1
|
Banibakhsh A, Sidhu D, Khan S, Haime H, Foster PA. Sex steroid metabolism and action in colon health and disease. J Steroid Biochem Mol Biol 2023; 233:106371. [PMID: 37516405 DOI: 10.1016/j.jsbmb.2023.106371] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 07/31/2023]
Abstract
The colon is the largest hormonally active tissue in the human body. It has been known for over a hundred years that various hormones and bioactive peptides play important roles in colon function. More recently there is a growing interest in the role the sex steroids, oestrogens and androgens, may play in both normal colon physiology and colon pathophysiology. In this review, we examine the potential role oestrogens and androgens play in the colon. The metabolism and subsequent action of sex steroids in colonic tissue is discussed and how these hormones impact colon motility is investigated. Furthermore, we also determine how oestrogens and androgens influence colorectal cancer incidence and development and highlight potential new therapeutic targets for this malignancy. This review also examines how sex steroids potentially impact the severity and progression of other colon disease, such as diverticulitis, irritable bowel syndrome, and polyp formation.
Collapse
Affiliation(s)
- Afnan Banibakhsh
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, Birmingham B15 2TT, UK
| | - Daljit Sidhu
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, Birmingham B15 2TT, UK
| | - Sunera Khan
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, Birmingham B15 2TT, UK
| | - Hope Haime
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, Birmingham B15 2TT, UK
| | - Paul A Foster
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes, and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK.
| |
Collapse
|
2
|
Bouras E, Papandreou C, Tzoulaki I, Tsilidis KK. Endogenous sex steroid hormones and colorectal cancer risk: a systematic review and meta-analysis. Discov Oncol 2021; 12:8. [PMID: 35201467 PMCID: PMC8777537 DOI: 10.1007/s12672-021-00402-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/02/2021] [Indexed: 12/24/2022] Open
Abstract
Preclinical data suggest that endogenous sex steroid hormones may be implicated in colorectal cancer (CRC) development, however, findings from epidemiological studies are conflicting. The aim of this systematic review and meta-analysis was to investigate the associations between endogenous concentrations of sex hormones and CRC risk. PubMed and Scopus were searched until June 2020 for prospective studies evaluating the association between pre-diagnostic plasma/serum concentrations of estradiol, testosterone and sex-hormone binding globulin (SHBG) and CRC risk. Summary relative risks (RRs) and 95% confidence intervals (CIs) were estimated using the inverse-variance weighted random-effects model based on the DerSimonian-Laird estimator. Eight studies were included in the meta-analysis after evaluating 3,859 non-duplicate records. Four of the eight studies had a nested case-control design, one study was a case-cohort and the rest three studies were cohort studies, and they included on average 295 cases (range:48-732) and 2,105 controls. No associations were found for endogenous sex steroid hormones in men or post-menopausal women with CRC risk, with evidence for substantial heterogeneity observed among women. Findings from this meta-analysis do not support presence of associations between pre-diagnostic concentrations of testosterone, estradiol and SHBG with incident CRC risk in men and post-menopausal women.
Collapse
Affiliation(s)
- Emmanouil Bouras
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | | | - Ioanna Tzoulaki
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Konstantinos K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece.
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, London, W2 1PG, UK.
| |
Collapse
|
3
|
Bouguen G, Dubuquoy L, Desreumaux P, Brunner T, Bertin B. Intestinal steroidogenesis. Steroids 2015; 103:64-71. [PMID: 25560486 DOI: 10.1016/j.steroids.2014.12.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 12/23/2014] [Indexed: 12/26/2022]
Abstract
Steroids are fundamental hormones that control a wide variety of physiological processes such as metabolism, immune functions, and sexual characteristics. Historically, steroid synthesis was considered a function restricted to the adrenals and the gonads. In the past 20 years, a significant number of studies have demonstrated that steroids could also be synthesized or metabolized by other organs. According to these studies, the intestine appears to be a major source of de novo produced glucocorticoids as well as a tissue capable of producing and metabolizing sex steroids. This finding is based on the detection of steroidogenic enzyme expression as well as the presence of bioactive steroids in both the rodent and human gut. Within the intestinal mucosa, the intestinal epithelial cell layer is one of the main cellular sources of steroids. Glucocorticoid synthesis regulation in the intestinal epithelial cells is unique in that it does not involve the classical positive regulator steroidogenic factor-1 (SF-1) but a closely related homolog, namely the liver receptor homolog-1 (LRH-1). This local production of immunoregulatory glucocorticoids contributes to intestinal homeostasis and has been linked to pathophysiology of inflammatory bowel diseases. Intestinal epithelial cells also possess the ability to metabolize sex steroids, notably estrogen; this mechanism may impact colorectal cancer development. In this review, we contextualize and discuss what is known about intestinal steroidogenesis and regulation as well as the key role these functions play both in physiological and pathological conditions.
Collapse
Affiliation(s)
- Guillaume Bouguen
- Service des Maladies de l'Appareil digestif, CHU Pontchaillou, Rennes, France; UMR991, Liver Metabolism and Cancer, France; Université de Rennes 1, France
| | - Laurent Dubuquoy
- Université de Lille, F-59000 Lille, France; Inserm U995, F-59045 Lille, France
| | - Pierre Desreumaux
- Université de Lille, F-59000 Lille, France; Inserm U995, F-59045 Lille, France; CHU Lille, Service des Maladies de l'Appareil Digestif et de la Nutrition, Hôpital Claude Huriez, F-59037 Lille, France
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Germany
| | - Benjamin Bertin
- Université de Lille, F-59000 Lille, France; Inserm U995, F-59045 Lille, France; Faculté des Sciences Pharmaceutiques et Biologiques, F-59006 Lille, France.
| |
Collapse
|
4
|
Basu A, Seth S, Arora K, Verma M. Evaluating estradiol levels in male patients with colorectal carcinoma. J Clin Diagn Res 2015; 9:BC08-10. [PMID: 25737973 PMCID: PMC4347064 DOI: 10.7860/jcdr/2015/10508.5397] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 11/13/2014] [Indexed: 11/24/2022]
Abstract
BACKGROUND Globally more than 1 million people suffer from colorectal cancer (CRC) per annum, resulting in about 0.5 million deaths. The role of estrogen in CRC is being researched with great interest; expression of estrogen receptors (alfa and beta) is being explored. AIMS AND OBJECTIVE Our objective was to compare the serum estradiol levels in diagnosed male patients of CRC, with age-matched controls; and to study the estradiol levels across the different stages of CRC. SETTING AND DESIGN A cross-sectional study was conducted from January, 2012 to March, 2013 at a tertiary care hospital in north India. MATERIALS AND METHODS Fifty one male preoperative CRC patients were enrolled along with 50 age-matched male controls. Ethical approval and informed written consent from each participant were duly obtained. CRC patients were staged as per TNM (T- Tumour, N- Node, M- Metastasis; I, II, III and IV) criteria. Serum estradiol level was measured by Chemiimmunofluroscence method (normal = 11.6 - 41.2 pg/ml). STATISTICAL ANALYSIS USED We used student's t test and ANOVA (analysis of variance) to analyse the data (SPSS version 17.0, SPSS, Inc., Chicago, Illinois) Result: The mean serum estradiol level among CRC patients (43.4, sd=27.1) was significantly more than that among controls (mean=24.7, sd=17.5), (p<0.0001). Across the four TNM stages of CRC patients, mean estradiol level was highest in Stage II (55.9, sd=15.5); followed by Stages III (44.1, sd=24.9), IV (36.3, sd=30.0) and I (26.4, sd=38.8). However, significant difference was obtained only between Stages I and II. CONCLUSION Our study revealed increased levels of serum estradiol in Indian male CRC patients. Further research is warranted to corroborate this finding, and to understand the role of estradiol across different TNM stages of CRC.
Collapse
Affiliation(s)
- Atreyee Basu
- Senior Resident, Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India
| | - Shashi Seth
- Senior Professor, Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India
| | - Kanchan Arora
- Senior Resident, Department of Biochemistry, VMMC and Safdarjung Hospital, New Delhi, India
| | - Monica Verma
- Senior Resident, Department of Biochemistry, Post Graduate Institute of Medical Sciences (PGIMS), Rohtak, Haryana, India
| |
Collapse
|
5
|
Janakiram NB, Mohammed A, Brewer M, Bryant T, Biddick L, Lightfoot S, Pathuri G, Gali H, Rao CV. Raloxifene and antiestrogenic gonadorelin inhibits intestinal tumorigenesis by modulating immune cells and decreasing stem-like cells. Cancer Prev Res (Phila) 2014; 7:300-9. [PMID: 24431404 DOI: 10.1158/1940-6207.capr-13-0345] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Studies suggest that estrogen plays a contributing role in colorectal cancer. This project examined the preventive effects of raloxifene, a selective estrogen receptor modulator (SERM), and gonadorelin, an antiestrogenic drug, in female Apc(Min/+) mouse intestinal tumorigenesis. Six-week-old Apc(Min/+)mice were fed diet containing 1 ppm raloxifene or control diet. Gonadorelin (150 ng/mouse) was injected subcutaneously into one treatment group. Intestinal tumors were evaluated for tumor multiplicity and size. Mice treated with raloxifene and gonadorelin showed colon tumor inhibition of 80% and 75%, respectively. Both drugs significantly inhibited small intestinal tumor multiplicity and size (75%-65%, P < 0.0001). Raloxifene and gonadorelin showed significant tumor inhibition with 98% and 94% inhibition of polyps >2 mm in size. In mice fed with raloxifene or injected with gonadorelin, tumors showed significantly reduced proliferating cell nuclear antigen expression (58%-65%, P < 0.0001). Raloxifene treatment decreased β-catenin, cyclin D1, laminin 1β, Ccl6, and stem-like cells (Lgr 5, EpCAM, CD44/CD24), as well as suppressed inflammatory genes (COX-2, mPGES-1, 5-LOX,). Gonadorelin showed significant decrease in COX-2, mPGES-1, iNOS, and stem-like cells or increased NK cells and chemokines required for NK cells. Both drugs were effective in suppressing tumor growth albeit with different mechanisms. These observations show that either suppression of estrogen levels or modulation of estrogen receptor dramatically suppresses small intestinal and colonic tumor formation in female Apc(Min/+) mice. These results support the concept of chemoprevention by these agents in reducing endogenous levels of estrogen or modulating ER signaling.
Collapse
Affiliation(s)
- Naveena B Janakiram
- Center for Cancer Prevention and Drug Development (CCPDD), Department of Medicine, Hematology Oncology Section, 975 NE 10th Street, BRC 1209, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104.
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Foster PA. Oestrogen and colorectal cancer: mechanisms and controversies. Int J Colorectal Dis 2013; 28:737-49. [PMID: 23319136 DOI: 10.1007/s00384-012-1628-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2012] [Indexed: 02/06/2023]
Abstract
AIM The role of oestrogen metabolism and action in colorectal cancer (CRC) is controversial. An extensive review of the current literature, encompassing epidemiological evidence, systemic and peripheral oestrogen concentrations, 17β-hydroxysteroid dehydrogenase (17β-HSD) and aromatase in CRC, steroid sulphatase (STS)/oestrone sulphotransferase (EST) and in vitro and in vivo genomic effects was therefore undertaken. METHODS A literature search (key words: colorectal cancer, oestrogen, oestrogen receptor, 17β-HSD, STS, organic anion transporter) was performed using Embase, Medline, and Pubmed and papers were evaluated on scientific relevance on an individual basis. RESULTS Epidemiological data highlights that premenopausal women, or postmenopausal women taking hormone replacement therapy, are significantly less likely than males to develop CRC. This implies that oestrogen signalling is most likely involved in CRC physiology and aetiology. Little is known about oestrogen metabolism in the colon. However, the expression of 17β-HSD, STS, and EST, enzymes involved in oestrogen metabolism, have shown prognostic significance. Evidence also suggests that protective effects are modulated through oestrogen receptor beta, although which metabolite of oestrogen, oestradiol (E2) or oestrone (E1), is more active remains undefined. To complicate matters, the changes in the peripheral ratios of these enzymes, oestrogens and receptors most likely influences CRC progression. CONCLUSION Epidemiological evidence, now supported by in vitro and in vivo studies, strongly associates oestrogen action and metabolism with CRC. Initially protective against CRC, once developed, results suggests that oestrogens increase proliferation. Consequently, hormone-ablation therapy, already successful against breast and prostate cancer, may be effective against CRC.
Collapse
Affiliation(s)
- Paul A Foster
- Centre for Endocrinology, Diabetes and Metabolism, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
7
|
Estrogen in obesity-associated colon cancer: friend or foe? Protecting postmenopausal women but promoting late-stage colon cancer. Cancer Causes Control 2012; 23:1767-73. [PMID: 23011535 DOI: 10.1007/s10552-012-0066-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/11/2012] [Indexed: 12/12/2022]
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.
Collapse
|
8
|
Singh P, Sarkar S, Kantara C, Maxwell C. Progastrin Peptides Increase the Risk of Developing Colonic Tumors: Impact on Colonic Stem Cells. CURRENT COLORECTAL CANCER REPORTS 2012; 8:277-289. [PMID: 23226720 DOI: 10.1007/s11888-012-0144-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pre-neoplastic lesions (ACF, aberrant-crypt-foci; Hp, hyperplastic/dysplastic polyps) are believed to be precursors of sporadic colorectal-tumors (Ad, adenomas; AdCA, adenocarcinomas). ACF/Hp likely originate due to abnormal growth of colonic-crypts in response to aberrant queues in the microenvironment of colonic-crypts. Thus identifying factors which regulate homeostatic vs aberrant proliferation/apoptosis of colonocytes, especially stem/progenitor cells, may lead to effective preventative/treatment strategies. Based on this philosophy, role of growth-factors/peptide-hormones, potentially available in the circulation/microenvironment of colonic-crypts is being examined extensively. Since the time gastrins were discovered as trophic (growth) factors for gastrointestinal-cells, the effect of gastrins on the growth of normal/cancer cells has been investigated, leading to many discoveries. Seminal discoveries made in the area of gastrins and colon-cancer, as it relates to molecular pathways associated with formation of colonic tumors will be reviewed, and possible impact on diagnostic/preventative/treatment strategies will be discussed.
Collapse
Affiliation(s)
- Pomila Singh
- Department of Neuroscience and Cell Biology, UTMB, Galveston TX 77555
| | | | | | | |
Collapse
|
9
|
Zervoudakis A, Strickler HD, Park Y, Xue X, Hollenbeck A, Schatzkin A, Gunter MJ. Reproductive history and risk of colorectal cancer in postmenopausal women. J Natl Cancer Inst 2011; 103:826-34. [PMID: 21447807 DOI: 10.1093/jnci/djr101] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND There are conflicting data regarding the role of sex hormones in colorectal cancer development. Whereas clinical trials data indicate that hormone therapy use reduces the risk of colorectal cancer, data from prospective cohort studies suggest that circulating estrogen levels are positively associated with colorectal cancer risk. A surrogate measure of lifetime estrogen exposure is reproductive history. We investigated the relationship between reproductive factors and the risk of colorectal cancer. METHODS Subjects were postmenopausal women enrolled in the National Institutes of Health-American Association of Retired Persons Diet and Health Study, a cohort of 214,162 individuals (aged 50-71 years) that included 2014 incident cases of colorectal cancer that occurred over a mean follow-up of 8.2 years. Questionnaires were used to collect data on reproductive factors, including ages at menarche, birth of first child, and menopause; parity, and use of oral contraceptives. Multivariable Cox proportional hazards models were constructed to examine associations between these reproductive factors and the risk of colorectal cancer, with adjustment for established colorectal cancer risk factors. All statistical tests were two-sided. RESULTS Age at menopause (≥ 55 vs < 40 years: hazard ratio [HR] = 1.50, 95% confidence interval [CI] = 1.23 to 1.83; P(trend) = .008) and age at birth of first child (≥ 30 vs ≤ 19 years: HR = 1.26, 95% CI = 1.01 to 1.58; P(trend) = .05) were positively associated with the risk of colorectal cancer. Among women with no history of hormone therapy use, age at menarche (≥ 15 vs 11-12 years: HR = 0.73, 95% CI = 0.57 to 0.94; P(trend) = .02) and parity (≥ 5 children vs no children: HR = 0.80, 95% CI = 0.63 to 1.02; P(trend) = .10) were inversely associated with the risk of colorectal cancer. CONCLUSION These data support a role for sex hormones in colorectal tumorigenesis and suggest that greater endogenous estrogen exposure may increase the risk of colorectal cancer in postmenopausal women.
Collapse
|
10
|
Grivas PD, Tzelepi V, Sotiropoulou-Bonikou G, Kefalopoulou Z, Papavassiliou AG, Kalofonos H. Estrogen receptor alpha/beta, AIB1, and TIF2 in colorectal carcinogenesis: do coregulators have prognostic significance? Int J Colorectal Dis 2009; 24:613-22. [PMID: 19198856 DOI: 10.1007/s00384-009-0647-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/09/2009] [Indexed: 02/06/2023]
Abstract
PURPOSE Estrogen receptor beta (ER beta) is abundantly expressed in colorectal tissue, but its role in colorectal carcinogenesis remains elusive. Estrogen receptor coregulators, amplified in breast cancer 1 (AIB1) and transcription intermediary factor 2 (TIF2), have been well-characterized, but their expression in colorectal carcinomas has not been investigated. MATERIALS AND METHODS Estrogen receptor alpha (ER alpha), ER beta, AIB1, and TIF2 protein expression were evaluated by immunohistochemistry in colorectal normal mucosa, adenomas, and adenocarcinomas from 110 patients with colorectal cancer. RESULTS ER alpha expression was rare in colorectal tissue and its expression does not appear to be associated with colorectal carcinogenesis. ER beta, AIB1, and TIF2 were detected in the nucleus of epithelial, endothelial, inflammatory, smooth muscle cells, and myofibroblasts. The expression of the three proteins was significantly increased in epithelial cells of carcinomas compared to normal mucosa. In carcinomas, a significant correlation between the levels of expression of AIB1 and TIF2 was noted. Although AIB1 overexpression was associated with local tumor invasion, it was also found to correlate independently with prolonged overall survival. CONCLUSIONS ER beta, AIB1, and TIF2 appear to be involved in colorectal tumorigenesis and might have prognostic significance.
Collapse
Affiliation(s)
- Petros D Grivas
- Division of Oncology and Clinical Oncology Laboratory, Medical School, University of Patras, 26504, Patras, Greece
| | | | | | | | | | | |
Collapse
|
11
|
Clendenen TV, Koenig KL, Shore RE, Levitz M, Arslan AA, Zeleniuch-Jacquotte A. Postmenopausal levels of endogenous sex hormones and risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev 2009; 18:275-81. [PMID: 19124509 DOI: 10.1158/1055-9965.epi-08-0777] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Observational epidemiologic studies and randomized trials have reported a protective effect of oral hormonal replacement therapy on risk of colorectal cancer. Only one previous prospective study, the Women's Health Initiative Observational Study, has reported on the relationship between endogenous hormones and incident colorectal cancer. Contrary to expectation, the investigators found that women with higher circulating estradiol levels were at increased risk of developing colorectal cancer. We conducted a case-control study nested within the New York University Women's Health Study prospective cohort to evaluate the association between endogenous levels of estrone, estradiol, and sex hormone-binding globulin (SHBG) with risk of colorectal cancer. We measured hormones and SHBG in serum samples collected at enrollment from a total of 148 women who subsequently developed colorectal cancer and 293 matched controls. Circulating estrone levels were positively associated with risk of colorectal cancer: The odds ratio for the highest versus lowest quartile of estrone was 1.8 (95% confidence interval, 1.0-3.3). We found a nonsignificant inverse association between SHBG and colorectal cancer, which disappeared after adjusting for body mass index. We did not find an association between estradiol and colorectal cancer risk, but we cannot rule out a potential association because of substantial laboratory error in the measurement. Our results suggest that endogenous estrone is associated with increased risk of colorectal cancer in postmenopausal women.
Collapse
Affiliation(s)
- Tess V Clendenen
- Division of Epidemiology, Department of Environmental Medicine, New York University School of Medicine, 650 First Avenue, 5th Floor, New York, NY 10016-3240, USA
| | | | | | | | | | | |
Collapse
|
12
|
Motylewska E, Mełeń-Mucha G. Estrone and progesterone inhibit the growth of murine MC38 colon cancer line. J Steroid Biochem Mol Biol 2009; 113:75-9. [PMID: 19073257 DOI: 10.1016/j.jsbmb.2008.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Revised: 11/16/2008] [Accepted: 11/19/2008] [Indexed: 10/21/2022]
Abstract
The unsatisfactory effectiveness of reference chemotherapy in colon cancer (fluorouracil - FU) results in continuous search for agents, which could enhance the action of FU. Some epidemiological data such as a decreased risk of colorectal cancer among menopausal women receiving hormonal replacement therapy indicate the role of female sex hormones in the pathogenesis of this disease. The aim of this study was to examine the direct effects of various concentrations of estrone and progesterone (10(-4) to 10(-12)M) applied alone or together with FU on the growth of murine MC38 colon cancer in vitro. Estrone inhibited MC38 cancer growth in a wide range of concentrations (10(-12) to 10(-4)M) with similar potency and at some concentrations (10(-6) and 10(-4)M) augmented also the cytotoxic action of FU. Progesterone induced MC38 cancer growth inhibition at high concentrations (10(-5) to 10(-4)M) in dose- and time-dependent manner but it did not intensify antineoplastic effect of FU. A weak inhibitory effect of progesterone was also observed for lower concentrations (10(-5) to 10(-10)M) in long lasting cultures (72h). The results indicate that estrone and progesterone inhibit the MC38 cancer growth and that estrone increases also the cytotoxic effect of FU, what confirms the role of female sex steroids in modulation of colon cancer growth.
Collapse
Affiliation(s)
- Ewelina Motylewska
- Department of Immunoendocrinology, Chair of Endocrinology, Medical University, Sterling Street 1/3, 91 425 Lodz, Poland.
| | | |
Collapse
|
13
|
Martineti V, Silvestri S, Tonelli F, Brandi ML. Control of colon cancer development and progression by selected estrogen receptor modulators. Expert Rev Endocrinol Metab 2008; 3:503-511. [PMID: 30290437 DOI: 10.1586/17446651.3.4.503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Estrogens behave as protective agents on the development of colorectal cancer, and hormonal-replacement therapy is associated with an increased survival rate in women with this disease, indicating that estrogenic therapy correlates with a better prognosis. The protective effect of estrogens on Fcolorectal cancer development and progression is presumably related to the expression of estrogen receptors in colon mucosa, with the estrogen receptor-β isoform being the predominant one. This observation suggests that estrogen receptor-β could have an inhibitory effect on colorectal cancer cell proliferation and a regulatory effect on colonic mucosa cell growth, opening the discussion on a pharmacologic approach to colorectal cancer prevention and therapy based on estrogenic compounds.
Collapse
Affiliation(s)
- Valentina Martineti
- a Department of Internal Medicine, School of Medicine, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Sandra Silvestri
- b Specialist in Endocrinology and Metabolic Disease, Expert Clinical Research Physician, Internal Medicine, Eli Lilly Italia S.p.A., Via Gramsci 731, 50019 Sesto Fiorentino, FI, Italy.
| | - Francesco Tonelli
- c Department of Clinical Physiopathology, School of Medicine - University of Florence, Viale Morgagni 85, 50134 Firenze, Italy.
| | - Maria Luisa Brandi
- d Department of Internal Medicine, School of Medicine - University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
14
|
Gunter MJ, Hoover DR, Yu H, Wassertheil-Smoller S, Rohan TE, Manson JE, Howard BV, Wylie-Rosett J, Anderson GL, Ho GYF, Kaplan RC, Li J, Xue X, Harris TG, Burk RD, Strickler HD. Insulin, insulin-like growth factor-I, endogenous estradiol, and risk of colorectal cancer in postmenopausal women. Cancer Res 2008; 68:329-37. [PMID: 18172327 DOI: 10.1158/0008-5472.can-07-2946] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity is a risk factor for colorectal cancer, and hyperinsulinemia, a common condition in obese patients, may underlie this relationship. Insulin, in addition to its metabolic effects, has promitotic and antiapoptotic activity that may be tumorigenic. Insulin-like growth factor (IGF)-I, a related hormone, shares sequence homology with insulin, and has even stronger mitogenic effects. However, few prospective colorectal cancer studies directly measured fasting insulin, and none evaluated free IGF-I, or endogenous estradiol, a potential cofactor in postmenopausal women. Therefore, we conducted a case-cohort investigation of colorectal cancer among nondiabetic subjects enrolled in the Women's Health Initiative Observational Study, a prospective cohort of 93,676 postmenopausal women. Fasting baseline serum specimens from all incident colorectal cancer cases (n = 438) and a random subcohort (n = 816) of Women's Health Initiative Observational Study subjects were tested for insulin, glucose, total IGF-I, free IGF-I, IGF binding protein-3, and estradiol. Comparing extreme quartiles, insulin [hazard ratio (HR)(q4-q1), 1.73; 95% confidence interval (CI), 1.16-2.57; P(trend) = 0.005], waist circumference (HR(q4-q1), 1.82; 95% CI, 1.22-2.70; P(trend) = 0.001), and free IGF-I (HR(q4-q1), 1.35; 95% CI, 0.92-1.98; P(trend) = 0.05) were each associated with colorectal cancer incidence in multivariate models. However, these associations each became nonsignificant when adjusted for one another. Endogenous estradiol levels, in contrast, were positively associated with risk of colorectal cancer (HR comparing high versus low levels, 1.53; 95% CI, 1.05-2.22), even after control for insulin, free IGF-I, and waist circumference. These data suggest the existence of at least two independent biological pathways that are related to colorectal cancer: one that involves endogenous estradiol, and a second pathway broadly associated with obesity, hyperinsulinemia, and free IGF-I.
Collapse
Affiliation(s)
- Marc J Gunter
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Colorectal cancer is the third most prevalent cancer in the world. If detected at an early stage, treatment often might lead to cure. As prevention is better than cure, epidemiological studies reveal that having a healthy diet often protects from promoting/ developing cancer. An important consideration in evaluating new drugs and devices is determining whether a product can effectively treat a targeted disease. There are quite a number of biomarkers making their way into clinical trials and few are awaiting the preclinical efficacy and safety results to enter into clinical trials. Researchers are facing challenges in modifying trial design and defining the right control population, validating biomarker assays from the biological and analytical perspective and using biomarker data as a guideline for decision making. In spite of following all guidelines, the results are disappointing from many of the large clinical trials. To avoid these disappointments, selection of biomarkers and its target drug needs to be evaluated in appropriate animal models for its toxicities and efficacies. The focus of this review is on the few of the potential molecular targets and their biomarkers in colorectal cancers. Strengths and limitations of biomarkers/surrogate endpoints are also discussed. Various pathways involved in tumor cells and the specific agents to target the altered molecular biomarker in biomolecular pathway are elucidated. Importance of emerging new platforms siRNAs and miRNAs technology for colorectal cancer therapeutics is reviewed.
Collapse
Affiliation(s)
- Naveena B Janakiram
- Department of Medicine, Hem-Onc Section, OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
16
|
Wuttke W, Jarry H, Seidlová-Wuttke D. Isoflavones--safe food additives or dangerous drugs? Ageing Res Rev 2007; 6:150-88. [PMID: 17604235 DOI: 10.1016/j.arr.2007.05.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 04/26/2007] [Accepted: 05/02/2007] [Indexed: 01/12/2023]
Abstract
The sales volume of products containing isoflavone has increased since the publication of the Women's Health Initiative. The many apparently contradictory results published on the effects of isoflavones on a variety of estrogen-regulated organs point to both beneficial as well as adverse effects on human health. It is of particular importance that psychovegetative climacteric complaints such as hot flushes are, if at all, only slightly influenced by isoflavones. The substances appear to have weak anti-osteoporotic effect. Their anti-atherosclerotic action is debatable, as not all authors find any beneficial effect on lipids. Most importantly, there is dispute as to whether isoflavones derived from soy or red clover have negative, positive or any effect at all on the mammary gland or endometrium. It is beyond any doubt that soy products may have cancer preventing properties in a variety of organs including the mammary gland. However, these properties may only be exerted if the developing organ was under the influence of isoflavones during childhood and puberty. This may also explain the often quoted "Japanese Phenomenon", the fact that breast cancer occurs to a lesser extent in Japanese women. When administered to isoflavone "inexperienced" women at the time of menopause, the phytoestrogens appear to share the same effects as estrogen used in classical preparations for hormone replacement therapy, i.e. they may stimulate the proliferation of endometrial and mammary gland tissue with at present unknown and unpredictable risk to these organs. Therefore, the following question arises for the clinician: Why should soy or red clover products containing isoflavone be recommended, if the positive effects are only negligible but the adverse effects serious?
Collapse
Affiliation(s)
- Wolfgang Wuttke
- Department of Clinical and Experimental Endocrinology, University of Goettingen, Robert-Koch-Str. 40, 37075 Goettingen, Germany.
| | | | | |
Collapse
|
17
|
Abstract
This article represents the first comprehensive assessment of hormetic effects of chemotherapeutic agents. Hormetic dose-response relationships were reported for a wide range of chemotherapeutics, including antibiotics, antiviral, and antitumor agents as well as substances that affect hair growth, prostate function, cognitive performance, and numerous other endpoints. Particular attention was given to assessing the quantitative features of the dose response, the underlying mechanistic features of the biphasic nature of the dose response, and the clinical implications of hormetic responses. Recognition of the hormetic-like biphasic nature of the dose response is expected to have an important impact on the design of experiments to assess chemotherapeutics and how such agents may be employed more successfully in clinical applications.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, Morrill Science Center, University of Massachusetts, Amherst, MA 01003, USA.
| | | |
Collapse
|
18
|
Carothers AM, Hughes SA, Ortega D, Bertagnolli MM. 2-Methoxyestradiol induces p53-associated apoptosis of colorectal cancer cells. Cancer Lett 2002; 187:77-86. [PMID: 12359354 DOI: 10.1016/s0304-3835(02)00409-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Menopausal estrogen replacement therapy is thought to be responsible for the recent decline in colorectal cancer (CRC) incidence among women. In the C57BL/6J-Min/+ mouse, an animal model of CRC, 17beta-estradiol (E(2)) prevents tumor formation in ovariectomized females. We examined human CRC intestinal cell lines to determine whether particular E(2) metabolites produced anti-tumor effects. Treatment of CRC cells with 2-methoxyestradiol (2-MeOE(2)) increased expression of p53 and p21(WAF1/CIP1) proteins and induced apoptosis, but did not produce changes in expression of estrogen receptor (ER)alpha or ERbeta. The finding that 2-MeOE(2) induces p53-mediated colon cell apoptosis in vitro supports a role for 2-MeOE(2) as an endogenous mediator of intestinal tumor suppression.
Collapse
Affiliation(s)
- A M Carothers
- Department of Surgery, Weill College of Medicine of Cornell University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
19
|
Cobb S, Wood T, Tessarollo L, Velasco M, Given R, Varro A, Tarasova N, Singh P. Deletion of functional gastrin gene markedly increases colon carcinogenesis in response to azoxymethane in mice. Gastroenterology 2002; 123:516-30. [PMID: 12145805 DOI: 10.1053/gast.2002.34754] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS We recently reported that transgenic mice overexpressing progastrin were at a higher risk for developing colon cancers in response to azoxymethane (AOM), whereas mice overexpressing gastrin-17 were at a reduced risk. To examine further the role of gastrins in colon carcinogenesis, we generated gastrin gene knockout mice (GAS-KO). METHODS The height and proliferative index (PI) of colonic crypts were similar in GAS-KO and wild-type (WT) mice, suggesting that the absence of gastrins in GAS-KO mice did not significantly affect the growth of colonic mucosa. GAS-KO and WT mice were treated with AOM for 3-4 weeks; control mice received saline. RESULTS Colonic proliferation in response to AOM was significantly increased in GAS-KO vs. WT mice. Aberrant crypt foci (ACFs) were similarly increased significantly by approximately 2-5-fold in GAS-KO vs. WT mice after 2 weeks of AOM treatment. Female GAS-KO mice developed adenomas (Ads) and adenocarcinomas (AdCAs) at earlier times ( approximately 10 months) than the male GAS-KO mice and the male and female WT mice ( approximately 12 months). The total numbers of Ads and AdCAs were significantly higher in GAS-KO than in WT mice. CONCLUSIONS These results suggest the novel possibility that loss of gastrin expression (and hence amidated gastrins) significantly increases susceptibility to colon carcinogenesis in response to AOM. Previous studies with FVB/N transgenic mice similarly suggested a protective role of amidated gastrins against colon carcinogenesis, which supports the present findings of an increase in colon carcinogenesis in GAS-KO mice lacking normal physiological levels of amidated gastrins.
Collapse
Affiliation(s)
- Stephanie Cobb
- Department of Anatomy and Neurosciences, University of Texas Medical Branch, Galveston, Texas 77555-1043, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Witte D, Chirala M, Younes A, Li Y, Younes M. Estrogen receptor beta is expressed in human colorectal adenocarcinoma. Hum Pathol 2001; 32:940-4. [PMID: 11567223 DOI: 10.1053/hupa.2001.27117] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Estrogen receptor beta (ER-beta) has recently been detected in a human colon cancer cell line. The aim of this work was to determine whether ER-beta is expressed in human colorectal carcinoma (CRC) tissue and the extent of this expression. ER-beta expression in CRC was investigated by immunohistochemical staining of sections of formalin-fixed, paraffin-embedded tissue from 55 CRC. The percent of positive cells was recorded. ER-beta immunoreactivity was always present in normal epithelium and adenomas in the same sections of some CRC and was always nuclear. In CRC, nuclear ER-beta immunoreactivity was detected in >10% of the cancer cells in 67% of the cases and was almost always associated with cytoplasmic immunoreactivity. There were no statistically significant differences between the ER-beta-positive and -negative groups in regard to depth of invasion, nodal metastases, or survival, regardless of the cut-off value used. We conclude that (1) a significant number of CRCs are positive for ER-beta. (2) estrogen may play an important role in the proliferation of normal colonic epithelium, and (3) there is differential localization of ER-beta immunoreactivity between normal colon, adenomas, and CRCs. Whether different ER-beta isoforms are differentially expressed in CRCs, and whether human CRCs respond to treatment with antiestrogens, is the subject of studies currently in progress.
Collapse
Affiliation(s)
- D Witte
- Department of Pathology, Baylor College of Medicine and the Methodist Hospital, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
21
|
English MA, Stewart PM, Hewison M. Estrogen metabolism and malignancy: analysis of the expression and function of 17beta-hydroxysteroid dehydrogenases in colonic cancer. Mol Cell Endocrinol 2001; 171:53-60. [PMID: 11165011 DOI: 10.1016/s0303-7207(00)00418-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Age and sex differences in the incidence of gastrointestinal cancers suggest the involvement of sex steroids. Post-menopausal loss of estrogen in women appears to be associated with a lower risk of colonic cancer, and studies in vitro have shown that estradiol (E2) stimulates the growth of colonic cancer cell lines. Paradoxically more recent epidemiological data have shown that hormone replacement therapy (HRT) is associated with a lower risk of colonic cancer, although this may reflect differences in the composition and route of administration of HRT regimes. The precise mechanism by which estrogens influence colonic cancer in vivo remains unclear, although E2-induced growth of colonic cancer cells in vitro appears to be dependent on estrogen receptor (ER) expression. We have previously demonstrated differential responses to E2 in pre-malignant and malignant colonic cancer cell lines, without any apparent difference in ER expression. Analogous to well documented studies in breast cancer, we have postulated that local steroid metabolism in the colon may play a key role in modulating the effects of oestrogens by determining the tissue availability of active E2. Using biopsy material we have shown that the normal colonic mucosa has a high level of 17beta-hydroxysteroid dehydrogenase (17beta-HSD)-mediated E2 metabolism. Furthermore, the predominant enzyme activity, inactivation of E2 to estrone (E1), was significantly decreased in paired tumor biopsies. The presence of 17beta-HSD activity in the colon appears to be due to expression of the type 2 and 4 isozymes of 17beta-HSD (17beta-HSD2 and 4), and expression of mRNA for the latter was shown to be significantly decreased in tumours compared to normal mucosa. Further studies have characterised the expression of 17beta-HSD2 and 4 in colonic epithelial cells and in colonic cancer cell lines, and have suggested a link between estrogen metabolism and colonic cell proliferation. Data reviewed here provide evidence for the importance of 17beta-HSD isozymes as attenuators of E2 bioavailability in the colon, and emphasise a possible role for 17beta-HSD2 and 4 in the pathogenesis of colon cancer.
Collapse
Affiliation(s)
- M A English
- Division of Medical Sciences, Queen Elizabeth Hospital, The University of Birmingham, Edgbaston, B15 2TH, Birmingham, UK
| | | | | |
Collapse
|
22
|
Kaklamanos IG, Bathe OF, Franceschi D, Lazaris AC, Davaris P, Glinatsis M, Golematis BC. Expression of receptors for estrogen and progesterone in malignant colonic mucosa as a prognostic factor for patient survival. J Surg Oncol 1999; 72:225-9. [PMID: 10589038 DOI: 10.1002/(sici)1096-9098(199912)72:4<225::aid-jso8>3.0.co;2-s] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND AND OBJECTIVES Estrogen receptors (ER) and progesterone receptors (PR) have been detected in both normal and malignant colonic mucosa, but the prognostic value of this observation is unknown. We aimed to define the prognostic significance of the presence of ER and PR in malignant cells from colorectal adenocarcinoma specimens. METHODS An immunohistochemical assay for ER and PR was performed on paraffinized sections from 65 colorectal adenocarcinoma specimens. Survival curves were analyzed to define the prognostic implications of ER and PR. RESULTS Twenty nine (45%) tumors tested receptor positive (32% for ER and 23% for PR). Tumors of advanced stage were more likely to express receptors than early stage tumors (56% vs. 32%; P = 0.01). Median survival of patients with neoplasms expressing PR was 30 months. For patients whose tumors did not express any receptors, median survival had not been reached at the time of follow-up (P = 0.04). Similarly, patients with tumors expressing both receptors had significantly reduced survival (median survival = 20 months; P = 0.003). CONCLUSIONS Expression of receptors for sex steroids correlates with advanced stage disease. Expression of PR by the tumor cells is associated with a shorter patient survival. The results suggest that sex steroids may play a role in carcinogenesis and tumor progression.
Collapse
Affiliation(s)
- I G Kaklamanos
- Division of Surgical Oncology, Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, FL 33136, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Manjer J, Janzon L. Covariance of breast cancer incidence with smoking-, oestrogen- and diet-related cancers in pre- and postmenopausal women in Sweden. Med Hypotheses 1999; 52:561-8. [PMID: 10459839 DOI: 10.1054/mehy.1997.0694] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Effects of smoking on breast cancer risk remains controversial. Tar products have been claimed to increase risk, antioestrogenic effects to reduce risk. Another possibility is that associations to smoking have been confounded by diet. The increasing incidence of breast cancer from 1960 to 1994 in Sweden is parallel to that of lung cancer and the increasing proportion of female smokers. The incidence of endometrial and colon cancer was in premenopausal women negatively and in postmenopausal women positively related to the incidence of breast cancer. Possible explanations and hypotheses to the different co-variance between breast cancer and lung, endometrial and colon cancer in pre- and postmenopausal women are discussed from the perspectives of smoking, sex hormones and diet. It is concluded that the strong and specific positive relationship between breast and lung cancer in premenopausal women is compatible with the hypothesis that aromatic hydrocarbons may be involved in the causation of disease.
Collapse
Affiliation(s)
- J Manjer
- Department of Community Medicine, Malmö University Hospital, Lund University, Sweden
| | | |
Collapse
|
24
|
Messina M, Bennink M. Soyfoods, isoflavones and risk of colonic cancer: a review of the in vitro and in vivo data. BAILLIERE'S CLINICAL ENDOCRINOLOGY AND METABOLISM 1998; 12:707-28. [PMID: 10384821 DOI: 10.1016/s0950-351x(98)80012-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Soy foods and soybean components have received considerable attention of late for their potential role in reducing cancer risk. Although the relationship between soy intake and the risk of breast and prostate cancer has been the focus of most interest, the relationship between soy intake and other cancers, including colorectal cancer, has also been studied. Several anti-carcinogens have been identified in soybeans, but most enthusiasm for the potential anti-cancer effects of soy undoubtedly stems from work involving soybean isoflavones. Isoflavones have a limited distribution in nature, and, for practical purposes, soyfoods are the only nutritionally relevant dietary source of these phytochemicals. Isoflavones are weak oestrogens but possess other potentially important biological attributes independent of their ability to bind to the oestrogen receptor. The isoflavone genistein inhibits the growth of most types of hormone-dependent and hormone-independent cancer cells in vitro, including colonic cancer cells. Several mechanisms for the in vitro anti-cancer effects of genistein have been proposed, including effects on signal transduction. A number of epidemiological studies, primarily of Asian origin, have examined the relationship between soy intake and the risk of colorectal cancer. Although these studies provide little support for a protective effect of soy, concerns have been raised about the completeness of the soy intake data, since soy was not the focus of these studies and most of this research was conducted prior to the recent interest in the anti-cancer effects of soy. The effect of soy/isoflavone intake has also been studied in rodents, but again these data are conflicting and provide only modest support for a protective effect. Although the relationship between soy intake and colonic cancer risk is certainly worthy of further investigation, there is, at the moment, very limited support for soy exerting a protective effect against this type of cancer.
Collapse
Affiliation(s)
- M Messina
- Loma Linda University, California, USA
| | | |
Collapse
|
25
|
Xu X, Thomas ML. Biphasic actions of estrogen on colon cancer cell growth: possible mediation by high- and low-affinity estrogen binding sites. Endocrine 1995; 3:661-5. [PMID: 21153224 DOI: 10.1007/bf02746342] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/1995] [Accepted: 06/20/1995] [Indexed: 12/23/2022]
Abstract
The present experiments were carried out to investigate the possible direct effects of estrogens (E) on the growth of colon cancer cells. Estradiol exhibited a concentration-dependent biphasic growth effect on a mouse colon cancer cell line (MC-26). Low concentrations of estradiol (10(-10) M: to 10(-8) M: ) had a growth-stimulatory effect, while higher concentrations (10(-7) M: to 10(-6) M: ) were growth-inhibitory. Estrogen receptor (ER) mRNA as well as specific, saturable binding of estradiol to ER (K(d)=0.3NM: , B(max)=0.72 fmol/μg DNA) was identified in these cells. In addition to the classical high affinity ER, lower affinity, higher capacity estrogen binding sites (K(d)=35MM: , B(max)=30 fmol/μg DNA) were also characterized in MC-26 cells. These two types of estrogen binding sites exhibited distinct binding specificities for E and antiestrogens. Treatment of MC-26 cells with an oligodeoxy-nucleotide antisense to the translation start codon of ER mRNA did not alter the grown-inhibitory effect of 10(-6) M: estradiol, demonstrating that the growth-inhibitory effect of high concentrations of E was not mediated by ER; we have previously shown that under the same conditions, ER antisense oligonucleotides do block the growth-stimulatory effects of 10(-9) M: E(2) in MC-26 cells. The data suggest that physiological concentrations of estradiol acting via the classical ER may have a proliferative effect on the growth of colon cancer cells. However, in situations where there are high luminal concentrations of estrogenic compounds, they may act on low affinity estrogen binding sites that mediate the growth-inhibitory effect.
Collapse
Affiliation(s)
- X Xu
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 77555-1031, Galveston, TX
| | | |
Collapse
|
26
|
Abstract
In vivo and epidemiological data suggest a mitogenic role for estrogens (E) in colon cancer. The presence of estrogen receptor (ER) and ER mRNA in colonic epithelium and colon cancer cells, make it necessary to explore the possible direct effects of E on colon cancer growth. In this study, a 15-mer oligodoxynucleotide (oligo) antisense to the region of the translation start codon of estrogen receptor mRNA inhibited ER expression in a mouse colon cancer cell line (MC-26), as determined by receptor binding assay. Antisense oligo also decreased ER mRNA levels in MC-26 cells. The growth-stimulatory effect of E was abolished by antisense oligo treatment, demonstrating that the ER is directly involved in the regulation of colon cancer cell growth.
Collapse
MESH Headings
- Adenocarcinoma/chemistry
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Animals
- Antineoplastic Agents/pharmacology
- Base Sequence
- Blotting, Northern
- Cell Division/physiology
- Colonic Neoplasms/chemistry
- Colonic Neoplasms/genetics
- Colonic Neoplasms/pathology
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Estrogen Antagonists/pharmacology
- Estrogens/metabolism
- Estrogens/pharmacology
- Fulvestrant
- Mice
- Molecular Sequence Data
- Oligonucleotides, Antisense/analysis
- Oligonucleotides, Antisense/chemistry
- Oligonucleotides, Antisense/genetics
- RNA, Messenger/analysis
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- Receptors, Estrogen/analysis
- Receptors, Estrogen/genetics
- Receptors, Estrogen/physiology
- Tamoxifen/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- X Xu
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston 77555
| | | |
Collapse
|