1
|
Zhao Y, Tan H, Zhang J, Zhan D, Yang B, Hong S, Pan B, Wang N, Chen T, Shi Y, Wang Z. Developing liver-targeted naringenin nanoparticles for breast cancer endocrine therapy by promoting estrogen metabolism. J Nanobiotechnology 2024; 22:122. [PMID: 38504208 PMCID: PMC10953142 DOI: 10.1186/s12951-024-02356-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Endocrine therapy is standard for hormone receptor-positive (HR+) breast cancer treatment. However, current strategies targeting estrogen signaling pay little attention to estradiol metabolism in the liver and is usually challenged by treatment failure. In a previous study, we demonstrated that the natural compound naringenin (NAR) inhibited HR+ breast cancer growth by activating estrogen sulfotransferase (EST) expression in the liver. Nevertheless, the poor water solubility, low bio-barrier permeability, and non-specific distribution limited its clinical application, particularly for oral administration. Here, a novel nano endocrine drug NAR-cell penetrating peptide-galactose nanoparticles (NCG) is reported. We demonstrated that NCG presented specific liver targeting and increased intestinal barrier permeability in both cell and zebrafish xenotransplantation models. Furthermore, NCG showed liver targeting and enterohepatic circulation in mouse breast cancer xenografts following oral administration. Notably, the cancer inhibition efficacy of NCG was superior to that of both NAR and the positive control tamoxifen, and was accompanied by increased hepatic EST expression and reduced estradiol levels in the liver, blood, and tumor tissue. Moreover, few side effects were observed after NCG treatment. Our findings reveal NCG as a promising candidate for endocrine therapy and highlight hepatic EST targeting as a novel therapeutic strategy for HR+ breast cancer.
Collapse
Affiliation(s)
- Yuying Zhao
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hanxu Tan
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Juping Zhang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dandan Zhan
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bowen Yang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shicui Hong
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bo Pan
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Neng Wang
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Yafei Shi
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Zhiyu Wang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Clinical Research On Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Wang J, Feng Y, Liu B, Xie W. Estrogen sulfotransferase and sulfatase in steroid homeostasis, metabolic disease, and cancer. Steroids 2024; 201:109335. [PMID: 37951289 PMCID: PMC10842091 DOI: 10.1016/j.steroids.2023.109335] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
Sulfation and desulfation of steroids are opposing processes that regulate the activation, metabolism, excretion, and storage of steroids, which account for steroid homeostasis. Steroid sulfation and desulfation are catalyzed by cytosolic sulfotransferase and steroid sulfatase, respectively. By modifying and regulating steroids, cytosolic sulfotransferase (SULT) and steroid sulfatase (STS) are also involved in the pathophysiology of steroid-related diseases, such as hormonal dysregulation, metabolic disease, and cancer. The estrogen sulfotransferase (EST, or SULT1E1) is a typical member of the steroid SULTs. This review is aimed to summarize the roles of SULT1E1 and STS in steroid homeostasis and steroid-related diseases.
Collapse
Affiliation(s)
- Jingyuan Wang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ye Feng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Brian Liu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
3
|
Yu S, You J, Shi X, Zou X, Lu Z, Wang Y, Tan J, Sun Z, Li Z, Ji Z, Song C. Rapid Analysis of Estrogens in Meat Samples by High Performance Liquid Chromatography with Fluorescence Detection. J Fluoresc 2024; 34:425-436. [PMID: 37284963 DOI: 10.1007/s10895-023-03248-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/18/2023] [Indexed: 06/08/2023]
Abstract
A novel reagent named 4-(N-methyl-1,3-dioxo-benzoisoquinolin-6-yl-oxy)benzene sulfonyl chloride (MBIOBS-Cl) for the determination of estrogens in food samples by high-performance liquid chromatography (HPLC) with fluorescence detection has been developed. Estrogens could be easily labeled by MBIOBS-Cl in Na2CO3-NaHCO3 buffer solution at pH 10.0. The complete labeling reaction for estrogens could be accomplished within five minutes, the corresponding derivatives exhibited strong fluorescence with the maximum excitation and emission wavelengths at 249 nm and 443 nm, respectively. The derivatization conditions, such as the molar ratio of reagent to estrogens, derivatization time, pH, temperature, and buffers were optimized. Derivatives were sufficiently stable to be efficiently analyzed by HPLC with a reversed-phase Agilent ZORBAX 300SB-C18 column with a good baseline resolution. Excellent linear correlations were obtained for all estrogen derivatives with correlation coefficients greater than 0.9998. Ultrasonic-Assisted extraction was used to optimize the extraction of estrogens from meat samples with a recovery higher than 82%. The detection limits (LOD, S/N = 3) of the method ranged from 0.95 to 3.3 μg· kg-1. The established method, which is fast, simple, inexpensive, and environment friendly, can be successfully applied for the detection of four steroidal estrogens from meat samples with little matrix interference.
Collapse
Affiliation(s)
- Shuiqiang Yu
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Jinmao You
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China.
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, People's Republic of China.
| | - Xinxin Shi
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Xiaocong Zou
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Zhihao Lu
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Yu Wang
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Jiangkun Tan
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Zhiwei Sun
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Zan Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Zhongyin Ji
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Cuihua Song
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| |
Collapse
|
4
|
Wenxuan L, Liu L, Zhang L, Qiu Z, Wu Z, Deng W. Role of gonadally synthesized steroid hormones in the colorectal cancer microenvironment. Front Oncol 2023; 13:1323826. [PMID: 38115900 PMCID: PMC10728810 DOI: 10.3389/fonc.2023.1323826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023] Open
Abstract
Objective To understand the relationship between steroid hormones synthesized by the gonads and colorectal cancer as well as its tumor microenvironment, in the expectation of providing new ideas in order to detect and treat colorectal cancer. Methods Through reviewing the relevant literature at home and abroad, we summarized that androgens promote the growth of colorectal cancer, and estrogens and progesterone help prevent bowel cancer from developing; these three hormones also have a relevant role in the cellular and other non-cellular components of the tumor microenvironment of colorectal cancer. Conclusion The current literature suggests that androgens, estrogens, and progesterone are valuable in diagnosing and treating colorectal cancer, and that androgens promote the growth of colorectal cancer whereas estrogens and progesterone inhibit colorectal cancer, and that, in addition, the receptors associated with them are implicated in the modulation of a variety of cellular components of the microenvironment of colorectal cancer.
Collapse
Affiliation(s)
- Liu Wenxuan
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Li Liu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhendong Qiu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhongkai Wu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
5
|
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
|
6
|
Refaat B, Aslam A, Idris S, Almalki AH, Alkhaldi MY, Asiri HA, Almaimani RA, Mujalli A, Minshawi F, Alamri SA, AlHussain MI, Baltow BA, Alqasmi MH, Basfar GT, Alosaimi OM, Muhayya IA. Profiling estrogen, progesterone, and androgen receptors in colorectal cancer in relation to gender, menopausal status, clinical stage, and tumour sidedness. Front Endocrinol (Lausanne) 2023; 14:1187259. [PMID: 37206439 PMCID: PMC10190606 DOI: 10.3389/fendo.2023.1187259] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Background Although estrogen (ERα/ERβ), progesterone (PGR), and androgen (AR) receptors are pathologically altered in colorectal cancer (CRC), their simultaneous expression within the same cohort of patients was not previously measured. Methods ERα/ERβ/PGR/AR proteins were measured in archived paired normal and malignant colon specimens (n =120 patients) by immunohistochemistry, and results were analyzed by gender, age (≤50 vs. ≥60 years), clinical stages (early-stage I/II vs. late-stage III/IV), and anatomical location (right; RSCs vs. left; LSCs). Effects of 17β-estradiol (E2), progesterone (P4), and testosterone alone or combined with the specific blockers of ERα (MPP dihydrochloride), ERβ (PHTPP), PGR (mifepristone), and AR (bicalutamide) on cell cycle and apoptosis were also measured in the SW480 male and HT29 female CRC cell lines. Results ERα and AR proteins increased, whilst ERβ and PGR declined markedly in malignant specimens. Moreover, male neoplastic tissues showed highest AR expression, whilst ERβ and PGR weakest alongside ERα strongest expression was seen in cancerous tissues from women aged ≥60 years. Late-stage neoplasms also revealed maximal alterations in the expression of sex steroid receptors. By tumor location, LSCs disclosed significant elevations in ERα with marked declines in PGR compared with RSCs, and ERα strongest alongside PGR weakest expression was detected in advanced LSCs from women aged ≥60 years. Late-stage LSCs from females aged ≥60 years also showed weakest ERβ and strongest AR expression. In contrast, male RSC and LSC tissues exhibited equal ERβ and AR expression in all clinical stages. ERα and AR proteins also correlated positively, whereas ERβ and PGR inversely, with tumor characteristics. Concomitantly, E2 and P4 monotherapies triggered cell cycle arrest and apoptosis in the SW480 and HT29 cells, and while pre-treatment with ERα-blocker enhanced the effects of E2, ERβ-blocker and PGR-blocker suppressed the E2 and P4 anti-cancer actions, respectively. In contrast, treatment with the AR-blocker induced apoptosis, whilst co-treatment with testosterone hindered the effects. Conclusions This study advocates that protein expression of sex steroid receptors in malignant tissues could represent prognostic markers, as well as hormonal therapy could provide an alternative strategy against CRC, and their efficacies could be dependent on gender, clinical stage, and tumor location.
Collapse
Affiliation(s)
- Bassem Refaat
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- *Correspondence: Bassem Refaat, ;
| | - Akhmed Aslam
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Shakir Idris
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmed H. Almalki
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Regional Laboratory and Central Blood Bank, Ministry of Health, Jizan, Saudi Arabia
| | - Mofareh Y. Alkhaldi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Laboratory And Blood Bank Department, Asir Central Hospital, Abha, Saudi Arabia
| | - Hassan A. Asiri
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Forensic Medicine Department, Health Affairs General Directorate in Assir, Abha, Saudi Arabia
| | - Riyad A. Almaimani
- Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulrahman Mujalli
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Faisal Minshawi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sara A. Alamri
- Histopathology Department, King Abdullah Medical City, Makkah, Saudi Arabia
| | - Mona I. AlHussain
- Histopathology Department, King Abdullah Medical City, Makkah, Saudi Arabia
| | - Badee A. Baltow
- Histopathology Department, King Abdullah Medical City, Makkah, Saudi Arabia
| | - Mansour H. Alqasmi
- Clinical Laboratories, Al-Noor Specialist Hospital, Makkah, Saudi Arabia
| | - Ghaiyda T. Basfar
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Clinical Laboratories, Al-Noor Specialist Hospital, Makkah, Saudi Arabia
| | - Ohoud M. Alosaimi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Clinical Laboratories, Eradah and Mental Health Complex, Ministry of Health, Taif, Saudi Arabia
| | - Ibrahim A. Muhayya
- Laboratory And Blood Bank Department, Asir Central Hospital, Abha, Saudi Arabia
| |
Collapse
|
7
|
Honma N, Arai T, Matsuda Y, Fukunaga Y, Akishima-Fukasawa Y, Yamamoto N, Kawachi H, Ishikawa Y, Takeuchi K, Mikami T. Estrogen concentration and estrogen receptor-β expression in postmenopausal colon cancer considering patient/tumor background. J Cancer Res Clin Oncol 2022; 148:1063-1071. [PMID: 35032217 DOI: 10.1007/s00432-021-03889-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE A large number of studies have suggested an inhibitory role of estrogens against colorectal cancer (CRC), but persistent controversy exists. CRC characteristics are affected by sex, age, and tumor locus, suggesting the need for a systematic study considering these factors. The purpose of this study was to verify the difference in the pathobiological role of estrogens in CRC according to patient/tumor backgrounds. METHODS Surgical specimens from 116 postmenopausal women (≥ 70 years/o, n = 74; < 70 years/o, n = 42) were studied. Estrogen receptor-β (ER-β), the main ER in the colorectal epithelium, was immunohistochemically examined. The concentrations of estradiol (E2) and estrone (E1) were examined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). These factors were compared according to the tissue type (cancerous or non-cancerous), patients' age, tumor backgrounds (locus, histology, pathological stage, status of mismatch repair protein = MMR), and clinical outcome. RESULTS ER-β-positivity, higher E2 concentration, deficient-MMR, and medullary/mucinous histology (Med/Muc) were closely related to right-sided tumors in women who were aged ≥ 70 years /o (R-Ca ≥ 70) and also closely related to each other. ER-β reduction compared with non-cancerous counterparts was observed only in left-sided tumors of patients < 70 years /o (L-Ca < 70), non-Med/Muc, or proficient-MMR tumors. CONCLUSION The present results suggest that estrogens do not suppress, but rather promote, R-Ca ≥ 70, Med/Muc, or deficient-MMR tumors, whereas estrogens suppress L-Ca < 70, non-Med/Muc, or proficient-MMR tumors, confirming the difference in pathobiological role of estrogens in postmenopausal colon cancer according to the patients' age and tumor background. This may at least partly explain the controversy regarding the association between estrogens and CRC.
Collapse
Affiliation(s)
- Naoko Honma
- Department of Pathology, Faculty of Medicine, Toho University, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan. .,Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Sakaecho 35-2, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yoko Matsuda
- Oncology Pathology, Department of Pathology and Host-Defense, Faculty of Medicine, Kagawa University, Ikenobe 1750-1, Kita-gun, Miki, Kagawa, 761-0793, Japan
| | - Yosuke Fukunaga
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Yuri Akishima-Fukasawa
- Department of Pathology, Faculty of Medicine, Toho University, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan
| | - Noriko Yamamoto
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiroshi Kawachi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Yuichi Ishikawa
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, International University of Health and Welfare, Mita 1-4-3, Minato-ku, Tokyo, 108-8329, Japan
| | - Kengo Takeuchi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Tetuo Mikami
- Department of Pathology, Faculty of Medicine, Toho University, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan
| |
Collapse
|
8
|
Mahbub AA, Aslam A, Elzubier ME, El-Boshy M, Abdelghany AH, Ahmad J, Idris S, Almaimani R, Alsaegh A, El-Readi MZ, Baghdadi MA, Refaat B. Enhanced anti-cancer effects of oestrogen and progesterone co-therapy against colorectal cancer in males. Front Endocrinol (Lausanne) 2022; 13:941834. [PMID: 36263327 PMCID: PMC9574067 DOI: 10.3389/fendo.2022.941834] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/14/2022] [Indexed: 12/24/2022] Open
Abstract
Although ovarian sex steroids could have protective roles against colorectal cancer (CRC) in women, little is currently known about their potential anti-tumorigenic effects in men. Hence, this study measured the therapeutic effects of 17β-oestradiol (E2) and/or progesterone (P4) against azoxymethane-induced CRC in male mice that were divided into (n = 10 mice/group): negative (NC) and positive (PC) controls, E2 (580 µg/Kg/day; five times/week) and P4 (2.9 mg/Kg/day; five times/week) monotherapies, and concurrent (EP) and sequential (E/P) co-therapy groups. Both hormones were injected intraperitoneally to the designated groups for four consecutive weeks. Similar treatment protocols with E2 (10 nM) and/or P4 (20 nM) were also used in the SW480 and SW620 human male CRC cell lines. The PC group showed abundant colonic tumours alongside increased colonic tissue testosterone levels and androgen (AR) and oestrogen (ERα) receptors, whereas E2 and P4 levels with ERβ and progesterone receptor (PGR) decreased significantly compared with the NC group. E2 and P4 monotherapies equally increased ERβ/PGR with p21/Cytochrome-C/Caspase-3, reduced testosterone levels, inhibited ERα/AR and CCND1/survivin and promoted apoptosis relative to the PC group. Both co-therapy protocols also revealed better anti-cancer effects with enhanced modulation of colonic sex steroid hormones and their receptors, with E/P the most prominent protocol. In vitro, E/P regimen showed the highest increases in the numbers of SW480 (2.1-fold) and SW620 (3.5-fold) cells in Sub-G1 phase of cell cycle. The E/P co-therapy also disclosed the lowest percentages of viable SW480 cells (2.8-fold), whilst both co-therapy protocols equally showed the greatest SW620 apoptotic cell numbers (5.2-fold) relative to untreated cells. Moreover, both co-therapy regimens revealed maximal inhibitions of cell cycle inducers, cell survival markers, and AR/ERα alongside the highest expression of cell cycle suppressors, pro-apoptotic molecules, and ERβ/PGR in both cell lines. In conclusion, CRC was associated with abnormal levels of colonic sex steroid hormones alongside aberrant protein expression of their receptors. While the anti-cancer effects of E2 and P4 monotherapies were equal, their combination protocols showed boosted tumoricidal actions against CRC in males, possibly by promoting ERβ and PGR-mediated androgen deprivation together with inhibition of ERα-regulated oncogenic pathways.
Collapse
Affiliation(s)
- Amani A. Mahbub
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Akhmed Aslam
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohamed E. Elzubier
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
- Biochemistry Department, Faculty of Medicine and Surgery, National University, Khartoum, Sudan
| | - Mohamed El-Boshy
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Clinical Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Abdelghany H. Abdelghany
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Jawwad Ahmad
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Shakir Idris
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Riyad Almaimani
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Aiman Alsaegh
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mahmoud Zaki El-Readi
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, Assuit, Egypt
| | - Mohammed A. Baghdadi
- Research Centre, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Bassem Refaat
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- *Correspondence: Bassem Refaat, ;
| |
Collapse
|
9
|
Simin J, Liu Q, Wang X, Fall K, Williams C, Callens S, Engstrand L, Brusselaers N. Prediagnostic use of estrogen-only therapy is associated with improved colorectal cancer survival in menopausal women: a Swedish population-based cohort study. Acta Oncol 2021; 60:881-887. [PMID: 33861686 DOI: 10.1080/0284186x.2021.1909747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Menopausal hormone therapy (MHT) reduces the risk of developing colorectal cancer (CRC), yet it is largely unclear whether it could also influence survival in women with CRC. Therefore, we aimed to investigate the influence of prediagnostic MHT use on CRC-specific and all-cause mortality in women with CRC. METHODS This nationwide nested cohort study, within a large population-based matched cohort, included all women diagnosed with incident CRC between January 2006 and December 2012 (N = 7814). A total of 1529 women had received at least one dispensed prescription of systemic MHT before CRC diagnosis, and 6285 CRC women with CRC did not receive MHT during the study period, as ascertained from the Swedish Prescribed Drug Registry. Multivariable Cox regression models provided adjusted hazard ratios (HRs) with 95% confidence intervals (CIs) for CRC-specific mortality and all-cause mortality. RESULTS Past use of prediagnostic estrogen-only therapy (E-MHT) was associated with lower CRC-specific (HR = 0.67, 95%CI 0.44-0.99) and all-cause mortality (HR = 0.68, 95%CI 0.59-0.93). However, all-cause mortality (HR = 1.23, 95%CI 1.02-1.48) was elevated among current prediagnostic E-MHT users who were 70+ years at diagnosis. Current estrogen combined progestin therapy (EP-MHT) was associated with higher CRC-specific mortality (HR = 1.61, 95%CI 1.06-2.44) in older women, but no association was shown for all-cause mortality. CONCLUSIONS Our findings suggest that E-MHT, but not EP-MHT use, might be associated with improved CRC survival, indicating a potential role of estrogens in sex hormone-related cancers. However, association of MHT use with grade of cancer remains unclear.
Collapse
Affiliation(s)
- Johanna Simin
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Reseaarch (CTMR), Stockholm, Sweden
- Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
| | - Qing Liu
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Reseaarch (CTMR), Stockholm, Sweden
- Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
| | - Xinchen Wang
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Reseaarch (CTMR), Stockholm, Sweden
| | - Katja Fall
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Epidemiology and Biostatistics School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Cecilia Williams
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Solna, Sweden
| | - Steven Callens
- Department of General Internal Medicine, Ghent University Hospital, Gent, Belgium
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Reseaarch (CTMR), Stockholm, Sweden
- Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
| | - Nele Brusselaers
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Reseaarch (CTMR), Stockholm, Sweden
- Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
- Global Health Institute, Antwerp University, Antwerp, Belgium
- Department of Head and Skin, Ghent University, Ghent, Belgium
| |
Collapse
|
10
|
Foster PA. Steroid Sulphatase and Its Inhibitors: Past, Present, and Future. Molecules 2021; 26:2852. [PMID: 34064842 PMCID: PMC8151039 DOI: 10.3390/molecules26102852] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/22/2022] Open
Abstract
Steroid sulphatase (STS), involved in the hydrolysis of steroid sulphates, plays an important role in the formation of both active oestrogens and androgens. Since these steroids significantly impact the proliferation of both oestrogen- and androgen-dependent cancers, many research groups over the past 30 years have designed and developed STS inhibitors. One of the main contributors to this field has been Prof. Barry Potter, previously at the University of Bath and now at the University of Oxford. Upon Prof. Potter's imminent retirement, this review takes a look back at the work on STS inhibitors and their contribution to our understanding of sulphate biology and as potential therapeutic agents in hormone-dependent disease. A number of potent STS inhibitors have now been developed, one of which, Irosustat (STX64, 667Coumate, BN83495), remains the only one to have completed phase I/II clinical trials against numerous indications (breast, prostate, endometrial). These studies have provided new insights into the origins of androgens and oestrogens in women and men. In addition to the therapeutic role of STS inhibition in breast and prostate cancer, there is now good evidence to suggest they may also provide benefits in patients with colorectal and ovarian cancer, and in treating endometriosis. To explore the potential of STS inhibitors further, a number of second- and third-generation inhibitors have been developed, together with single molecules that possess aromatase-STS inhibitory properties. The further development of potent STS inhibitors will allow their potential therapeutic value to be explored in a variety of hormone-dependent cancers and possibly other non-oncological conditions.
Collapse
Affiliation(s)
- Paul A. Foster
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK; ; Tel.: +44-121-414-3776
- Centre for Endocrinology, Metabolism and Diabetes, University of Birmingham, Birmingham Health Partners, Birmingham B15 2TT, UK
| |
Collapse
|
11
|
Sato N, Ise K, Hata S, Yamashita S, Ito A, Sasano H, Nakamura Y. Clinicopathological Significance of Estrogen Receptor β and Estrogen Synthesizing/Metabolizing Enzymes in Urothelial Carcinoma of Urinary Bladder. Pathol Oncol Res 2021; 27:589649. [PMID: 34257538 PMCID: PMC8262212 DOI: 10.3389/pore.2021.589649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 02/10/2021] [Indexed: 11/13/2022]
Abstract
Sex-specific differences in the incidence of urinary bladder carcinomas are well known, and the possible involvement of sex steroids has been proposed. We previously reported the association of the loss of androgen receptors and androgen-producing enzymes with tumor progression of urinary bladder cancer patients. Clinically, the selective estrogen receptor modulators (SERMs) were reported to suppress the progression of these tumors but the status of estrogen receptors (ERs) has not been well studied in patients with bladder urinary cancer. Moreover, not only ERs but also estrogen-related enzymes, such as aromatase, steroid sulfatase (STS), and estrogen sulfotransferase (EST), have been reported in the biological/clinical behavior of various hormone-dependent carcinomas but not studied in urinary bladder carcinoma. Therefore, in this study, we immunolocalized ERs as well as estrogen metabolizing enzymes in urinary bladder carcinoma and performed immunoblotting and cell proliferation assays using the bladder urothelial carcinoma cell line, T24. The results revealed that the loss of STS and aromatase was significantly correlated with advanced stages of the carcinoma. In vitro studies also revealed that T24 cell proliferation rates were significantly ameliorated after treatment with estradiol or diarylpropionitrile (DPN). EST and aromatase were also significantly correlated with the nuclear grade of the carcinoma. The results of our present study, for the first time, demonstrated that biologically active estrogens that bind to ERs could suppress tumor progression and the inactive ones could promote its progression and the potential clinical utility of SERM treatment in selective patients with urinary bladder carcinoma.
Collapse
Affiliation(s)
- Naomi Sato
- Division of Pathology, Sendai City Hospital, Sendai, Japan
| | - Kazue Ise
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.,Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuko Hata
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.,Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinichi Yamashita
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihiro Ito
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.,Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
12
|
Abancens M, Bustos V, Harvey H, McBryan J, Harvey BJ. Sexual Dimorphism in Colon Cancer. Front Oncol 2020; 10:607909. [PMID: 33363037 PMCID: PMC7759153 DOI: 10.3389/fonc.2020.607909] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
A higher incidence of colorectal cancer (CRC) is found in males compared to females. Young women (18-44 years) with CRC have a better survival outcome compared to men of the same age or compared to older women (over 50 years), indicating a global incidence of sexual dimorphism in CRC rates and survival. This suggests a protective role for the sex steroid hormone estrogen in CRC development. Key proliferative pathways in CRC tumorigenesis exhibit sexual dimorphism, which confer better survival in females through estrogen regulated genes and cell signaling. Estrogen regulates the activity of a class of Kv channels (KCNQ1:KCNE3), which control fundamental ion transport functions of the colon and epithelial mesenchymal transition through bi-directional interactions with the Wnt/β-catenin signalling pathway. Estrogen also modulates CRC proliferative responses in hypoxia via the novel membrane estrogen receptor GPER and HIF1A and VEGF signaling. Here we critically review recent clinical and molecular insights into sexual dimorphism of CRC biology modulated by the tumor microenvironment, estrogen, Wnt/β-catenin signalling, ion channels, and X-linked genes.
Collapse
Affiliation(s)
- Maria Abancens
- Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
| | - Viviana Bustos
- Departamento de Acuicultura y Recursos Agroalimentarios, Programa Fitogen, Universidad de Los Lagos, Osorno, Chile
| | - Harry Harvey
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - Jean McBryan
- Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
| | - Brian J. Harvey
- Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, Ireland
- Centro de Estudios Cientificos CECs, Valdivia, Chile
| |
Collapse
|
13
|
Pace F, Watnick PI. The Interplay of Sex Steroids, the Immune Response, and the Intestinal Microbiota. Trends Microbiol 2020; 29:849-859. [PMID: 33257138 DOI: 10.1016/j.tim.2020.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022]
Abstract
The role of sex steroids in mammalian maturation is well established. Recently, it has been increasingly appreciated that sex steroids also play an important role in the propensity of adults to develop a myriad of diseases. The exposure and responsiveness of tissues to sex steroids varies among individuals and between the sexes, and this has been correlated with gender-specific differences in the composition of the intestinal microbiota and in susceptibility to metabolic, autoimmune, and neoplastic diseases. Here we focus on recent studies that demonstrate an interplay between sex steroids, the intestinal immune response, and the intestinal microbiota. While correlations between biological sex, the intestinal innate immune response, intestinal inflammation, and intestinal microbiota have been established, many gaps in our knowledge prevent the emergence of an overarching model for this complex interaction. Such a model could aid in the development of prebiotic, probiotic, or synthetic therapeutics that decrease the risk of autoimmune, metabolic, neoplastic, and infectious diseases of the intestine and mitigate the particular health risks faced by individuals receiving sex steroid treatment.
Collapse
Affiliation(s)
- Fernanda Pace
- Division of Infectious Diseases, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Paula I Watnick
- Division of Infectious Diseases, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Matsunaga T, Okumura N, Saito H, Morikawa Y, Suenami K, Hisamatsu A, Endo S, Ikari A. Significance of aldo-keto reductase 1C3 and ATP-binding cassette transporter B1 in gain of irinotecan resistance in colon cancer cells. Chem Biol Interact 2020; 332:109295. [PMID: 33096057 DOI: 10.1016/j.cbi.2020.109295] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 11/29/2022]
Abstract
Irinotecan (CPT11) is widely prescribed for treatment of various intractable cancers such as advanced and metastatic colon cancer cells, but its continuous treatment promotes the resistance development. In this study, we established CPT11-resistant variants of three human colon cancer (DLD1, RKO and LoVo) cell lines, and found that gain of the resistance elicited an up-regulation of aldo-keto reductase (AKR) 1C3 in the cells. Additionally, the sensitivity to CPT11 toxicity was decreased and increased by overexpression and knockdown, respectively, of the enzyme. Moreover, the resistant cells suppressed formation of reactive 4-hydroxy-2-nonenal by CPT11 treatment, and the suppressive effect was almost completely abolished by addition of an AKR1C3 inhibitor. These results suggest that up-regulated AKR1C3 contributes to promotion of the chemoresistance by detoxifying the reactive aldehyde. Western blot and real-time polymerase-chain reaction analyses and ATP-binding cassette (ABC) B1-functional assay revealed that, among three ABC transporters, ABCB1 was the most highly up-regulated by development of the CPT11 resistance, inferring a significant contribution of pregnane-X receptor-dependent signaling to the ABCB1 up-regulation. The combined treatment with inhibitors of AKR1C3 and ABCB1 potently sensitized the resistant cells to CPT11 and its active metabolite SN38. Taken together, our results suggest that combination of AKR1C3 and ABCB1 inhibitors is effective as adjuvant therapy to enhance CPT11 sensitivity of intractable colon cancer cells.
Collapse
Affiliation(s)
- Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 502-8585, Japan.
| | - Naoko Okumura
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Haruhi Saito
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Yoshifumi Morikawa
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Koichi Suenami
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Aki Hisamatsu
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 502-8585, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| |
Collapse
|
15
|
Konings G, Brentjens L, Delvoux B, Linnanen T, Cornel K, Koskimies P, Bongers M, Kruitwagen R, Xanthoulea S, Romano A. Intracrine Regulation of Estrogen and Other Sex Steroid Levels in Endometrium and Non-gynecological Tissues; Pathology, Physiology, and Drug Discovery. Front Pharmacol 2018; 9:940. [PMID: 30283331 PMCID: PMC6157328 DOI: 10.3389/fphar.2018.00940] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022] Open
Abstract
Our understanding of the intracrine (or local) regulation of estrogen and other steroid synthesis and degradation expanded in the last decades, also thanks to recent technological advances in chromatography mass-spectrometry. Estrogen responsive tissues and organs are not passive receivers of the pool of steroids present in the blood but they can actively modify the intra-tissue steroid concentrations. This allows fine-tuning the exposure of responsive tissues and organs to estrogens and other steroids in order to best respond to the physiological needs of each specific organ. Deviations in such intracrine control can lead to unbalanced steroid hormone exposure and disturbances. Through a systematic bibliographic search on the expression of the intracrine enzymes in various tissues, this review gives an up-to-date view of the intracrine estrogen metabolisms, and to a lesser extent that of progestogens and androgens, in the lower female genital tract, including the physiological control of endometrial functions, receptivity, menopausal status and related pathological conditions. An overview of the intracrine regulation in extra gynecological tissues such as the lungs, gastrointestinal tract, brain, colon and bone is given. Current therapeutic approaches aimed at interfering with these metabolisms and future perspectives are discussed.
Collapse
Affiliation(s)
- Gonda Konings
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Linda Brentjens
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Bert Delvoux
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Karlijn Cornel
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Marlies Bongers
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Roy Kruitwagen
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Sofia Xanthoulea
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Andrea Romano
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
16
|
Fashe M, Hashiguchi T, Yi M, Moore R, Negishi M. Phenobarbital-induced phosphorylation converts nuclear receptor RORα from a repressor to an activator of the estrogen sulfotransferase gene Sult1e1 in mouse livers. FEBS Lett 2018; 592:2760-2768. [PMID: 30025153 PMCID: PMC10445657 DOI: 10.1002/1873-3468.13199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/22/2018] [Accepted: 07/13/2018] [Indexed: 01/01/2023]
Abstract
The estrogen sulfotransferase SULT1E1 sulfates and inactivates estrogen, which is reactivated via desulfation by steroid sulfatase, thus regulating estrogen homeostasis. Phenobarbital (PB), a clinical sedative, activates Sult1e1 gene transcription in mouse livers. Here, the molecular mechanism by which the nuclear receptors CAR, which is targeted by PB, and RORα communicate through phosphorylation to regulate Sult1e1 activation has been studied. RORα, a basal activity repressor of the Sult1e1 promoter, becomes phosphorylated at serine 100 and converts to an activator of the Sult1e1 promoter in response to PB. CAR regulates both the RORα phosphorylation and conversion. Our findings suggest that PB signals CAR to communicate with RORα via serine 100 phosphorylation, converting RORα from transcription repressor to activator of the Sult1e1 gene and inducing SULT1E1 expression in mouse livers.
Collapse
Affiliation(s)
- Muluneh Fashe
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Takuyu Hashiguchi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - MyeongJin Yi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Rick Moore
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Masahiko Negishi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| |
Collapse
|
17
|
Foster PA, Mueller JW. SULFATION PATHWAYS: Insights into steroid sulfation and desulfation pathways. J Mol Endocrinol 2018; 61:T271-T283. [PMID: 29764919 DOI: 10.1530/jme-18-0086] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/18/2022]
Abstract
Sulfation and desulfation pathways represent highly dynamic ways of shuttling, repressing and re-activating steroid hormones, thus controlling their immense biological potency at the very heart of endocrinology. This theme currently experiences growing research interest from various sides, including, but not limited to, novel insights about phospho-adenosine-5'-phosphosulfate synthase and sulfotransferase function and regulation, novel analytics for steroid conjugate detection and quantification. Within this review, we will also define how sulfation pathways are ripe for drug development strategies, which have translational potential to treat a number of conditions, including chronic inflammatory diseases and steroid-dependent cancers.
Collapse
Affiliation(s)
- Paul A Foster
- Institute of Metabolism and Systems Research (IMSR)University of Birmingham, Birmingham, UK
- Centre for EndocrinologyDiabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| | - Jonathan Wolf Mueller
- Institute of Metabolism and Systems Research (IMSR)University of Birmingham, Birmingham, UK
- Centre for EndocrinologyDiabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| |
Collapse
|
18
|
Significance of intratissue estrogen concentration coupled with estrogen receptors levels in colorectal cancer prognosis. Oncotarget 2017; 8:115546-115560. [PMID: 29383180 PMCID: PMC5777792 DOI: 10.18632/oncotarget.23309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of estrogen related pathways is implicated colorectal cancer (CRC) development. However, significance of intratissue concentration of estrone (E1) and 17β-estradiol (E2) in relation to estrogen receptor (ESR) expression level was not addressed so far. Herein, we measured E1 and E2 intratissue concentration using liquid chromatography electrospray ionization tandem mass spectrometry (ESI LC/MS) and mRNA levels of ESR1 and ESR2 using RT-qPCR in cancerous and histopathologically unchanged tissue from 75 and 110 CRC patients, respectively. The obtained results were associated with clinicopathological factors, expression of estrogen dependent genes (CTNNB1, CCND1) and prognostic significance. We found no statistically significant differences in E1 or E2 concentration between cancerous tissue and histopathologically unchanged counterparts. Moreover, mRNA levels of ESR1 and ESR2 were significantly decreased in cancerous tissue compared with histopathologically unchanged (p=0.00001). Log rank analysis revealed no benefit of low E1 to E2 ratio, high E1, E2 concentration or ESR1, ESR2 mRNA level for patients’ overall (OS) and disease free survival (DFS). Interestingly, we have observed that patients with low ESR1 mRNA level coupled with low E1 intratissue concentration had a significant decrease in DFS compared with group of patients with high ESR1 mRNA level and high E1 concentration (HR=0.16, 95% CI 0.02-1.05; p=0.06). Furthermore, patients with low E1 concentration and low ESR1 transcript had significantly higher CTNNB1 and CCND1 mRNA level compare with subgroup with high level of both grouping factors. Our study indicates a potential value of estrogen intratissue concentration and its receptor expression level for CRC patients’ prognosis.
Collapse
|
19
|
Gilligan LC, Rahman HP, Hewitt AM, Sitch AJ, Gondal A, Arvaniti A, Taylor AE, Read ML, Morton DG, Foster PA. Estrogen Activation by Steroid Sulfatase Increases Colorectal Cancer Proliferation via GPER. J Clin Endocrinol Metab 2017; 102:4435-4447. [PMID: 28945888 PMCID: PMC5718700 DOI: 10.1210/jc.2016-3716] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 08/24/2017] [Indexed: 02/08/2023]
Abstract
CONTEXT Estrogens affect the incidence and progression of colorectal cancer (CRC), although the precise molecular mechanisms remain ill-defined. OBJECTIVE The present study investigated prereceptor estrogen metabolism through steroid sulphatase (STS) and 17β-hydroxysteroid dehydrogenase activity and subsequent nongenomic estrogen signaling in human CRC tissue, in The Cancer Genome Atlas colon adenocarcinoma data set, and in in vitro and in vivo CRC models. We aimed to define and therapeutically target pathways through which estrogens alter CRC proliferation and progression. DESIGN, SETTING, PATIENTS, AND INTERVENTIONS Human CRC samples with normal tissue-matched controls were collected from postmenopausal female and age-matched male patients. Estrogen metabolism enzymes and nongenomic downstream signaling pathways were determined. CRC cell lines were transfected with STS and cultured for in vitro and in vivo analysis. Estrogen metabolism was determined using an ultra-performance liquid chromatography-tandem mass spectrometry method. PRIMARY OUTCOME MEASURE The proliferative effects of estrogen metabolism were evaluated using 5-bromo-2'-deoxyuridine assays and CRC mouse xenograft studies. RESULTS Human CRC exhibits dysregulated estrogen metabolism, favoring estradiol synthesis. The activity of STS, the fundamental enzyme that activates conjugated estrogens, is significantly (P < 0.001) elevated in human CRC compared with matched controls. STS overexpression accelerates CRC proliferation in in vitro and in vivo models, with STS inhibition an effective treatment. We defined a G-protein-coupled estrogen receptor (GPER) proproliferative pathway potentially through increased expression of connective tissue growth factor in CRC. CONCLUSION Human CRC favors estradiol synthesis to augment proliferation via GPER stimulation. Further research is required regarding whether estrogen replacement therapy should be used with caution in patients at high risk of developing CRC.
Collapse
Affiliation(s)
- Lorna C. Gilligan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Habibur P. Rahman
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Anne-Marie Hewitt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Alice J. Sitch
- Institute of Applied Health Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Ali Gondal
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Anastasia Arvaniti
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Angela E. Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Martin L. Read
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Dion G. Morton
- Institute of Cancer and Genomic Sciences, Academic Department of Surgery, University of Birmingham, Birmingham B15 2TH, United Kingdom
| | - Paul A. Foster
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom
| |
Collapse
|
20
|
Al-Mukaynizi FB, Alanazi M, Al-Daihan S, Parine NR, Almadi M, Aljebreen A, Azzam N, Alharbi O, Arafah M, Warsy A. CYP19A1 gene polymorphism and colorectal cancer etiology in Saudi population: case-control study. Onco Targets Ther 2017; 10:4559-4567. [PMID: 29066910 PMCID: PMC5604566 DOI: 10.2147/ott.s121557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Considerable interest is directed toward the enzyme aromatase (CYP19A1) and the development of cancer, due to CYP19A1's role in estrogen biosynthesis. Several cancers display excessive intra-tumor accumulation of estrogens, and aromatase inhibitors are used for treatment. The CYP19A1 gene exhibits polymorphism and mutations that can alter its expression or aromatase activity and influence estrogen production. We designed this study to investigate the link between CYP19A1 polymorphism and susceptibility to colorectal cancer (CRC) development in Saudis. PATIENTS AND METHODS Blood samples from 100 CRC patients and 100 healthy controls were drawn for DNA extractions. Three polymorphic sites, rs4774585, rs936308, and rs4775936, were genotyped using Taqman genotyping by real-time polymerase chain reaction. Allelic and genotype frequencies were calculated and compared in the two groups. RESULTS All single nucleotide polymorphisms (SNPs) were polymorphic in Saudis, and comparison of allele frequencies showed several differences when compared to other populations. None of the SNPs were associated with the risk of CRC development in Saudis (P>0.05). Some gender and location (colon or rectal) differences were observed. DISCUSSION The results of this study highlighted the genetic heterogeneity existing between populations in the prevalence of different SNPs and their relation to disease state. It showed that, although rs4774585, rs936308, and rs4775936 are involved in CRC development in several populations, their role is not significant in the etiology of CRC in Saudis; however, some SNPs do increase susceptibility or resistance to CRC development as judged from the odds ratio. Further large-scale studies are warranted to clarify the role of the CYP19A1 development in CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maha Arafah
- Department of Pathology, College of Medicine
| | - Arjumand Warsy
- Central Laboratory, Female Center for Scientific & Medical Colleges, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
21
|
Yang X, Huang H, Wang M, Zheng X, Xu J, Xie M. Effect of nonylphenol on the regulation of cell growth in colorectal cancer cells. Mol Med Rep 2017; 16:2211-2216. [PMID: 28656208 DOI: 10.3892/mmr.2017.6817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 04/06/2017] [Indexed: 11/06/2022] Open
Abstract
Nonylphenol (NP) is a well-known endocrine-disrupting chemical (EDC), which can enhance the progression of cancer by functioning as an estrogen‑like factor. In the present study, the effects of different concentrations of NP on COLO205 colorectal cancer (CRC) cells were examined. The results of flow cytometric analysis revealed that NP significantly decreased the proportion of cells in the G0/G1 phase in a dose‑dependent manner, which was accompanied by a marginal increase in the proportions of cells in S and G2/M phases. NP did not induce apoptosis, whereas estradiol (E2) did induce apoptosis. To elucidate the mechanisms underlying the action of NP on COLO205 cells, the transcriptional levels of extracellular signal‑regulated kinase (ERK)1, ERK2 and phosphoinositide 3‑kinase (PI3K) were assessed using reverse transcription‑quantitative polymerase chain reaction analysis. The expressions levels of ERK1, ERK2 and PI3K were increased by treatment with NP in a dose‑dependent manner. On examining protein levels, the expression of PI3K p38 was increased by NP and E2, and the expression of ERK1/2 was increased by NP. The phosphorylation of the ERK protein was significantly increased by treatment with NP at a high concentration (10‑4 M; P<0.01), but significantly decreased by E2 (P<0.01). Two key proteins in the transforming growth factor (TGF)β pathway (c‑Fos and SnoN) were selected for analysis using western blot analysis in the COLO205 cells treated with NP and E2. The expression levels of c‑Fos and SnoN were significantly increased by treatment with E2 (10‑7 M; P<0.01) and NP (10‑7‑10‑4 M; P<0.01). Taken together, these results indicated that NP affected the development of CRC via the ERK signaling pathway and TGFβ pathway.
Collapse
Affiliation(s)
- Xuefeng Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Handong Huang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Maijian Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xingbin Zheng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jie Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Ming Xie
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
22
|
Ye DJ, Kwon YJ, Shin S, Baek HS, Shin DW, Chun YJ. Induction of Integrin Signaling by Steroid Sulfatase in Human Cervical Cancer Cells. Biomol Ther (Seoul) 2017; 25:321-328. [PMID: 27956712 PMCID: PMC5424643 DOI: 10.4062/biomolther.2016.155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/02/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
Steroid sulfatase (STS) is an enzyme responsible for the hydrolysis of aryl and alkyl sulfates. STS plays a pivotal role in the regulation of estrogens and androgens that promote the growth of hormone-dependent tumors, such as those of breast or prostate cancer. However, the molecular function of STS in tumor growth is still not clear. To elucidate the role of STS in cancer cell proliferation, we investigated whether STS is able to regulate the integrin signaling pathway. We found that overexpression of STS in HeLa cells increases the protein and mRNA levels of integrin β1 and fibronectin, a ligand of integrin α5β1. Dehydroepiandrosterone (DHEA), one of the main metabolites of STS, also increases mRNA and protein expression of integrin β1 and fibronectin. Further, STS expression and DHEA treatment enhanced phosphorylation of focal adhesion kinase (FAK) at the Tyr 925 residue. Moreover, increased phosphorylation of ERK at Thr 202 and Tyr 204 residues by STS indicates that STS activates the MAPK/ERK pathway. In conclusion, these results suggest that STS expression and DHEA treatment may enhance MAPK/ERK signaling through up-regulation of integrin β1 and activation of FAK.
Collapse
Affiliation(s)
- Dong-Jin Ye
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sangyun Shin
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hyoung-Seok Baek
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dong-Won Shin
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
23
|
Ban MJ, Ji SH, Lee CK, Bae SB, Kim HJ, Ahn TS, Lee MS, Baek MJ, Jeong D. Solute carrier organic anion transporter family member 4A1 (SLCO4A1) as a prognosis marker of colorectal cancer. J Cancer Res Clin Oncol 2017; 143:1437-1447. [DOI: 10.1007/s00432-017-2393-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/12/2017] [Indexed: 12/29/2022]
|
24
|
Gilligan LC, Gondal A, Tang V, Hussain MT, Arvaniti A, Hewitt AM, Foster PA. Estrone Sulfate Transport and Steroid Sulfatase Activity in Colorectal Cancer: Implications for Hormone Replacement Therapy. Front Pharmacol 2017; 8:103. [PMID: 28326039 PMCID: PMC5339229 DOI: 10.3389/fphar.2017.00103] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/20/2017] [Indexed: 12/31/2022] Open
Abstract
Hormone replacement therapy (HRT) affects the incidence and potential progression of colorectal cancer (CRC). As HRT primarily consists of estrone sulfate (E1S), understanding whether this conjugated estrogen is transported and metabolized in CRC will define its potential effect in this malignancy. Here, we show that a panel of CRC cell lines (Colo205, Caco2, HCT116, HT-29) have steroid sulfatase (STS) activity, and thus can hydrolyze E1S. STS activity is significantly higher in CRC cell lysate, suggesting the importance of E1S transport in intracellular STS substrate availability. As E1S transport is regulated by the expression pattern of certain solute carrier organic anion transporter polypeptides, we show that in CRC OATP4A1 is the most abundantly expressed transporter. All four CRC cell lines rapidly transported E1S into cells, with this effect significantly inhibited by the competitive OATP inhibitor BSP. Transient knockdown of OATP4A1 significantly disrupted E1S uptake. Examination of estrogen receptor status showed ERα was present in Colo205 and Caco2 cells. None of the cells expressed ERβ. Intriguingly, HCT116 and HT29 cells strongly expressed the G protein coupled estrogen receptor (GPER), and that stimulation of this receptor with estradiol (E2) and G1, a GPER agonist, significantly (p < 0.01) increased STS activity. Furthermore, tamoxifen and fulvestrant, known GPER agonist, also increased CRC STS activity, with this effect inhibited by the GPER antagonist G15. These results suggest that CRC can take up and hydrolyze E1S, and that subsequent GPER stimulation increases STS activity in a potentially novel positive feedback loop. As elevated STS expression is associated with poor prognosis in CRC, these results suggest HRT, tamoxifen and fulvestrant may negatively impact CRC patient outcomes.
Collapse
Affiliation(s)
- Lorna C Gilligan
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham Birmingham, UK
| | - Ali Gondal
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham Birmingham, UK
| | - Vivien Tang
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham Birmingham, UK
| | - Maryam T Hussain
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham Birmingham, UK
| | - Anastasia Arvaniti
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham Birmingham, UK
| | - Anne-Marie Hewitt
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham Birmingham, UK
| | - Paul A Foster
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, University of BirminghamBirmingham, UK; Centre for Endocrinology, Diabetes, and Metabolism, Birmingham Health PartnersBirmingham, UK
| |
Collapse
|
25
|
Shahbazi Y, Malekinejad H, Tajik H. Determination of naturally occurring estrogenic hormones in cow's and river buffalo's meat by HPLC-FLD method. J Food Drug Anal 2016; 24:457-463. [PMID: 28911549 PMCID: PMC9336673 DOI: 10.1016/j.jfda.2016.02.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 01/16/2016] [Accepted: 02/15/2016] [Indexed: 11/30/2022] Open
Abstract
This study was performed to measure and compare the levels of steroid hormones [estrone (E1), 17β-estradiol (E2), and estriol (E3)] and their conjugated metabolites in cow’s and river buffalo’s meat in two distinct follicular and luteal phases. Moreover, the possible effect of a heating process on steroid hormone concentration was also investigated. The collected meat (biceps femoris muscle) samples were subjected to liquid extraction, enzymatical deconjugation, and C18 solid-phase extraction. Estrogens were analyzed using high performance liquid chromatography equipped with a fluorescence detector. In the follicular phase the levels of steroid hormones (E1 and E2) in either tested species were higher than the luteal phase. Moreover, in the present study, E1 concentration (free and deconjugated value, 16.2 ± 1.1 ng/L) was found to be the highest phenolic estrogen in beef, while the dominant estrogen in muscle of river buffalo was E2 (free and deconjucated value, 23.3 ± 1.3 ng/L). The study revealed that animal species influenced the concentration of hormones (E1 and E2) in the samples. The heating process did not significantly change (p > 0.05) the levels of estrogens. The further findings of the present study showed that E3 (deconjugated form) was only detected in the buffalo’s meat (15.8 ± 1.9 ng/L). These data suggest that although meat is one of the valuable nutrient sources for humans, there are, however, increasing concerns about the safety of meat due to the excessive presence of steroid hormones.
Collapse
Affiliation(s)
- Yasser Shahbazi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Razi University, Kermanshah, Islamic Republic of
Iran
- Corresponding author. Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Razi University, Shaykh Tusi Av., P.O. Box: 671568-5414, Kermanshah, Islamic Republic of Iran. E-mail address: (Y. Shahbazi)
| | - Hassan Malekinejad
- Food and Beverages Safety Research Center, Urmia University of Medical Sciences, Urmia, Islamic Republic of
Iran
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Urmia University, Urmia, Islamic Republic of
Iran
| | - Hossein Tajik
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Urmia University, Urmia, Islamic Republic of
Iran
| |
Collapse
|
26
|
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
|
27
|
Thomas MP, Potter BVL. Discovery and Development of the Aryl O-Sulfamate Pharmacophore for Oncology and Women's Health. J Med Chem 2015; 58:7634-58. [PMID: 25992880 PMCID: PMC5159624 DOI: 10.1021/acs.jmedchem.5b00386] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In 1994, following work from this laboratory, it was reported that estrone-3-O-sulfamate irreversibly inhibits a new potential hormone-dependent cancer target steroid sulfatase (STS). Subsequent drug discovery projects were initiated to develop the core aryl O-sulfamate pharmacophore that, over some 20 years, have led to steroidal and nonsteroidal drugs in numerous preclinical and clinical trials, with promising results in oncology and women's health, including endometriosis. Drugs have been designed to inhibit STS, e.g., Irosustat, as innovative dual-targeting aromatase-steroid sulfatase inhibitors (DASIs) and as multitargeting agents for hormone-independent tumors, such as the steroidal STX140 and nonsteroidal counterparts, acting inter alia through microtubule disruption. The aryl sulfamate pharmacophore is highly versatile, operating via three distinct mechanisms of action, and imbues attractive pharmaceutical properties. This Perspective gives a personal view of the work leading both to the therapeutic concepts and these drugs, their current status, and how they might develop in the future.
Collapse
Affiliation(s)
- Mark P. Thomas
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Barry V. L. Potter
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| |
Collapse
|
28
|
Mueller JW, Gilligan LC, Idkowiak J, Arlt W, Foster PA. The Regulation of Steroid Action by Sulfation and Desulfation. Endocr Rev 2015; 36:526-63. [PMID: 26213785 PMCID: PMC4591525 DOI: 10.1210/er.2015-1036] [Citation(s) in RCA: 285] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Steroid sulfation and desulfation are fundamental pathways vital for a functional vertebrate endocrine system. After biosynthesis, hydrophobic steroids are sulfated to expedite circulatory transit. Target cells express transmembrane organic anion-transporting polypeptides that facilitate cellular uptake of sulfated steroids. Once intracellular, sulfatases hydrolyze these steroid sulfate esters to their unconjugated, and usually active, forms. Because most steroids can be sulfated, including cholesterol, pregnenolone, dehydroepiandrosterone, and estrone, understanding the function, tissue distribution, and regulation of sulfation and desulfation processes provides significant insights into normal endocrine function. Not surprisingly, dysregulation of these pathways is associated with numerous pathologies, including steroid-dependent cancers, polycystic ovary syndrome, and X-linked ichthyosis. Here we provide a comprehensive examination of our current knowledge of endocrine-related sulfation and desulfation pathways. We describe the interplay between sulfatases and sulfotransferases, showing how their expression and regulation influences steroid action. Furthermore, we address the role that organic anion-transporting polypeptides play in regulating intracellular steroid concentrations and how their expression patterns influence many pathologies, especially cancer. Finally, the recent advances in pharmacologically targeting steroidogenic pathways will be examined.
Collapse
Affiliation(s)
- Jonathan W Mueller
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Lorna C Gilligan
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jan Idkowiak
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Wiebke Arlt
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Paul A Foster
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
29
|
Honma N, Hosoi T, Arai T, Takubo K. Estrogen and cancers of the colorectum, breast, and lung in postmenopausal women. Pathol Int 2015; 65:451-9. [PMID: 26126901 DOI: 10.1111/pin.12326] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 05/28/2015] [Indexed: 01/01/2023]
Abstract
As estrogens play an important role in maintaining physiological function in various organs, the estrogen decrease after menopause is thought to cause various diseases frequently observed in postmenopausal or elderly women. With the aging of society and a decrease in infectious or vascular diseases, neoplasms have now become the most frequent cause of death in Japan. Cancers of the colorectum, breast, and lung have been rapidly increasing both in incidence and death, especially among postmenopausal women. Interestingly, all three of these cancers are associated with estrogens. In premenopausal women, ovarian estrogens plays major roles in the female reproductive organs through the classic estrogen receptor, ER-α. In postmenopausal women, however, estrogens produced/activated by peripherally localized estrogen-metabolizing enzymes such as aromatase, which converts androgen into estrogens, are thought to play physiologically and pathobiologically important roles in various organs through second ER, namely ER-β, distributing systemically. In this article, the association of estrogens with these cancers in postmenopausal or elderly women are reviewed, especially focusing on the role of ER-β and peripheral estrogen metabolism. The possibility of prevention or treatment of these diseases through estrogenic control is also discussed.
Collapse
Affiliation(s)
- Naoko Honma
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Takayuki Hosoi
- Kenkoin Clinic, Institute for Preventive Medicine, Kenkoin Medical Corporation, Tokyo, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Kaiyo Takubo
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
30
|
Reproductive and hormonal factors and mortality among women with colorectal cancer in the NIH-AARP Diet and Health Study. Br J Cancer 2015; 113:562-8. [PMID: 26103572 PMCID: PMC4522633 DOI: 10.1038/bjc.2015.224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/12/2015] [Accepted: 05/21/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Although use of menopausal hormone therapy (MHT) and some reproductive factors have been associated with colorectal cancer (CRC) risk, relations between these factors and survival after CRC diagnosis are unclear. METHODS Among 2053 post-menopausal women diagnosed with incident CRC in the NIH-AARP Diet and Health Study, we calculated hazard ratios (HRs) and 95% confidence intervals (CIs) using multivariable Cox proportional hazards regression to test associations between oral contraceptive (OC) use, menarche age, age at first birth, parity, menopausal age, and MHT use with all-cause and CRC-specific mortality. RESULTS There were 759 deaths (332 CRC-related deaths) over a median follow-up of 7.7 years. We observed no statistically significant associations between OC use, menarche age, age at first birth, parity, menopausal age, and mortality. Compared with never MHT use, former use was not associated with mortality, but we found an inverse association among baseline current users, for both all-cause (HR=0.79, 95% CI 0.66-0.94) and CRC mortality (0.76, 0.59-0.99). CONCLUSION Future studies should further focus on the mechanisms by which exogenous oestrogen exposure might affect tumour progression and CRC survival.
Collapse
|
31
|
Li CY, Song B, Wang YY, Meng H, Guo SB, Liu LN, Lv HC, Wu QJ. Age at menarche and risk of colorectal cancer: a meta-analysis. PLoS One 2013; 8:e65645. [PMID: 23762403 PMCID: PMC3675201 DOI: 10.1371/journal.pone.0065645] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 04/25/2013] [Indexed: 12/30/2022] Open
Abstract
Background Various observational studies have focused on the relationship between menarcheal age and the risk of colorectal cancer (CRC). However, the association is still controversial because of inconsistent results. Therefore, we performed a meta-analysis to assess this issue from epidemiological studies. Methods After a literature search in MEDLINE, EMBASE, and Web of Science for studies of menarcheal age and CRC risk published through the end of January 2013, we pooled the relative risks (RRs) from included studies using a fixed- or random-effects model and performed heterogeneity and publication bias analyses. All statistical tests were two-sided. Results Eleven case-control and 11 cohort studies were eligible for inclusion in our analysis. The random-effects pooled RR for oldest versus youngest menarcheal age was 0.95 [95% confidence intervals (CIs) = 0.85–1.06], with significant heterogeneity (Q = 61.03, P<0.001, I2 = 65.6%). When separately analyzed, case-control (RR = 0.95, 95% CI = 0.75–1.21) and cohort studies (RR = 0.97, 95% CI = 0.90–1.04) yielded similar results. Moreover, similar results were also observed among the subgroup analyses by study quality, population, exposure assessment, anatomic cancer site, subsite of colon cancer, and several potential important confounders and risk factors. There was no evidence of publication bias and significant heterogeneity between subgroups detected by meta-regression analyses. Conclusions Findings from this meta-analysis demonstrated that menarcheal age was not associated with the risk of CRC in humans. Further studies are warranted to stratify results by the subsite of colon cancer and menopause status in the future.
Collapse
Affiliation(s)
- Chun-Yan Li
- Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
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
|
33
|
Rawłuszko AA, Antoniucci M, Horbacka K, Lianeri M, Krokowicz P, Jagodziński PP. Reduced expression of steroid sulfatase in primary colorectal cancer. Biomed Pharmacother 2013; 67:577-82. [PMID: 23916543 DOI: 10.1016/j.biopha.2013.04.007] [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: 03/25/2013] [Accepted: 04/19/2013] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is considered an estrogen-dependent malignancy, and intratissue estrogen concentration can be controlled by steroid sulfatase (STS). Little is known about changes in the expression of STS during the development of CRC. Therefore, we analysed the STS mRNA levels in primary colonic adenocarcinoma tissues and adjacent histopathologically unchanged colonic mucosa from patients who underwent radical colon resection (n=90). We found a statistically significant decrease in STS transcript levels in CRC (P=0.0453). Moreover, we found that sodium butyrate (NaBu) significantly upregulated STS transcript levels in DLD-1 and HCT116 CRC cells. Our results suggest that STS expression can be decreased in the process of large intestinal carcinogenesis. Moreover, we observed that NaBu might increase STS expression in CRC cells.
Collapse
Affiliation(s)
- Agnieszka Anna Rawłuszko
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Poznań, Poland
| | | | | | | | | | | |
Collapse
|
34
|
Steroid sulphatase and oestrogen sulphotransferase in human non-small-cell lung carcinoma. Br J Cancer 2013; 108:1415-24. [PMID: 23531699 PMCID: PMC3629433 DOI: 10.1038/bjc.2013.84] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background: Steroid sulphatase (STS) is one of the steroid-metabolising enzymes involved in desulphating inactive steroid sulphates and oestrogen sulphotransferase (EST) sulphates active oestrogen. The roles of both STS and EST have not been examined in oestrogen-dependent non-small-cell lung cancer (NSCLC). Methods: We evaluated the immunoreactivity of STS and EST in NSCLC cases using immunohistochemistry. The function of STS and EST was further demonstrated using NSCLC cell lines. Results: The immunoreactivity of STS and EST was detected in 49.5% and 27.8% of NSCLC cases, respectively. The immunoreactivity of STS was significantly higher in female adenocarcinoma cases. The STS-positive NSCLCs were also significantly correlated in an inversed manner with tumour size and cell proliferation and tended to be associated with better clinical outcome. However, the immunoreactivity of EST was significantly correlated with intracellular oestradiol concentration. Results of in vitro analysis demonstrated that oestrone sulphate (E1-S) induced and pregnenolone sulphate (Preg-S) inhibited the proliferation in STS-expressing cell lines. The inhibition by Preg-S was reversed by a specific progesterone receptor blocker. Simultaneous addition of E1-S and Preg-S significantly suppressed the proliferation. Conclusion: In NSCLC patients, STS is considered a good prognostic factor. Results of our present study also indicated the benefits of potential progesterone therapy for NSCLC patients.
Collapse
|
35
|
Secky L, Svoboda M, Klameth L, Bajna E, Hamilton G, Zeillinger R, Jäger W, Thalhammer T. The sulfatase pathway for estrogen formation: targets for the treatment and diagnosis of hormone-associated tumors. JOURNAL OF DRUG DELIVERY 2013; 2013:957605. [PMID: 23476785 PMCID: PMC3586502 DOI: 10.1155/2013/957605] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 12/17/2012] [Indexed: 12/15/2022]
Abstract
The extragonadal synthesis of biological active steroid hormones from their inactive precursors in target tissues is named "intracrinology." Of particular importance for the progression of estrogen-dependent cancers is the in situ formation of the biological most active estrogen, 17beta-estradiol (E2). In cancer cells, conversion of inactive steroid hormone precursors to E2 is accomplished from inactive, sulfated estrogens in the "sulfatase pathway" and from androgens in the "aromatase pathway." Here, we provide an overview about expression and function of enzymes of the "sulfatase pathway," particularly steroid sulfatase (STS) that activates estrogens and estrogen sulfotransferase (SULT1E1) that converts active estrone (E1) and other estrogens to their inactive sulfates. High expression of STS and low expression of SULT1E1 will increase levels of active estrogens in malignant tumor cells leading to the stimulation of cell proliferation and cancer progression. Therefore, blocking the "sulfatase pathway" by STS inhibitors may offer an attractive strategy to reduce levels of active estrogens. STS inhibitors either applied in combination with aromatase inhibitors or as novel, dual aromatase-steroid sulfatase inhibiting drugs are currently under investigation. Furthermore, STS inhibitors are also suitable as enzyme-based cancer imaging agents applied in the biomedical imaging technique positron emission tomography (PET) for cancer diagnosis.
Collapse
Affiliation(s)
- Lena Secky
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Martin Svoboda
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Lukas Klameth
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Cluster Translational Oncology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Erika Bajna
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gerhard Hamilton
- Ludwig Boltzmann Cluster Translational Oncology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Robert Zeillinger
- Ludwig Boltzmann Cluster Translational Oncology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, 1090 Vienna, Austria
| | - Theresia Thalhammer
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
36
|
Passarelli MN, Phipps AI, Potter JD, Makar KW, Coghill AE, Wernli KJ, White E, Chan AT, Hutter CM, Peters U, Newcomb PA. Common single-nucleotide polymorphisms in the estrogen receptor β promoter are associated with colorectal cancer survival in postmenopausal women. Cancer Res 2013; 73:767-75. [PMID: 23149914 PMCID: PMC3588850 DOI: 10.1158/0008-5472.can-12-2484] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Loss of estrogen receptor β (ERβ) expression in the gut is associated with colorectal cancer (CRC) initiation and progression. Germline single-nucleotide polymorphisms (SNP) in genes for the sex-steroid hormone receptors are not strongly associated with CRC risk; however, these SNPs have not previously been evaluated in relation to survival after diagnosis. We enrolled 729 women, ages 50 to 74, diagnosed with invasive CRC between 1997 and 2002 in 13 counties covered by the Seattle-Puget Sound Surveillance Epidemiology and End Results cancer registry. Participants provided germline DNA. We selected 99 tag-SNPs for the androgen receptor (AR), ERα (ESR1), ERβ (ESR2), and progesterone receptor (PGR) genes. Mortality outcomes were ascertained from the National Death Index. During a median of 6.6 years of follow-up, 244 deaths occurred (161 from CRC). We identified 20 SNPs (12 of ESR2 and 8 of PGR) for replication in 1,729 women diagnosed with incident invasive CRC (555 deaths; 405 from CRC) from three prospective cohort studies that participate in the Genetics and Epidemiology of Colorectal Cancer Consortium. Three correlated SNPs in the promoter of ESR2 (rs2987983, rs3020443, and rs2978381) were statistically significant predictors of CRC-specific and overall survival. Minor alleles of each were associated with improved survival [for rs2987983, CRC-specific HR, 0.77; 95% confidence interval (CI), 0.60-0.99 in the initial study, and HR, 0.79; CI, 0.64-0.98 in replication]. No associations were noted for SNPs of AR, ESR1, or PGR. SNPs in the promoter of ESR2 may be important to pathways related to the association between ERβ and tumor progression and metastasis.
Collapse
Affiliation(s)
- Michael N. Passarelli
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | - Amanda I. Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - John D. Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
- Centre for Public Health Research, Massey University, Wellington, New Zealand
| | - Karen W. Makar
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Anna E. Coghill
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | | | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | - Andrew T. Chan
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Carolyn M. Hutter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | - Polly A. Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| |
Collapse
|
37
|
Peerless Y, Simon E, Sabo E, Ben-Izhak O, Hershkovitz D. Normal colon tissue and colon carcinoma show no difference in heparanase promoter methylation. Exp Mol Pathol 2013; 94:309-13. [PMID: 23313782 DOI: 10.1016/j.yexmp.2013.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 01/02/2013] [Accepted: 01/02/2013] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Heparanase, the sole heparan sulfate degrading enzyme, has a role in cellular invasion. Accordingly, a large number of studies have demonstrated an association between heparanase expression and tumor stage and patients' prognosis. In colon carcinoma, heparanase shows increased expression in tumor compared to normal tissue and its expression correlates with the presence of metastasis. One of the regulatory mechanisms of heparanase expression is de-methylation on its promoter. In the present study we evaluated the role of heparanase promoter methylation in colon carcinoma. MATERIAL AND METHODS Analysis of heparanase promoter methylation was done on 32 samples of colon carcinoma as well as 30 samples of normal colonic mucosa. DNA was extracted from FFPE tissue and subjected to bisulfite conversion. The relative fraction of methylated and unmethylated DNA was evaluated using quantitative real-time PCR. RESULTS The fraction of methylated DNA was 1 ± 3.4% in the colon carcinoma group, and 2.5 ± 3.3% in the normal colon group (P=0.11). Only one case in the normal group and one case in the tumor group showed more than 10% methylation in the heparanase promoter. CONCLUSION We did not find any significant difference in heparanase promoter methylation between colon carcinoma and normal colonic mucosa, suggesting that heparanase overexpression in colon carcinoma is mediated by other mechanisms.
Collapse
Affiliation(s)
- Yehudit Peerless
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | | | | | | | | |
Collapse
|
38
|
Honma N, Yamamoto K, Ohnaka K, Morita M, Toyomura K, Kono S, Muramatsu M, Arai T, Ueki T, Tanaka M, Kakeji Y, Maehara Y, Okamura T, Ikejiri K, Futami K, Maekawa T, Yasunami Y, Takenaka K, Ichimiya H, Terasaka R. Estrogen receptor-β gene polymorphism and colorectal cancer risk: effect modified by body mass index and isoflavone intake. Int J Cancer 2012; 132:951-8. [PMID: 22729816 DOI: 10.1002/ijc.27688] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 05/10/2012] [Indexed: 12/27/2022]
Abstract
Estrogen receptor (ER)-β signaling has generally been implicated in protection against colorectal cancer. The ER-β gene cytosine-adenine (ESR2 CA) repeat polymorphism was reported to be associated with colorectal cancer, although showing contradicting results probably caused by ethnicity or age distribution of the subjects. We investigated the association between this polymorphism and the colorectal cancer risk in a community-based case-control study in Japan (685 cases/778 controls), including only subjects younger than 75. The effect modifications of the body mass index (BMI) and isoflavone intake were also examined. ESR2 CA repeat polymorphism was determined by polymerase chain reaction using fluorescein-labeled primers. CA repeat alleles were classified into short (S) allele (<22 repeats) and long (L) allele (≥ 22 repeats). Subjects were divided into three genotype groups (SS/SL/LL). The risk of colon cancer, but not of rectal cancer, was increased with an increasing number of L alleles among postmenopausal women; age-adjusted odds ratio (OR) for SL and LL genotypes compared with the SS genotype were 1.78 and 2.91, respectively (trend p = 0.002). Increased risks of colon cancer associated with the L allele were more evident among postmenopausal women with low BMI (<25 kg m(-2)) or with high isoflavone intake. Such associations were not observed among men or premenopausal women. Having longer ESR2 CA repeat increases colon cancer risk among postmenopausal women younger than 75, possibly with modification of BMI and isoflavone intake. Aging and estrogenic condition may be important in the colon cancer pathogenesis associated with ESR2 CA repeat polymorphism.
Collapse
Affiliation(s)
- Naoko Honma
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zuguchi M, Miki Y, Onodera Y, Fujishima F, Takeyama D, Okamoto H, Miyata G, Sato A, Satomi S, Sasano H. Estrogen receptor α and β in esophageal squamous cell carcinoma. Cancer Sci 2012; 103:1348-55. [PMID: 22463081 DOI: 10.1111/j.1349-7006.2012.02288.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 03/21/2012] [Accepted: 03/22/2012] [Indexed: 12/21/2022] Open
Abstract
A gender difference has been reported in the morbidity of esophageal squamous cell carcinoma (ESCC). Estrogens have been proposed to play a role in this difference but the details have not yet been clarified. Therefore, in the present study, we examined the status of estrogen receptor (ER)α and ERβ in 90 Japanese ESCC patients. ERα and ERβ immunoreactivity was detected in the nuclei of ESCC cells (41.1 and 97.8%, respectively). There was a significant positive association between the ERβ H score and histological differentiation (P = 0.0403), TNM-pM (LYM) (P = 0.00164) and Ki67/MIB1 LI of carcinoma cells (P = 0.0497, r = 0.207). In addition, the ERβ status of carcinoma cells was significantly correlated with unfavorable clinical outcome of the patients. Multivariate analysis further revealed the ERβ status in carcinoma cells as an independent unfavorable prognostic factor of these patients. We further examined the effects of estrogen treatment on ESCC cell line (ECGI-10) transfected with ERα or ERβ in vitro. The number of ECGI-10 transfected with ERβ was increased by estradiol or ERβ specific agonist but estradiol did not exert any effect upon the cell number of ECGI-10 transfected with ERα. In summary, the results of the present study clearly demonstrate that the status of ERβ in ESCC was closely associated with the unfavorable prognosis, possibly through altering cell proliferation of carcinoma cells.
Collapse
Affiliation(s)
- Masashi Zuguchi
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sase T, Suzuki T, Miura K, Shiiba K, Sato I, Nakamura Y, Takagi K, Onodera Y, Miki Y, Watanabe M, Ishida K, Ohnuma S, Sasaki H, Sato R, Karasawa H, Shibata C, Unno M, Sasaki I, Sasano H. Runt-related transcription factor 2 in human colon carcinoma: a potent prognostic factor associated with estrogen receptor. Int J Cancer 2012; 131:2284-93. [PMID: 22396198 DOI: 10.1002/ijc.27525] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 02/15/2012] [Indexed: 12/31/2022]
Abstract
Runt-related transcription factor 2 (RUNX2) belongs to the RUNX family of heterodimeric transcription factors, and is mainly associated with osteogenesis. Previous in vitro studies demonstrated that RUNX2 increased the cell proliferation of mouse and rat colon carcinoma cells but the status of RUNX2 has remained unknown in human colon carcinoma. Therefore, we examined clinical significance and biological functions of RUNX2 in colon carcinoma. RUNX2 immunoreactivity was examined in 157 colon carcinoma tissues using immunohistochemistry. RUNX2 immunoreactivity was evaluated as percentage of positive carcinoma cells [i.e., labeling index (LI)]. We used SW480 and DLD-1 human colon carcinoma cells, expressing estrogen receptor-β (ER) in subsequent in vitro studies. RUNX2 immunoreactivity was detected in colon carcinoma cells, and the median value of RUNX2 LI was 67%. RUNX2 LI was significantly associated with Dukes' stage, liver metastasis and ERβ status. In addition, RUNX2 LI was significantly associated with adverse clinical outcome of the colon carcinoma patients, and turned out an independent prognostic factor following multivariate analysis. Results of in vitro studies demonstrated that both SW480 and DLD-1 cells transfected with small interfering RNA against RUNX2 significantly decreased their cell proliferation, migration and invasive properties. In addition, RUNX2 mRNA level was significantly decreased by ER antagonist in these two cells. These findings all suggest that RUNX2 is a potent prognostic factor in human colon carcinoma patients through the promotion of cell proliferation and invasion properties, and is at least partly upregulated by estrogen signals through ERβ of carcinoma cells.
Collapse
Affiliation(s)
- Tomohiko Sase
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Effect of butyrate on aromatase cytochrome P450 levels in HT29, DLD-1 and LoVo colon cancer cells. Biomed Pharmacother 2012; 66:77-82. [PMID: 22386365 DOI: 10.1016/j.biopha.2011.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/15/2011] [Indexed: 02/07/2023] Open
Abstract
Epidemiological studies suggest that colonic production of butyrate and estrogen may be involved in human susceptibility to colorectal cancer (CRC). Estrone (E1) can be produced by the aromatase pathway during the conversion of androstenedione (A) to E1. Therefore, we studied the effect of sodium butyrate (NaBu) on the CYP19A1 transcript and protein levels and on the conversion of A to E1 in HT29, DLD-1 and LoVo CRC cells. We found that NaBu significantly downregulated CYP19A1 transcript and protein levels, a phenomenon that was associated with reduced conversion of A to E1 in HT29, DLD-1 and LoVo cells. Our studies demonstrated that, although butyrate exhibited a protective role in CRC development, this compound may reduce aromatase activity and the production of E1 in colon cancer cells.
Collapse
|
42
|
Inoue T, Miki Y, Abe K, Hatori M, Hosaka M, Kariya Y, Kakuo S, Fujimura T, Hachiya A, Aiba S, Sasano H. The role of estrogen-metabolizing enzymes and estrogen receptors in human epidermis. Mol Cell Endocrinol 2011; 344:35-40. [PMID: 21740958 DOI: 10.1016/j.mce.2011.06.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 05/16/2011] [Accepted: 06/18/2011] [Indexed: 10/18/2022]
Abstract
Local estrogen metabolism and its sensitivities in the skin have been also suggested to contribute to skin homeostasis in addition to age- and/or gender-dependent circulating estrogen, even though their local mechanisms have been largely unknown. To characterize their potential correlations, age- and gender-dependencies were evaluated focusing on 5 pivotal estrogen-metabolizing enzymes including aromatase, estrogen sulfotransferase, steroid sulfatase, and 17β-hydroxysteroid dehydrogenases and estrogen receptors (ERα and ERβ) using immunohistochemistry of 100 human skin specimens. When their epidermal expression levels were compared among 7 age groups, ranging from the teens to the seventies, the highest expression in the teens group and the lowest expression in the seventies group were found in the expression of aromatase and ERβ, respectively, while no significant differences between the male and the female groups were found in the immunoreactivities of our interested proteins. Our results suggest that age-related differences in aromatase and ERβ expressions impact epidermal homeostasis.
Collapse
Affiliation(s)
- Takayoshi Inoue
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Suzuki T, Miki Y, Nakamura Y, Ito K, Sasano H. Steroid sulfatase and estrogen sulfotransferase in human carcinomas. Mol Cell Endocrinol 2011; 340:148-53. [PMID: 21073915 DOI: 10.1016/j.mce.2010.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2009] [Revised: 10/06/2010] [Accepted: 11/01/2010] [Indexed: 01/23/2023]
Abstract
Estrogens are closely involved in the development of hormone-dependent carcinomas. Estrone is locally produced from circulating inactive estrone sulfate by steroid sulfatase (STS), while estrone is inversely inactivated into estrone sulfate by estrogen sulfotransferase (EST). Recent studies suggested importance of this STS pathway in various human carcinomas. Therefore, in this review, we summarized recent results of STS and EST in several estrogen-dependent carcinomas. STS and EST expressions were detected in the breast and endometrial carcinomas, and activation of STS pathway due to increment in STS and/or decrement in EST expressions plays important role in their estrogen-dependent growth. STS expression was also reported in the ovarian and prostate carcinomas. STS/EST status was associated with intratumoral estrogen level in the colon carcinoma, and STS-negative/EST-positive colon carcinoma patients had longer survival. Therefore, STS pathway and estrogen actions may play an important role in the development of these carcinomas, and further investigations are required.
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University, Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | | | | | |
Collapse
|
44
|
Miki Y, Abe K, Suzuki S, Suzuki T, Sasano H. Suppression of estrogen actions in human lung cancer. Mol Cell Endocrinol 2011; 340:168-74. [PMID: 21354461 DOI: 10.1016/j.mce.2011.02.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 02/08/2011] [Accepted: 02/13/2011] [Indexed: 11/21/2022]
Abstract
Estrogen plays a critical role in female reproduction but has also been reported to have important roles in various target tissues expressing estrogen receptor (ER) α and/or ERβ in both male and female. ERs especially ERβ have been demonstrated to be present and functional in both normal human lung and its disorders including cancer. Non-small cell lung carcinomas (NSCLCs) are well-known to be composed of heterogeneous groups. Squamous cell carcinoma is the most common subtype in men, but adenocarcinoma is the most common histologic subtype in women. Therefore, sex steroid hormones such as estrogens have been considered to play some roles in NSCLC. In particular, results of several epidemiological analyses pointed out the association between physiological or artificial alterations of hormone status such as menstruation and postmenopausal administration of hormone replacement therapy and lung cancer risks or its development especially in female subjects. In NSCLC tissues, intratumoral estrogen synthesis via aromatase, which is a key enzyme in the estrogen synthesis involved in aromatization of androgens into estrogens, has recently become of clinical interest as a possible target of therapy. Therefore, in this review, we focused on the potential of an endocrine therapy in NSCLC using clinically available inhibitors of estrogen and aromatase actions.
Collapse
Affiliation(s)
- Yasuhiro Miki
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | | | | | | | | |
Collapse
|
45
|
Ahima RS, Stanley TL, Khor VK, Zanni MV, Grinspoon SK. Estrogen sulfotransferase is expressed in subcutaneous adipose tissue of obese humans in association with TNF-alpha and SOCS3. J Clin Endocrinol Metab 2011; 96:E1153-8. [PMID: 21543429 PMCID: PMC3135195 DOI: 10.1210/jc.2010-2903] [Citation(s) in RCA: 14] [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: 12/14/2022]
Abstract
CONTEXT AND OBJECTIVE Estrogen sulfotransferase (EST) catalyzes the inactivation of estrone and estradiol in numerous tissues. Animal studies suggest that EST modulates glucose and lipid metabolism in adipose tissue, but it is unknown whether EST is expressed in human adipose tissue and, if so, how its expression relates to features of the metabolic syndrome. DESIGN AND PARTICIPANTS Cross-sectional data from 16 obese men and women with metabolic dysregulation were collected as part of a larger randomized trial at an academic medical center. OUTCOME MEASURES Participants underwent assessment of body composition, oral glucose tolerance testing, measurement of serum hormones and inflammatory markers, and sc fat biopsy to assess adipose expression of TNF-α, suppressor of cytokine signaling 3 (SOCS3), leptin, adiponectin, and EST. RESULTS EST expression was detectable in sc adipose tissue from both men and women. Log(10) EST mRNA was not significantly associated with age, race, sex or menopausal status, or circulating levels of estrogen or testosterone. In univariate analysis, log(10) EST mRNA was significantly associated with visceral adipose tissue area (r = 0.57, P = 0.02) as well as adipose tissue expression of TNF-α (r = 0.94, P < 0.0001) and SOCS3 mRNA (r = 0.93, P < 0.0001). The associations between EST expression and TNF-α and SOCS3 held in multivariate modeling controlling for age, race, sex and menopausal status, and visceral adiposity. EST expression was not significantly associated with the adipose tissue levels of leptin or adiponectin expression. CONCLUSIONS EST is expressed in abdominal sc adipose tissue of both obese males and females in association with expression of TNF-α and SOCS3, suggesting potential roles in inflammation. Further studies are needed to determine the specific metabolic roles of EST expression in human adipose tissue.
Collapse
Affiliation(s)
- Rexford S Ahima
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | | | | | |
Collapse
|
46
|
Lin JH, Manson JE, Kraft P, Cochrane BB, Gunter MJ, Chlebowski RT, Zhang SM. Estrogen and progesterone-related gene variants and colorectal cancer risk in women. BMC MEDICAL GENETICS 2011; 12:78. [PMID: 21627810 PMCID: PMC3125237 DOI: 10.1186/1471-2350-12-78] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 05/31/2011] [Indexed: 12/11/2022]
Abstract
Background Observational studies and randomized trials have suggested that estrogens and/or progesterone may lower the risk for colorectal cancer. Inherited variation in the sex-hormone genes may be one mechanism by which sex hormones affect colorectal cancer, although data are limited. Method We conducted a comprehensive evaluation of single nucleotide polymorphisms (SNPs) in genes encoding 3 hormone receptors (ESR1, ESR2, PGR) and 5 hormone synthesizers (CYP19A1 and CYP17A1, HSD17B1, HSD17B2, HSD17B4) among 427 women with incident colorectal cancer and 871 matched controls who were Caucasians of European ancestry from 93676 postmenopausal women enrolled in the Women's Health Initiative Observational cohort. A total of 242 haplotype-tagging and functional SNPs in the 8 genes were included for analysis. Unconditional logistic regression with adjustment for age and hysterectomy status was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). Results We observed a weak association between the CYP17A1 rs17724534 SNP and colorectal cancer risk (OR per risk allele (A) = 1.39, 95% CI = 1.09-1.78, corrected p-value = 0.07). In addition, a suggestive interaction between rs17724534 and rs10883782 in 2 discrete LD blocks of CYP17A1 was observed in relation to colorectal cancer (empirical p value = 0.04). Moreover, one haplotype block of CYP19A1 was associated with colorectal cancer (corrected global p value = 0.02), which likely reflected the association with the tagging SNP, rs1902584, in the block. Conclusion Our findings offer some support for a suggestive association of CYP17A1 and CYP19A1 variants with colorectal cancer risk.
Collapse
Affiliation(s)
- Jennifer H Lin
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Variation in the CYP19A1 gene and risk of colon and rectal cancer. Cancer Causes Control 2011; 22:955-63. [PMID: 21479914 DOI: 10.1007/s10552-011-9768-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 03/31/2011] [Indexed: 12/23/2022]
Abstract
CYP19A1, or aromatase, influences estrogen-metabolizing enzymes and may influence cancer risk. We examine variation in the CYP19A1 gene and risk of colorectal cancer using data from population-based case-control studies (colon n = 1,574 cases, 1,970 controls; rectal n = 791 cases, 999 controls). Four SNPs were statistically significantly associated with colon cancer and four were associated with rectal cancer. After adjustment for multiple comparisons, the AA genotype of rs12591359 was associated with an increased risk of colon cancer (OR 1.44 95% CI 1.16-1.80) and the AA genotype of rs2470144 was associated with a reduced risk of rectal cancer (OR 0.65 95% CI 0.50-0.84). Variants of CYP19A1 were associated with CIMP+ and CIMP+/KRAS2-mutated tumors. CT/TT genotypes of rs1961177 were significantly associated with an increased likelihood of a MSI+ colon tumor (OR 1.77 95% CI 1.26-2.37). We observed statistically significant interactions between genetic variation in NFκB1 and CYP19A1 for both colon and rectal cancer. Our data suggest the importance of CYP19A1 in the development of colon and rectal cancer and that estrogen may influence risk through an inflammation-related mechanism.
Collapse
|
48
|
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
|
49
|
Miki Y, Suzuki T, Abe K, Suzuki S, Niikawa H, Iida S, Hata S, Akahira JI, Mori K, Evans DB, Kondo T, Yamada-Okabe H, Sasano H. Intratumoral localization of aromatase and interaction between stromal and parenchymal cells in the non-small cell lung carcinoma microenvironment. Cancer Res 2010; 70:6659-69. [PMID: 20710045 DOI: 10.1158/0008-5472.can-09-4653] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Estrogens produced as a result of intratumoral aromatization has been recently shown to play important roles in proliferation of human non-small cell lung carcinomas (NSCLC), but the details have remained largely unknown. Therefore, in this study, we evaluated the possible roles of intratumoral aromatase in NSCLCs as follows: (a) evaluation of intratumoral localization of aromatase mRNA/protein in six lung adenocarcinoma cases using laser capture microdissection combined with quantitative reverse transcriptase-PCR and immunohistochemistry; (b) examination of the possible effects of isolated stromal cells from lung carcinoma tissues on aromatase mRNA transcript expression in lung carcinoma cell lines (A549 and LK87) through a coculture system; and (c) screening of cytokines derived from stromal LK001S and LK002S cells using cytokine antibody arrays and subsequent evaluation of effects of these cytokines on aromatase expression in A549 and LK87. Both aromatase mRNA and protein were mainly detected in intratumoral carcinoma cells but not in stromal cells. Aromatase expression of A549 and LK87 was upregulated in the presence of LK001S or LK002S cells. Several cytokines such as interleukin-6 (IL-6), oncostatin M, and tumor necrosis factor-alpha, all known as inducible factors of aromatase gene, were detected in conditioned media of LK001S and LK002S cells. Treatment of both oncostatin M and IL-6 induced aromatase gene expression in A549 an LK87, respectively. These results all indicated that intratumoral microenvironments, especially carcinoma-stromal cell interactions, play a pivotal role in the regulation of intratumoral estrogen synthesis through aromatase expression in human lung adenocarcinomas.
Collapse
Affiliation(s)
- Yasuhiro Miki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sasaki Y, Miki Y, Hirakawa H, Onodera Y, Takagi K, Akahira JI, Honma S, Ishida T, Watanabe M, Sasano H, Suzuki T. Immunolocalization of estrogen-producing and metabolizing enzymes in benign breast disease: comparison with normal breast and breast carcinoma. Cancer Sci 2010; 101:2286-92. [PMID: 20682005 PMCID: PMC11159500 DOI: 10.1111/j.1349-7006.2010.01673.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
It is well known that estrogens play important roles in the cell proliferation of breast carcinoma. Benign breast disease (BBD) contains a wide spectrum of diseases, and some are considered an important risk factor for subsequent breast carcinoma development. However, the significance of estrogens in BBD has remained largely unknown. Therefore, in this study, we examined tissue concentrations of estrogens and immunolocalization of estrogen-producing/metabolizing enzymes in BBD, and compared these findings with those in the normal breast and ductal carcinoma in situ (DCIS). Tissue concentration of estradiol in BBD (n = 9) was significantly (3.4-fold) higher than normal breast (n = 9) and nearly the same (0.7-fold) as in DCIS (n = 9). Immunoreactivity of estrogen sulfotransferase in BBD was significantly lower (n = 82) than normal breast (n = 28) but was not significantly different from DCIS (n = 28). Aromatase and steroid sulfatase immunoreactivities tended to be higher (P = 0.07) in BBD than in normal breast, and 17β-hydroxysteroid dehydrogenase type 1 immunoreactivity was significantly higher in BBD than normal breast in the postmenopausal tissues. Immunoreactivity of estrogen and progesterone receptors was also significantly higher in BBD than normal breast. These results suggest that tissue concentration of estradiol is increased in BBD at a level similar to DCIS, which is considered mainly due to loss of estrogen sulfotransferase expression. Increased local estradiol concentration in BBD due to aberrant expression of estrogen-producing/metabolizing enzymes may play important roles in the accumulation of estradiol-mediated growth and/or subsequent development of breast carcinoma.
Collapse
Affiliation(s)
- Yoshie Sasaki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|