1
|
Liu T, Zhao M, Peng L, Chen J, Xing P, Gao P, Chen L, Qiao X, Wang Z, Di J, Qu H, Jiang B, Su X. WFDC3 inhibits tumor metastasis by promoting the ERβ-mediated transcriptional repression of TGFBR1 in colorectal cancer. Cell Death Dis 2023; 14:425. [PMID: 37443102 PMCID: PMC10345115 DOI: 10.1038/s41419-023-05956-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/20/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
Estrogen plays a protective role in colorectal cancer (CRC) and primarily functions through estrogen receptor β (ERβ). However, clinical strategies for CRC therapy associated with ERβ are still under investigation. Our discoveries identified WFDC3 as a tumor suppressor that facilitates estrogen-induced inhibition of metastasis through the ERβ/TGFBR1 signaling axis. WFDC3 interacts with ERβ and increases its protein stability by inhibiting its proteasome-dependent degradation. WFDC3 represses TGFBR1 expression through ERβ-mediated transcription. Blocking TGFβ signaling with galunisertib, a drug used in clinical trials that targets TGFBR1, impaired the migration of CRC cells induced by WFDC3 depletion. Moreover, there was clinical significance to WFDC3 in CRC, as CRC patients with high WFDC3 expression in tumor cells had favorable prognoses. Therefore, this work suggests that WFDC3 could be an indicator for therapies targeting the estrogen/ERβ pathway in CRC patients.
Collapse
Affiliation(s)
- Tianqi Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Min Zhao
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, Sunshine Coast, QLD, 4556, Australia
| | - Lin Peng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Jiangbo Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Pu Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Pin Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Lei Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiaowen Qiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Zaozao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Jiabo Di
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Hong Qu
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, 100871, Beijing, People's Republic of China.
| | - Beihai Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| | - Xiangqian Su
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| |
Collapse
|
2
|
Therapeutic Strategies and Potential Actions of Female Sex Steroid Hormones and Their Receptors in Colon Cancer Based on Preclinical Studies. Life (Basel) 2022; 12:life12040605. [PMID: 35455096 PMCID: PMC9032023 DOI: 10.3390/life12040605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Several epidemiological studies have reported that the use of female sex steroid hormones could reduce the risk of colon cancer (CRC). This review summarizes the available data related to estradiol (E2) and progesterone (P4) single and dual treatments in CRC male and female in vitro and in vivo models, mainly from preclinical studies, alongside their potential molecular mechanisms. Most of the studies showed that E2 exogenous treatment and/or reactivation of its beta receptor (ERβ) significantly inhibited cell proliferation, induced cell cycle arrest, and promoted apoptosis by modulating several molecular pathways. Likewise, the inhibition of ERα receptors produced similar antitumorigenic actions, both in vivo and in vitro, suggesting that E2 could have dual opposing roles in CRC that are dependent on the expression profile of its nuclear receptors. The available studies on P4 are scarce, and the results revealed that in vitro and in vivo treatments with natural and synthetic progesterone were also associated with promising tumoricidal actions. Nevertheless, the combination of E2 with P4 showed enhanced anticancer activities compared with their monotherapy protocols in male–female cell lines and animals. Collectively, the studies suggested that the female sex steroid hormones could provide a novel and effective therapeutic strategy against CRC.
Collapse
|
3
|
Ditonno I, Losurdo G, Rendina M, Pricci M, Girardi B, Ierardi E, Di Leo A. Estrogen Receptors in Colorectal Cancer: Facts, Novelties and Perspectives. Curr Oncol 2021; 28:4256-4263. [PMID: 34898546 PMCID: PMC8544350 DOI: 10.3390/curroncol28060361] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is the second cause of cancer-related death in both sexes worldwide. As pre-menopausal women are less likely to develop CRC compared to age-matched men, a protective role for estrogens has been hypothesized. Indeed, two isoforms of nuclear estrogen receptors (ER) have been described: ERα and ERβ. While the binding of 17beta-estradiol to ERα activates anti-apoptotic pathways, the interaction with ERβ activates caspase-3, inducing apoptosis. In this regard, several pieces of evidence show that ERβ tends to be under-regulated in advanced adenomas and CRC, with an opposite trend for ERα. Furthermore, ERβ stimulation slows adenomatous polyp growth and modulates relevant CRC pathways. Based on such considerations, dietary modulation of ER is promising, particularly in subjects with genetic predisposition for CRC. Nevertheless, the main limitation is the lack of clinical trials on a large population scale.
Collapse
Affiliation(s)
- Ilaria Ditonno
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University “Aldo Moro” of Bari, 70124 Bari, Italy; (I.D.); (G.L.); (M.R.); (E.I.)
| | - Giuseppe Losurdo
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University “Aldo Moro” of Bari, 70124 Bari, Italy; (I.D.); (G.L.); (M.R.); (E.I.)
- Ph.D. Course in Organs and Tissues Transplantation and Cellular Therapies, Department of Emergency and Organ Transplantation, University “Aldo Moro” of Bari, 70124 Bari, Italy
| | - Maria Rendina
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University “Aldo Moro” of Bari, 70124 Bari, Italy; (I.D.); (G.L.); (M.R.); (E.I.)
| | | | | | - Enzo Ierardi
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University “Aldo Moro” of Bari, 70124 Bari, Italy; (I.D.); (G.L.); (M.R.); (E.I.)
| | - Alfredo Di Leo
- Ph.D. Course in Organs and Tissues Transplantation and Cellular Therapies, Department of Emergency and Organ Transplantation, University “Aldo Moro” of Bari, 70124 Bari, Italy
| |
Collapse
|
4
|
Pozios I, Seel NN, Hering NA, Hartmann L, Liu V, Camaj P, Müller MH, Lee LD, Bruns CJ, Kreis ME, Seeliger H. Raloxifene inhibits pancreatic adenocarcinoma growth by interfering with ERβ and IL-6/gp130/STAT3 signaling. Cell Oncol (Dordr) 2021; 44:167-177. [PMID: 32940862 PMCID: PMC7906944 DOI: 10.1007/s13402-020-00559-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Currently, the exact role of estrogen receptor (ER) signaling in pancreatic cancer is unknown. Recently, we showed that expression of phosphorylated ERβ correlates with a poor prognosis in patients with pancreatic ductal adenocarcinoma (PDAC). Here, we hypothesized that raloxifene, a FDA-approved selective ER modulator (SERM), may suppress PDAC tumor growth by interfering with ERβ signaling. To test this hypothesis, we studied the impact of raloxifene on interleukin-6/glycoprotein-130/signal transducer and activator of transcription-3 (IL-6/gp130/STAT3) signaling. METHODS Human PDAC cell lines were exposed to raloxifene after which growth inhibition was assessed using a BrdU assay. ER knockdown was performed using siRNAs specific for ERα and ERβ. The effects of raloxifene on IL-6 expression and STAT3 phosphorylation in PDAC cells were assessed by ELISA and Western blotting, respectively. In addition, raloxifene was administered to an orthotopic PDAC tumor xenograft mouse model, after which tumor growth was monitored and immunohistochemistry was performed. RESULTS Raloxifene inhibited the in vitro growth of PDAC cells, and this effect was reversed by siRNA-mediated knockdown of ERβ, but not of ERα, indicating ER isotype-specific signaling. We also found that treatment with raloxifene inhibited the release of IL-6 and suppressed the phosphorylation of STAT3Y705 in PDAC cells. In vivo, we found that orthotopic PDAC tumor growth, lymph node and liver metastases as well as Ki-67 expression were reduced in mice treated with raloxifene. CONCLUSIONS Inhibition of ERβ and the IL-6/gp130/STAT3 signaling pathway by raloxifene leads to potent reduction of PDAC growth in vitro and in vivo. Our results suggest that ERβ signaling and IL-6/gp130 interaction may serve as promising drug targets for pancreatic cancer and that raloxifene may serve as an attractive therapeutic option for PDAC patients expressing the ERβ isotype.
Collapse
Affiliation(s)
- Ioannis Pozios
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Nina N Seel
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Munich, Germany
| | - Nina A Hering
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Lisa Hartmann
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Verena Liu
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
- Department of Minimal Invasive and Visceral Surgery, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Peter Camaj
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Munich, Germany
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Mario H Müller
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
- Department of Minimal Invasive and Visceral Surgery, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Lucas D Lee
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Christiane J Bruns
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Martin E Kreis
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Hendrik Seeliger
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
5
|
Zhang Y, Wu Y, Zhou X, Yi B, Wang L. Estrogen Receptor Beta Inhibits The Proliferation, Migration, And Angiogenesis Of Gastric Cancer Cells Through Inhibiting Nuclear Factor-Kappa B Signaling. Onco Targets Ther 2019; 12:9153-9164. [PMID: 31807000 PMCID: PMC6842292 DOI: 10.2147/ott.s214529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose This study aimed to investigate the regulatory roles of estrogen receptor beta (ERβ) on gastric cancer (GC) cells, and reveal the potential mechanisms relating to nuclear factor-kappa B (NF-κB) signaling. Methods GC cell lines SGC7901 and MKN45 were transfected with pEGFP-C1-ERβ to overexpress ERβ, and treated with PMA (a NF-κB activator) to activate NF-κB signaling. The cell proliferation and migration, as well as the formation of vessel-like structures in human venous endothelial cells (HUVECs) were detected. The expression of ERβ, NF-κB p65, p-NF-κB p65, Ki67 (a proliferation marker), vascular endothelial growth factor A (VEGF-A) and matrix metalloproteinase 2 (MMP-2), the DNA binding activity of NF-κB p65, the content of VEGF-A, and the activity of MMP-2 were detected in SGC7901 and MKN45 cells. Results The transfection of pEGFP-C1-ERβ significantly increased the expression of ERβ in SGC7901 and MKN45 cells (P < 0.05). Overexpression of ERβ in SGC7901 and MKN45 cells significantly decreased the cell activity, cell number in G2/M phase, cell migration, the expression of Ki67, VEGF-A and MMP-2, VEGF-A content, MMP-2 activity, as well as the number of vessel-like structures formed by HUVECs (P < 0.05). Overexpression of ERβ also significantly decreased the DNA binding activity and the expression of p-NF-κB p65 in SGC7901 and MKN45 cells (P < 0.05). The anti-tumor effect of ERβ overexpression on GC cells was reversed by the intervention of PMA (P < 0.05). Conclusion Overexpression of ERβ inhibited the proliferation, migration, and angiogenesis of GC cells through inhibiting NF-κB signaling.
Collapse
Affiliation(s)
- Yiping Zhang
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Yahua Wu
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Xufeng Zhou
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Benyi Yi
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Lili Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| |
Collapse
|
6
|
Zhang Y, Yin C, Zhou X, Wu Y, Wang L. Silencing of estrogen receptor β promotes the invasion and migration of osteosarcoma cells through activating Wnt signaling pathway. Onco Targets Ther 2019; 12:6779-6788. [PMID: 31692529 PMCID: PMC6710541 DOI: 10.2147/ott.s219222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/30/2019] [Indexed: 12/31/2022] Open
Abstract
Purpose This study aimed to evaluate the specific roles of estrogen receptor β (ERβ) on the invasion and migration of osteosarcoma (OS) cells and explore the regulatory mechanisms relating with Wnt signaling pathway. Methods The expression of ERβ was detected in human OS tissues by quantitative real-time PCR and immunohistochemistry. U2-OS cells were transfected with siRNA-ERβ (si-ERβ) to downregulate ERβ and treated with FH535 to inhibit Wnt signaling. The migration and invasion ability was detected by scratch and transwell assay, respectively. The expression of β-catenin, MMP-7, and MMP-9 was detected by Western blot. Subcutaneous tumor-bearing model was established by injection of U2-OS cells into mice, and the tumor volumes were measured. Orthotopic transplantation model was established by transplantation of tumor tissues into the liver of mice, and the metastatic tumors were counted. Results ERβ was downregulated in human OS tissues and U2-OS cells. The transfection of si-ERβ significantly increased the scratch healing rate; the number of invasion cells; and the expression of β-catenin, MMP-7, and MMP-9 in U2-OS cells. The injection of si-ERβ-transfected U2-OS cells into mice significantly increased the subcutaneous tumor volume; the expression of β-catenin, MMP-7, and MMP-9; and the number of metastatic tumors in liver tissues. The promoting effects of si-ERβ on the invasion and migration of U2-OS cells were significantly reversed by FH535 in vitro and in vivo. Conclusion Silencing of ERβ promotes the invasion and migration of OS cells via activating Wnt signaling pathway.
Collapse
Affiliation(s)
- Yiping Zhang
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Changchang Yin
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Xufeng Zhou
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Yahua Wu
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Lili Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| |
Collapse
|
7
|
Galal AMF, Soltan MM, Ahmed ER, Hanna AG. Synthesis and biological evaluation of novel 5-chloro- N-(4-sulfamoylbenzyl) salicylamide derivatives as tubulin polymerization inhibitors. MEDCHEMCOMM 2018; 9:1511-1528. [PMID: 30288225 PMCID: PMC6148682 DOI: 10.1039/c8md00214b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/15/2018] [Indexed: 12/28/2022]
Abstract
A novel series of sulfonamide derivatives, coupled with a salicylamide scaffold, was designed and synthesized. The structures of the synthesized compounds were established using 1H NMR, 13C NMR and high-resolution mass spectroscopy. The synthesized compounds were tested in vitro against five types of human cell lines. Two were breast adenocarcinoma, including the hormone-dependent MCF-7 and the hormone-independent MDA-MB-231. The others were the colorectal adenocarcinoma Caco-2, the carcinoma HCT-116 and the immortalized retinal-pigmented epithelium, hTERT-RPE1. Nine sulfonamides were able to inhibit the growth of the four tested cancer cells. Compound 33 was the most active against the selected colon cancer (Caco-2 and HCT-116) subtypes, while compound 24 showed the best efficacy against the examined breast cancer (MCF-7 and MDA-MB-231) cells. The selectivity index introduced compounds 24 and 33 as having the best selectivity among the breast and colon subtypes, respectively. In vitro tubulin polymerization experiments and flow cytometric assays showed that compounds 24 and 33 led to cell cycle arrest at the G2/M phase in a dose-dependent manner by effectively inhibiting tubulin polymerization. Furthermore, the results of the molecular docking studies indicate that this class of compounds can bind to the colchicine-binding site of tubulin.
Collapse
Affiliation(s)
- Alaaeldin M F Galal
- Chemistry of Natural Compounds Department , Pharmaceutical and Drug Industries Research Division , National Research Centre , 33 El Bohouth St. (former El Tahir St.) , Dokki , Giza , 12622 Egypt .
| | - Maha M Soltan
- Biology unit , Central Laboratory for Pharmaceutical and Drug Industries Research Division , Chemistry of Medicinal Plants Department , Pharmaceutical and Drug Industries Research Division , National Research Centre , 33 El Bohouth St. 33 , Dokki , Giza 12622 , Egypt
| | - Esam R Ahmed
- Confirmatory Diagnostic unit , Vacsera , Giza , Egypt
| | - Atef G Hanna
- Chemistry of Natural Compounds Department , Pharmaceutical and Drug Industries Research Division , National Research Centre , 33 El Bohouth St. (former El Tahir St.) , Dokki , Giza , 12622 Egypt .
| |
Collapse
|
8
|
Faknuam S, Assanasen T, Ruangvejvorachai P, Hanvivadhanakul P, Intragumtornchai T, Rojnuckarin P. Estrogen receptor beta expression and prognosis of diffuse large B cell lymphoma. ACTA ACUST UNITED AC 2017; 23:235-241. [PMID: 29032728 DOI: 10.1080/10245332.2017.1389508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Estrogen receptor beta (ERβ)-selective agonists inhibited B cell lymphoma growth in animal models. However, a recent study found that higher ERβ expression in tissue from diffuse large B cell lymphoma (DLBCL) patients indicated a poorer survival. This study aimed to determine the ERβ expression in DLBCL tissue using immunohistochemistry and correlate with clinical outcomes. METHODS Diagnostic tissues from newly diagnosed adult DLBCL patients treated with Rituximab-Cyclophosphamide/Doxorubicin/Vincristine/Prednisolone were counted for ERβ1-expressing cells. Nodal lymphoma (N = 41) was analyzed separately from extra-nodal DLBCL (N = 31). RESULTS On immunohistochemistry, ERβ1 was expressed in 73.6% of cases with the median expressing cells of 20%. For nodal lymphoma, high ERβ expression (≥25%) was associated with poorer event free survival (EFS) independent of the international prognostic index with the adjusted hazard ratio (HR) of 2.49 (95% Confidence interval (CI) 1.03-6.00, P = 0.042). On the contrary, high ERβ expression (≥25%) was associated with superior outcomes in extra-nodal DLBCL with the adjusted HR of 0.25 (95% CI 0.09-0.75, P = 0.013) for EFS and adjusted HR of 0.29 (95% CI 0.10-0.85, P = 0.024) for overall survival in multivariate analyses. CONCLUSION ERβ1 protein expression represented opposite prognostic factors in nodal vs. extra-nodal DLBCL.
Collapse
Affiliation(s)
- Saruta Faknuam
- a Department of Medicine, Faculty of Medicine , Chulalongkorn University and King Chulalongkorn Memorial Hospital (KCMH), Thai Red Cross Society , Bangkok , Thailand
| | - Thamathorn Assanasen
- b Department of Pathology, Faculty of Medicine , Chulalongkorn University and King Chulalongkorn Memorial Hospital (KCMH), Thai Red Cross Society , Bangkok , Thailand
| | - Preecha Ruangvejvorachai
- b Department of Pathology, Faculty of Medicine , Chulalongkorn University and King Chulalongkorn Memorial Hospital (KCMH), Thai Red Cross Society , Bangkok , Thailand
| | - Punchong Hanvivadhanakul
- c Department of Medicine, Faculty of Medicine , Thammasat University Hospital , Bangkok , Thailand
| | - Tanin Intragumtornchai
- a Department of Medicine, Faculty of Medicine , Chulalongkorn University and King Chulalongkorn Memorial Hospital (KCMH), Thai Red Cross Society , Bangkok , Thailand
| | - Ponlapat Rojnuckarin
- a Department of Medicine, Faculty of Medicine , Chulalongkorn University and King Chulalongkorn Memorial Hospital (KCMH), Thai Red Cross Society , Bangkok , Thailand
| |
Collapse
|
9
|
Iannone A, Losurdo G, Pricci M, Girardi B, Massaro A, Principi M, Barone M, Ierardi E, Di Leo A. Stool Investigations for Colorectal Cancer Screening: From Occult Blood Test to DNA Analysis. J Gastrointest Cancer 2016; 47:143-151. [PMID: 26922358 DOI: 10.1007/s12029-016-9810-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE We report an update of current methods for colorectal cancer (CRC) screening based on fecal sample analysis. METHODS A systematic review of the literature was performed in MEDLINE, EMBASE, and Science Direct electronic databases. RESULTS Blood in the stools is the first and most used strategy. Fecal occult blood test (FOBT) and fecal immunochemical test (FIT) are the main methods. Both are economic, easy to perform with high specificity, and low sensitivity. Based on CRC multi-step process with genetic and epigenetic alterations in large bowel cell DNA, single mutations or panels of alterations have been detected. These tests have the advantage of a marked improvement of the sensitivity when compared to fecal blood. However, high costs, poor availability, and correct choice of marker panel represent the major limits. A specific sDNA panel including aberrantly methylated BMP3 and NDRG4 promoter regions, mutant k-ras and β-actin (a reference gene for human DNA quantity), and an immunochemical assay for human hemoglobin has been recently approved by Food and Drug Administration. Novel promising biomarkers for CRC screening are represented by microRNAs (miRNAs), a group of 18-25 nucleotide non-coding RNA molecules that regulate gene expression. Reports on these fecal biomarkers are case-control studies, and each of them evaluates single miRNAs or multi-target panels. On the other hand, some fecal proteins have been studied as possible CRC screening markers, even though they demonstrated poor results. Finally, alterations of estrogen receptor-beta (i.e., dramatic reduction in the early stage of CRC) have been demonstrated in tissue samples. CONCLUSIONS Specific investigations are warranted in order to add further noninvasive markers to the panel of CRC screening tools.
Collapse
Affiliation(s)
- Andrea Iannone
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giuseppe Losurdo
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Maria Pricci
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Bruna Girardi
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Antonio Massaro
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Mariabeatrice Principi
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Michele Barone
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Enzo Ierardi
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Alfredo Di Leo
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy.
| |
Collapse
|
10
|
Williams C, DiLeo A, Niv Y, Gustafsson JÅ. Estrogen receptor beta as target for colorectal cancer prevention. Cancer Lett 2016; 372:48-56. [PMID: 26708506 PMCID: PMC4744541 DOI: 10.1016/j.canlet.2015.12.009] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/05/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is a leading cause of death in the United States. Despite its slow development and the capacity for early diagnosis, current preventive approaches are not sufficient. However, a role for estrogen has been demonstrated in multiple epidemiologic studies, which may benefit CRC prevention. A large body of evidence from preclinical studies indicates that expression of the estrogen receptor beta (ERβ/ESR2) demonstrates an inverse relationship with the presence of colorectal polyps and stage of tumors, and can mediate a protective response. Natural compounds, including phytoestrogens, or synthetic ERβ selective agonists, can activate or upregulate ERβ in the colon and promote apoptosis in preclinical models and in clinical experience. Importantly, this activity has been associated with a reduction in polyp formation and, in rodent models of CRC, has been shown to lower incidence of colon adenocarcinoma. Collectively, these findings indicate that targeted activation of ERβ may represent a novel clinical approach for management of colorectal adenomatous polyps and prevention of colorectal carcinoma in patients at risk for this condition. In this review, we discuss the potential of new chemopreventive or dietary approaches based on estrogen signaling.
Collapse
Affiliation(s)
- Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5056, USA; SciLifeLab, School of Biotechnology, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Biosciences and Nutrition, Novum, Karolinska Institutet, 141 83 Stockholm, Sweden.
| | - Alfredo DiLeo
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Yaron Niv
- Department of Gastroenterology, Rabin Medical Center, Tel Aviv University, Petach Tikva 49100, Israel
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5056, USA; Department of Biosciences and Nutrition, Novum, Karolinska Institutet, 141 83 Stockholm, Sweden
| |
Collapse
|
11
|
Park SB, Kim CH, Hong M, Yang HJ, Chung CK. Effect of a selective estrogen receptor modulator on bone formation in osteoporotic spine fusion using an ovariectomized rat model. Spine J 2016; 16:72-81. [PMID: 26343247 DOI: 10.1016/j.spinee.2015.08.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/18/2015] [Accepted: 08/25/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Selective estrogen receptor modulators (SERMs) are drugs used to treat osteoporosis. Selective estrogen receptor modulators improve bone mineral density and bone mechanical strength. However, there is a lack of data on the effect of SERMs on osteoporotic spinal fusion. PURPOSE This study aimed to investigate whether treatment with a SERM enhances spinal fusion in an ovariectomized (OVX) rat model. STUDY DESIGN An in vivo animal study was carried out. METHODS Female Sprague Dawley rats (n=90) were OVX or sham-operated, and randomized into three groups: control (sham-operated+fusion procedure+saline administration), OVX (OVX+fusion procedure+saline administration), and SERM (OVX+fusion procedure+administration of SERM). Eight weeks after OVX, a bilateral lumbar spinal fusion procedure was performed using autologous iliac bone. In each group, gene expression was evaluated at 2, 4, and 8 weeks after the fusion procedure, histologic analysis was performed at 4 and 8 weeks after the procedure, and bone parameters were measured by micro-computed tomography at 2 days, 4 weeks, and 8 weeks after the procedure. RESULTS Bone mineral density and trabecular quality of the vertebral body were significantly higher in the SERM group compared with the OVX group 16 weeks after OVX. The fusion rate and bone volume ratio were higher in the fusion bed of the SERM group compared with the OVX group 8 weeks after the fusion procedure. Real-time reverse transcription polymerase chain reaction at 4 and 8 weeks after the fusion procedure showed increased expression of the genes for osteoblast-related markers (alkaline phosphatase, osteocalcin, runt-related transcription factor 2, and transforming growth factor) in the SERM group compared with the OVX group. The OVX group showed sparse bone mass between transverse processes. By contrast, the SERM group exhibited a compact bridging fusion mass within the fusion bed at 8 weeks after the fusion procedure. CONCLUSIONS Selective estrogen receptor modulator treatment improved the trabecular quality of the vertebral body, enhanced spinal fusion, and increased the amount of compact bone mass within the fusion bed in rats that had received an ovariectomy. Therapeutic SERM treatment may be recommended to improve the fusion rate in osteoporotic patients who undergo spinal fusion.
Collapse
Affiliation(s)
- Sung Bae Park
- Department of Neurosurgery, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Chi Heon Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Minyoung Hong
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee-Jin Yang
- Department of Neurosurgery, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Chun Kee Chung
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, South Korea; Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, South Korea; Clinical Research Institute, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|
12
|
Pterostilbine, an active component of blueberries, sensitizes colon cancer cells to 5-fluorouracil cytotoxicity. Sci Rep 2015; 5:15239. [PMID: 26472352 PMCID: PMC4608003 DOI: 10.1038/srep15239] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/14/2015] [Indexed: 01/08/2023] Open
Abstract
Although colorectal cancer (CRC) treatment with 5-fluorouracil (5-FU) is the first line of therapy for this debilitating disease, treatment effectiveness is often hampered by the development of drug resistance and toxicity at high doses. ER-β can play an important role in CRC development and possibly in its response to therapy. Pterostilbene (PT) possesses antioxidant and anticancer effects that are mediated by ER-β. In the current study, we test the hypothesis that PT sensitizes colon cancer cells to 5-FU and we examine the underlying mechanism(s) by which PT exerts its cytotoxic effects in CRC cells. Our data indicate that PT exhibited a more potent cytotoxic effect in Caco-2 compared to HCT-116 cells. PT/5-FU co-treatment was more effective in Caco-2 cells. Our data indicate that ER-β is expressed at higher levels in Caco-2 cells and its levels are further boosted with PT treatment. PT significantly suppressed Akt and ERK phosphorylations, and enhanced FOXO-1 and p27kip1 levels in Caco-2 cells. PT also induced a significant increase in Caco-2 cells at pre-G phase coupled with increased Bax/Bcl-2 ratio and PARP cleavage. These results provide a rationale for novel combination treatment strategies, especially for patients with 5-FU-resistant tumors expressing ER-β protein.
Collapse
|
13
|
Lian JY, Tuo BG, Wen GR, Jin H, Liang T. Role of estrogen receptors in digestive system tumors. Shijie Huaren Xiaohua Zazhi 2015; 23:4227-4235. [DOI: 10.11569/wcjd.v23.i26.4227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Estrogen receptors are steroid hormone receptors that modulate the expression of target genes when bound to ligand. Humans have two ligand-activated transcription factors that bind to estrogen, encoded by separate genes, estrogen receptor α (ERα) and estrogen receptor β (ERβ). In addition, the membrane localized G protein-coupled estrogen receptor 1 (GPER1) can be activated by estradiol and mediate non-genomic signaling. Many studies have described the role of estrogen receptors in human cancers. Digestive system tumors account for a large proportion of all the tumors, and the mortality is very high in many digestive system tumors, such as esophageal cancer, gastric cancer, hepatocellular carcinoma, colorectal cancer, cholangiocarcinoma and pancreatic carcinoma. This review summarizes the role of estrogen receptors in digestive system tumors, aiming at finding new routes for the rational design of targeted anticancer therapies for digestive system tumors.
Collapse
|
14
|
Caiazza F, Ryan EJ, Doherty G, Winter DC, Sheahan K. Estrogen receptors and their implications in colorectal carcinogenesis. Front Oncol 2015; 5:19. [PMID: 25699240 PMCID: PMC4313613 DOI: 10.3389/fonc.2015.00019] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/16/2015] [Indexed: 12/31/2022] Open
Abstract
Upon binding their cognate receptors, ERα (ESR1) and ERβ (ESR2), estrogens activate intracellular signaling cascades that have important consequences for cellular behavior. Historically linked to carcinogenesis in reproductive organs, estrogens have also been implicated in the pathogenesis of different cancer types of non-reproductive tissues including the colon. ERβ is the predominant estrogen receptor expressed in both normal and malignant colonic epithelium. However, during colon cancer progression, ERβ expression is lost, suggesting that estrogen signaling may play a role in disease progression. Estrogens may in fact exert an anti-tumor effect through selective activation of pro-apoptotic signaling mediated by ERβ, inhibition of inflammatory signals and modulation of the tumor microenvironment. In this review, we analyze the estrogen pathway as a possible therapeutic avenue in colorectal cancer, we report the most recent experimental evidence to explain the cellular and molecular mechanisms of estrogen-mediated protection against colorectal tumorigenesis, and we discuss future challenges and potential avenues for targeted therapy.
Collapse
Affiliation(s)
- Francesco Caiazza
- Centre for Colorectal Disease, Saint Vincent's University Hospital , Dublin , Ireland ; School of Medicine and Medical Science, University College , Dublin , Ireland
| | - Elizabeth J Ryan
- Centre for Colorectal Disease, Saint Vincent's University Hospital , Dublin , Ireland ; School of Medicine and Medical Science, University College , Dublin , Ireland
| | - Glen Doherty
- Centre for Colorectal Disease, Saint Vincent's University Hospital , Dublin , Ireland ; School of Medicine and Medical Science, University College , Dublin , Ireland
| | - Desmond C Winter
- Centre for Colorectal Disease, Saint Vincent's University Hospital , Dublin , Ireland ; Department of Surgery, St. Vincent's University Hospital, Elm Park , Dublin , Ireland
| | - Kieran Sheahan
- Centre for Colorectal Disease, Saint Vincent's University Hospital , Dublin , Ireland ; School of Medicine and Medical Science, University College , Dublin , Ireland ; Department of Pathology, Saint Vincent's University Hospital , Dublin , Ireland
| |
Collapse
|
15
|
HEGER ZBYNEK, GUMULEC JAROMIR, CERNEI NATALIA, TMEJOVA KATERINA, KOPEL PAVEL, BALVAN JAN, MASARIK MICHAL, ZITKA ONDREJ, BEKLOVA MIROSLAVA, ADAM VOJTECH, KIZEK RENE. 17β-estradiol-containing liposomes as a novel delivery system for the antisense therapy of ER-positive breast cancer: An in vitro study on the MCF-7 cell line. Oncol Rep 2014; 33:921-9. [DOI: 10.3892/or.2014.3627] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/11/2014] [Indexed: 11/06/2022] Open
|
16
|
Vandamme TF. Use of rodents as models of human diseases. J Pharm Bioallied Sci 2014; 6:2-9. [PMID: 24459397 PMCID: PMC3895289 DOI: 10.4103/0975-7406.124301] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 11/20/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022] Open
Abstract
Advances in molecular biology have significantly increased the understanding of the biology of different diseases. However, these discoveries have not yet been fully translated into improved treatments for patients with diseases such as cancers. One of the factors limiting the translation of knowledge from preclinical studies to the clinic has been the limitations of in vivo diseases models. In this brief review, we will discuss the advantages and disadvantages of rodent models that have been developed to simulate human pathologies, focusing in models that employ xenografts and genetic modification. Within the framework of genetically engineered mouse (GEM) models, we will review some of the current genetic strategies for modeling diseases in the mouse and the preclinical studies that have already been undertaken. We will also discuss how recent improvements in imaging technologies may increase the information derived from using these GEMs during early assessments of potential therapeutic pathways. Furthermore, it is interesting to note that one of the values of using a mouse model is the very rapid turnover rate of the animal, going through the process of birth to death in a very short timeframe relative to that of larger mammalian species.
Collapse
Affiliation(s)
- Thierry F Vandamme
- University of Strasbourg, Faculty of Pharmacy, UMR 7199 CNRS, Laboratory of Concept and Application of Bioactive Molecules, Biogalenic Team, 74 Route du Rhin, 67400 Illkirch Graffenstaden, France
| |
Collapse
|
17
|
Tu Z, Ma Y, Akers W, Achilefu S, Gu Y. Therapeutic effect of the treatment for colorectal cancer with adenoviral vectors mediated estrogen receptor β gene therapy combined with thermotherapy. J Cancer Res Clin Oncol 2014; 140:623-32. [PMID: 24531912 DOI: 10.1007/s00432-014-1611-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 02/04/2014] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Our preliminary study on repressing colorectal tumors by recombinant adenoviruses (Ads) delivering the human ERβ gene (Ad-ERβ) has achieved positive result. METHODS In this study, hydrophobic fluorescent dyes ICG-Der-01 was entrapped into the N-succinyl-N'-octyl chitosan (SOC) micelles to form the near infrared absorbing dyes SOC-ICG-Der-01 and SOC-ICG-Der-01 mediated near infrared laser (SOC-ICG-Der-01/NIR) thermotherapy was combined with Ad-ERβ gene therapy to regress colon cancer in vivo. RESULTS Firstly, the antitumor efficacies of SOC-ICG-Der-01/NIR thermotherapy were investigated on S180 ascites tumor-bearing mice. Results indicated that, the average tumor volume of SOC-ICG-Der-01/NIR group was the smallest among the three treatment groups. Then, thermotherapy with SOC-ICG-Der-01/NIR combined with Ad-ERβ gene therapy to treat HCT-116 colon cancer xenograft model was investigated. Further results demonstrated that, SOC-ICG-Der-01/NIR thermotherapy showed the significantly inhibitory efficiency compared with control group and Ad-ERβ enhanced the therapeutic effect of SOC-ICG-Der-01/NIR. CONCLUSION These findings demonstrated that combined administration of Ad-ERβ with SOC-ICG-Der-01/NIR thermotherapy represents a promising colon cancer therapeutic strategy.
Collapse
Affiliation(s)
- Zhenzhen Tu
- Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023,, China
| | | | | | | | | |
Collapse
|
18
|
van Halteren H, Mulder D, Ruijter E. Oestrogen receptor-beta as a potential target for treatment in advanced colorectal cancer: a pilot study. Histopathology 2013; 64:787-790. [PMID: 24720253 DOI: 10.1111/his.12340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 11/30/2013] [Indexed: 11/27/2022]
Abstract
AIMS Oestrogen receptor-beta (ER-β) is expressed in colorectal cancer. Theoretically, ER-β stimulation could slow down tumour proliferation, and this is supported by preclinical research data. While preparing a Phase II trial for advanced colorectal cancer patients we performed a pilot study with three questions: (i) in what percentage of patients do metastases display strong ER-β1 expression; (ii) is there any concordance in expression between primary tumours and metastases; and (iii) is the immunohistochemical (IHC) scoring procedure reproducible? METHODS AND RESULTS Thirty patients were selected, 15 with locoregional lymph node metastases and 15 with either synchronous or metachronous hepatic metastases. All primary tumours and metastases were analysed for immunohistochemical ER-β1 expression according to a predefined scoring system. The scoring was performed independently by two pathologists in order to calculate the weighted kappa value. Strong ER-β1 expression was found in four of 15 hepatic metastases and four of 15 lymph node metastases. In 15 of 30 patients, the level of ER-β1 expression in the metastasis was concordant with that observed in the primary tumour. Weighted kappa values of IHC ER-β1 expression were satisfactory. CONCLUSIONS In twenty-five per cent of patients there was strong ER-β1 expression in metastases, biopsy of which will be considered mandatory for trial inclusion.
Collapse
Affiliation(s)
- Henk van Halteren
- Department of Internal Medicine, Admiraal de Ruijter Hospital, Goes, the Netherlands
| | | | | |
Collapse
|
19
|
Spiro AS, Khadem S, Jeschke A, Marshall RP, Pogoda P, Ignatius A, Amling M, Beil FT. The SERM raloxifene improves diaphyseal fracture healing in mice. J Bone Miner Metab 2013; 31:629-36. [PMID: 23546819 DOI: 10.1007/s00774-013-0461-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 03/14/2013] [Indexed: 11/26/2022]
Abstract
Although several studies reported that raloxifene treatment improves postmenopausal osteoporotic bone structure and reduces fracture risk, only a few animal and no human studies have examined its effects on the fracture healing process. Thus the aim of the present study was to determine, whether systemic application of the selective estrogen receptor modulator raloxifene promotes fracture healing compared to untreated control-, estrogen-deficient-, as well as estrogen-treated mice using a standardized femoral osteotomy model (n = 60 mice). Ten days after surgery, contact radiography and undecalcified histomorphometric analysis revealed that raloxifene administration significantly improved the early stage of fracture healing compared to all other groups. At day 20, raloxifene and estrogen treatment led to a significant increase in callus mineralization and trabecular thickness compared to control mice. μCT analyses revealed no evidence of complete bony bridging of the fracture site in any control-, nor estrogen-deficient mouse after 20 days, while all femoral fractures in the raloxifene and estrogen group already healed adequately at this time. These data indicate that raloxifene treatment significantly improves all phases of fracture healing at least in mice. Therefore, raloxifene could be a possible pharmaceutical to enhance fracture healing in women, without the known side effects of estrogen.
Collapse
Affiliation(s)
- Alexander S Spiro
- Department of Trauma, Hand, and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Yakimchuk K, Jondal M, Okret S. Estrogen receptor α and β in the normal immune system and in lymphoid malignancies. Mol Cell Endocrinol 2013; 375:121-9. [PMID: 23707618 DOI: 10.1016/j.mce.2013.05.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 05/14/2013] [Accepted: 05/18/2013] [Indexed: 02/07/2023]
Abstract
Estrogens regulate various normal and pathophysiological processes including cancers. Cellular signaling by estrogens is mediated by estrogen receptor α (ERα) and β (ERβ), respectively. Binding of agonists to the ERs affects gene transcription. The main endogenous estrogen, 17β-estradiol (E2), binds to both ERα and ERβ with similar affinity. However, the ligand-binding pocket of ERα and ERβ are slightly different which has allowed the development of selective ER ligands. Importantly, while estrogens via ERα stimulate proliferation, signaling via ERβ inhibits proliferation and promotes apoptosis. In both normal and cancer cells the ERs are co-expressed with ER splice variants which may modify the transcriptional activity of the wild-type receptors. Estrogens have prominent effects on immune functions and both ERα and ERβ are expressed in immune cells and lymphoid malignancies. With regard to lymphoid malignancies, most show estrogen influence as several epidemiological studies of lymphoid cancers demonstrate gender differences in incidence and prognosis with males being more affected. In line with these findings, recent results generated by us have shown that ERβ selective agonists inhibit growth and induce apoptosis in human and murine lymphomas in vivo in xenograft experiments. This suggests that ERβ selective agonists in the future may be useful in the treatment of lymphomas.
Collapse
Affiliation(s)
- Konstantin Yakimchuk
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, SE-141 83 Huddinge, Sweden
| | | | | |
Collapse
|
21
|
Kapoor S. Beyond breast carcinomas: the attenuating effect on tumor growth in other systemic malignancies by raloxifene. Eur J Clin Nutr 2013; 67:686. [PMID: 23462944 DOI: 10.1038/ejcn.2013.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Edvardsson K, Nguyen-Vu T, Kalasekar SM, Pontén F, Gustafsson JÅ, Williams C. Estrogen receptor β expression induces changes in the microRNA pool in human colon cancer cells. Carcinogenesis 2013; 34:1431-41. [PMID: 23436804 DOI: 10.1093/carcin/bgt067] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is epidemiological, animal and in vitro evidence that estrogen receptor β (ERβ) can mediate protective effects against colon cancer, but the mechanism is not completely understood. Previous research has indicated critical pathways whereby ERβ acts in an antitumorigenic fashion. In this study, we investigate ERβ's impact on the microRNA (miRNA) pool in colon cancer cells using large-scale genomic approaches, bioinformatics and focused functional studies. We detect and confirm 27 miRNAs to be significantly changed following ERβ expression in SW480 colon cancer cells. Among these, the oncogenic miR-17-92 cluster and miR-200a/b are strongly downregulated. Using target prediction and anticorrelation to gene expression data followed by focused mechanistic studies, we demonstrate that repression of miR-17 is a secondary event following ERβ's downregulatory effect on MYC. We show that re-introduction of miR-17 can reverse the antiproliferative effects of ERβ. The repression of miR-17 also influences cell death upon DNA damage and mediates regulation of NCOA3 (SRC-3) and CLU in colon cancer cells. We further determine that the downregulation of miR-200a/b mediates increased ZEB1 while decreasing E-cadherin levels in ERβ-expressing colon cancer cells. Changes in these genes correspond to significant alterations in morphology and migration. Our work contributes novel data of ERβ and miRNA in the colon. Elucidating the mechanism of ERβ and biomarkers of its activity has significant potential to impact colon cancer prevention and treatment.
Collapse
Affiliation(s)
- Karin Edvardsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA
| | | | | | | | | | | |
Collapse
|
23
|
Estrogen in obesity-associated colon cancer: friend or foe? Protecting postmenopausal women but promoting late-stage colon cancer. Cancer Causes Control 2012; 23:1767-73. [PMID: 23011535 DOI: 10.1007/s10552-012-0066-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/11/2012] [Indexed: 12/12/2022]
Abstract
Obesity is associated with the increased incidence of colon cancer. Many cancer risk factors have been identified including increased blood levels of insulin, leptin, interleukin-6, interleukin-17, tumor necrosis factor-alpha, and decreased blood levels of adiponectin. However, the role of blood levels of estrogen in obesity-associated colon cancer is controversial. Evidence showed that obesity affected men more strongly than women in the carcinogenesis of colon cancer, indicating protective effect of estrogen which is increased in obesity. However, an epidemiological study has also shown that endogenous estradiol level is an independent risk factor for colon cancer, positively associated with colon cancer after normalizing insulin, IGF-1. The controversial opinions may be caused by different effects of ER-alpha and ER-beta. ER-alpha can increase colon cancer cell proliferation and increase cancer incidence. ER-beta has the opposite effect to ER-alpha, and it causes apoptosis of colon cancer cells. The normal colonocytes mainly express ER-beta. Therefore, increased estrogen in obesity may have protective effect via ER-beta in obesity-associated colon cancer. However, with the development of colon cancer, ER-alpha is increased and ER-beta is decreased. In the late stage of colon cancer, estrogen may promote cancer development via ER-alpha. The different effects and expression of ER-alpha and ER-beta may explain the different results observed in several epidemiological studies as well as several animal experiments. Therefore, manipulation of estrogen-caused signal pathways to inhibit ER-alpha and stimulate ER-beta may have preventive and therapeutic effect for obesity-associated colon cancer.
Collapse
|