S, Alqahtani MS, Abbas M, Lee EHC, Yap KC-H, Sethi G, Kumar AP, Kunnumakkara AB. Demystifying the functional role of nuclear receptors in esophageal cancer. Int J Mol Sci. 2022;23:10952.PubMedPubMedCentralCrossRef Jayaprakash S, Hegde M, Girisa S, Alqahtani MS, Abbas M, Lee EHC, Yap KC-H, Sethi G, Kumar AP, Kunnumakkara AB. Demystifying the functional role of nuclear receptors in esophageal cancer. Int J Mol Sci. 2022;23:10952.PubMedPubMedCentralCrossRef
64.
go back to reference O’Malley BW, Tsai M-J. Molecular pathways of steroid receptor action. Biol Reprod. 1992;46:163–7.PubMedCrossRef O’Malley BW, Tsai M-J. Molecular pathways of steroid receptor action. Biol Reprod. 1992;46:163–7.PubMedCrossRef
65.
go back to reference De Bosscher K, Desmet SJ, Clarisse D, Estébanez-Perpiña E, Brunsveld L. Nuclear receptor crosstalk—defining the mechanisms for therapeutic innovation. Nat Rev Endocrinol. 2020;16:363–77.PubMedCrossRef De Bosscher K, Desmet SJ, Clarisse D, Estébanez-Perpiña E, Brunsveld L. Nuclear receptor crosstalk—defining the mechanisms for therapeutic innovation. Nat Rev Endocrinol. 2020;16:363–77.PubMedCrossRef
76.
go back to reference Zhou ZW, Wan DS, Wang GQ, Pan ZZ, Lu HP, Gao JH, Ding PR. Expression of estrogen receptor and progesterone receptor in colorectal cancer: a quantitative study. Ai Zheng. 2004;23:851–4.PubMed Zhou ZW, Wan DS, Wang GQ, Pan ZZ, Lu HP, Gao JH, Ding PR. Expression of estrogen receptor and progesterone receptor in colorectal cancer: a quantitative study. Ai Zheng. 2004;23:851–4.PubMed
77.
go back to reference Nüssler NC, Reinbacher K, Shanny N, Schirmeier A, Glanemann M, Neuhaus P, Nussler AK, Kirschner M. Sex-specific differences in the expression levels of estrogen receptor subtypes in colorectal cancer. Gend Med. 2008;5:209–17.PubMedCrossRef Nüssler NC, Reinbacher K, Shanny N, Schirmeier A, Glanemann M, Neuhaus P, Nussler AK, Kirschner M. Sex-specific differences in the expression levels of estrogen receptor subtypes in colorectal cancer. Gend Med. 2008;5:209–17.PubMedCrossRef
78.
go back to reference Jiang HP, Teng RY, Wang Q, Zhang X, Wang HH, Cao J, Teng LS. Estrogen receptor alpha variant ERalpha46 mediates growth inhibition and apoptosis of human HT-29 colon adenocarcinoma cells in the presence of 17beta-oestradiol. Chin Med J (Engl). 2008;121:1025–31.PubMedCrossRef Jiang HP, Teng RY, Wang Q, Zhang X, Wang HH, Cao J, Teng LS. Estrogen receptor alpha variant ERalpha46 mediates growth inhibition and apoptosis of human HT-29 colon adenocarcinoma cells in the presence of 17beta-oestradiol. Chin Med J (Engl). 2008;121:1025–31.PubMedCrossRef
79.
go back to reference Jiang L, Fei H, Yang A, Zhu J, Sun J, Liu X, Xu W, Yang J, Zhang S. Estrogen inhibits the growth of colon cancer in mice through reversing extracellular vesicle-mediated immunosuppressive tumor microenvironment. Cancer Lett. 2021;520:332–43.PubMedCrossRef Jiang L, Fei H, Yang A, Zhu J, Sun J, Liu X, Xu W, Yang J, Zhang S. Estrogen inhibits the growth of colon cancer in mice through reversing extracellular vesicle-mediated immunosuppressive tumor microenvironment. Cancer Lett. 2021;520:332–43.PubMedCrossRef
80.
go back to reference Pereira MA, Tao L, Wang W, Li Y, Umar A, Steele VE, Lubet RA. Modulation by celecoxib and difluoromethylornithine of the methylation of DNA and the estrogen receptor-α gene in rat colon tumors. Carcinogenesis. 2004;25:1917–23.PubMedCrossRef Pereira MA, Tao L, Wang W, Li Y, Umar A, Steele VE, Lubet RA. Modulation by celecoxib and difluoromethylornithine of the methylation of DNA and the estrogen receptor-α gene in rat colon tumors. Carcinogenesis. 2004;25:1917–23.PubMedCrossRef
81.
go back to reference Armstrong CM, Billimek AR, Allred KF, Sturino JM, Weeks BR, Allred CD. A novel shift in estrogen receptor expression occurs as estradiol suppresses inflammation-associated colon tumor formation. Endocr Relat Cancer. 2013;20:515–25.PubMedCrossRef Armstrong CM, Billimek AR, Allred KF, Sturino JM, Weeks BR, Allred CD. A novel shift in estrogen receptor expression occurs as estradiol suppresses inflammation-associated colon tumor formation. Endocr Relat Cancer. 2013;20:515–25.PubMedCrossRef
82.
go back to reference Topi G, Ghatak S, Satapathy SR, Ehrnström R, Lydrup M-L, Sjölander A. Combined estrogen alpha and beta receptor expression has a prognostic significance for colorectal cancer patients. Front Med. 2022;9:739620.CrossRef Topi G, Ghatak S, Satapathy SR, Ehrnström R, Lydrup M-L, Sjölander A. Combined estrogen alpha and beta receptor expression has a prognostic significance for colorectal cancer patients. Front Med. 2022;9:739620.CrossRef
83.
go back to reference Castiglione F, Taddei A, Degl’Innocenti DR, Buccoliero AM, Bechi P, Garbini F, Chiara FG, Moncini D, Cavallina G, Marascio L. Expression of estrogen receptor β in colon cancer progression. Diagn Mol Pathol. 2008;17:231–6.PubMedCrossRef Castiglione F, Taddei A, Degl’Innocenti DR, Buccoliero AM, Bechi P, Garbini F, Chiara FG, Moncini D, Cavallina G, Marascio L. Expression of estrogen receptor β in colon cancer progression. Diagn Mol Pathol. 2008;17:231–6.PubMedCrossRef
84.
go back to reference Edvardsson K, Nguyen-Vu T, Kalasekar SM, Pontén F, Gustafsson J-Å, Williams C. Estrogen receptor β expression induces changes in the microRNA pool in human colon cancer cells. Carcinogenesis. 2013;34:1431–41.PubMedCrossRef Edvardsson K, Nguyen-Vu T, Kalasekar SM, Pontén F, Gustafsson J-Å, Williams C. Estrogen receptor β expression induces changes in the microRNA pool in human colon cancer cells. Carcinogenesis. 2013;34:1431–41.PubMedCrossRef
86.
go back to reference Hartman J, Edvardsson K, Lindberg K, Zhao C, Williams C, Ström A, Gustafsson J-A. Tumor repressive functions of estrogen receptor β in SW480 colon cancer cells. Can Res. 2009;69:6100–6.CrossRef Hartman J, Edvardsson K, Lindberg K, Zhao C, Williams C, Ström A, Gustafsson J-A. Tumor repressive functions of estrogen receptor β in SW480 colon cancer cells. Can Res. 2009;69:6100–6.CrossRef
87.
go back to reference López-Calderero I, Carnero A, Astudillo A, Palacios J, Chaves M, Benavent M, Limón ML, Garcia-Carbonero R. Prognostic relevance of estrogen receptor-α Ser167 phosphorylation in stage II-III colon cancer patients. Hum Pathol. 2014;45:2437–46.PubMedCrossRef López-Calderero I, Carnero A, Astudillo A, Palacios J, Chaves M, Benavent M, Limón ML, Garcia-Carbonero R. Prognostic relevance of estrogen receptor-α Ser167 phosphorylation in stage II-III colon cancer patients. Hum Pathol. 2014;45:2437–46.PubMedCrossRef
88.
go back to reference Motylewska E, Stasikowska O, Mełeń-Mucha G. The inhibitory effect of diarylpropionitrile, a selective agonist of estrogen receptor beta, on the growth of MC38 colon cancer line. Cancer Lett. 2009;276:68–73.PubMedCrossRef Motylewska E, Stasikowska O, Mełeń-Mucha G. The inhibitory effect of diarylpropionitrile, a selective agonist of estrogen receptor beta, on the growth of MC38 colon cancer line. Cancer Lett. 2009;276:68–73.PubMedCrossRef
89.
go back to reference Ya G, Wang H, Ma Y, Hu A, Ma Y, Hu J, Yu Y. Serum miR-129 functions as a biomarker for colorectal cancer by targeting estrogen receptor (ER) β. Die Pharmazie Intl J Pharm Sci. 2017;72:107–12. Ya G, Wang H, Ma Y, Hu A, Ma Y, Hu J, Yu Y. Serum miR-129 functions as a biomarker for colorectal cancer by targeting estrogen receptor (ER) β. Die Pharmazie Intl J Pharm Sci. 2017;72:107–12.
90.
go back to reference Mostafaie N, Kállay E, Sauerzapf E, Bonner E, Kriwanek S, Cross HS, Huber KR, Krugluger W. Correlated downregulation of estrogen receptor beta and the circadian clock gene Per1 in human colorectal cancer. Mol Carcinog. 2009;48:642–7.PubMedCrossRef Mostafaie N, Kállay E, Sauerzapf E, Bonner E, Kriwanek S, Cross HS, Huber KR, Krugluger W. Correlated downregulation of estrogen receptor beta and the circadian clock gene Per1 in human colorectal cancer. Mol Carcinog. 2009;48:642–7.PubMedCrossRef
91.
go back to reference Topi G, Ehrnström R, Jirström K, Palmquist I, Lydrup M-L, Sjölander A. Association of the oestrogen receptor beta with hormone status and prognosis in a cohort of female patients with colorectal cancer. Eur J Cancer. 2017;83:279–89.PubMedCrossRef Topi G, Ehrnström R, Jirström K, Palmquist I, Lydrup M-L, Sjölander A. Association of the oestrogen receptor beta with hormone status and prognosis in a cohort of female patients with colorectal cancer. Eur J Cancer. 2017;83:279–89.PubMedCrossRef
92.
go back to reference Konstantinopoulos P, Kominea A, Vandoros G, Sykiotis G, Andricopoulos P, Varakis I, Sotiropoulou-Bonikou G, Papavassiliou A. Oestrogen receptor beta (ERβ) is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer. 2003;39:1251–8.PubMedCrossRef Konstantinopoulos P, Kominea A, Vandoros G, Sykiotis G, Andricopoulos P, Varakis I, Sotiropoulou-Bonikou G, Papavassiliou A. Oestrogen receptor beta (ERβ) is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer. 2003;39:1251–8.PubMedCrossRef
93.
go back to reference Hases L, Indukuri R, Birgersson M, Nguyen-Vu T, Lozano R, Saxena A, Hartman J, Frasor J, Gustafsson J-Å, Katajisto P. Intestinal estrogen receptor beta suppresses colon inflammation and tumorigenesis in both sexes. Cancer Lett. 2020;492:54–62.PubMedCrossRef Hases L, Indukuri R, Birgersson M, Nguyen-Vu T, Lozano R, Saxena A, Hartman J, Frasor J, Gustafsson J-Å, Katajisto P. Intestinal estrogen receptor beta suppresses colon inflammation and tumorigenesis in both sexes. Cancer Lett. 2020;492:54–62.PubMedCrossRef
94.
go back to reference Janakiram NB, Mohammed A, Zhang Y, Brewer M, Bryant T, Lightfoot S, Steele VE, Rao CV. Chemopreventive efficacy of raloxifene, bexarotene, and their combination on the progression of chemically induced colon adenomas to adenocarcinomas in rats. Cancer Prev Res. 2013;6:1251–61.CrossRef Janakiram NB, Mohammed A, Zhang Y, Brewer M, Bryant T, Lightfoot S, Steele VE, Rao CV. Chemopreventive efficacy of raloxifene, bexarotene, and their combination on the progression of chemically induced colon adenomas to adenocarcinomas in rats. Cancer Prev Res. 2013;6:1251–61.CrossRef
100.
go back to reference Lax S, Schauer G, Prein K, Kapitan M, Silbert D, Berghold A, Berger A, Trauner M. Expression of the nuclear bile acid receptor/farnesoid X receptor is reduced in human colon carcinoma compared to nonneoplastic mucosa independent from site and may be associated with adverse prognosis. Int J Cancer. 2012;130:2232–9.PubMedCrossRef Lax S, Schauer G, Prein K, Kapitan M, Silbert D, Berghold A, Berger A, Trauner M. Expression of the nuclear bile acid receptor/farnesoid X receptor is reduced in human colon carcinoma compared to nonneoplastic mucosa independent from site and may be associated with adverse prognosis. Int J Cancer. 2012;130:2232–9.PubMedCrossRef
101.
go back to reference De Gottardi A, Touri F, Maurer CA, Perez A, Maurhofer O, Ventre G, Bentzen CL, Bentzen EJ, Dufour J-F. The bile acid nuclear receptor FXR and the bile acid binding protein IBABP are differently expressed in colon cancer. Dig Dis Sci. 2004;49:982–9.PubMedCrossRef De Gottardi A, Touri F, Maurer CA, Perez A, Maurhofer O, Ventre G, Bentzen CL, Bentzen EJ, Dufour J-F. The bile acid nuclear receptor FXR and the bile acid binding protein IBABP are differently expressed in colon cancer. Dig Dis Sci. 2004;49:982–9.PubMedCrossRef
112.
go back to reference Ouyang N, Williams JL, Rigas B. NO-donating aspirin isomers downregulate peroxisome proliferator-activated receptor (PPAR) δ expression in APC min/+ mice proportionally to their tumor inhibitory effect: implications for the role of PPARδ in carcinogenesis. Carcinogenesis. 2006;27:232–9.PubMedCrossRef Ouyang N, Williams JL, Rigas B. NO-donating aspirin isomers downregulate peroxisome proliferator-activated receptor (PPAR) δ expression in APC min/+ mice proportionally to their tumor inhibitory effect: implications for the role of PPARδ in carcinogenesis. Carcinogenesis. 2006;27:232–9.PubMedCrossRef
114.
go back to reference Zuo X, Xu M, Yu J, Wu Y, Moussalli MJ, Manyam GC, Lee SI, Liang S, Gagea M, Morris JS. Potentiation of colon cancer susceptibility in mice by colonic epithelial PPAR-δ/β overexpression. J Natl Cancer Inst. 2014;106:052.CrossRef Zuo X, Xu M, Yu J, Wu Y, Moussalli MJ, Manyam GC, Lee SI, Liang S, Gagea M, Morris JS. Potentiation of colon cancer susceptibility in mice by colonic epithelial PPAR-δ/β overexpression. J Natl Cancer Inst. 2014;106:052.CrossRef
119.
go back to reference Ouyang N, Ke S, Eagleton N, Xie Y, Chen G, Laffins B, Yao H, Zhou B, Tian Y. Pregnane X receptor suppresses proliferation and tumourigenicity of colon cancer cells. Br J Cancer. 2010;102:1753–61.PubMedPubMedCentralCrossRef Ouyang N, Ke S, Eagleton N, Xie Y, Chen G, Laffins B, Yao H, Zhou B, Tian Y. Pregnane X receptor suppresses proliferation and tumourigenicity of colon cancer cells. Br J Cancer. 2010;102:1753–61.PubMedPubMedCentralCrossRef
120.
go back to reference Raynal C, Pascussi J-M, Leguelinel G, Breuker C, Kantar J, Lallemant B, Poujol S, Bonnans C, Joubert D, Hollande F. Pregnane× Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Mol Cancer. 2010;9:1–13.CrossRef Raynal C, Pascussi J-M, Leguelinel G, Breuker C, Kantar J, Lallemant B, Poujol S, Bonnans C, Joubert D, Hollande F. Pregnane× Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Mol Cancer. 2010;9:1–13.CrossRef
121.
go back to reference Jiang H, Chen K, He J, Pan F, Li J, Chen J, Chen W, Liang H. Association of pregnane X receptor with multidrug resistance-related protein 3 and its role in human colon cancer chemoresistance. J Gastrointest Surg. 2009;13:1831–8.PubMedCrossRef Jiang H, Chen K, He J, Pan F, Li J, Chen J, Chen W, Liang H. Association of pregnane X receptor with multidrug resistance-related protein 3 and its role in human colon cancer chemoresistance. J Gastrointest Surg. 2009;13:1831–8.PubMedCrossRef
124.
go back to reference Yamazaki K, Shimizu M, Okuno M, Matsushima-Nishiwaki R, Kanemura N, Araki H, Tsurumi H, Kojima S, Weinstein IB, Moriwaki H. Synergistic effects of RXRα and PPARγ ligands to inhibit growth in human colon cancer cells—phosphorylated RXRα is a critical target for colon cancer management. Gut. 2007;56:1557–63.PubMedPubMedCentralCrossRef Yamazaki K, Shimizu M, Okuno M, Matsushima-Nishiwaki R, Kanemura N, Araki H, Tsurumi H, Kojima S, Weinstein IB, Moriwaki H. Synergistic effects of RXRα and PPARγ ligands to inhibit growth in human colon cancer cells—phosphorylated RXRα is a critical target for colon cancer management. Gut. 2007;56:1557–63.PubMedPubMedCentralCrossRef
125.
go back to reference Modarai SR, Gupta A, Opdenaker LM, Kowash R, Masters G, Viswanathan V, Zhang T, Fields JZ, Boman BM. The anti-cancer effect of retinoic acid signaling in CRC occurs via decreased growth of ALDH+ colon cancer stem cells and increased differentiation of stem cells. Oncotarget. 2018;9:34658.PubMedPubMedCentralCrossRef Modarai SR, Gupta A, Opdenaker LM, Kowash R, Masters G, Viswanathan V, Zhang T, Fields JZ, Boman BM. The anti-cancer effect of retinoic acid signaling in CRC occurs via decreased growth of ALDH+ colon cancer stem cells and increased differentiation of stem cells. Oncotarget. 2018;9:34658.PubMedPubMedCentralCrossRef
128.
go back to reference Uchuya-Castillo J, Aznar N, Frau C, Martinez P, Le Nevé C, Marisa L, Penalva LO, Laurent-Puig P, Puisieux A, Scoazec J-Y. Increased expression of the thyroid hormone nuclear receptor TRα1 characterizes intestinal tumors with high Wnt activity. Oncotarget. 2018;9:30979.PubMedPubMedCentralCrossRef Uchuya-Castillo J, Aznar N, Frau C, Martinez P, Le Nevé C, Marisa L, Penalva LO, Laurent-Puig P, Puisieux A, Scoazec J-Y. Increased expression of the thyroid hormone nuclear receptor TRα1 characterizes intestinal tumors with high Wnt activity. Oncotarget. 2018;9:30979.PubMedPubMedCentralCrossRef
129.
133.
go back to reference Long MD, Campbell MJ. Integrative genomic approaches to dissect clinically-significant relationships between the VDR cistrome and gene expression in primary colon cancer. J Steroid Biochem Mol Biol. 2017;173:130–8.PubMedCrossRef Long MD, Campbell MJ. Integrative genomic approaches to dissect clinically-significant relationships between the VDR cistrome and gene expression in primary colon cancer. J Steroid Biochem Mol Biol. 2017;173:130–8.PubMedCrossRef
134.
go back to reference Pena C, Garcia J, Larriba M, Barderas R, Gomez I, Herrera M, García V, Silva J, Domínguez G, Rodríguez R. SNAI1 expression in colon cancer related with CDH1 and VDR downregulation in normal adjacent tissue. Oncogene. 2009;28:4375–85.PubMedCrossRef Pena C, Garcia J, Larriba M, Barderas R, Gomez I, Herrera M, García V, Silva J, Domínguez G, Rodríguez R. SNAI1 expression in colon cancer related with CDH1 and VDR downregulation in normal adjacent tissue. Oncogene. 2009;28:4375–85.PubMedCrossRef
135.
go back to reference Wada K, Tanaka H, Maeda K, Inoue T, Noda E, Amano R, Kubo N, Muguruma K, Yamada N, Yashiro M. Vitamin D receptor expression is associated with colon cancer in ulcerative colitis. Oncol Rep. 2009;22:1021–5.PubMed Wada K, Tanaka H, Maeda K, Inoue T, Noda E, Amano R, Kubo N, Muguruma K, Yamada N, Yashiro M. Vitamin D receptor expression is associated with colon cancer in ulcerative colitis. Oncol Rep. 2009;22:1021–5.PubMed
136.
go back to reference Murillo G, Matusiak D, Benya RV, Mehta RG. Chemopreventive efficacy of 25-hydroxyvitamin D3 in colon cancer. J Steroid Biochem Mol Biol. 2007;103:763–7.PubMedPubMedCentralCrossRef Murillo G, Matusiak D, Benya RV, Mehta RG. Chemopreventive efficacy of 25-hydroxyvitamin D3 in colon cancer. J Steroid Biochem Mol Biol. 2007;103:763–7.PubMedPubMedCentralCrossRef
137.
go back to reference Larriba MJ, Martín-Villar E, García JM, Pereira F, Pena C, de Garcia Herreros A, Bonilla F, Munoz A. Snail2 cooperates with Snail1 in the repression of vitamin D receptor in colon cancer. Carcinogenesis. 2009;30:1459–68.PubMedCrossRef Larriba MJ, Martín-Villar E, García JM, Pereira F, Pena C, de Garcia Herreros A, Bonilla F, Munoz A. Snail2 cooperates with Snail1 in the repression of vitamin D receptor in colon cancer. Carcinogenesis. 2009;30:1459–68.PubMedCrossRef
138.
go back to reference Anderson MG, Nakane M, Ruan X, Kroeger PE, Wu-Wong JR. Expression of VDR and CYP24A1 mRNA in human tumors. Cancer Chemother Pharmacol. 2006;57:234–40.PubMedCrossRef Anderson MG, Nakane M, Ruan X, Kroeger PE, Wu-Wong JR. Expression of VDR and CYP24A1 mRNA in human tumors. Cancer Chemother Pharmacol. 2006;57:234–40.PubMedCrossRef
143.
go back to reference Picariello L, Fiorelli G, Martineti V, Tognarini I, Pampaloni B, Tonelli F, Brandi ML. Growth response of colon cancer cell lines to selective estrogen receptor modulators. Anticancer Res. 2003;23:2419–24.PubMed Picariello L, Fiorelli G, Martineti V, Tognarini I, Pampaloni B, Tonelli F, Brandi ML. Growth response of colon cancer cell lines to selective estrogen receptor modulators. Anticancer Res. 2003;23:2419–24.PubMed
144.
go back to reference Yang L, Allred KF, Dykes L, Allred CD, Awika JM. Enhanced action of apigenin and naringenin combination on estrogen receptor activation in non-malignant colonocytes: implications on sorghum-derived phytoestrogens. Food Funct. 2015;6:749–55.PubMedCrossRef Yang L, Allred KF, Dykes L, Allred CD, Awika JM. Enhanced action of apigenin and naringenin combination on estrogen receptor activation in non-malignant colonocytes: implications on sorghum-derived phytoestrogens. Food Funct. 2015;6:749–55.PubMedCrossRef
145.
go back to reference Fiorelli G, Picariello L, Martineti V, Tonelli F, Brandi ML. Functional estrogen receptor β in colon cancer cells. Biochem Biophys Res Commun. 1999;261:521–7.PubMedCrossRef Fiorelli G, Picariello L, Martineti V, Tonelli F, Brandi ML. Functional estrogen receptor β in colon cancer cells. Biochem Biophys Res Commun. 1999;261:521–7.PubMedCrossRef
146.
go back to reference Janakiram NB, Steele VE, Rao CV. Estrogen receptor-β as a potential target for colon cancer prevention: chemoprevention of azoxymethane-induced colon carcinogenesis by raloxifene in F344 rats. Cancer Prev Res. 2009;2:52–9.CrossRef Janakiram NB, Steele VE, Rao CV. Estrogen receptor-β as a potential target for colon cancer prevention: chemoprevention of azoxymethane-induced colon carcinogenesis by raloxifene in F344 rats. Cancer Prev Res. 2009;2:52–9.CrossRef
147.
go back to reference Topi G, Satapathy SR, Dash P, Fred Mehrabi S, Ehrnström R, Olsson R, Lydrup ML, Sjölander A. Tumour-suppressive effect of oestrogen receptor β in colorectal cancer patients, colon cancer cells, and a zebrafish model. J Pathol. 2020;251:297–309.PubMedCrossRef Topi G, Satapathy SR, Dash P, Fred Mehrabi S, Ehrnström R, Olsson R, Lydrup ML, Sjölander A. Tumour-suppressive effect of oestrogen receptor β in colorectal cancer patients, colon cancer cells, and a zebrafish model. J Pathol. 2020;251:297–309.PubMedCrossRef
148.
go back to reference Hsu H-H, Cheng S-F, Chen L-M, Liu J-Y, Chu C-H, Weng Y-J, Li Z-Y, Lin C-S, Lee S-D, Kuo W-W. Over-expressed estrogen receptor-α up-regulates hTNF-α gene expression and down-regulates β-catenin signaling activity to induce the apoptosis and inhibit proliferation of LoVo colon cancer cells. Mol Cell Biochem. 2006;289:101–9.PubMedCrossRef Hsu H-H, Cheng S-F, Chen L-M, Liu J-Y, Chu C-H, Weng Y-J, Li Z-Y, Lin C-S, Lee S-D, Kuo W-W. Over-expressed estrogen receptor-α up-regulates hTNF-α gene expression and down-regulates β-catenin signaling activity to induce the apoptosis and inhibit proliferation of LoVo colon cancer cells. Mol Cell Biochem. 2006;289:101–9.PubMedCrossRef
149.
go back to reference Jin P, Wang D-z, Lyu C-x, Wang Y-t, He Y-q, Sheng J-q, Li X. Mismatch repair protein hMLH1, but not hMSH2, enhances estrogen-induced apoptosis of colon cancer cells. J Cancer. 2017;8:3232.PubMedPubMedCentralCrossRef Jin P, Wang D-z, Lyu C-x, Wang Y-t, He Y-q, Sheng J-q, Li X. Mismatch repair protein hMLH1, but not hMSH2, enhances estrogen-induced apoptosis of colon cancer cells. J Cancer. 2017;8:3232.PubMedPubMedCentralCrossRef
150.
go back to reference Wei Y, Huang C, Wu H, Huang J. Estrogen receptor beta (ERβ) mediated-CyclinD1 degradation via autophagy plays an anti-proliferation role in colon cells. Int J Biol Sci. 2019;15:942–52.PubMedPubMedCentralCrossRef Wei Y, Huang C, Wu H, Huang J. Estrogen receptor beta (ERβ) mediated-CyclinD1 degradation via autophagy plays an anti-proliferation role in colon cells. Int J Biol Sci. 2019;15:942–52.PubMedPubMedCentralCrossRef
151.
go back to reference Nakayama Y, Sakamoto H, Satoh K, Yamamoto T. Tamoxifen and gonadal steroids inhibit colon cancer growth in association with inhibition of thymidylate synthase, survivin and telomerase expression through estrogen receptor beta mediated system. Cancer Lett. 2000;161:63–71.PubMedCrossRef Nakayama Y, Sakamoto H, Satoh K, Yamamoto T. Tamoxifen and gonadal steroids inhibit colon cancer growth in association with inhibition of thymidylate synthase, survivin and telomerase expression through estrogen receptor beta mediated system. Cancer Lett. 2000;161:63–71.PubMedCrossRef
152.
go back to reference Bustos V, Nolan ÁM, Nijhuis A, Harvey H, Parker A, Poulsom R, McBryan J, Thomas W, Silver A, Harvey BJ. GPER mediates differential effects of estrogen on colon cancer cell proliferation and migration under normoxic and hypoxic conditions. Oncotarget. 2017;8:84258.PubMedPubMedCentralCrossRef Bustos V, Nolan ÁM, Nijhuis A, Harvey H, Parker A, Poulsom R, McBryan J, Thomas W, Silver A, Harvey BJ. GPER mediates differential effects of estrogen on colon cancer cell proliferation and migration under normoxic and hypoxic conditions. Oncotarget. 2017;8:84258.PubMedPubMedCentralCrossRef
153.
go back to reference Raju J, Bielecki A, Caldwell D, Lok E, Taylor M, Kapal K, Curran I, Cooke GM, Bird RP, Mehta R. Soy isoflavones modulate azoxymethane-induced rat colon carcinogenesis exposed pre-and postnatally and inhibit growth of DLD-1 human colon adenocarcinoma cells by increasing the expression of estrogen receptor-β. J Nutr. 2009;139:474–81.PubMedCrossRef Raju J, Bielecki A, Caldwell D, Lok E, Taylor M, Kapal K, Curran I, Cooke GM, Bird RP, Mehta R. Soy isoflavones modulate azoxymethane-induced rat colon carcinogenesis exposed pre-and postnatally and inhibit growth of DLD-1 human colon adenocarcinoma cells by increasing the expression of estrogen receptor-β. J Nutr. 2009;139:474–81.PubMedCrossRef
154.
go back to reference Giroux V, Lemay F, Bernatchez G, Robitaille Y, Carrier JC. Estrogen receptor β deficiency enhances small intestinal tumorigenesis in ApcMin/+ mice. Int J Cancer. 2008;123:303–11.PubMedCrossRef Giroux V, Lemay F, Bernatchez G, Robitaille Y, Carrier JC. Estrogen receptor β deficiency enhances small intestinal tumorigenesis in ApcMin/+ mice. Int J Cancer. 2008;123:303–11.PubMedCrossRef
155.
go back to reference He Y-q, Sheng J-q, Ling X-l, Fu L, Jin P, Yen L, Rao J. Estradiol regulates miR-135b and mismatch repair gene expressions via estrogen receptor-β in colorectal cells. Exp Mol Med. 2012;44:723–32.PubMedPubMedCentralCrossRef He Y-q, Sheng J-q, Ling X-l, Fu L, Jin P, Yen L, Rao J. Estradiol regulates miR-135b and mismatch repair gene expressions via estrogen receptor-β in colorectal cells. Exp Mol Med. 2012;44:723–32.PubMedPubMedCentralCrossRef
156.
go back to reference Fang Y-J, Pan Z-Z, Li L-R, Lu Z-H, Zhang L-Y, Wan D-S. MMP7 expression regulated by endocrine therapy in ERβ-positive colon cancer cells. J Exp Clin Cancer Res. 2009;28:1–8.CrossRef Fang Y-J, Pan Z-Z, Li L-R, Lu Z-H, Zhang L-Y, Wan D-S. MMP7 expression regulated by endocrine therapy in ERβ-positive colon cancer cells. J Exp Clin Cancer Res. 2009;28:1–8.CrossRef
157.
go back to reference Caiazza F, Galluzzo P, Lorenzetti S, Marino M. 17β-estradiol induces ERβ up-regulation via p38/MAPK activation in colon cancer cells. Biochem Biophys Res Commun. 2007;359:102–7.PubMedCrossRef Caiazza F, Galluzzo P, Lorenzetti S, Marino M. 17β-estradiol induces ERβ up-regulation via p38/MAPK activation in colon cancer cells. Biochem Biophys Res Commun. 2007;359:102–7.PubMedCrossRef
158.
go back to reference Qiu Y, Waters C, Lewis A, Langman M, Eggo M. Oestrogen-induced apoptosis in colonocytes expressing oestrogen receptor beta. J Endocrinol. 2002;174:369–77.PubMedCrossRef Qiu Y, Waters C, Lewis A, Langman M, Eggo M. Oestrogen-induced apoptosis in colonocytes expressing oestrogen receptor beta. J Endocrinol. 2002;174:369–77.PubMedCrossRef
159.
go back to reference Fiocchetti M, Camilli G, Acconcia F, Leone S, Ascenzi P, Marino M. ERβ-dependent neuroglobin up-regulation impairs 17β-estradiol-induced apoptosis in DLD-1 colon cancer cells upon oxidative stress injury. J Steroid Biochem Mol Biol. 2015;149:128–37.PubMedCrossRef Fiocchetti M, Camilli G, Acconcia F, Leone S, Ascenzi P, Marino M. ERβ-dependent neuroglobin up-regulation impairs 17β-estradiol-induced apoptosis in DLD-1 colon cancer cells upon oxidative stress injury. J Steroid Biochem Mol Biol. 2015;149:128–37.PubMedCrossRef
160.
go back to reference Sasso CV, Santiano FE, Arboccó FCV, Zyla LE, Semino SN, Guerrero-Gimenez ME, Creydt VP, Fontana CML, Carón RW. Estradiol and progesterone regulate proliferation and apoptosis in colon cancer. Endocr Connect. 2019;8:217–29.PubMedPubMedCentralCrossRef Sasso CV, Santiano FE, Arboccó FCV, Zyla LE, Semino SN, Guerrero-Gimenez ME, Creydt VP, Fontana CML, Carón RW. Estradiol and progesterone regulate proliferation and apoptosis in colon cancer. Endocr Connect. 2019;8:217–29.PubMedPubMedCentralCrossRef
164.
go back to reference Bernatchez G, Giroux V, Lassalle T, Carpentier AC, Rivard N, Carrier JC. ERRα metabolic nuclear receptor controls growth of colon cancer cells. Carcinogenesis. 2013;34:2253–61.PubMedCrossRef Bernatchez G, Giroux V, Lassalle T, Carpentier AC, Rivard N, Carrier JC. ERRα metabolic nuclear receptor controls growth of colon cancer cells. Carcinogenesis. 2013;34:2253–61.PubMedCrossRef
167.
go back to reference Maran RR, Thomas A, Roth M, Sheng Z, Esterly N, Pinson D, Gao X, Zhang Y, Ganapathy V, Gonzalez FJ. Farnesoid X receptor deficiency in mice leads to increased intestinal epithelial cell proliferation and tumor development. J Pharmacol Exp Ther. 2009;328:469–77.PubMedCrossRef Maran RR, Thomas A, Roth M, Sheng Z, Esterly N, Pinson D, Gao X, Zhang Y, Ganapathy V, Gonzalez FJ. Farnesoid X receptor deficiency in mice leads to increased intestinal epithelial cell proliferation and tumor development. J Pharmacol Exp Ther. 2009;328:469–77.PubMedCrossRef
168.
go back to reference Peng Z, Raufman J-P, Xie G. Src-mediated cross-talk between farnesoid X and epidermal growth factor receptors inhibits human intestinal cell proliferation and tumorigenesis. PLoS ONE. 2012;7:e48461.ADSPubMedPubMedCentralCrossRef Peng Z, Raufman J-P, Xie G. Src-mediated cross-talk between farnesoid X and epidermal growth factor receptors inhibits human intestinal cell proliferation and tumorigenesis. PLoS ONE. 2012;7:e48461.ADSPubMedPubMedCentralCrossRef
169.
go back to reference Yang F, Hu Y, Liu H-X, Wan Y-JY. MiR-22-silenced cyclin A expression in colon and liver cancer cells is regulated by bile acid receptor. J Biol Chem. 2015;290:6507–15.PubMedPubMedCentralCrossRef Yang F, Hu Y, Liu H-X, Wan Y-JY. MiR-22-silenced cyclin A expression in colon and liver cancer cells is regulated by bile acid receptor. J Biol Chem. 2015;290:6507–15.PubMedPubMedCentralCrossRef
170.
go back to reference Hotta M, Sakatani T, Ishino K, Wada R, Kudo M, Yokoyama Y, Yamada T, Yoshida H, Naito Z. Farnesoid X receptor induces cell death and sensitizes to TRAIL-induced inhibition of growth in colorectal cancer cells through the up-regulation of death receptor 5. Biochem Biophys Res Commun. 2019;519:824–31.PubMedCrossRef Hotta M, Sakatani T, Ishino K, Wada R, Kudo M, Yokoyama Y, Yamada T, Yoshida H, Naito Z. Farnesoid X receptor induces cell death and sensitizes to TRAIL-induced inhibition of growth in colorectal cancer cells through the up-regulation of death receptor 5. Biochem Biophys Res Commun. 2019;519:824–31.PubMedCrossRef
171.
go back to reference Selmin OI, Fang C, Lyon AM, Doetschman TC, Thompson PA, Martinez JD, Smith JW, Lance PM, Romagnolo DF. Inactivation of adenomatous polyposis coli reduces bile acid/farnesoid X receptor expression through Fxr gene CpG methylation in mouse colon tumors and human colon cancer cells. J Nutr. 2016;146:236–42.PubMedCrossRef Selmin OI, Fang C, Lyon AM, Doetschman TC, Thompson PA, Martinez JD, Smith JW, Lance PM, Romagnolo DF. Inactivation of adenomatous polyposis coli reduces bile acid/farnesoid X receptor expression through Fxr gene CpG methylation in mouse colon tumors and human colon cancer cells. J Nutr. 2016;146:236–42.PubMedCrossRef
172.
go back to reference Qiao P, Li S, Zhang H, Yao L, Wang F. Farnesoid X receptor inhibits proliferation of human colorectal cancer cells via the miR-135A1/CCNG2 signaling pathway. Oncol Rep. 2018;40:2067–78.PubMed Qiao P, Li S, Zhang H, Yao L, Wang F. Farnesoid X receptor inhibits proliferation of human colorectal cancer cells via the miR-135A1/CCNG2 signaling pathway. Oncol Rep. 2018;40:2067–78.PubMed
173.
go back to reference Modica S, Murzilli S, Salvatore L, Schmidt DR, Moschetta A. Nuclear bile acid receptor FXR protects against intestinal tumorigenesis. Can Res. 2008;68:9589–94.CrossRef Modica S, Murzilli S, Salvatore L, Schmidt DR, Moschetta A. Nuclear bile acid receptor FXR protects against intestinal tumorigenesis. Can Res. 2008;68:9589–94.CrossRef
174.
go back to reference Peng Z, Chen J, Drachenberg CB, Raufman J-P, Xie G. Farnesoid X receptor represses matrix metalloproteinase 7 expression, revealing this regulatory axis as a promising therapeutic target in colon cancer. J Biol Chem. 2019;294:8529–42.PubMedPubMedCentralCrossRef Peng Z, Chen J, Drachenberg CB, Raufman J-P, Xie G. Farnesoid X receptor represses matrix metalloproteinase 7 expression, revealing this regulatory axis as a promising therapeutic target in colon cancer. J Biol Chem. 2019;294:8529–42.PubMedPubMedCentralCrossRef
177.
go back to reference Algamas-Dimantov A, Yehuda-Shnaidman E, Peri I, Schwartz B. Epigenetic control of HNF-4α in colon carcinoma cells affects MUC4 expression and malignancy. Cell Oncol. 2013;36:155–67.CrossRef Algamas-Dimantov A, Yehuda-Shnaidman E, Peri I, Schwartz B. Epigenetic control of HNF-4α in colon carcinoma cells affects MUC4 expression and malignancy. Cell Oncol. 2013;36:155–67.CrossRef
186.
189.
196.
go back to reference Hu Y, French SW, Chau T, Liu HX, Sheng L, Wei F, Stondell J, Garcia JC, Du Y, Bowlus CL, et al. RARbeta acts as both an upstream regulator and downstream effector of miR-22, which epigenetically regulates NUR77 to induce apoptosis of colon cancer cells. FASEB J. 2019;33:2314–26. https://doi.org/10.1096/fj.201801390R.CrossRefPubMed Hu Y, French SW, Chau T, Liu HX, Sheng L, Wei F, Stondell J, Garcia JC, Du Y, Bowlus CL, et al. RARbeta acts as both an upstream regulator and downstream effector of miR-22, which epigenetically regulates NUR77 to induce apoptosis of colon cancer cells. FASEB J. 2019;33:2314–26. https://​doi.​org/​10.​1096/​fj.​201801390R.CrossRefPubMed
202.
go back to reference Hu Y, Chau T, Liu H-X, Liao D, Keane R, Nie Y, Yang H, Wan Y-JY. Bile acids regulate nuclear receptor (Nur77) expression and intracellular location to control proliferation and apoptosis. Mol Cancer Res. 2015;13:281–92.PubMedCrossRef Hu Y, Chau T, Liu H-X, Liao D, Keane R, Nie Y, Yang H, Wan Y-JY. Bile acids regulate nuclear receptor (Nur77) expression and intracellular location to control proliferation and apoptosis. Mol Cancer Res. 2015;13:281–92.PubMedCrossRef
206.
go back to reference Lachal S, Ford J, Shulkes A, Baldwin GS. PPARα agonists stimulate progastrin production in human colorectal carcinoma cells. Regul Pept. 2004;120:243–51.PubMedCrossRef Lachal S, Ford J, Shulkes A, Baldwin GS. PPARα agonists stimulate progastrin production in human colorectal carcinoma cells. Regul Pept. 2004;120:243–51.PubMedCrossRef
207.
go back to reference Grau R, Iñiguez MA, Fresno M. Inhibition of activator protein 1 activation, vascular endothelial growth factor, and cyclooxygenase-2 expression by 15-deoxy-Δ12, 14-prostaglandin J2 in colon carcinoma cells: evidence for a redox-sensitive peroxisome proliferator-activated receptor-γ-independent mechanism. Can Res. 2004;64:5162–71.CrossRef Grau R, Iñiguez MA, Fresno M. Inhibition of activator protein 1 activation, vascular endothelial growth factor, and cyclooxygenase-2 expression by 15-deoxy-Δ12, 14-prostaglandin J2 in colon carcinoma cells: evidence for a redox-sensitive peroxisome proliferator-activated receptor-γ-independent mechanism. Can Res. 2004;64:5162–71.CrossRef
208.
go back to reference Kohno H, Yoshitani Si, Takashima S, Okumura A, Hosokawa M, Yamaguchi N, Tanaka T. Troglitazone, a Ligand for Peroxisome Proliferator-activated Receptor γ Inhibits Chemically-induced Aberrant Crypt Foci in Rats. Jpn J Cancer Res. 2001;92:396–403.PubMedPubMedCentralCrossRef Kohno H, Yoshitani Si, Takashima S, Okumura A, Hosokawa M, Yamaguchi N, Tanaka T. Troglitazone, a Ligand for Peroxisome Proliferator-activated Receptor γ Inhibits Chemically-induced Aberrant Crypt Foci in Rats. Jpn J Cancer Res. 2001;92:396–403.PubMedPubMedCentralCrossRef
209.
go back to reference Gupta RA, Brockman JA, Sarraf P, Willson TM, DuBois RN. Target genes of peroxisome proliferator-activated receptor γ in colorectal cancer cells. J Biol Chem. 2001;276:29681–7.PubMedCrossRef Gupta RA, Brockman JA, Sarraf P, Willson TM, DuBois RN. Target genes of peroxisome proliferator-activated receptor γ in colorectal cancer cells. J Biol Chem. 2001;276:29681–7.PubMedCrossRef
211.
go back to reference Lin MS, Chen WC, Bai X, Wang YD. Activation of peroxisome proliferator-activated receptor γ inhibits cell growth via apoptosis and arrest of the cell cycle in human colorectal cancer. J Dig Dis. 2007;8:82–8.PubMedCrossRef Lin MS, Chen WC, Bai X, Wang YD. Activation of peroxisome proliferator-activated receptor γ inhibits cell growth via apoptosis and arrest of the cell cycle in human colorectal cancer. J Dig Dis. 2007;8:82–8.PubMedCrossRef
212.
go back to reference Moon CM, Kwon JH, Kim JS, Oh SH, Jin Lee K, Park JJ, Pil Hong S, Cheon JH, Kim TI, Kim WH. Nonsteroidal anti-inflammatory drugs suppress cancer stem cells via inhibiting PTGS2 (cyclooxygenase 2) and NOTCH/HES1 and activating PPARG in colorectal cancer. Int J Cancer. 2014;134:519–29.PubMedCrossRef Moon CM, Kwon JH, Kim JS, Oh SH, Jin Lee K, Park JJ, Pil Hong S, Cheon JH, Kim TI, Kim WH. Nonsteroidal anti-inflammatory drugs suppress cancer stem cells via inhibiting PTGS2 (cyclooxygenase 2) and NOTCH/HES1 and activating PPARG in colorectal cancer. Int J Cancer. 2014;134:519–29.PubMedCrossRef
213.
go back to reference Kohno H, Yasui Y, Suzuki R, Hosokawa M, Miyashita K, Tanaka T. Dietary seed oil rich in conjugated linolenic acid from bitter melon inhibits azoxymethane-induced rat colon carcinogenesis through elevation of colonic PPARγ expression and alteration of lipid composition. Int J Cancer. 2004;110:896–901.PubMedCrossRef Kohno H, Yasui Y, Suzuki R, Hosokawa M, Miyashita K, Tanaka T. Dietary seed oil rich in conjugated linolenic acid from bitter melon inhibits azoxymethane-induced rat colon carcinogenesis through elevation of colonic PPARγ expression and alteration of lipid composition. Int J Cancer. 2004;110:896–901.PubMedCrossRef
214.
go back to reference Wang J-B, Qi L-L, Zheng S-D, Wang H-Z, Wu T-X. Curcumin suppresses PPARδ expression and related genes in HT-29 cells. World J Gastroenterol: WJG. 2009;15:1346.PubMedPubMedCentralCrossRef Wang J-B, Qi L-L, Zheng S-D, Wang H-Z, Wu T-X. Curcumin suppresses PPARδ expression and related genes in HT-29 cells. World J Gastroenterol: WJG. 2009;15:1346.PubMedPubMedCentralCrossRef
221.
go back to reference Yang W-L, Frucht H. Activation of the PPAR pathway induces apoptosis and COX-2 inhibition in HT-29 human colon cancer cells. Carcinogenesis. 2001;22:1379–83.PubMedCrossRef Yang W-L, Frucht H. Activation of the PPAR pathway induces apoptosis and COX-2 inhibition in HT-29 human colon cancer cells. Carcinogenesis. 2001;22:1379–83.PubMedCrossRef
223.
go back to reference Yoshizumi T, Ohta T, Ninomiya I, Terada I, Fushida S, Fujimura T, Nishimura G, Shimizu K, Yi S, Miwa K. Thiazolidinedione, a peroxisome proliferator-activated receptor-gamma ligand, inhibits growth and metastasis of HT-29 human colon cancer cells through differentiation-promoting effects. Int J Oncol. 2004;25:631–9.PubMed Yoshizumi T, Ohta T, Ninomiya I, Terada I, Fushida S, Fujimura T, Nishimura G, Shimizu K, Yi S, Miwa K. Thiazolidinedione, a peroxisome proliferator-activated receptor-gamma ligand, inhibits growth and metastasis of HT-29 human colon cancer cells through differentiation-promoting effects. Int J Oncol. 2004;25:631–9.PubMed
224.
go back to reference Chintharlapalli S, Papineni S, Liu S, Jutooru I, Chadalapaka G, Cho S-d, Murthy RS, You Y, Safe S. 2-Cyano-lup-1-en-3-oxo-20-oic acid, a cyano derivative of betulinic acid, activates peroxisome proliferator-activated receptor γ in colon and pancreatic cancer cells. Carcinogenesis. 2007;28:2337–46.PubMedCrossRef Chintharlapalli S, Papineni S, Liu S, Jutooru I, Chadalapaka G, Cho S-d, Murthy RS, You Y, Safe S. 2-Cyano-lup-1-en-3-oxo-20-oic acid, a cyano derivative of betulinic acid, activates peroxisome proliferator-activated receptor γ in colon and pancreatic cancer cells. Carcinogenesis. 2007;28:2337–46.PubMedCrossRef
229.
go back to reference Tsukahara T, Haniu H, Matsuda Y. PTB-associated splicing factor (PSF) is a PPARγ-binding protein and growth regulator of colon cancer cells. PLoS ONE. 2013;8: e58749.ADSPubMedPubMedCentralCrossRef Tsukahara T, Haniu H, Matsuda Y. PTB-associated splicing factor (PSF) is a PPARγ-binding protein and growth regulator of colon cancer cells. PLoS ONE. 2013;8: e58749.ADSPubMedPubMedCentralCrossRef
231.
go back to reference Yaacob NS, Darus HM, Norazmi MN. Modulation of cell growth and PPARγ expression in human colorectal cancer cell lines by ciglitazone. Exp Toxicol Pathol. 2008;60:505–12.PubMedCrossRef Yaacob NS, Darus HM, Norazmi MN. Modulation of cell growth and PPARγ expression in human colorectal cancer cell lines by ciglitazone. Exp Toxicol Pathol. 2008;60:505–12.PubMedCrossRef
234.
go back to reference Weidner C, Rousseau M, Micikas RJ, Fischer C, Plauth A, Wowro SJ, Siems K, Hetterling G, Kliem M, Schroeder FC. Amorfrutin C induces apoptosis and inhibits proliferation in colon cancer cells through targeting mitochondria. J Nat Prod. 2016;79:2–12.PubMedCrossRef Weidner C, Rousseau M, Micikas RJ, Fischer C, Plauth A, Wowro SJ, Siems K, Hetterling G, Kliem M, Schroeder FC. Amorfrutin C induces apoptosis and inhibits proliferation in colon cancer cells through targeting mitochondria. J Nat Prod. 2016;79:2–12.PubMedCrossRef
236.
go back to reference Papi A, Rocchi P, Ferreri AM, Orlandi M. RXRγ and PPARγ ligands in combination to inhibit proliferation and invasiveness in colon cancer cells. Cancer Lett. 2010;297:65–74.PubMedCrossRef Papi A, Rocchi P, Ferreri AM, Orlandi M. RXRγ and PPARγ ligands in combination to inhibit proliferation and invasiveness in colon cancer cells. Cancer Lett. 2010;297:65–74.PubMedCrossRef
237.
go back to reference Bansard L, Bouvet O, Moutin E, Le Gall G, Giammona A, Pothin E, Bacou M, Hassen-Khodja C, Bordignon B, Bourgaux JF. Niclosamide induces miR-148a to inhibit PXR and sensitize colon cancer stem cells to chemotherapy. Stem Cell Reports. 2022;17:835–48.PubMedPubMedCentralCrossRef Bansard L, Bouvet O, Moutin E, Le Gall G, Giammona A, Pothin E, Bacou M, Hassen-Khodja C, Bordignon B, Bourgaux JF. Niclosamide induces miR-148a to inhibit PXR and sensitize colon cancer stem cells to chemotherapy. Stem Cell Reports. 2022;17:835–48.PubMedPubMedCentralCrossRef
238.
go back to reference Planque C, Rajabi F, Grillet F, Finetti P, Bertucci F, Gironella M, Lozano JJ, Beucher B, Giraud J, Garambois V. Pregnane X-receptor promotes stem cell-mediated colon cancer relapse. Oncotarget. 2016;7:56558.PubMedPubMedCentralCrossRef Planque C, Rajabi F, Grillet F, Finetti P, Bertucci F, Gironella M, Lozano JJ, Beucher B, Giraud J, Garambois V. Pregnane X-receptor promotes stem cell-mediated colon cancer relapse. Oncotarget. 2016;7:56558.PubMedPubMedCentralCrossRef
239.
go back to reference Cheng J, Fang Z-Z, Nagaoka K, Okamoto M, Qu A, Tanaka N, Kimura S, Gonzalez FJ. Activation of intestinal human pregnane X receptor protects against azoxymethane/dextran sulfate sodium-induced colon cancer. J Pharmacol Exp Ther. 2014;351:559–67.PubMedPubMedCentralCrossRef Cheng J, Fang Z-Z, Nagaoka K, Okamoto M, Qu A, Tanaka N, Kimura S, Gonzalez FJ. Activation of intestinal human pregnane X receptor protects against azoxymethane/dextran sulfate sodium-induced colon cancer. J Pharmacol Exp Ther. 2014;351:559–67.PubMedPubMedCentralCrossRef
240.
go back to reference Zimmermann C, van Waterschoot RA, Harmsen S, Maier A, Gutmann H, Schinkel AH. PXR-mediated induction of human CYP3A4 and mouse Cyp3a11 by the glucocorticoid budesonide. Eur J Pharm Sci. 2009;36:565–71.PubMedCrossRef Zimmermann C, van Waterschoot RA, Harmsen S, Maier A, Gutmann H, Schinkel AH. PXR-mediated induction of human CYP3A4 and mouse Cyp3a11 by the glucocorticoid budesonide. Eur J Pharm Sci. 2009;36:565–71.PubMedCrossRef
241.
go back to reference Cheng J, Shah YM, Ma X, Pang X, Tanaka T, Kodama T, Krausz KW, Gonzalez FJ. Therapeutic role of rifaximin in inflammatory bowel disease: clinical implication of human pregnane X receptor activation. J Pharmacol Exp Ther. 2010;335:32–41.PubMedPubMedCentralCrossRef Cheng J, Shah YM, Ma X, Pang X, Tanaka T, Kodama T, Krausz KW, Gonzalez FJ. Therapeutic role of rifaximin in inflammatory bowel disease: clinical implication of human pregnane X receptor activation. J Pharmacol Exp Ther. 2010;335:32–41.PubMedPubMedCentralCrossRef
242.
go back to reference Dou W, Mukherjee S, Li H, Venkatesh M, Wang H, Kortagere S, Peleg A, Chilimuri SS, Wang Z-T, Feng Y. Alleviation of gut inflammation by Cdx2/Pxr pathway in a mouse model of chemical colitis. PLoS ONE. 2012;7:e36075.ADSPubMedPubMedCentralCrossRef Dou W, Mukherjee S, Li H, Venkatesh M, Wang H, Kortagere S, Peleg A, Chilimuri SS, Wang Z-T, Feng Y. Alleviation of gut inflammation by Cdx2/Pxr pathway in a mouse model of chemical colitis. PLoS ONE. 2012;7:e36075.ADSPubMedPubMedCentralCrossRef
243.
go back to reference Liu C-L, Lim Y-P, Hu M-L. Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Mar Drugs. 2012;10:242–57.PubMedPubMedCentralCrossRef Liu C-L, Lim Y-P, Hu M-L. Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Mar Drugs. 2012;10:242–57.PubMedPubMedCentralCrossRef
244.
go back to reference Esposito G, Gigli S, Seguella L, Nobile N, D’alessandro A, Pesce M, Capoccia E, Steardo L, Cirillo C, Cuomo R. Rifaximin, a non-absorbable antibiotic, inhibits the release of pro-angiogenic mediators in colon cancer cells through a pregnane X receptor-dependent pathway. Int J Oncol. 2016;49:639–45.PubMedCrossRef Esposito G, Gigli S, Seguella L, Nobile N, D’alessandro A, Pesce M, Capoccia E, Steardo L, Cirillo C, Cuomo R. Rifaximin, a non-absorbable antibiotic, inhibits the release of pro-angiogenic mediators in colon cancer cells through a pregnane X receptor-dependent pathway. Int J Oncol. 2016;49:639–45.PubMedCrossRef
245.
go back to reference Harmsen S, Meijerman I, Febus C, Maas-Bakker R, Beijnen J, Schellens J. PXR-mediated induction of P-glycoprotein by anticancer drugs in a human colon adenocarcinoma-derived cell line. Cancer Chemother Pharmacol. 2010;66:765–71.PubMedCrossRef Harmsen S, Meijerman I, Febus C, Maas-Bakker R, Beijnen J, Schellens J. PXR-mediated induction of P-glycoprotein by anticancer drugs in a human colon adenocarcinoma-derived cell line. Cancer Chemother Pharmacol. 2010;66:765–71.PubMedCrossRef
246.
go back to reference Habano W, Gamo T, Terashima J, Sugai T, Otsuka K, Wakabayashi G, Ozawa S. Involvement of promoter methylation in the regulation of Pregnane X receptor in colon cancer cells. BMC Cancer. 2011;11:1–10.CrossRef Habano W, Gamo T, Terashima J, Sugai T, Otsuka K, Wakabayashi G, Ozawa S. Involvement of promoter methylation in the regulation of Pregnane X receptor in colon cancer cells. BMC Cancer. 2011;11:1–10.CrossRef
248.
go back to reference Cesario RM, Stone J, Yen W-C, Bissonnette RP, Lamph WW. Differentiation and growth inhibition mediated via the RXR: PPARγ heterodimer in colon cancer. Cancer Lett. 2006;240:225–33.PubMedCrossRef Cesario RM, Stone J, Yen W-C, Bissonnette RP, Lamph WW. Differentiation and growth inhibition mediated via the RXR: PPARγ heterodimer in colon cancer. Cancer Lett. 2006;240:225–33.PubMedCrossRef
249.
go back to reference Dillard AC, Lane MA. Retinol decreases β-catenin protein levels in retinoic acid-resistant colon cancer cell lines. Mol Carcinog. 2007;46:315–29.PubMedCrossRef Dillard AC, Lane MA. Retinol decreases β-catenin protein levels in retinoic acid-resistant colon cancer cell lines. Mol Carcinog. 2007;46:315–29.PubMedCrossRef
251.
go back to reference Janakiram NB, Mohammed A, Qian L, Choi C-I, Steele VE, Rao CV. Chemopreventive effects of RXR-selective rexinoid bexarotene on intestinal neoplasia of ApcMin/+ mice. Neoplasia. 2012;14:159–68.PubMedPubMedCentralCrossRef Janakiram NB, Mohammed A, Qian L, Choi C-I, Steele VE, Rao CV. Chemopreventive effects of RXR-selective rexinoid bexarotene on intestinal neoplasia of ApcMin/+ mice. Neoplasia. 2012;14:159–68.PubMedPubMedCentralCrossRef
252.
go back to reference Ruan H, Zhan Y, Hou J, Xu B, Chen B, Tian Y, Wu D, Zhao Y, Zhang Y, Chen X. Berberine binds RXRα to suppress β-catenin signaling in colon cancer cells. Oncogene. 2017;36:6906–18.PubMedPubMedCentralCrossRef Ruan H, Zhan Y, Hou J, Xu B, Chen B, Tian Y, Wu D, Zhao Y, Zhang Y, Chen X. Berberine binds RXRα to suppress β-catenin signaling in colon cancer cells. Oncogene. 2017;36:6906–18.PubMedPubMedCentralCrossRef
253.
go back to reference Xu B, Jiang X, Xiong J, Lan J, Tian Y, Zhong L, Wang X, Xu N, Cao H, Zhang W. Structure–activity relationship study enables the discovery of a novel berberine analogue as the RXRα activator to inhibit colon cancer. J Med Chem. 2020;63:5841–55.PubMedCrossRef Xu B, Jiang X, Xiong J, Lan J, Tian Y, Zhong L, Wang X, Xu N, Cao H, Zhang W. Structure–activity relationship study enables the discovery of a novel berberine analogue as the RXRα activator to inhibit colon cancer. J Med Chem. 2020;63:5841–55.PubMedCrossRef
254.
go back to reference Papi A, Ferreri A, Guerra F, Orlandi M. Anti-invasive effects and proapoptotic activity induction by the rexinoid IIF and valproic acid in combination on colon cancer cell lines. Anticancer Res. 2012;32:2855–62.PubMed Papi A, Ferreri A, Guerra F, Orlandi M. Anti-invasive effects and proapoptotic activity induction by the rexinoid IIF and valproic acid in combination on colon cancer cell lines. Anticancer Res. 2012;32:2855–62.PubMed
258.
269.
go back to reference Ishizawa M, Hirayu A, Makishima M. Zinc Inhibits Cadherin 1 Expression Induced by 1α, 25-Dihydroxyvitamin D3 in Colon Cancer Cells. Anticancer Res. 2021;41:5453–9.PubMedCrossRef Ishizawa M, Hirayu A, Makishima M. Zinc Inhibits Cadherin 1 Expression Induced by 1α, 25-Dihydroxyvitamin D3 in Colon Cancer Cells. Anticancer Res. 2021;41:5453–9.PubMedCrossRef
270.
go back to reference Thompson PD, Jurutka PW, Whitfield GK, Myskowski SM, Eichhorst KR, Dominguez CE, Haussler CA, Haussler MR. Liganded VDR induces CYP3A4 in small intestinal and colon cancer cells via DR3 and ER6 vitamin D responsive elements. Biochem Biophys Res Commun. 2002;299:730–8.PubMedCrossRef Thompson PD, Jurutka PW, Whitfield GK, Myskowski SM, Eichhorst KR, Dominguez CE, Haussler CA, Haussler MR. Liganded VDR induces CYP3A4 in small intestinal and colon cancer cells via DR3 and ER6 vitamin D responsive elements. Biochem Biophys Res Commun. 2002;299:730–8.PubMedCrossRef
271.
go back to reference Egan JB, Thompson PA, Vitanov MV, Bartik L, Jacobs ET, Haussler MR, Gerner EW, Jurutka PW. Vitamin D receptor ligands, adenomatous polyposis coli, and the vitamin D receptor FokI polymorphism collectively modulate β-catenin activity in colon cancer cells. Mol Carcinog. 2010;49:337–52.PubMedPubMedCentralCrossRef Egan JB, Thompson PA, Vitanov MV, Bartik L, Jacobs ET, Haussler MR, Gerner EW, Jurutka PW. Vitamin D receptor ligands, adenomatous polyposis coli, and the vitamin D receptor FokI polymorphism collectively modulate β-catenin activity in colon cancer cells. Mol Carcinog. 2010;49:337–52.PubMedPubMedCentralCrossRef
272.
go back to reference Bartik L, Whitfield GK, Kaczmarska M, Lowmiller CL, Moffet EW, Furmick JK, Hernandez Z, Haussler CA, Haussler MR, Jurutka PW. Curcumin: a novel nutritionally derived ligand of the vitamin D receptor with implications for colon cancer chemoprevention. J Nutr Biochem. 2010;21:1153–61.PubMedPubMedCentralCrossRef Bartik L, Whitfield GK, Kaczmarska M, Lowmiller CL, Moffet EW, Furmick JK, Hernandez Z, Haussler CA, Haussler MR, Jurutka PW. Curcumin: a novel nutritionally derived ligand of the vitamin D receptor with implications for colon cancer chemoprevention. J Nutr Biochem. 2010;21:1153–61.PubMedPubMedCentralCrossRef
273.
go back to reference Qi X, Tang J, Pramanik R, Schultz RM, Shirasawa S, Sasazuki T, Han J, Chen G. p38 MAPK activation selectively induces cell death in K-ras-mutated human colon cancer cells through regulation of vitamin D receptor. J Biol Chem. 2004;279:22138–44.PubMedCrossRef Qi X, Tang J, Pramanik R, Schultz RM, Shirasawa S, Sasazuki T, Han J, Chen G. p38 MAPK activation selectively induces cell death in K-ras-mutated human colon cancer cells through regulation of vitamin D receptor. J Biol Chem. 2004;279:22138–44.PubMedCrossRef
274.
go back to reference Beildeck ME, Islam M, Shah S, Welsh J, Byers SW. Control of TCF-4 expression by VDR and vitamin D in the mouse mammary gland and colorectal cancer cell lines. PLoS ONE. 2009;4: e7872.ADSPubMedPubMedCentralCrossRef Beildeck ME, Islam M, Shah S, Welsh J, Byers SW. Control of TCF-4 expression by VDR and vitamin D in the mouse mammary gland and colorectal cancer cell lines. PLoS ONE. 2009;4: e7872.ADSPubMedPubMedCentralCrossRef
277.
go back to reference Davey RA, Grossmann M. Androgen Receptor Structure, Function and Biology: From Bench to Bedside. Clin Biochem Rev. 2016;37:3–15.PubMedPubMedCentral Davey RA, Grossmann M. Androgen Receptor Structure, Function and Biology: From Bench to Bedside. Clin Biochem Rev. 2016;37:3–15.PubMedPubMedCentral
280.
go back to reference Buchanan G, Birrell SN, Peters AA, Bianco-Miotto T, Ramsay K, Cops EJ, Yang M, Harris JM, Simila HA, Moore NL. Decreased androgen receptor levels and receptor function in breast cancer contribute to the failure of response to medroxyprogesterone acetate. Can Res. 2005;65:8487–96.CrossRef Buchanan G, Birrell SN, Peters AA, Bianco-Miotto T, Ramsay K, Cops EJ, Yang M, Harris JM, Simila HA, Moore NL. Decreased androgen receptor levels and receptor function in breast cancer contribute to the failure of response to medroxyprogesterone acetate. Can Res. 2005;65:8487–96.CrossRef
281.
go back to reference Castellano I, Allia E, Accortanzo V, Vandone AM, Chiusa L, Arisio R, Durando A, Donadio M, Bussolati G, Coates AS. Androgen receptor expression is a significant prognostic factor in estrogen receptor positive breast cancers. Breast Cancer Res Treat. 2010;124:607–17.PubMedCrossRef Castellano I, Allia E, Accortanzo V, Vandone AM, Chiusa L, Arisio R, Durando A, Donadio M, Bussolati G, Coates AS. Androgen receptor expression is a significant prognostic factor in estrogen receptor positive breast cancers. Breast Cancer Res Treat. 2010;124:607–17.PubMedCrossRef
282.
go back to reference Gucalp A, Traina TA. Triple-negative breast cancer: role of the androgen receptor. Cancer J. 2010;16:62–5.PubMedCrossRef Gucalp A, Traina TA. Triple-negative breast cancer: role of the androgen receptor. Cancer J. 2010;16:62–5.PubMedCrossRef
283.
go back to reference Lønning PE. Additive endocrine therapy for advanced breast cancer–back to the future. Acta Oncol. 2009;48:1092–101.PubMedCrossRef Lønning PE. Additive endocrine therapy for advanced breast cancer–back to the future. Acta Oncol. 2009;48:1092–101.PubMedCrossRef
284.
go back to reference Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Harris JM, Jindal S, Segara D, Jia L, Moore NL. Androgen receptor inhibits estrogen receptor-α activity and is prognostic in breast cancer. Can Res. 2009;69:6131–40.CrossRef Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Harris JM, Jindal S, Segara D, Jia L, Moore NL. Androgen receptor inhibits estrogen receptor-α activity and is prognostic in breast cancer. Can Res. 2009;69:6131–40.CrossRef
294.
296.
go back to reference Kim K, Kumagai J, Eishi Y, Ishige I, Ishige Y, Koike M. Clonality analysis for normal and cancerous colon tissues with human androgen receptor gene polymerase chain reaction. J Med Dent Sci. 2005;52:163–70.PubMed Kim K, Kumagai J, Eishi Y, Ishige I, Ishige Y, Koike M. Clonality analysis for normal and cancerous colon tissues with human androgen receptor gene polymerase chain reaction. J Med Dent Sci. 2005;52:163–70.PubMed
300.
go back to reference Bharathkumar H, Mohan CD, Ananda H, Fuchs JE, Li F, Rangappa S, Surender M, Bulusu KC, Girish KS, Sethi G. Microwave-assisted synthesis, characterization and cytotoxic studies of novel estrogen receptor α ligands towards human breast cancer cells. Bioorg Med Chem Lett. 2015;25:1804–7.PubMedCrossRef Bharathkumar H, Mohan CD, Ananda H, Fuchs JE, Li F, Rangappa S, Surender M, Bulusu KC, Girish KS, Sethi G. Microwave-assisted synthesis, characterization and cytotoxic studies of novel estrogen receptor α ligands towards human breast cancer cells. Bioorg Med Chem Lett. 2015;25:1804–7.PubMedCrossRef
307.
go back to reference Berner C, Aumüller E, Gnauck A, Nestelberger M, Just A, Haslberger AG. Epigenetic control of estrogen receptor expression and tumor suppressor genes is modulated by bioactive food compounds. Ann Nutr Metab. 2010;57:183–9.PubMedCrossRef Berner C, Aumüller E, Gnauck A, Nestelberger M, Just A, Haslberger AG. Epigenetic control of estrogen receptor expression and tumor suppressor genes is modulated by bioactive food compounds. Ann Nutr Metab. 2010;57:183–9.PubMedCrossRef
316.
go back to reference Trauner M, Gulamhusein A, Hameed B, Caldwell S, Shiffman ML, Landis C, Eksteen B, Agarwal K, Muir A, Rushbrook S, et al. The nonsteroidal farnesoid X receptor agonist cilofexor (GS-9674) improves markers of cholestasis and liver injury in patients with primary sclerosing cholangitis. Hepatology. 2019;70:788–801. https://doi.org/10.1002/hep.30509.CrossRefPubMed Trauner M, Gulamhusein A, Hameed B, Caldwell S, Shiffman ML, Landis C, Eksteen B, Agarwal K, Muir A, Rushbrook S, et al. The nonsteroidal farnesoid X receptor agonist cilofexor (GS-9674) improves markers of cholestasis and liver injury in patients with primary sclerosing cholangitis. Hepatology. 2019;70:788–801. https://​doi.​org/​10.​1002/​hep.​30509.CrossRefPubMed
321.
go back to reference Giaginis C, Karandrea D, Alexandrou P, Giannopoulou I, Tsourouflis G, Troungos C, Danas E, Keramopoulos A, Patsouris E, Nakopoulou L, et al. High Farnesoid X Receptor (FXR) expression is a strong and independent prognosticator in invasive breast carcinoma. Neoplasma. 2017;64:633–9. https://doi.org/10.4149/neo_2017_420.CrossRefPubMed Giaginis C, Karandrea D, Alexandrou P, Giannopoulou I, Tsourouflis G, Troungos C, Danas E, Keramopoulos A, Patsouris E, Nakopoulou L, et al. High Farnesoid X Receptor (FXR) expression is a strong and independent prognosticator in invasive breast carcinoma. Neoplasma. 2017;64:633–9. https://​doi.​org/​10.​4149/​neo_​2017_​420.CrossRefPubMed
322.
go back to reference Girisa S, Parama D, Harsha C, Banik K, Kunnumakkara AB. Potential of guggulsterone, a farnesoid X receptor antagonist, in the prevention and treatment of cancer. Explor Target Antitumor Ther. 2020;1:313–42.PubMedPubMedCentralCrossRef Girisa S, Parama D, Harsha C, Banik K, Kunnumakkara AB. Potential of guggulsterone, a farnesoid X receptor antagonist, in the prevention and treatment of cancer. Explor Target Antitumor Ther. 2020;1:313–42.PubMedPubMedCentralCrossRef
340.
go back to reference Gao BC, Chen Q, Sun T, Wang DM, Xiao J. Research progress of the relationship between liver X receptor and inflammatory-related diseases. Sheng Li Xue Bao. 2018;70:335–41.PubMed Gao BC, Chen Q, Sun T, Wang DM, Xiao J. Research progress of the relationship between liver X receptor and inflammatory-related diseases. Sheng Li Xue Bao. 2018;70:335–41.PubMed
352.
go back to reference Wilson AJ, Arango D, Mariadason JM, Heerdt BG, Augenlicht LH. TR3/Nur77 in colon cancer cell apoptosis. Cancer Res. 2003;63:5401–7.PubMed Wilson AJ, Arango D, Mariadason JM, Heerdt BG, Augenlicht LH. TR3/Nur77 in colon cancer cell apoptosis. Cancer Res. 2003;63:5401–7.PubMed
355.
go back to reference Sriraman V, Rudd MD, Lohmann SM, Mulders SM, Richards JS. Cyclic guanosine 5′-monophosphate-dependent protein kinase II is induced by luteinizing hormone and progesterone receptor-dependent mechanisms in granulosa cells and cumulus oocyte complexes of ovulating follicles. Mol Endocrinol. 2006;20:348–61. https://doi.org/10.1210/me.2005-0317.CrossRefPubMed Sriraman V, Rudd MD, Lohmann SM, Mulders SM, Richards JS. Cyclic guanosine 5′-monophosphate-dependent protein kinase II is induced by luteinizing hormone and progesterone receptor-dependent mechanisms in granulosa cells and cumulus oocyte complexes of ovulating follicles. Mol Endocrinol. 2006;20:348–61. https://​doi.​org/​10.​1210/​me.​2005-0317.CrossRefPubMed
356.
go back to reference Mirza AZ, Althagafi II, Shamshad H. Role of PPAR receptor in different diseases and their ligands: physiological importance and clinical implications. Eur J Med Chem. 2019;166:502–13.PubMedCrossRef Mirza AZ, Althagafi II, Shamshad H. Role of PPAR receptor in different diseases and their ligands: physiological importance and clinical implications. Eur J Med Chem. 2019;166:502–13.PubMedCrossRef
359.
go back to reference Strosznajder AK, Wójtowicz S, Jeżyna MJ, Sun GY, Strosznajder JB. Recent insights on the role of PPAR-β/δ in neuroinflammation and neurodegeneration, and its potential target for therapy. NeuroMol Med. 2021;23:86–98.CrossRef Strosznajder AK, Wójtowicz S, Jeżyna MJ, Sun GY, Strosznajder JB. Recent insights on the role of PPAR-β/δ in neuroinflammation and neurodegeneration, and its potential target for therapy. NeuroMol Med. 2021;23:86–98.CrossRef
364.
go back to reference Sarraf P, Mueller E, Jones D, King FJ, DeAngelo DJ, Partridge JB, Holden SA, Chen LB, Singer S, Fletcher C. Differentiation and reversal of malignant changes in colon cancer through PPARγ. Nat Med. 1998;4:1046–52.PubMedCrossRef Sarraf P, Mueller E, Jones D, King FJ, DeAngelo DJ, Partridge JB, Holden SA, Chen LB, Singer S, Fletcher C. Differentiation and reversal of malignant changes in colon cancer through PPARγ. Nat Med. 1998;4:1046–52.PubMedCrossRef
366.
go back to reference Pishvaian MJ, Marshall JL, Wagner AJ, Hwang JJ, Malik S, Cotarla I, Deeken JF, He AR, Daniel H, Halim AB, et al. A phase 1 study of efatutazone, an oral peroxisome proliferator-activated receptor gamma agonist, administered to patients with advanced malignancies. Cancer. 2012;118:5403–13. https://doi.org/10.1002/cncr.27526.CrossRefPubMed Pishvaian MJ, Marshall JL, Wagner AJ, Hwang JJ, Malik S, Cotarla I, Deeken JF, He AR, Daniel H, Halim AB, et al. A phase 1 study of efatutazone, an oral peroxisome proliferator-activated receptor gamma agonist, administered to patients with advanced malignancies. Cancer. 2012;118:5403–13. https://​doi.​org/​10.​1002/​cncr.​27526.CrossRefPubMed
367.
go back to reference Komatsu Y, Yoshino T, Yamazaki K, Yuki S, Machida N, Sasaki T, Hyodo I, Yachi Y, Onuma H, Ohtsu A. Phase 1 study of efatutazone, a novel oral peroxisome proliferator-activated receptor gamma agonist, in combination with FOLFIRI as second-line therapy in patients with metastatic colorectal cancer. Invest New Drugs. 2014;32:473–80. https://doi.org/10.1007/s10637-013-0056-3.CrossRefPubMed Komatsu Y, Yoshino T, Yamazaki K, Yuki S, Machida N, Sasaki T, Hyodo I, Yachi Y, Onuma H, Ohtsu A. Phase 1 study of efatutazone, a novel oral peroxisome proliferator-activated receptor gamma agonist, in combination with FOLFIRI as second-line therapy in patients with metastatic colorectal cancer. Invest New Drugs. 2014;32:473–80. https://​doi.​org/​10.​1007/​s10637-013-0056-3.CrossRefPubMed
368.
go back to reference Xing Y, Yan J, Niu Y. PXR: a center of transcriptional regulation in cancer. Acta Pharmaceutica Sinica B. 2020;10:197–206.PubMedCrossRef Xing Y, Yan J, Niu Y. PXR: a center of transcriptional regulation in cancer. Acta Pharmaceutica Sinica B. 2020;10:197–206.PubMedCrossRef
380.
go back to reference Fathi N, Ahmadian E, Shahi S, Roshangar L, Khan H, Kouhsoltani M, Dizaj SM, Sharifi S. Role of vitamin D and vitamin D receptor (VDR) in oral cancer. Biomed Pharmacother. 2019;109:391–401.PubMedCrossRef Fathi N, Ahmadian E, Shahi S, Roshangar L, Khan H, Kouhsoltani M, Dizaj SM, Sharifi S. Role of vitamin D and vitamin D receptor (VDR) in oral cancer. Biomed Pharmacother. 2019;109:391–401.PubMedCrossRef
381.
go back to reference Jeon S-M, Shin E. Exploring vitamin D metabolism and function in cancer. Exp Mol Med. 2018;50:1–14.PubMed Jeon S-M, Shin E. Exploring vitamin D metabolism and function in cancer. Exp Mol Med. 2018;50:1–14.PubMed
Metadata
Title
Delineating the role of nuclear receptors in colorectal cancer, a focused review
Authors
Mukesh Kumar Manickasamy
Sujitha Jayaprakash
Sosmitha Girisa
Aviral Kumar
Hiu Yan Lam
Elena Okina
Huiyan Eng
Mohammed S. Alqahtani
Mohamed Abbas
Gautam Sethi
Alan Prem Kumar
Ajaikumar B. Kunnumakkara
Publication date
01-12-2024
Publisher
Springer US
Published in
Discover Oncology / Issue 1/2024
Print ISSN: 1868-8497
Electronic ISSN: 2730-6011
DOI
https://doi.org/10.1007/s12672-023-00808-x

Other articles of this Issue 1/2024

Discover Oncology 1/2024 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine