1
|
Cheng JN, Frye JB, Whitman SA, Ehsani S, Ali S, Funk JL. Interrogating Estrogen Signaling Pathways in Human ER-Positive Breast Cancer Cells Forming Bone Metastases in Mice. Endocrinology 2024; 165:bqae038. [PMID: 38715255 PMCID: PMC11076418 DOI: 10.1210/endocr/bqae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Indexed: 05/12/2024]
Abstract
Breast cancer bone metastases (BMET) are incurable, primarily osteolytic, and occur most commonly in estrogen receptor-α positive (ER+) breast cancer. ER+ human breast cancer BMET modeling in mice has demonstrated an estrogen (E2)-dependent increase in tumor-associated osteolysis and bone-resorbing osteoclasts, independent of estrogenic effects on tumor proliferation or bone turnover, suggesting a possible mechanistic link between tumoral ERα-driven osteolysis and ER+ bone progression. To explore this question, inducible secretion of the osteolytic factor, parathyroid hormone-related protein (PTHrP), was utilized as an in vitro screening bioassay to query the osteolytic potential of estrogen receptor- and signaling pathway-specific ligands in BMET-forming ER+ human breast cancer cells expressing ERα, ERß, and G protein-coupled ER. After identifying genomic ERα signaling, also responsibility for estrogen's proliferative effects, as necessary and sufficient for osteolytic PTHrP secretion, in vivo effects of a genomic-only ER agonist, estetrol (E4), on osteolytic ER+ BMET progression were examined. Surprisingly, while pharmacologic effects of E4 on estrogen-dependent tissues, including bone, were evident, E4 did not support osteolytic BMET progression (vs robust E2 effects), suggesting an important role for nongenomic ER signaling in ER+ metastatic progression at this site. Because bone effects of E4 did not completely recapitulate those of E2, the relative importance of nongenomic ER signaling in tumor vs bone cannot be ascertained here. Nonetheless, these intriguing findings suggest that targeted manipulation of estrogen signaling to mitigate ER+ metastatic progression in bone may require a nuanced approach, considering genomic and nongenomic effects of ER signaling on both sides of the tumor/bone interface.
Collapse
Affiliation(s)
- Julia N Cheng
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA
| | - Jennifer B Frye
- Department of Medicine, University of Arizona, Tucson, AZ 86724, USA
| | - Susan A Whitman
- Department of Medicine, University of Arizona, Tucson, AZ 86724, USA
| | - Sima Ehsani
- Department of Medicine, University of Arizona, Tucson, AZ 86724, USA
| | - Simak Ali
- Department of Surgery & Cancer, Imperial College London, London W12 0NN, UK
| | - Janet L Funk
- Department of Medicine, University of Arizona, Tucson, AZ 86724, USA
| |
Collapse
|
2
|
Zhang D, Chen H, Wang J, Ji J, Imam M, Zhang Z, Yan S. Current progress and prospects for G protein-coupled estrogen receptor in triple-negative breast cancer. Front Cell Dev Biol 2024; 12:1338448. [PMID: 38476263 PMCID: PMC10928007 DOI: 10.3389/fcell.2024.1338448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a biologically and clinically heterogeneous disease. The G protein-coupled estrogen receptor (GPER) plays a crucial role in mediating the effect of estrogen and estrogen-like compounds in TNBC cells. Compared with other subtypes, GPER has a higher expression in TNBC. The GPER mechanisms have been thoroughly characterized and analyzed in estrogen receptor α (ERα) positive breast cancer, but not in TNBC. Our previous work revealed that a higher expression of GPER mRNA indicates a better prognosis for ERα-positive breast cancer; however, its effects in TNBC differ. Whether GPER could serve as a predictive prognostic marker or therapeutic target for TNBC remains unclear. In this review, we provide a detailed introduction to the subcellular localization of GPER, the different effects of various ligands, and the interactions between GPER and closely associated factors in TNBC. We focused on the internal molecular mechanisms specific to TNBC and thoroughly explored the role of GPER in promoting tumor development. We also discussed the interaction of GPER with specific cytokines and chemokines, and the relationship between GPER and immune evasion. Additionally, we discussed the feasibility of using GPER as a therapeutic target in the context of existing studies. This comprehensive review highlights the effects of GPER on TNBC, providing a framework and directions for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Khan AS, Campbell KJ, Cameron ER, Blyth K. The RUNX/CBFβ Complex in Breast Cancer: A Conundrum of Context. Cells 2023; 12:641. [PMID: 36831308 PMCID: PMC9953914 DOI: 10.3390/cells12040641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023] Open
Abstract
Dissecting and identifying the major actors and pathways in the genesis, progression and aggressive advancement of breast cancer is challenging, in part because neoplasms arising in this tissue represent distinct diseases and in part because the tumors themselves evolve. This review attempts to illustrate the complexity of this mutational landscape as it pertains to the RUNX genes and their transcription co-factor CBFβ. Large-scale genomic studies that characterize genetic alterations across a disease subtype are a useful starting point and as such have identified recurring alterations in CBFB and in the RUNX genes (particularly RUNX1). Intriguingly, the functional output of these mutations is often context dependent with regards to the estrogen receptor (ER) status of the breast cancer. Therefore, such studies need to be integrated with an in-depth understanding of both the normal and corrupted function in mammary cells to begin to tease out how loss or gain of function can alter the cell phenotype and contribute to disease progression. We review how alterations to RUNX/CBFβ function contextually ascribe to breast cancer subtypes and discuss how the in vitro analyses and mouse model systems have contributed to our current understanding of these proteins in the pathogenesis of this complex set of diseases.
Collapse
Affiliation(s)
- Adiba S. Khan
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; (A.S.K.); (K.J.C.)
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Kirsteen J. Campbell
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; (A.S.K.); (K.J.C.)
| | - Ewan R. Cameron
- School of Biodiversity One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK;
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; (A.S.K.); (K.J.C.)
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
4
|
Khan MZI, Uzair M, Nazli A, Chen JZ. An overview on Estrogen receptors signaling and its ligands in breast cancer. Eur J Med Chem 2022; 241:114658. [PMID: 35964426 DOI: 10.1016/j.ejmech.2022.114658] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 02/07/2023]
Abstract
Estrogen governs the regulations of various pathological and physiological actions throughout the body in both males and females. Generally, 17β-estradiol an endogenous estrogen is responsible for different health problems in pre and postmenopausal women. The major activities of endogenous estrogen are executed by nuclear estrogen receptors (ERs) ERα and ERβ while non-genomic cytoplasmic pathways also govern cell growth and apoptosis. Estrogen accomplished a fundamental role in the formation and progression of breast cancer. In this review, we have hyphenated different studies regarding ERs and a thorough and detailed study of estrogen receptors is presented. This review highlights different aspects of estrogens ranging from receptor types, their isoforms, structures, signaling pathways of ERα, ERβ and GPER along with their crystal structures, pathological roles of ER, ER ligands, and therapeutic strategies to overcome the resistance.
Collapse
Affiliation(s)
| | - Muhammad Uzair
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Adila Nazli
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Jian-Zhong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
5
|
Togo S, Imanishi H, Hayashi M, Koyama M, Kira Y, Sugawara K, Tsuruta D. Exploring the impact of ovariectomy on hair growth: can ovariectomized mouse serve as a model for investigating female pattern hair loss in humans? Med Mol Morphol 2022; 55:210-226. [PMID: 35486188 DOI: 10.1007/s00795-022-00320-1] [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: 01/06/2022] [Accepted: 04/06/2022] [Indexed: 11/29/2022]
Abstract
Female pattern hair loss (FPHL), a type of hair disease common in pre- and postmenopausal women, is characterized by thinning of hair to O-type, mainly at the crown. Although a mouse model of this disease has recently been established, its details are still unknown, and thus, warrants further analysis. In this study, 3 week-old and 7- to 8 week-old C57BL/6 female mice were divided into two groups: one group underwent ovariectomy (OVX), while the other underwent sham surgery. In the 3 week-old mice, the dorsal skin was collected at seven weeks of age, while in the 7- to 8 week-old mice, it was collected at 12 and 24 weeks of age. In the former group, both the pore size of the hair follicles (HFs) and diameter of the hair shaft of telogen HFs decreased upon OVX; while in the latter group, these factors increased significantly. Notably, the thickness of the dermis and subcutis increased significantly in the OVX group. It needs to be further elucidated whether OVX mouse could serve as an ideal mouse model for FPHL, but our results upon evaluation of skin thickness indicate that it could be used to establish a novel treatment for non-hair-related diseases, such as post-menopause-related skin condition.
Collapse
Affiliation(s)
- Sayaka Togo
- Department of Dermatology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Hisayoshi Imanishi
- Department of Dermatology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Masami Hayashi
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masayasu Koyama
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yukimi Kira
- Department of Research Support Platform, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Sugawara
- Department of Dermatology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
6
|
Marcos X, Méndez-Luna D, Fragoso-Vázquez M, Rosales-Hernández M, Correa-Basurto J. Anti-breast cancer activity of novel compounds loaded in polymeric mixed micelles: Characterization and in vitro studies. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
7
|
Bertoni APS, Manfroi PDA, Tomedi J, Assis-Brasil BM, de Souza Meyer EL, Furlanetto TW. The gene expression of GPER1 is low in fresh samples of papillary thyroid carcinoma (PTC), and in silico analysis. Mol Cell Endocrinol 2021; 535:111397. [PMID: 34273443 DOI: 10.1016/j.mce.2021.111397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/08/2021] [Accepted: 07/13/2021] [Indexed: 11/26/2022]
Abstract
Papillary thyroid cancer (PTC), whose incidence has been increasing in the last years, occurs more frequently in women. Experimental studies suggested that estrogen could be an important risk factor for the higher female incidence. In fact, it has been demonstrated that 17β-estradiol (E2) could increase proliferation and dedifferentiation in thyroid follicular cells. Genomic estrogen responses are typically mediated through classical estrogen receptors, the α and β isoforms, which have been described in normal and abnormal human thyroid tissue. Nevertheless, effects mediated through G protein estrogen receptor 1 (GPR30/GPER/GPER1), described in some thyroid cancer cell lines, could be partially responsible for the regulation of growth in normal cells. In this study, GPER1 gene and protein expression are described in non-malignant and in papillary thyroid cancer (PTC), as well as its association with clinical features of patients with PTC. The GPER1 expression was lower in PTC as compared to paired non-malignant thyroid tissues in fresh samples of PTC and in silico analysis of GEO and TCGA databases. In PTC cases of TCGA database, low GPER1 mRNA expression was independently associated with metastatic lymph nodes, female gender, and BRAF mutation. Besides, GPER1 mRNA levels were positively correlated with mRNA levels of thyroid differentiation genes. These results support the hypothesis that GPER1 have a role in PTC tumorigenesis and might be a potential target for its therapy. Further studies are needed to determine the functionality of these receptors in normal and diseased thyroid.
Collapse
Affiliation(s)
- Ana Paula Santin Bertoni
- Departamento de Ciências Básicas da Saúde (DCBS) e Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Brazil
| | - Patrícia de Araujo Manfroi
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil
| | - Joelson Tomedi
- Serviço de Patologia, Hospital de Clínicas de Porto Alegre (HCPA), UFRGS, Brazil
| | | | | | - Tania Weber Furlanetto
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil.
| |
Collapse
|
8
|
Cheng JN, Frye JB, Whitman SA, Kunihiro AG, Pandey R, Funk JL. A Role for TGFβ Signaling in Preclinical Osteolytic Estrogen Receptor-Positive Breast Cancer Bone Metastases Progression. Int J Mol Sci 2021; 22:4463. [PMID: 33923316 PMCID: PMC8123146 DOI: 10.3390/ijms22094463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/25/2022] Open
Abstract
While tumoral Smad-mediated transforming growth factor β (TGFβ) signaling drives osteolytic estrogen receptor α-negative (ER-) breast cancer bone metastases (BMETs) in preclinical models, its role in ER+ BMETs, representing the majority of clinical BMETs, has not been documented. Experiments were undertaken to examine Smad-mediated TGFβ signaling in human ER+ cells and bone-tropic behavior following intracardiac inoculation of estrogen (E2)-supplemented female nude mice. While all ER+ tumor cells tested (ZR-75-1, T47D, and MCF-7-derived) expressed TGFβ receptors II and I, only cells with TGFβ-inducible Smad signaling (MCF-7) formed osteolytic BMETs in vivo. Regulated secretion of PTHrP, an osteolytic factor expressed in >90% of clinical BMETs, also tracked with osteolytic potential; TGFβ and E2 each induced PTHrP in bone-tropic or BMET-derived MCF-7 cells, with the combination yielding additive effects, while in cells not forming BMETs, PTHrP was not induced. In vivo treatment with 1D11, a pan-TGFβ neutralizing antibody, significantly decreased osteolytic ER+ BMETs in association with a decrease in bone-resorbing osteoclasts at the tumor-bone interface. Thus, TGFβ may also be a driver of ER+ BMET osteolysis. Moreover, additive pro-osteolytic effects of tumoral E2 and TGFβ signaling could at least partially explain the greater propensity for ER+ tumors to form BMETs, which are primarily osteolytic.
Collapse
Affiliation(s)
- Julia N. Cheng
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA;
| | - Jennifer B. Frye
- Department of Medicine, University of Arizona, Tucson, AZ 85724, USA; (J.B.F.); (S.A.W.)
| | - Susan A. Whitman
- Department of Medicine, University of Arizona, Tucson, AZ 85724, USA; (J.B.F.); (S.A.W.)
| | - Andrew G. Kunihiro
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85724, USA;
| | - Ritu Pandey
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA;
| | - Janet L. Funk
- Department of Medicine, University of Arizona, Tucson, AZ 85724, USA; (J.B.F.); (S.A.W.)
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85724, USA;
| |
Collapse
|
9
|
Niță AR, Knock GA, Heads RJ. Signalling mechanisms in the cardiovascular protective effects of estrogen: With a focus on rapid/membrane signalling. Curr Res Physiol 2021; 4:103-118. [PMID: 34746830 PMCID: PMC8562205 DOI: 10.1016/j.crphys.2021.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/22/2022] Open
Abstract
In modern society, cardiovascular disease remains the biggest single threat to life, being responsible for approximately one third of worldwide deaths. Male prevalence is significantly higher than that of women until after menopause, when the prevalence of CVD increases in females until it eventually exceeds that of men. Because of the coincidence of CVD prevalence increasing after menopause, the role of estrogen in the cardiovascular system has been intensively researched during the past two decades in vitro, in vivo and in observational studies. Most of these studies suggested that endogenous estrogen confers cardiovascular protective and anti-inflammatory effects. However, clinical studies of the cardioprotective effects of hormone replacement therapies (HRT) not only failed to produce proof of protective effects, but also revealed the potential harm estrogen could cause. The "critical window of hormone therapy" hypothesis affirms that the moment of its administration is essential for positive treatment outcomes, pre-menopause (3-5 years before menopause) and immediately post menopause being thought to be the most appropriate time for intervention. Since many of the cardioprotective effects of estrogen signaling are mediated by effects on the vasculature, this review aims to discuss the effects of estrogen on vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) with a focus on the role of estrogen receptors (ERα, ERβ and GPER) in triggering the more recently discovered rapid, or membrane delimited (non-genomic), signaling cascades that are vital for regulating vascular tone, preventing hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Ana-Roberta Niță
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
| | - Greg A. Knock
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Richard J. Heads
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- Cardiovascular Research Section, King’s BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s College London, UK
| |
Collapse
|
10
|
Hernández-Silva CD, Villegas-Pineda JC, Pereira-Suárez AL. Expression and Role of the G Protein-Coupled Estrogen Receptor (GPR30/GPER) in the Development and Immune Response in Female Reproductive Cancers. Front Endocrinol (Lausanne) 2020; 11:544. [PMID: 32973677 PMCID: PMC7468389 DOI: 10.3389/fendo.2020.00544] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/06/2020] [Indexed: 12/22/2022] Open
Abstract
Cancer is a major public health issue and represents the second leading cause of death in women worldwide, as female reproductive-related neoplasms are the main cause of incidence and mortality. Female reproductive cancers have a close relationship to estrogens, the principal female sex steroid hormones. Estrogens exert their actions by the nuclear estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). ERα, and ERβ act as transcription factors mediating genomic effects. Besides, the G protein-coupled estrogen receptor (GPER, formerly known as GPR30) was recently described as a seven-transmembrane receptor that mediates non-genomic estrogenic signaling, including calcium mobilization, cAMP synthesis, cleavage of matrix metalloproteinases, transactivation of epidermal growth factor receptor (EGFR), and the subsequent activation of PI3K and MAPK signaling pathways, which are the reasons why it is related to cellular processes, such as cell-cycle progression, cellular proliferation, differentiation, apoptosis, migration, and invasion. Since its discovery, selective agonists and antagonists have been found and developed. GPER has been implicated in a variety of hormone-responsiveness tumors, such as breast, endometrial, ovarian, cervical, prostate, and testicular cancer as well as lung, hepatic, thyroid, colorectal, and adrenocortical cancers. Nevertheless, GPER actions in cancer are still debatable due to the conflicting information that has been reported to date, since many reports indicate that activation of this receptor can modulate carcinogenesis. In contrast, many others show that its activation inhibits tumor activity. Besides, estrogens play an essential role in the regulation of the immune system, but little information exists about the role of GPER activation on its modulation within cancer context. This review focuses on the role that the stimulation of GPER plays in female reproductive neoplasms, specifically breast, endometrial, ovarian, and cervical cancers, in its tumor activity and immune response regulation.
Collapse
Affiliation(s)
- Christian David Hernández-Silva
- Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Julio César Villegas-Pineda
- Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Laboratorio de Inmunología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Ana Laura Pereira-Suárez
- Laboratorio de Inmunología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- *Correspondence: Ana Laura Pereira-Suárez
| |
Collapse
|
11
|
Manfroi PDA, Bertoni APS, Furlanetto TW. GPER1 in the thyroid: A systematic review. Life Sci 2020; 241:117112. [DOI: 10.1016/j.lfs.2019.117112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 11/26/2022]
|
12
|
Martínez-Muñoz A, Prestegui-Martel B, Méndez-Luna D, Fragoso-Vázquez MJ, García-Sánchez JR, Bello M, Martínez-Archundia M, Chávez-Blanco A, Dueñas-González A, Mendoza-Lujambio I, Trujillo-Ferrara J, Correa-Basurto J. Selection of a GPER1 Ligand via Ligand-based Virtual Screening Coupled to Molecular Dynamics Simulations and Its Anti-proliferative Effects on Breast Cancer Cells. Anticancer Agents Med Chem 2019; 18:1629-1638. [PMID: 29745344 DOI: 10.2174/1871520618666180510121431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/28/2018] [Accepted: 04/28/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recent reports have demonstrated the role of the G Protein-Coupled Estrogen Receptor 1 (GPER1) on the proliferation of breast cancer. The coupling of GPER1 to estrogen triggers cellular signaling pathways related to cell proliferation. OBJECTIVE Develop new therapeutic strategies against breast cancer. METHOD We performed in silico studies to explore the binding mechanism of a set of G15 /G1 analogue compounds. We included a carboxyl group instead of the acetyl group from G1 to form amides with several moieties to increase affinity on GPER1. The designed ligands were submitted to ligand-based and structure-based virtual screening to get insights into the binding mechanism of the best designed compound and phenol red on GPER1. RESULTS According to the in silico studies, the best molecule was named G1-PABA ((3aS,4R,9bR)-4-(6- bromobenzo[d][1,3]dioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinoline-8-carboxylic acid). It was synthesized and assayed in vitro in breast cancer (MCF-7 and MDA-MB-231) and normal (MCF-10A) cell lines. Experimental studies showed that the target compound was able to decrease cell proliferation, IC50 values of 15.93 µM, 52.92 µM and 32.45 µM in the MCF-7, MDA-MB-231 and MCF-10A cell lines, respectively, after 72 h of treatment. The compound showed better IC50 values without phenol red, suggesting that phenol red interfere with the G1-PABA action at GPER1, as observed through in silico studies, which is present in MCF-7 cells according to PCR studies and explains the cell proliferation effects. CONCLUSION Concentration-dependent inhibition of cell proliferation occurred with G1-PABA in the assayed cell lines and could be due to its action on GPER1.
Collapse
Affiliation(s)
- Alberto Martínez-Muñoz
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - Berenice Prestegui-Martel
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - David Méndez-Luna
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - Manuel J Fragoso-Vázquez
- Departamento de Quimica Organica, Escuela Nacional de Ciencias, Biologicas, Instituto Politecnico Nacional, Prolongacion de Carpio y Plan de Ayala, 11340, Ciudad de Mexico, Mexico
| | - José Rubén García-Sánchez
- Laboratorio de Oncologia Molecular y estres oxidativo, Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - Martiniano Bello
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - Marlet Martínez-Archundia
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - Alma Chávez-Blanco
- Instituto Nacional de Cancerologia, Ciudad de Mexico, Tlalpan 14080, Mexico
| | - Alfonso Dueñas-González
- Unidad de Investigacion Biomedica en Cancer, Instituto de Investigaciones Biomedicas UNAM/Instituto Nacional de Cancerologia, Ciudad de Mexico, Mexico
| | - Irene Mendoza-Lujambio
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - José Trujillo-Ferrara
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| | - José Correa-Basurto
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovación Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Díaz Miron, 11340 Ciudad de Mexico, Mexico
| |
Collapse
|
13
|
Xie M, Liang JL, Huang HD, Wang MJ, Zhang T, Yang XF. Low Doses of Nonylphenol Promote Growth of Colon Cancer Cells through Activation of ERK1/2 via G Protein‒Coupled Receptor 30. Cancer Res Treat 2019; 51:1620-1631. [PMID: 31096733 PMCID: PMC6790866 DOI: 10.4143/crt.2018.340] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 04/12/2019] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Nonylphenol (NP) is an endocrine disruptor found in products such as cleaners, plastics, and detergents. It exerts actions similar to endogenous 17β-estradiol (E2) and is reported to influence various cancers. However, its role in colon cancer remains elusive. Materials and Methods Colon cancer cell lines COLO 205 and SW480 were employed in our study. The cells were treated with NP or E2 followed by measurement of apoptosis and proliferation using flow cytometry and MTT assays, respectively. G protein-coupled estrogen receptor 30 (GPR30) expression was visualized using immunofluorescence and Western blot. To investigate the underlying mechanism, the expression levels of GPR30, p-protein kinase A (PKA), c-myc, cyclin D1, and ERK1/2 were analyzed using Western blot. Meanwhile, the GPR30 antagonist G15 was utilized to validate the role of GPR30 in colon cancer progression. Finally, the effect of a GPR30 inhibitor on tumor growth was determined in vivo using tumor xenograft mouse models. RESULTS NP facilitated the proliferation of colon cancer cells and induced apoptosis failure in vitro. Western blot revealed increased GPR30 expression levels in response to NP treatment. Cyclin D1, p-PKA, c-myc, and proliferating cell nuclear antigen, proteins that regulate the cell cycle, were all upregulated by NP, and NP-mediated ERK1/2 activation and subsequent cell proliferation were abrogated by the GPR30 inhibitor G15. Moreover, colon cancer mice that received G15 administration demonstrated impaired tumor growth in vivo. CONCLUSION Low dose NP promotes the growth of colon tumors through GPR30-mediated activation of ERK1/2 signaling.
Collapse
Affiliation(s)
- Ming Xie
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Jin-Long Liang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Han-Dong Huang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Mai-Jian Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Tao Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Xue-Feng Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| |
Collapse
|
14
|
Varennes J, Moon HR, Saha S, Mugler A, Han B. Physical constraints on accuracy and persistence during breast cancer cell chemotaxis. PLoS Comput Biol 2019; 15:e1006961. [PMID: 30970018 PMCID: PMC6476516 DOI: 10.1371/journal.pcbi.1006961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 04/22/2019] [Accepted: 03/18/2019] [Indexed: 01/19/2023] Open
Abstract
Directed cell motion in response to an external chemical gradient occurs in many biological phenomena such as wound healing, angiogenesis, and cancer metastasis. Chemotaxis is often characterized by the accuracy, persistence, and speed of cell motion, but whether any of these quantities is physically constrained by the others is poorly understood. Using a combination of theory, simulations, and 3D chemotaxis assays on single metastatic breast cancer cells, we investigate the links among these different aspects of chemotactic performance. In particular, we observe in both experiments and simulations that the chemotactic accuracy, but not the persistence or speed, increases with the gradient strength. We use a random walk model to explain this result and to propose that cells’ chemotactic accuracy and persistence are mutually constrained. Our results suggest that key aspects of chemotactic performance are inherently limited regardless of how favorable the environmental conditions are. One of the most ubiquitous and important cell behaviors is chemotaxis: the ability to move in the direction of a chemical gradient. Due to its importance, key aspects of chemotaxis have been quantified for a variety of cells, including the accuracy, persistence, and speed of cell motion. However, whether these aspects are mutually constrained is poorly understood. Can a cell be accurate but not persistent, or vice versa? Here we use theory, simulations, and experiments on cancer cells to uncover mutual constraints on the properties of chemotaxis. Our results suggest that accuracy and persistence are mutually constrained.
Collapse
Affiliation(s)
- Julien Varennes
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana, United States of America
| | - Hye-ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette Indiana, United States of America
| | - Soutick Saha
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana, United States of America
| | - Andrew Mugler
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail: (AM); (BH)
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette Indiana, United States of America
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail: (AM); (BH)
| |
Collapse
|
15
|
Smith LC, Moreno S, Robertson L, Robinson S, Gant K, Bryant AJ, Sabo-Attwood T. Transforming growth factor beta1 targets estrogen receptor signaling in bronchial epithelial cells. Respir Res 2018; 19:160. [PMID: 30165855 PMCID: PMC6117929 DOI: 10.1186/s12931-018-0861-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/13/2018] [Indexed: 02/08/2023] Open
Abstract
Background Sex differences in idiopathic pulmonary fibrosis (IPF) suggest a protective role for estrogen (E2); however, mechanistic studies in animal models have produced mixed results. Reports using cell lines have investigated molecular interactions between transforming growth factor beta1 (TGF-β1) and estrogen receptor (ESR) pathways in breast, prostate, and skin cells, but no such interactions have been described in human lung cells. To address this gap in the literature, we investigated a role for E2 in modulating TGF-β1-induced signaling mechanisms and identified novel pathways impacted by estrogen in bronchial epithelial cells. Methods We investigated a role for E2 in modulating TGF-β1-induced epithelial to mesenchymal transition (EMT) in bronchial epithelial cells (BEAS-2Bs) and characterized the effect of TGF-β1 on ESR mRNA and protein expression in BEAS-2Bs. We also quantified mRNA expression of ESRs in lung tissue from individuals with IPF and identified potential downstream targets of E2 signaling in BEAS-2Bs using RNA-Seq and gene set enrichment analysis. Results E2 negligibly modulated TGF-β1-induced EMT; however, we report the novel observation that TGF-β1 repressed ESR expression, most notably estrogen receptor alpha (ESR1). Results of the RNA-Seq analysis showed that TGF-β1 and E2 inversely modulated the expression of several genes involved in processes such as extracellular matrix (ECM) turnover, airway smooth muscle cell contraction, and calcium flux regulation. We also report that E2 specifically modulated the expression of genes involved in chromatin remodeling pathways and that this regulation was absent in the presence of TGF-β1. Conclusions Collectively, these results suggest that E2 influences unexplored pathways that may be relevant to pulmonary disease and highlights potential roles for E2 in the lung that may contribute to sex-specific differences. Electronic supplementary material The online version of this article (10.1186/s12931-018-0861-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- L Cody Smith
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA.,Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Santiago Moreno
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Lauren Robertson
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA.,Department of Environmental and Global Health, Center for Environmental and Human Toxicology, University of Florida, Box 110885, 2187 Mowry Rd, Gainesville, FL, 32611, USA
| | - Sarah Robinson
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA.,Department of Environmental and Global Health, Center for Environmental and Human Toxicology, University of Florida, Box 110885, 2187 Mowry Rd, Gainesville, FL, 32611, USA
| | - Kristal Gant
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA.,Department of Environmental and Global Health, Center for Environmental and Human Toxicology, University of Florida, Box 110885, 2187 Mowry Rd, Gainesville, FL, 32611, USA
| | - Andrew J Bryant
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Tara Sabo-Attwood
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA. .,Department of Environmental and Global Health, Center for Environmental and Human Toxicology, University of Florida, Box 110885, 2187 Mowry Rd, Gainesville, FL, 32611, USA.
| |
Collapse
|
16
|
Hadjimarkou MM, Vasudevan N. GPER1/GPR30 in the brain: Crosstalk with classical estrogen receptors and implications for behavior. J Steroid Biochem Mol Biol 2018; 176:57-64. [PMID: 28465157 DOI: 10.1016/j.jsbmb.2017.04.012] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/16/2017] [Accepted: 04/23/2017] [Indexed: 12/31/2022]
Abstract
The GPER1/GPR30 is a membrane estrogen receptor (mER) that binds 17β-estradiol (17β-E) with high affinity and is thought to play a role in cancer progression and cardiovascular health. Though widespread in the central nervous system, less is known about this receptor's function in the brain. GPER1 has been shown to activate kinase cascades and calcium flux within cells rapidly, thus fitting in with the idea of being a mER that mediates non-genomic signaling by estrogens. Signaling from GPER1 has been shown to improve spatial memory, possibly via release of neurotransmitters and generation of new spines on neurons in the hippocampus. In addition, GPER1 activation contributes to behaviors that denote anxiety and to social behaviors such as social memory and lordosis behavior in mice. In the male hippocampus, GPER1 activation has also been shown to phosphorylate the classical intracellular estrogen receptor (ER)α, suggesting that crosstalk with ERα is important in the display of these behaviors, many of which are absent in ERα-null mice. In this review, we present a number of categories of such crosstalk, using examples from literature. The function of GPER1 as an ERα collaborator or as a mER in different tissues is relevant to understanding both normal physiology and abnormal pathology, mediated by estrogen signaling.
Collapse
Affiliation(s)
- Maria M Hadjimarkou
- School of Humanities and Social Sciences, University of Nicosia, 1700 Nicosia, Cyprus.
| | - Nandini Vasudevan
- School of Biological Sciences, University of Reading, Reading, United Kingdom RG6 6AS, United Kingdom.
| |
Collapse
|
17
|
Zhou Q, Wu X, Dai X, Yuan R, Qi H. The different dosages of estrogen affect endometrial fibrosis and receptivity, but not SDF-1/CXCR4 axis in the treatment of intrauterine adhesions. Gynecol Endocrinol 2018; 34:49-55. [PMID: 28531361 DOI: 10.1080/09513590.2017.1328050] [Citation(s) in RCA: 15] [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] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE The study was to evaluate whether fibrotic markers, endometrial receptivity markers and SDF-1/CXCR4 had been changed in the treatment of intrauterine adhesions (IUAs) by different dosages of estrogen. STUDY DESIGN A total of 39 patients with IUAs were treated with EV 4 mg or 9 mg randomly post-surgery. TGF-β1/MMP-9, VEGF/αvβ3 and SDF-1/CXCR4 were detected in endometrial tissue before and after treatment by real-time PCR and Western blot. RESULTS TGF-β1 and MMP-9 expression significantly decreased after treatment for 3 months than before (p < .05), the falling range was larger with EV 4 mg than 9 mg in the mild-moderate degree IUAs (p < .05); Integrin avβ3 expression significantly increased after treatment for 3 months than before (p < .05), the variation range was larger with EV 4 mg than 9 mg (p < .05); CXCR4 expression had no significant change after treatment 3 months compared to that before treatment (p > .05). SDF-1 presented an upward tendency at early phase, and it came back to the level of pre-surgery. But there were no significant difference between treatment with 4 mg and 9 mg in the rate of menstrual restoration and pregnancy follow-up 3 months after the treatment. CONCLUSIONS Endometrium fibrosis may be inhibited and endometrium receptivity may be improved by estrogen with moderate dosage therapy. Compared to the large one, it seems to be advantageous.
Collapse
Affiliation(s)
- Qin Zhou
- a Department of Obstetrics and Gynecology , First Affiliated Hospital, Chongqing Medical University , Chongqing , China
| | - Xixi Wu
- b First Affiliated Hospital, Chongqing Medical University , Chongqing , China
| | - Xuelin Dai
- b First Affiliated Hospital, Chongqing Medical University , Chongqing , China
| | - Rui Yuan
- a Department of Obstetrics and Gynecology , First Affiliated Hospital, Chongqing Medical University , Chongqing , China
| | - Hongbo Qi
- b First Affiliated Hospital, Chongqing Medical University , Chongqing , China
| |
Collapse
|
18
|
Intrauterine exposure to 17β-oestradiol (E2) impairs postnatal development in both female and male prostate in gerbil. Reprod Toxicol 2017; 73:30-40. [DOI: 10.1016/j.reprotox.2017.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022]
|
19
|
Sousa C, Ribeiro M, Rufino AT, Leitão AJ, Mendes AF. Assessment of cell line competence for studies of pharmacological GPR30 modulation. J Recept Signal Transduct Res 2016; 37:181-188. [PMID: 27401115 DOI: 10.1080/10799893.2016.1203943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
CONTEXT/OBJECTIVE Cell lines used to study the role of the G protein-coupled receptor 30 (GPR30) or G protein-coupled estrogen receptor (GPER) as a mediator of estrogen responses have yielded conflicting results. This work identified a simple assay to predict cell line competence for pharmacological studies of GPR30. MATERIALS AND METHODS The phosphorylation or expression levels of ERK1/2, Akt, c-Fos and eNOS were evaluated to assess GPR30 activation in response to known agonists (17β-estradiol and G-1) in MCF-7 and T-47D breast cancer cell lines and in bovine aortic endothelial cells. GPR30 expression was analyzed by qRT-PCR and Western blot with two distinct antibodies directed at its carboxy and amino terminals. RESULTS None of the agonists, at any of the concentrations tested, activated any of those target proteins. Additional experiments excluded the disruption of the signaling pathway, interference of phenol red in the culture medium and constitutive proteasome degradation of GPR30 as possible causes for the lack of response of the three cell lines. Analysis of receptor expression showed the absence of clearly detectable GPR30 species of 44 and 50-55 kDa previously identified in cell lines that respond to 17β-estradiol and G-1. DISCUSSION AND CONCLUSION Cells that do not express the 44 and 50-55 kDa species do not respond to GPR30 agonists. Thus, the presence or absence of these GPR30 species is a simple and rapid manner to determine whether a given cell line is suitable for pharmacological or molecular studies of GPR30 modulation.
Collapse
Affiliation(s)
- Cátia Sousa
- a Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal
| | - Madalena Ribeiro
- a Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal
| | - Ana Teresa Rufino
- a Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal
| | - Alcino Jorge Leitão
- a Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal
| | - Alexandrina Ferreira Mendes
- a Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal
| |
Collapse
|
20
|
The complex nature of oestrogen signalling in breast cancer: enemy or ally? Biosci Rep 2016; 36:BSR20160017. [PMID: 27160081 PMCID: PMC5293589 DOI: 10.1042/bsr20160017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
The pleiotropic nature of oestradiol, the main oestrogen found in women, has been well described in the literature. Oestradiol is positioned to play a unique role since it can respond to environmental, genetic and non-genetic cues to affect genetic expression and cellular signalling. In breast cancer, oestradiol signalling has a dual effect, promoting or inhibiting cancer growth. The potential impact of oestradiol on tumorigenesis depends on the molecular and cellular characteristics of the breast cancer cell. In this review, we provide a broad survey discussing the cellular and molecular consequences of oestrogen signalling in breast cancer. First, we review the structure of the classical oestrogen receptors and resultant transcriptional (genomic) and non-transcriptional (non-genomic) signalling. We then discuss the nature of oestradiol signalling in breast cancer including the specific receptors that initiate these signalling cascades as well as potential outcomes, such as cancer growth, proliferation and angiogenesis. Finally, we examine cellular and molecular mechanisms underlying the dimorphic effect of oestrogen signalling in breast cancer.
Collapse
|
21
|
Peek GW, Tollefsbol TO. Down-regulation of hTERT and Cyclin D1 transcription via PI3K/Akt and TGF-β pathways in MCF-7 Cancer cells with PX-866 and Raloxifene. Exp Cell Res 2016; 344:95-102. [PMID: 27017931 DOI: 10.1016/j.yexcr.2016.03.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/16/2016] [Accepted: 03/22/2016] [Indexed: 11/26/2022]
Abstract
Human telomerase reverse transcriptase (hTERT) is the catalytic and limiting component of telomerase and also a transcription factor. It is critical to the integrity of the ends of linear chromosomes and to the regulation, extent and rate of cell cycle progression in multicellular eukaryotes. The level of hTERT expression is essential to a wide range of bodily functions and to avoidance of disease conditions, such as cancer, that are mediated in part by aberrant level and regulation of cell cycle proliferation. Value of a gene in regulation depends on its ability to both receive input from multiple sources and transmit signals to multiple effectors. The expression of hTERT and the progression of the cell cycle have been shown to be regulated by an extensive network of gene products and signaling pathways, including the PI3K/Akt and TGF-β pathways. The PI3K inhibitor PX-866 and the competitive estrogen receptor ligand raloxifene have been shown to modify progression of those pathways and, in combination, to decrease proliferation of estrogen receptor positive (ER+) MCF-7 breast cancer cells. We found that combinations of modulators of those pathways decreased not only hTERT transcription but also transcription of additional essential cell cycle regulators such as Cyclin D1. By evaluating known expression profile signatures for TGF-β pathway diversions, we confirmed additional genes such as heparin-binding epidermal growth factor-like growth factor (HB EGF) by which those pathways and their perturbations may also modify cell cycle progression.
Collapse
Affiliation(s)
- Gregory W Peek
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA; Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, USA; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA; Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
22
|
Busch S, Sims AH, Stål O, Fernö M, Landberg G. Loss of TGFβ Receptor Type 2 Expression Impairs Estrogen Response and Confers Tamoxifen Resistance. Cancer Res 2016; 75:1457-69. [PMID: 25833830 DOI: 10.1158/0008-5472.can-14-1583] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One third of the patients with estrogen receptor α (ERα)-positive breast cancer who are treated with the antiestrogen tamoxifen will either not respond to initial therapy or will develop drug resistance. Endocrine response involves crosstalk between ERα and TGFβ signaling, such that tamoxifen nonresponsiveness or resistance in breast cancer might involve aberrant TGFβ signaling. In this study, we analyzed TGFβ receptor type 2 (TGFBR2) expression and correlated it with ERα status and phosphorylation in a cohort of 564 patients who had been randomized to tamoxifen or no-adjuvant treatment for invasive breast carcinoma. We also evaluated an additional four independent genetic datasets in invasive breast cancer. In all the cohorts we analyzed, we documented an association of low TGFBR2 protein and mRNA expression with tamoxifen resistance. Functional investigations confirmed that cell cycle or apoptosis responses to estrogen or tamoxifen in ERα-positive breast cancer cells were impaired by TGFBR2 silencing, as was ERα phosphorylation, tamoxifen-induced transcriptional activation of TGFβ, and upregulation of the multidrug resistance protein ABCG2. Acquisition of low TGFBR2 expression as a contributing factor to endocrine resistance was validated prospectively in a tamoxifen-resistant cell line generated by long-term drug treatment. Collectively, our results established a central contribution of TGFβ signaling in endocrine resistance in breast cancer and offered evidence that TGFBR2 can serve as an independent biomarker to predict treatment outcomes in ERα-positive forms of this disease.
Collapse
Affiliation(s)
- Susann Busch
- Sahlgrenska Cancer Center, Gothenburg University, Gothenburg, Sweden
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh, Cancer Research UK Centre, United Kingdom
| | - Olle Stål
- Department of Clinical and Experimental Medicine, Institution of Surgery and Clinical Oncology, Linköpings Universitet, Linköping, Sweden
| | - Mårten Fernö
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Göran Landberg
- Sahlgrenska Cancer Center, Gothenburg University, Gothenburg, Sweden. Molecular Pathology, Breakthrough Breast Cancer Research Unit, University of Manchester, United Kingdom.
| |
Collapse
|
23
|
Kasap B, Öztürk Turhan N, Edgünlü T, Duran M, Akbaba E, Öner G. G-protein-coupled estrogen receptor-30 gene polymorphisms are associated with uterine leiomyoma risk. Bosn J Basic Med Sci 2016; 16:39-45. [PMID: 26773178 DOI: 10.17305/bjbms.2016.683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/12/2015] [Accepted: 10/12/2015] [Indexed: 12/20/2022] Open
Abstract
The G-protein-coupled estrogen receptor (GPR30, GPER-1) is a member of the G-protein-coupled receptor 1 family and is expressed significantly in uterine leiomyomas. To understand the relationship between GPR30 single nucleotide polymorphisms and the risk of leiomyoma, we measured the follicle-stimulating hormone (FSH) and estradiol (E2) levels of 78 perimenopausal healthy women and 111 perimenopausal women with leiomyomas. The participants' leiomyoma number and volume were recorded. DNA was extracted from whole blood with a GeneJET Genomic DNA Purification Kit. An amplification-refractory mutation system polymerase chain reaction approach was used for genotyping of the GPR30 gene (rs3808350, rs3808351, and rs11544331). The differences in genotype and allele frequencies between the leiomyoma and control groups were calculated using the chi-square (χ2) and Fischer's exact test. The median FSH level was higher in controls (63 vs. 10 IU/L, p=0.000), whereas the median E2 level was higher in the leiomyoma group (84 vs. 9.1 pg/mL, p=0.000). The G allele of rs3808351 and the GG genotype of both the rs3808350 and rs3808351 polymorphisms and the GGC haplotype increased the risk of developing leiomyoma. There was no significant difference in genotype frequencies or leiomyoma volume. However, the GG genotype of the GPR30 rs3808351 polymorphism and G allele of the GPR30 rs3808351 polymorphism were associated with the risk of having a single leiomyoma. Our results suggest that the presence of the GG genotype of the GPR30 rs3808351 polymorphism and the G allele of the GPR30 rs3808351 polymorphism affect the characteristics and development of leiomyomas in the Turkish population.
Collapse
Affiliation(s)
- Burcu Kasap
- Department of Obstetrics and Gynecology, School of Medicine, Mugla Sitki Kocman University, Mugla, Turkey.
| | | | | | | | | | | |
Collapse
|
24
|
Peek GW, Tollefsbol TO. Combinatorial PX-866 and Raloxifene Decrease Rb Phosphorylation, Cyclin E2 Transcription, and Proliferation of MCF-7 Breast Cancer Cells. J Cell Biochem 2015; 117:1688-96. [PMID: 26660119 DOI: 10.1002/jcb.25462] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/10/2015] [Indexed: 01/03/2023]
Abstract
As a potential means to reduce proliferation of breast cancer cells, a multiple-pathway approach with no effect on control cells was explored. The human interactome being constructed by the Center for Cancer Systems Biology will prove indispensable to understanding composite effects of multiple pathways, but its discovered protein-protein interactions require characterization. Accordingly, we explored the effects of regulators of one protein on downstream targets of the other protein. MCF-7 estrogen receptor-positive (ER+) breast cancer cells were treated with raloxifene to upregulate the TGF-β pathway and PX-866 to down-regulate the PI3K/Akt pathway. This resulted in highly significant downstream reduction of cell cycle proliferation in breast cancer cells with no significant proliferation reduction following similar treatment of noncancerous MCF10A breast epithelial cells. Reduced phosphorylation of p107 and substantial reduction of Rb phosphorylation were observed in response. The effects of reduced Rb and p107 phosphorylation were reflected in significant decline in E2F-1 transcriptional activity, which is dependent on pocket protein phosphorylation status. The reduced proliferation was related to decreased expression of cyclins, including E2F-1-regulated Cyclin E2, which was also in response to raloxifene and PX-866. All combinations of raloxifene and PX-866 produced significant or highly significant results for reduced MCF-7 cell proliferation, reduced Cyclin E2 transcription, and reduced Rb phosphorylation. These studies demonstrated that uncontrolled proliferation of ER+ breast cancer cells can be significantly reduced by combinational targeting of two relevant pathways. J. Cell. Biochem. 117: 1688-1696, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gregory W Peek
- Department of Biology, University of Alabama, Birmingham, Alabama
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama, Birmingham, Alabama.,Comprehensive Cancer Center, University of Alabama, Birmingham, Alabama.,Comprehensive Center for Healthy Aging, University of Alabama, Birmingham, Alabama.,Comprehensive Diabetes Center, University of Alabama, Birmingham, Alabama.,Nutrition Obesity Research Center, University of Alabama, Birmingham, Alabama
| |
Collapse
|
25
|
Down-regulation of osteopontin mediates a novel mechanism underlying the cytostatic activity of TGF-β. Cell Oncol (Dordr) 2015; 39:119-28. [PMID: 26584547 DOI: 10.1007/s13402-015-0257-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2015] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Loss of a cytostatic response to TGF-β has been implicated in multiple hyper-proliferative disorders, including cancer. Although several key genes involved in the cytostatic activity of TGF-β have in the past been identified, its exact mode of action is yet to be elucidated. A comprehensive understanding of the mechanisms underlying the cytostatic activity of TGF-β may open up new avenues for the development of therapeutic strategies. METHODS Quantitative real-time RT-PCR was used to assess osteopontin (OPN) gene expression in human hepatoma-derived Huh-7 and lung adenocarcinoma-derived A549 cells. Reporter assays using an OPN promoter-luciferase construct and its mutated counterparts were performed to assess its transcriptional activity. Binding of Smad4 to the OPN gene promoter was investigated using chromatin immunoprecipitation (CHIP). The putative role of Smad4 in OPN gene expression down-regulation was also assessed using a shRNA-mediated knockdown strategy. The anti-proliferative effect of TGF-β on different cancer-derived cell lines was determined using the cell proliferation reagent WST-1. RESULTS We found that the OPN expression levels dose-dependently decreased in TGF-β-treated Huh-7 and A549 cells. Our reporter assays indicated that this TGF-β-induced repression occurred at the transcriptional level, and could largely be abrogated by disruption of an element (TIE2) similar to the TGF-β inhibitory element found in other TGF-β-repressed genes. Our CHIP assay revealed that the Smad protein complex specifically binds to the OPN gene promoter, and that the TGF-β-mediated inhibition of OPN was lost upon shRNA-mediated knockdown of Smad4. Moreover, we found that the deregulation of OPN gene expression by TGF-β occurred concomitantly with loss of the TGF-β anti-proliferative response, whereas a neutralizing anti-OPN antibody partially restored this response. CONCLUSIONS Our results indicate that the OPN gene is a direct target of Smad-mediated TGF-β signaling, implying that OPN expression inhibition serves as a novel mechanism underlying the cytostatic activity of TGF-β.
Collapse
|
26
|
Popli P, Sirohi VK, Manohar M, Shukla V, Kaushal JB, Gupta K, Dwivedi A. Regulation of cyclooxygenase-2 expression in rat oviductal epithelial cells: Evidence for involvement of GPR30/Src kinase-mediated EGFR signaling. J Steroid Biochem Mol Biol 2015; 154:130-41. [PMID: 26241029 DOI: 10.1016/j.jsbmb.2015.07.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/13/2015] [Accepted: 07/29/2015] [Indexed: 11/26/2022]
Abstract
The oviduct plays a crucial role in female reproduction by regulating gamete transport, providing a specific microenvironment for fertilization and early embryonic development. Cyclooxygenase (COX)-derived prostaglandins play essential role in carrying out these oviduct-specific functions. Estrogen upregulates COX-2 expression in rat oviduct; however, the mechanisms responsible for regulation of COX-2 expression in rat oviductal epithelial cells (OECs) remain unclear. In the present study, we proposed that estrogen induces COX-2 expression via G-protein coupled receptor i.e., GPR30 in OECs. To investigate this hypothesis, we examined the effects of E2-BSA, ICI 182,780, GPR30 agonist and GPR30 antagonist on COX-2 expression and explored potential signaling pathway leading to COX-2 expression. Co-localization experiments revealed GPR30 to be primarily located in the peri-nuclear space, which was also the site of E2-BSA-fluorescein isothiocyanate (E2-BSA-FITC) binding. The E2-BSA induced-COX-2 and prostaglandin release were subjected to regulation by both EGFR and PI3K signaling as inhibitors of c-Src kinase (PP2), EGFR (EGFR inhibitor) and PI-3 kinase (LY294002) attenuated E2-BSA mediated effect. These results suggest that EGFR transactivation leading to activation of PI-3K/Akt pathway participates in COX-2 expression in rat OECs. Interestingly, E2-BSA induced COX-2 expression and subsequent prostaglandin release were abolished by NF-κB inhibitor. In addition, E2-BSA induced the nuclear translocation of p65-NF-κB and up-regulated the NF-κB promoter activity in rat OECs. Taken together, results demonstrated that E2-BSA induced the COX-2 expression and consequent PGE2 and PGF2α release in rat OECs. These effects are mediated through GPR30-derived EGFR transactivation and PI-3K/Akt cascade leading to NF-κB activation.
Collapse
Affiliation(s)
- Pooja Popli
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Vijay Kumar Sirohi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Murli Manohar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Vinay Shukla
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Jyoti Bala Kaushal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Kanchan Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Anila Dwivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India.
| |
Collapse
|
27
|
Kim YS, Choi KC, Hwang KA. Genistein suppressed epithelial-mesenchymal transition and migration efficacies of BG-1 ovarian cancer cells activated by estrogenic chemicals via estrogen receptor pathway and downregulation of TGF-β signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:993-999. [PMID: 26407941 DOI: 10.1016/j.phymed.2015.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/20/2015] [Accepted: 08/03/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT), which is activated by 17β-estradiol (E2) in estrogen-responsive cancers, is an important process in tumor migration or progression. As typical endocrine disrupting chemicals (EDCs), bisphenol A (BPA) and nonylphenol (NP) have a potential to promote EMT and migration of estrogen-responsive cancers. On the contrary, genistein (GEN) as a phytoestrogen is known to have chemopreventive effects in diverse cancers. METHODS In the present study, the effects of BPA and GEN on EMT and the migration of BG-1 ovarian cancer cells and the underlying mechanism were investigated. ICI 182,780, an estrogen receptor (ER) antagonist, was co-treated with E2 or BPA or NP to BG-1 cells to identify the relevance of ER signaling in EMT and migration. RESULTS As results, E2 and BPA upregulated the protein expression of vimentin, cathepsin D, and MMP-2, but downregulated the protein expression of E-cadherin via ER signaling pathway, suggesting that E2 and BPA promote EMT and cell migration related gene expressions. However, the increased protein expressions of vimentin, cathepsin D, and MMP-2 by E2, BPA, or NP were reduced by the co-treatment of GEN. In a scratch assay, the migration capability of BG-1 cells was enhanced by E2, BPA, and NP via ER signaling but reversed by the co-treatment of GEN. In the protein expression of SnoN and Smad3, E2, BPA, and NP upregulated SnoN, a negative regulator of TGF-β signaling, and downregulated pSmad3, a transcription factor in the downstream pathway of TGF-β signaling pathway, suggesting that E2, BPA, and NP simultaneously lead to the downregualtion of TGF-β signaling in the process of induction of EMT and migration of BG-1 cells via ER signaling. On the other hand, the co-treatment of GEN reversed the downregulation of TGF-β signaling by estrogenic chemicals. CONCLUSION Taken together, GEN suppressed EMT and migration capacities of BG-1 ovarian cancer cells enhanced by E2, BPA, and NP via ER signaling and the downregulation of TGF-β signal.
Collapse
Affiliation(s)
- Ye-Seul Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, Republic of Korea.
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, Republic of Korea.
| |
Collapse
|
28
|
Mata KM, Li W, Reslan OM, Siddiqui WT, Opsasnick LA, Khalil RA. Adaptive increases in expression and vasodilator activity of estrogen receptor subtypes in a blood vessel-specific pattern during pregnancy. Am J Physiol Heart Circ Physiol 2015; 309:H1679-96. [PMID: 26408543 DOI: 10.1152/ajpheart.00532.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/09/2015] [Indexed: 11/22/2022]
Abstract
Normal pregnancy is associated with adaptive hemodynamic, hormonal, and vascular changes, and estrogen (E2) may promote vasodilation during pregnancy; however, the specific E2 receptor (ER) subtype, post-ER signaling mechanism, and vascular bed involved are unclear. We tested whether pregnancy-associated vascular adaptations involve changes in the expression/distribution/activity of distinct ER subtypes in a blood vessel-specific manner. Blood pressure (BP) and plasma E2 were measured in virgin and pregnant (day 19) rats, and the thoracic aorta, carotid artery, mesenteric artery, and renal artery were isolated for measurements of ERα, ERβ, and G protein-coupled receptor 30 [G protein-coupled ER (GPER)] expression and tissue distribution in parallel with relaxation responses to E2 (all ERs) and the specific ER agonist 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)-tris-phenol (PPT; ERα), diarylpropionitrile (DPN; ERβ), and G1 (GPER). BP was slightly lower and plasma E2 was higher in pregnant versus virgin rats. Western blots revealed increased ERα and ERβ in the aorta and mesenteric artery and GPER in the aorta of pregnant versus virgin rats. Immunohistochemistry revealed that the increases in ERs were mainly in the intima and media. In phenylephrine-precontracted vessels, E2 and PPT caused relaxation that was greater in the aorta and mesenteric artery but similar in the carotid and renal artery of pregnant versus virgin rats. DPN- and G1-induced relaxation was greater in the mesenteric and renal artery than in the aorta and carotid artery, and aortic relaxation to G1 was greater in pregnant versus virgin rats. The nitric oxide synthase inhibitor N(ω)-nitro-l-arginine methyl ester with or without the cyclooxygenase inhibitor indomethacin with or without the EDHF blocker tetraethylammonium or endothelium removal reduced E2, PPT, and G1-induced relaxation in the aorta of pregnant rats, suggesting an endothelium-dependent mechanism, but did not affect E2-, PPT-, DPN-, or G1-induced relaxation in other vessels, suggesting endothelium-independent mechanisms. E2, PPT, DPN, and G1 caused relaxation of Ca(2+) entry-dependent KCl contraction, and the effect of PPT was greater in the mesenteric artery of pregnant versus virgin rats. Thus, during pregnancy, an increase in ERα expression in endothelial and vascular smooth muscle layers of the aorta and mesenteric artery is associated with increased ERα-mediated relaxation via endothelium-derived vasodilators and inhibition of Ca(2+) entry into vascular smooth muscle, supporting a role of aortic and mesenteric arterial ERα in pregnancy-associated vasodilation. GPER may contribute to aortic relaxation while enhanced ERβ expression could mediate other genomic vascular effects during pregnancy.
Collapse
Affiliation(s)
- Karina M Mata
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ossama M Reslan
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Waleed T Siddiqui
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lauren A Opsasnick
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Mazzuca MQ, Mata KM, Li W, Rangan SS, Khalil RA. Estrogen receptor subtypes mediate distinct microvascular dilation and reduction in [Ca2+]I in mesenteric microvessels of female rat. J Pharmacol Exp Ther 2014; 352:291-304. [PMID: 25472954 DOI: 10.1124/jpet.114.219865] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Estrogen interacts with estrogen receptors (ERs) to induce vasodilation, but the ER subtype and post-ER relaxation pathways are unclear. We tested if ER subtypes mediate distinct vasodilator and intracellular free Ca(2+) concentration ([Ca(2+)]i) responses via specific relaxation pathways in the endothelium and vascular smooth muscle (VSM). Pressurized mesenteric microvessels from female Sprague-Dawley rats were loaded with fura-2, and the changes in diameter and [Ca(2+)]i in response to 17β-estradiol (E2) (all ERs), PPT (4,4',4''-[4-propyl-(1H)-pyrazole-1,3,5-triyl]-tris-phenol) (ERα), diarylpropionitrile (DPN) (ERβ), and G1 [(±)-1-[(3aR*,4S*,9bS*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro:3H-cyclopenta(c)quinolin-8-yl]-ethanon] (GPR30) were measured. In microvessels preconstricted with phenylephrine, ER agonists caused relaxation and decrease in [Ca(2+)]i that were with E2 = PPT > DPN > G1, suggesting that E2-induced vasodilation involves ERα > ERβ > GPR30. Acetylcholine caused vasodilation and decreased [Ca(2+)]i, which were abolished by endothelium removal or treatment with the nitric oxide synthase blocker Nω-nitro-l-arginine methyl ester (L-NAME) and the K(+) channel blockers tetraethylammonium (nonspecific) or apamin (small conductance Ca(2+)-activated K(+) channel) plus TRAM-34 (1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole) (intermediate conductance Ca(2+)-activated K(+) channel), suggesting endothelium-derived hyperpolarizing factor-dependent activation of KCa channels. E2-, PPT-, DPN-, and G1-induced vasodilation and decreased [Ca(2+)]i were not blocked by L-NAME, TEA, apamin plus TRAM-34, iberiotoxin (large conductance Ca(2+)- and voltage-activated K(+) channel), 4-aminopyridine (voltage-dependent K(+) channel), glibenclamide (ATP-sensitive K(+) channel), or endothelium removal, suggesting an endothelium- and K(+) channel-independent mechanism. In endothelium-denuded vessels preconstricted with phenylephrine, high KCl, or the Ca(2+) channel activator Bay K 8644 (1,4-dihydro-2,6-dimethyl-5-nitro-4-[2-(trifluoromethyl)phenyl]-3-pyridinecarboxylic acid methyl ester), ER agonist-induced relaxation and decreased [Ca(2+)]i were with E2 = PPT > DPN > G1 and not inhibited by the guanylate cyclase inhibitor ODQ [1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one], and showed a similar relationship between decreased [Ca(2+)]i and vasorelaxation, supporting direct effects on Ca(2+) entry in VSM. Immunohistochemistry revealed ERα, ERβ, and GPR30 mainly in the vessel media and VSM. Thus, in mesenteric microvessels, ER subtypes mediate distinct vasodilation and decreased [Ca(2+)]i (ERα > ERβ > GPR30) through endothelium- and K(+) channel-independent inhibition of Ca(2+) entry mechanisms of VSM contraction.
Collapse
Affiliation(s)
- Marc Q Mazzuca
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Karina M Mata
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sridhar S Rangan
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
30
|
Li YC, Ding XS, Li HM, Zhang Y, Bao J. Role of G protein-coupled estrogen receptor 1 in modulating transforming growth factor-β stimulated mesangial cell extracellular matrix synthesis and migration. Mol Cell Endocrinol 2014; 391:50-9. [PMID: 24793639 DOI: 10.1016/j.mce.2014.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 04/18/2014] [Accepted: 04/23/2014] [Indexed: 12/25/2022]
Abstract
Estrogen has been demonstrated to exert beneficial effects on kidney; however, the role of G protein-coupled estrogen receptor 1 (GPER) is still uncertain. In the present study, we investigated the effect of 17β-estradiol and GPER agonist Fulvestrant on extracellular matrix production under transforming growth factor-β1 (TGF-β1) stimulation in human and rat mesangial cells. As a result, 17β-estradiol and Fulvestrant inhibit TGF-β1-induced type IV collagen and fibronectin expression in a dose-dependent manner, by suppressing acute Smad2/3 phosphorylation and Smad4 complex formation. Furthermore, estrogen and Fulvestrant also down-regulate Smad signaling by promoting ubiquitin/proteasome-dependent Smad2 degradation. These effects could be abrogated by receptor antagonist G-15 or GPER gene knockdown. GPER is also required for estrogen and Fulvestrant to regulate mesangial cell migration in response to TGF-β1. To conclude, GPER is crucial in modulating glomerular mesangial cell function including extracellular matrix production and migration.
Collapse
Affiliation(s)
- Yi-Chen Li
- Department of Pharmacy, Drum Tower Hospital Affiliated to Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu Province 210008, PR China; Department of Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu Province 210009, PR China.
| | - Xuan-Sheng Ding
- Department of Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu Province 210009, PR China.
| | - Hui-Mei Li
- Department of Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu Province 210009, PR China.
| | - Ying Zhang
- Department of Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu Province 210009, PR China.
| | - Jing Bao
- Department of Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu Province 210009, PR China.
| |
Collapse
|
31
|
Li YC, Ding XS, Li HM, Zhang C. Icariin attenuates high glucose-induced type IV collagen and fibronectin accumulation in glomerular mesangial cells by inhibiting transforming growth factor-β production and signalling through G protein-coupled oestrogen receptor 1. Clin Exp Pharmacol Physiol 2014; 40:635-43. [PMID: 23772748 DOI: 10.1111/1440-1681.12143] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/05/2013] [Accepted: 06/11/2013] [Indexed: 12/31/2022]
Abstract
Icariin has been shown to attenuate diabetic nephropathy in rats by decreasing transforming growth factor-β (TGF-β) and type IV collagen expression, but its mode of action in glomerular mesangial cells is uncertain. The present study aimed to investigate the effect of icariin on excess mesangial type IV collagen and fibronectin accumulation induced by high glucose, and to determine the mechanism underlying its protective effects. Under high-glucose conditions, icariin diminished type IV collagen and fibronectin accumulation, as well as TGF-β production in human and rat mesangial cells. Mesangial cells treated with icariin after TGF-β1 exposure expressed less type IV collagen and fibronectin than those without icariin treatment, suggesting inhibition by icariin of TGF-β1 downstream pathways. On TGF-β1 stimulation, icariin inhibited TGF-β canonical Smad signalling and extracellular signal-regulated kinase (ERK)1/2 signalling by decreasing Smad2/3 and ERK1/2 phosphorylation in a dose-dependent manner. U0126, which blocked the ERK1/2 pathway, exerted an additive effect on the icariin suppression of type IV collagen and fibronectin expression, enhancing the beneficial effects of icariin. The G protein-coupled oestrogen receptor 1 (GPER) antagonist, G-15, abolished the icariin-induced inhibition of type IV collagen, and fibronectin overproduction and TGF-β signalling. Treatment of cells with fulvestrant, a downregulator of the oestrogen receptor, enhanced the action of icariin. In conclusion, icariin decreased type IV collagen and fibronectin accumulation induced by high glucose in mesangial cells by inhibiting TGF-β production, as well as Smad and ERK signalling in a GPER-dependent manner.
Collapse
Affiliation(s)
- Yi-Chen Li
- Department of Clinical Pharmacy, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | | | | | | |
Collapse
|
32
|
Retinoids and breast cancer: from basic studies to the clinic and back again. Cancer Treat Rev 2014; 40:739-49. [PMID: 24480385 DOI: 10.1016/j.ctrv.2014.01.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/09/2014] [Accepted: 01/10/2014] [Indexed: 11/23/2022]
Abstract
All-trans retinoic acid (ATRA) is the most important active metabolite of vitamin A controlling segmentation in the developing organism and the homeostasis of various tissues in the adult. ATRA as well as natural and synthetic derivatives, collectively known as retinoids, are also promising agents in the treatment and chemoprevention of different types of neoplasia including breast cancer. The major aim of the present article is to review the basic knowledge acquired on the anti-tumor activity of classic retinoids, like ATRA, in mammary tumors, focusing on the underlying cellular and molecular mechanisms and the determinants of retinoid sensitivity/resistance. In the first part, an analysis of the large number of pre-clinical studies available is provided, stressing the point that this has resulted in a limited number of clinical trials. This is followed by an overview of the knowledge acquired on the role played by the retinoid nuclear receptors in the anti-tumor responses triggered by retinoids. The body of the article emphasizes the potential of ATRA and derivatives in modulating and in being influenced by some of the most relevant cellular pathways involved in the growth and progression of breast cancer. We review the studies centering on the cross-talk between retinoids and some of the growth-factor pathways which control the homeostasis of the mammary tumor cell. In addition, we consider the cross-talk with relevant intra-cellular second messenger pathways. The information provided lays the foundation for the development of rational and retinoid-based therapeutic strategies to be used for the management of breast cancer.
Collapse
|
33
|
Samartzis EP, Noske A, Meisel A, Varga Z, Fink D, Imesch P. The G protein-coupled estrogen receptor (GPER) is expressed in two different subcellular localizations reflecting distinct tumor properties in breast cancer. PLoS One 2014; 9:e83296. [PMID: 24421881 PMCID: PMC3885421 DOI: 10.1371/journal.pone.0083296] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/11/2013] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION The G protein-coupled estrogen receptor (GPER) is a novel estrogen receptor that mediates proliferative effects induced by estrogen but also by tamoxifen. The aim of our study was to analyze the frequency of GPER in a large collective of primary invasive breast carcinomas, with special emphasis on the subcellular expression and to evaluate the association with clinicopathological parameters and patient overall survival. METHODS The tissue microarrays from formalin-fixed, paraffin embedded samples of primary invasive breast carcinomas (n = 981) were analyzed for GPER expression using immunohistochemistry. Expression data were compared to the clinicopathological parameters and overall survival. GPER localization was also analyzed in two immortalized breast cancer cell lines T47D and MCF7 by confocal immunofluorescence microscopy. RESULTS A predominantly cytoplasmic GPER expression was found in 189 carcinomas (19.3%), whereas a predominantly nuclear expression was observed in 529 cases (53.9%). A simultaneous comparable positive expression of both patterns was found in 32 of 981 cases (3.2%), and negative staining was detected in 295 cases (30%). Confocal microscopy confirmed the occurrence of cytoplasmic and nuclear GPER expression in T47D and MCF7. Cytoplasmic GPER expression was significantly associated with non-ductal histologic subtypes, low tumor stage, better histologic differentiation, as well as Luminal A and B subtypes. In contrast, nuclear GPER expression was significantly associated with poorly differentiated carcinomas and the triple-negative subtype. In univariate analysis, cytoplasmic GPER expression was associated with better overall survival (p = 0.012). CONCLUSION Our data suggest that predominantly cytoplasmic and/or nuclear GPER expression are two distinct immunohistochemical patterns in breast carcinomas and may reflect different biological features, reason why these patterns should be clearly distinguished in histological evaluations. Prospective studies will be needed to assess whether the expression status of GPER in breast carcinomas should be routinely observed by clinicians, for instance, before implementing endocrine breast cancer treatment.
Collapse
Affiliation(s)
| | - Aurelia Noske
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Alexander Meisel
- Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Zsuzsanna Varga
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Daniel Fink
- Department of Gynecology, University Hospital Zurich, Zurich, Switzerland
| | - Patrick Imesch
- Department of Gynecology, University Hospital Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
34
|
Subtype-specific estrogen receptor-mediated vasodilator activity in the cephalic, thoracic, and abdominal vasculature of female rat. J Cardiovasc Pharmacol 2013; 62:26-40. [PMID: 23429596 DOI: 10.1097/fjc.0b013e31828bc88a] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Estrogen receptors (ERs) mediate genomic and nongenomic vasodilator effects, but estrogen therapy may not provide systemic vascular protection. To test whether this is because of regional differences in ER distribution or vasodilator activity, cephalic (carotid artery), thoracic (thoracic aorta and pulmonary artery), and abdominal arteries (abdominal aorta, mesenteric artery, and renal artery) from female Sprague-Dawley rats were prepared to measure contraction to phenylephrine and relaxation to acetylcholine (ACh) and the ER activators 17β-estradiol (E2) (all ERs), 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)-tris-phenol (PPT) (ERα), diarylpropionitrile (DPN) (ERβ), and (±)-1-[(3aR*,4S*,9bS*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone (G1) (GPR30). Phenylephrine caused contraction that was enhanced in endothelium-denuded aorta, supporting endothelial release of vasodilators. In cephalic and thoracic arteries, ACh relaxation was abolished by the nitric oxide (NO) synthase inhibitor Nω-nitro-L-arginine methyl ester (L-NAME), suggesting a role of NO. In mesenteric vessels, ACh-induced relaxation was partly inhibited by the L-NAME + cyclooxygenase inhibitor indomethacin and blocked by the K+ channel blocker tetraethylammonium, suggesting a hyperpolarization pathway. E2 and PPT caused similar relaxation in all vessels. DPN and G1 caused smaller relaxation that was more prominent in abdominal vessels. Reverse transcription-polymerase chain reaction revealed variable ERα messenger RNA expression and increased ERβ in carotid artery and GPR30 in abdominal arteries. Western blots revealed greater amounts of ERα, ERβ, and GPR30 in abdominal arteries. In thoracic aorta, E2-, PPT-, and DPN-induced relaxation was blocked by L-NAME and was associated with increased nitrite/nitrate production, suggesting a role of NO. In abdominal vessels, E2-, PPT-, DPN-, and G1-induced relaxation persisted in L-NAME + indomethacin + tetraethylammonium-treated or endothelium-denuded arteries, suggesting direct effect on vascular smooth muscle. E2, PPT, DPN, and G1 caused greater relaxation of KCl-induced contraction in abdominal vessels, suggesting inhibitory effects on Ca2+ entry. Thus, E2 and ERα stimulation produces similar relaxation of the cephalic, thoracic, and abdominal arteries. In the cephalic and thoracic arteries, particularly the thoracic aorta, E2-induced and ERα- and ERβ-mediated vasodilation involves NO production. ERβ- and GPR30-mediated relaxation is greater in the abdominal arteries and seems to involve hyperpolarization and inhibition of vascular smooth muscle Ca2+ entry. Specific ER agonists could produce vasodilation in specific vascular beds without affecting other vessels in the systemic circulation.
Collapse
|
35
|
Mo Z, Liu M, Yang F, Luo H, Li Z, Tu G, Yang G. GPR30 as an initiator of tamoxifen resistance in hormone-dependent breast cancer. Breast Cancer Res 2013; 15:R114. [PMID: 24289103 PMCID: PMC3978564 DOI: 10.1186/bcr3581] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 11/14/2013] [Indexed: 01/23/2023] Open
Abstract
Introduction Tamoxifen is widely used to treat hormone-dependent breast cancer, but its therapeutic benefit is limited by the development of drug resistance. Here, we investigated the role of estrogen G-protein coupled receptor 30 (GPR30) on Tamoxifen resistance in breast cancer. Methods Primary tumors (PTs) of breast cancer and corresponding metastases (MTs) were used to evaluate the expression of GPR30 and epidermal growth factor receptor (EGFR) immunohistochemically. Tamoxifen-resistant (TAM-R) subclones derived from parent MCF-7 cells were used to investigate the role of GPR30 in the development of tamoxifen resistance, using MTT assay, western blot, RT-PCR, immunofluorescence, ELISA and flow cytometry. TAM-R xenografts were established to assess anti-tumor effects of combination therapy with GPR30 antagonist G15 plus 4-hydroxytamoxifen (Tam), using tumor volume measurement and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). Results In 53 human breast cancer specimens, GPR30 expression in MTs increased compared to matched PTs; in MTs, the expression patterns of GPR30 and EGFR were closely related. Compared to parent MCF-7 cells, TAM-R cells had greater growth responses to 17β-estradiol (E2), GPR30 agonist G1 and Tam, and significantly higher activation of Mitogen-activated protein (MAP) kinases; but this increased activity was abolished by G15 or AG1478. In TAM-R cells, GPR30 cell-surface translocation facilitated crosstalk with EGFR, and reduced cAMP generation, attenuating inhibition of EGFR signaling. Combination therapy both promoted apoptosis in TAM-R cells and decreased drug-resistant tumor progression. Conclusions Long-term endocrine treatment facilitates the translocation of GPR30 to cell surfaces, which interferes with the EGFR signaling pathway; GPR30 also attenuates the inhibition of MAP kinases. These factors contribute to tamoxifen resistance development in breast cancer. Combination therapy with GPR30 inhibitors and tamoxifen may provide a new therapeutic option for drug-resistant breast cancer.
Collapse
|
36
|
Organista-Juárez D, Carretero-Ortega J, Vicente-Fermín O, Vázquez-Victorio G, Sosa-Garrocho M, Vázquez-Prado J, Macías-Silva M, Reyes-Cruz G. Calcium-sensing receptor inhibits TGF-β-signaling by decreasing Smad2 phosphorylation. IUBMB Life 2013; 65:1035-42. [DOI: 10.1002/iub.1232] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/12/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Diana Organista-Juárez
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Jorge Carretero-Ortega
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Onasis Vicente-Fermín
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Genaro Vázquez-Victorio
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Marcela Sosa-Garrocho
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - José Vázquez-Prado
- Department of Pharmacology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| |
Collapse
|
37
|
Bai LY, Weng JR, Hu JL, Wang D, Sargeant AM, Chiu CF. G15, a GPR30 antagonist, induces apoptosis and autophagy in human oral squamous carcinoma cells. Chem Biol Interact 2013; 206:375-84. [DOI: 10.1016/j.cbi.2013.10.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/14/2013] [Accepted: 10/14/2013] [Indexed: 01/11/2023]
|
38
|
Skrzypczak M, Schüler S, Lattrich C, Ignatov A, Ortmann O, Treeck O. G protein-coupled estrogen receptor (GPER) expression in endometrial adenocarcinoma and effect of agonist G-1 on growth of endometrial adenocarcinoma cell lines. Steroids 2013; 78:1087-91. [PMID: 23921077 DOI: 10.1016/j.steroids.2013.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/04/2013] [Accepted: 07/23/2013] [Indexed: 11/21/2022]
Abstract
The G protein-coupled estrogen receptor (GPER, GPR30) is suggested to be involved in non-nuclear estrogen signaling and is expressed in a variety of hormone dependent cancer entities. This study was performed to further elucidate the role of this receptor in endometrial adenocarcinoma. We first analyzed GPER expression at the mRNA level in 88 endometrial cancer or normal endometrial tissue samples and compared it to those of nuclear steroid hormone receptors. GPER transcript levels were found to be about 6-fold reduced, but still present in endometrial cancer. Expression of this receptor was decreased in all grading subgroups when compared to pre- or postmenopausal endometrium. GPER mRNA expression was associated with PR mRNA levels (Spearman's rho 0.4610, p<0.001). We then tested the effect of the GPER ligand G-1 on growth of three endometrial cancer cell lines with different GPER expression. GPER protein levels were highest in RL95-2 cells, moderate in HEC-1A cells and not detectable in HEC-1B cells. The moderate expression level in HEC-1A cells was similar to average tumor tissue expression. Treatment with G-1 significantly inhibited growth of the GPER-positive cell lines RL95-2 and HEC-1A in a dose-dependent manner, whereas the GPER-negative line HEC-1B was not affected. Though GPER transcript levels were found to be reduced in endometrial cancer, our in vitro data suggest that moderate GPER expression might be sufficient to mediate growth-inhibitory effects triggered by its agonist G-1.
Collapse
Affiliation(s)
- Maciej Skrzypczak
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Regensburg, Germany; Second Department of Gynecology, Medical University of Lublin, Poland
| | | | | | | | | | | |
Collapse
|
39
|
Rabi T, Huwiler A, Zangemeister-Wittke U. AMR-Me inhibits PI3K/Akt signaling in hormone-dependent MCF-7 breast cancer cells and inactivates NF-κB in hormone-independent MDA-MB-231 cells. Mol Carcinog 2013; 53:578-88. [PMID: 23475563 DOI: 10.1002/mc.22012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 01/07/2013] [Accepted: 01/13/2013] [Indexed: 11/06/2022]
Abstract
AMR-Me, a C-28 methylester derivative of triterpenoid compound Amooranin isolated from Amoora rohituka stem bark and the plant has been reported to possess multitude of medicinal properties. Our previous studies have shown that AMR-Me can induce apoptosis through mitochondrial apoptotic and MAPK signaling pathways by regulating the expression of apoptosis related genes in human breast cancer MCF-7 cells. However, the molecular mechanism of AMR-Me induced apoptotic cell death remains unclear. Our results showed that AMR-Me dose-dependently inhibited the proliferation of MCF-7 and MDA-MB-231 cells under serum-free conditions supplemented with 1 nM estrogen (E2) with an IC50 value of 0.15 µM, 0.45 µM, respectively. AMR-Me had minimal effects on human normal breast epithelial MCF-10A + ras and MCF-10A cells with IC50 value of 6 and 6.5 µM, respectively. AMR-Me downregulated PI3K p85, Akt1, and p-Akt in an ERα-independent manner in MCF-7 cells and no change in expression levels of PI3K p85 and Akt were observed in MDA-MB-231 cells treated under similar conditions. The PI3K inhibitor LY294002 suppressed Akt activation similar to AMR-Me and potentiated AMR-Me induced apoptosis in MCF-7 cells. EMSA revealed that AMR-Me inhibited nuclear factor-kappaB (NF-κB) DNA binding activity in MDA-MB-231 cells in a time-dependent manner and abrogated EGF induced NF-κB activation. From these studies we conclude that AMR-Me decreased ERα expression and effectively inhibited Akt phosphorylation in MCF-7 cells and inactivate constitutive nuclear NF-κB and its regulated proteins in MDA-MB-231 cells. Due to this multifactorial effect in hormone-dependent and independent breast cancer cells AMR-Me deserves attention for use in breast cancer prevention and therapy.
Collapse
Affiliation(s)
- Thangaiyan Rabi
- Institute of Pharmacology, University of Bern, Bern, Switzerland; K.R. Sterling Cancer and Aids Hospital, Chennai, Tamilnadu, India
| | | | | |
Collapse
|
40
|
Manavathi B, Dey O, Gajulapalli VNR, Bhatia RS, Bugide S, Kumar R. Derailed estrogen signaling and breast cancer: an authentic couple. Endocr Rev 2013; 34:1-32. [PMID: 22947396 PMCID: PMC3565105 DOI: 10.1210/er.2011-1057] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 07/09/2012] [Indexed: 02/06/2023]
Abstract
Estrogen or 17β-estradiol, a steroid hormone, plays a critical role in the development of mammary gland via acting through specific receptors. In particular, estrogen receptor-α (ERα) acts as a transcription factor and/or a signal transducer while participating in the development of mammary gland and breast cancer. Accumulating evidence suggests that the transcriptional activity of ERα is altered by the action of nuclear receptor coregulators and might be responsible, at least in part, for the development of breast cancer. In addition, this process is driven by various posttranslational modifications of ERα, implicating active participation of the upstream receptor modifying enzymes in breast cancer progression. Emerging studies suggest that the biological outcome of breast cancer cells is also influenced by the cross talk between microRNA and ERα signaling, as well as by breast cancer stem cells. Thus, multiple regulatory controls of ERα render mammary epithelium at risk for transformation upon deregulation of normal homeostasis. Given the importance that ERα signaling has in breast cancer development, here we will highlight how the activity of ERα is controlled by various regulators in a spatial and temporal manner, impacting the progression of the disease. We will also discuss the possible therapeutic value of ERα modulators as alternative drug targets to retard the progression of breast cancer.
Collapse
Affiliation(s)
- Bramanandam Manavathi
- Department of Biochemistry, School of Life Sciences, Gachibowli, Prof. CR Rao Road, University of Hyderabad, Hyderabad 500046, India.
| | | | | | | | | | | |
Collapse
|
41
|
WANG LIJUAN, HAN SUXIA, BAI E, ZHOU XIA, LI MENG, JING GUIHUA, ZHAO JING, YANG ANGANG, ZHU QING. Dose-dependent effect of tamoxifen in tamoxifen-resistant breast cancer cells via stimulation by the ERK1/2 and AKT signaling pathways. Oncol Rep 2013; 29:1563-9. [DOI: 10.3892/or.2013.2245] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 12/20/2012] [Indexed: 11/06/2022] Open
|
42
|
Gigoux V, Fourmy D. Acting on Hormone Receptors with Minimal Side Effect on Cell Proliferation: A Timely Challenge Illustrated with GLP-1R and GPER. Front Endocrinol (Lausanne) 2013; 4:50. [PMID: 23641235 PMCID: PMC3638125 DOI: 10.3389/fendo.2013.00050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 04/10/2013] [Indexed: 12/18/2022] Open
Abstract
G protein-coupled receptors (GPCRs) constitute a large family of receptors that sense molecules outside the cell and activate inside signal transduction pathways and cellular responses. GPCR are involved in a wide variety of physiological processes, including in the neuroendocrine system. GPCR are also involved in many diseases and are the target of 30% of marketed medicinal drugs. Whereas the majority of the GPCR-targeting drugs have proved their therapeutic benefit, some of them were associated with undesired effects. We develop two examples of used drugs whose therapeutic benefits are tarnished by carcinogenesis risks. The chronic administration of glucagon-like peptide-1 (GLP-1) analogs widely used to treat type-2 diabetes was associated with an increased risk of pancreatic or thyroid cancers. The long-term treatment with the estrogen antagonist tamoxifen, developed to target breast cancer overexpressing estrogen receptors ER, presents agonist activity on the G protein-coupled estrogen receptor which is associated with an increased incidence of endometrial cancer and breast cancer resistance to hormonotherapy. We point out and discuss the need of pharmacological studies to understand and overcome the undesired effects associated with the chronic administration of GPCR ligands. In fact, biological effects triggered by GPCR often result from the activation of multiple intracellular signaling pathways. Deciphering which signaling networks are engaged following GPCR activation appears to be primordial to unveil their contribution in the physiological and physiopathological processes. The development of biased agonists to elucidate the role of the different signaling mechanisms mediated by GPCR activation will allow the generation of new therapeutic agents with improved efficacy and reduced side effects. In this regard, the identification of GLP-1R biased ligands promoting insulin secretion without inducing pro-tumoral effects would offer therapeutic benefit.
Collapse
Affiliation(s)
- Véronique Gigoux
- Université de Toulouse, Université Paul SabatierToulouse, France
- *Correspondence: Véronique Gigoux, CHU Rangueil – INSERM, Université de Toulouse, Université Paul Sabatier, EA4552, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France. e-mail:
| | - Daniel Fourmy
- Université de Toulouse, Université Paul SabatierToulouse, France
| |
Collapse
|
43
|
Fujiwara S, Terai Y, Kawaguchi H, Takai M, Yoo S, Tanaka Y, Tanaka T, Tsunetoh S, Sasaki H, Kanemura M, Tanabe A, Yamashita Y, Ohmichi M. GPR30 regulates the EGFR-Akt cascade and predicts lower survival in patients with ovarian cancer. J Ovarian Res 2012; 5:35. [PMID: 23163984 PMCID: PMC3543193 DOI: 10.1186/1757-2215-5-35] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 11/09/2012] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED OBJECTIVES G protein-coupled receptor 30 (GPR30) is a 7-transmembrane estrogen receptor that functions alongside traditional estrogen receptors to regulate the cellular responses to estrogen. Recent studies suggest that GPR30 expression is associated with a poor prognosis, and that this is due to the GPR30-mediated transactivation of the EGFR in breast cancer. However, the biological contribution of GPR30 in ovarian cancer remains unclear. The purpose of this study was to elucidate the relationships between GPR30 expression and the clinicopathological findings, and to determine how the signaling cascade influences the prognosis of ovarian cancer. METHODS The expression levels of GPR30, EGFR, ERα, and ERβ were analyzed using an immunohistochemical analysis, and their correlations with the clinicopathological features were examined in 10 patients with borderline malignant tumors and 152 patients with epithelial ovarian cancer. We also examined whether GPR30 signaling activates the EGFR-Akt pathway in an ovarian cancer cell line (Caov-3) by a Western blotting analysis. RESULTS The GPR30 expression in ovarian carcinomas was significantly higher than that in borderline malignancies (p=0.0016), and was not associated with the expression of the EGFR, ERα, or ERβ. The expression of GPR30 in clear cell carcinomas was significantly lower than that in other subtypes of cancer (P <; 0.001). The expression of both GPR30 and EGFR was significantly associated with a poor prognosis in terms of the progression-free survival rate. The phosphorylation of the EGFR and Akt could be significantly enhanced by G1 (p <; 0.05) and inhibited by a Src family kinase inhibitor. CONCLUSION The expression of both GPR30 and EGFR is associated with a poor outcome in ovarian cancer, and GPR30 increases the phosphorylation of Akt via the EGFR in ovarian cancer cells. The regulation of GPR30 might be a potentially useful new therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Satoe Fujiwara
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7, Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Transmembrane estrogen receptor GPR30 is more frequently expressed in malignant than benign ovarian endometriotic cysts and correlates with MMP-9 expression. Int J Gynecol Cancer 2012; 22:539-45. [PMID: 22495744 DOI: 10.1097/igc.0b013e318247323d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Molecular studies supporting the idea of malignant transformation of endometriosis are sparse and not well substantiated. The aims of this study were to detect expression levels of the novel estrogen-responsive receptor G protein-coupled estrogen receptor 1 GPER, also termed GPR30, and to determine its correlation with matrix metalloproteinase-9 (MMP-9) in benign and malignant ovarian endometriotic cysts and to explore the significance of GPR30. METHODS Immunohistochemical staining with the streptavidin-peroxidase method was conducted to determine the expression of GPR30 and MMP-9 in 24 cases of endometriosis-associated ovarian carcinoma (EAOC) and 32 specimens of ovarian endometriosis without malignant transformation. Reverse transcriptase polymerase chain reaction was performed to determine messenger RNA expression of GPR30 and MMP-9 in benign and malignant ovarian endometriotic cysts. We also investigated their associations with known clinic pathological parameters and the interrelationship between the expressions of the 2 proteins. RESULTS The positive staining ratio of GPR30 was 95.8% (23/24) in EAOC cases, and the HScore was 268; whereas the positive ratio was 25% (8/32) in benign endometriotic cysts, and the Hscore was 95. Matrix metalloproteinase-9 was expressed in all 24 EAOC cases and 87.5% (28/32) of the benign samples, and the Hscores were 280 and 260, respectively (P > 0.05). The receptor GPR30 was significantly higher in EAOCs than in benign endometriotic cysts (P < 0.05). The expression of GPR30 messenger RNA was also significantly higher in malignant ovarian endometriotic cysts than in the benign group. The receptor GPR30 was positively related to tumor size, tumor stage, and lymph node metastasis. A positive relationship between GPR30 and MMP-9 was found (P = 0.002). CONCLUSIONS The results suggest that the abnormal expression of GPR30 may be involved in malignant transformation, invasion, and metastasis of EAOCs. Testing of GPR30 expression levels may present both diagnostic and therapeutic options for the treatment of ovarian malignancies.
Collapse
|
45
|
Voutsadakis IA. Epithelial to mesenchymal transition in the pathogenesis of uterine malignant mixed Müllerian tumours: the role of ubiquitin proteasome system and therapeutic opportunities. Clin Transl Oncol 2012; 14:243-53. [PMID: 22484631 DOI: 10.1007/s12094-012-0792-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malignant mixed Müllerian tumours (malignant mixed mesodermal tumours, MMMT) of the uterus are metaplastic carcinomas with a sarcomatous component and thus they are also called carcinosarcomas. It has now been accepted that the sarcomatous component is derived from epithelial elements that have undergone metaplasia. The process that produces this metaplasia is epithelial to mesenchymal transition (EMT), which has recently been described as a neoplasia-associated programme shared with embryonic development and enabling neoplastic cells to move and metastasise. The ubiquitin proteasome system (UPS) regulates the turnover and functions of hundreds of cellular proteins. It plays important roles in EMT by being involved in the regulation of several pathways participating in the execution of this metastasis-associated programme. In this review the specifi c role of UPS in EMT of MMMT is discussed and therapeutic opportunities from UPS manipulations are proposed.
Collapse
Affiliation(s)
- I A Voutsadakis
- Centre Pluridisciplinaire d'Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|
46
|
Khajah MA, Al Saleh S, Mathew PM, Luqmani YA. Differential effect of growth factors on invasion and proliferation of endocrine resistant breast cancer cells. PLoS One 2012; 7:e41847. [PMID: 22860018 PMCID: PMC3408495 DOI: 10.1371/journal.pone.0041847] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 06/29/2012] [Indexed: 01/27/2023] Open
Abstract
We have established several breast cancer cell lines that exhibit a permanent ER-depleted phenotype, induced by shRNA transfection of MCF-7 cells, which afford a useful model for studying acquired endocrine resistance. Previously we showed that MDA-231 as well as ER-silenced cells could invade through simulated extracellular matrix components. However, the contribution of individual serum components responsible for cell invasion was not determined. In the present study, an under-agarose gel assay was used to quantitatively assess the invasive movement of two ER-silenced cell lines (pII and YS2.5) in comparison to the parental MCF-7, the ER negative MDA-231, and normal HBL100 cells, as well as a line that was ER-shRNA transfected but failed to exhibit ER down-regulation (YS1.2). We also examined the effect of the growth factors EGF, IGF-1, TGFβ, PDGFC and RANTES on pII cell invasion and proliferation. All breast cancer cell lines which had reduced ER expression exhibited a serum-dependent invasive ability related to the degree of induced ER loss. TGFβ treatment inhibited pII cell proliferation and enhanced their invasive ability but at a relatively high dose. IGF-1 and EGF enhanced pII cell proliferation, with the latter playing the major role in promoting cell invasion. PDGFC did not affect either process although it is highly expressed in pII cells. Differential effects were observed on activation of Akt and ERK1/2 suggesting their involvement as intracellular mediators of EGF induced invasion, in part through the regulation of matrix metalloproteinase activity. Targeting EGF receptor tyrosine kinase activity by erlotinib resulted in significant inhibition of both pII cell proliferation and directional invasion towards EGF suggesting that this drug has potential therapeutic usefulness for preventing spread of particularly endocrine resistant breast cancer.
Collapse
|
47
|
Landkocz Y, Poupin P, Atienzar F, Vasseur P. Transcriptomic effects of di-(2-ethylhexyl)-phthalate in Syrian hamster embryo cells: an important role of early cytoskeleton disturbances in carcinogenesis? BMC Genomics 2011; 12:524. [PMID: 22026506 PMCID: PMC3218109 DOI: 10.1186/1471-2164-12-524] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 10/25/2011] [Indexed: 01/18/2023] Open
Abstract
Background Di-(2-ethylhexyl)-phthalate (DEHP) is a commonly used plasticizer in polyvinylchloride (PVC) formulations and a potentially non-genotoxic carcinogen. The aim of this study was to identify genes whose level of expression is altered by DEHP by using a global wide-genome approach in Syrian hamster embryo (SHE) cells, a model similar to human cells regarding their responses to this type of carcinogen. With mRNA Differential Display (DD), we analysed the transcriptional regulation of SHE cells exposed to 0, 12.5, 25 and 50 μM of DEHP for 24 hrs, conditions which induced neoplastic transformation of these cells. A real-time quantitative polymerase chain reaction (qPCR) was used to confirm differential expression of genes identified by DD. Results Gene expression profiling showed 178 differentially-expressed fragments corresponding to 122 genes after tblastx comparisons, 79 up-regulated and 43 down-regulated. The genes of interest were involved in many biological pathways, including signal transduction, regulation of the cytoskeleton, xenobiotic metabolism, apoptosis, lipidogenesis, protein conformation, transport and cell cycle. We then focused particularly on genes involved in the regulation of the cytoskeleton, one of the processes occurring during carcinogenesis and in the early steps of neoplastic transformation. Twenty one cytoskeleton-related genes were studied by qPCR. The down-regulated genes were involved in focal adhesion or cell junction. The up-regulated genes were involved in the regulation of the actin cytoskeleton and this would suggest a role of cellular plasticity in the mechanism of chemical carcinogenesis. The gene expression changes identified in the present study were PPAR-independent. Conclusion This study identified a set of genes whose expression is altered by DEHP exposure in mammalian embryo cells. This is the first study that elucidates the genomic changes of DEHP involved in the organization of the cytoskeleton. The latter genes may be candidates as biomarkers predictive of early events in the multistep carcinogenic process.
Collapse
Affiliation(s)
- Yann Landkocz
- CNRS UMR7146, Laboratoire I.E.B.E., Rue General Delestraint, 57070 Metz, France.
| | | | | | | |
Collapse
|
48
|
Zu X, Zhang Q, Cao R, Liu J, Zhong J, Wen G, Cao D. Transforming growth factor-β signaling in tumor initiation, progression and therapy in breast cancer: an update. Cell Tissue Res 2011; 347:73-84. [PMID: 21845401 DOI: 10.1007/s00441-011-1225-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 07/22/2011] [Indexed: 01/06/2023]
Abstract
Transforming growth factor-β (TGF-β) is a ubiquitous cytokine playing an essential role in cell proliferation, differentiation, apoptosis, adhesion and invasion, as well as in cellular microenvironment. In malignant diseases, TGF-β signaling features a growth inhibitory effect at an early stage but aggressive oncogenic activity at the advanced malignant state. Here, we update the current understanding of TGF-β signaling in cancer development and progression with a focus on breast cancer. We also review the current approaches of TGF-β signaling-targeted therapeutics for human malignancies.
Collapse
Affiliation(s)
- Xuyu Zu
- Clinical Research Institution, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
49
|
Fussell KC, Udasin RG, Smith PJ, Gallo MA, Laskin JD. Catechol metabolites of endogenous estrogens induce redox cycling and generate reactive oxygen species in breast epithelial cells. Carcinogenesis 2011; 32:1285-93. [PMID: 21665890 PMCID: PMC3149209 DOI: 10.1093/carcin/bgr109] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 05/04/2011] [Accepted: 06/01/2011] [Indexed: 12/21/2022] Open
Abstract
Estrogens are major risk factors for the development of breast cancer; they can be metabolized to catechols, which are further oxidized to DNA-reactive quinones and semiquinones (SQs). These metabolites are mutagenic and may contribute to the carcinogenic activity of estrogens. Redox cycling of the SQs and subsequent generation of reactive oxygen species (ROS) is also an important mechanism leading to DNA damage. The SQs of exogenous estrogens have been shown to redox cycle, however, redox cycling and the generation of ROS by endogenous estrogens has never been characterized. In the present studies, we determined whether the catechol metabolites of endogenous estrogens, including 2-hydroxyestradiol, 4-hydroxyestradiol, 4-hydroxyestrone and 2-hydroxyestriol, can redox cycle in breast epithelial cells. These catechol estrogens, but not estradiol, estrone, estriol or 2-methoxyestradiol, were found to redox cycle and generate hydrogen peroxide (H(2)O(2)) and hydroxyl radicals in lysates of three different breast epithelial cell lines: MCF-7, MDA-MB-231 and MCF-10A. The generation of ROS required reduced nicotinamide adenine dinucleotide phosphate as a reducing equivalent and was inhibited by diphenyleneiodonium, a flavoenzyme inhibitor, indicating that redox cycling is mediated by flavin-containing oxidoreductases. Using extracellular microsensors, catechol estrogen metabolites stimulated the release of H(2)O(2) by adherent cells, indicating that redox cycling occurs in viable intact cells. Taken together, these data demonstrate that catechol metabolites of endogenous estrogens undergo redox cycling in breast epithelial cells, resulting in ROS production. Depending on the localized concentrations of catechol estrogens and enzymes that mediate redox cycling, this may be an important mechanism contributing to the development of breast cancer.
Collapse
Affiliation(s)
| | | | - Peter J.S. Smith
- Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | | | - Jeffrey D. Laskin
- To whom correspondence should be addressed. Tel: +1 732 445 0170; Fax: +1 732 445 0119;
| |
Collapse
|
50
|
Drabsch Y, ten Dijke P. TGF-β signaling in breast cancer cell invasion and bone metastasis. J Mammary Gland Biol Neoplasia 2011; 16:97-108. [PMID: 21494783 PMCID: PMC3095797 DOI: 10.1007/s10911-011-9217-1] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 03/14/2011] [Indexed: 12/16/2022] Open
Abstract
The contribution of transforming growth factor β (TGF-β) signaling to breast cancer has been studied for more than two decades. In an early phase TGF-β may act as a tumour suppressor, while later, when cells have become resistant to its anti-mitogenic effects, the role of TGF-β switches towards malignant conversion and progression. TGF-β stimulates cell invasion and modifies the microenvironment to the advantage of cancer cells. Studies have shown that TGF-β promotes bone and lung metastasis via different mechanisms. The therapeutic strategies to target the TGF-β pathway in breast cancer are becoming increasingly clear. This review will focus on the role TGF-β in breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Yvette Drabsch
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Peter ten Dijke
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
- Ludwig Institute for Cancer Research and Uppsala University, Box 595, 75124 Uppsala, Sweden
| |
Collapse
|