1
|
Namba N, Fujii S. Hydroboration of vinylsilanes providing diversity-oriented hydrophobic building blocks for biofunctional molecules. Org Biomol Chem 2024. [PMID: 38826124 DOI: 10.1039/d4ob00632a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Hydroboration of vinylsilanes with BH3 affords two silylethanol regioisomers. Herein, we investigated the regioisomeric ratio of hydroboration products from various vinylsilanes, focusing on the characteristic reaction profile. All investigated vinylsilanes afforded both regioisomers, and greater bulkiness increased the proportion of the Markovnikov products. The obtained silylethanols were used as hydrophobic building blocks for constructing nuclear progesterone receptor (PR) modulators. Notably, structural conversions from an α-isomer (silylethan-1-oxy derivative) to a β-isomer (2-silylethoxy derivative) caused complete activity-switching from a PR agonist to an antagonist. Our results indicate that silylethanols are useful for structural development, and vinylsilanes are a versatile source of hydrophobic building blocks for obtaining biofunctional molecules.
Collapse
Affiliation(s)
- Nao Namba
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| |
Collapse
|
2
|
Fu S, Ke H, Yuan H, Xu H, Chen W, Zhao L. Dual role of pregnancy in breast cancer risk. Gen Comp Endocrinol 2024; 352:114501. [PMID: 38527592 DOI: 10.1016/j.ygcen.2024.114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Reproductive history is one of the strongest risk factors for breast cancer in women. Pregnancy can promote short-term breast cancer risk, but also reduce a woman's lifetime risk of breast cancer. Changes in hormone levels before and after pregnancy are one of the key factors in breast cancer risk. This article summarizes the changes in hormone levels before and after pregnancy, and the roles of hormones in mammary gland development and breast cancer progression. Other factors, such as changes in breast morphology and mammary gland differentiation, changes in the proportion of mammary stem cells (MaSCs), changes in the immune and inflammatory environment, and changes in lactation before and after pregnancy, also play key roles in the occurrence and development of breast cancer. This review discusses the dual effects and the potential mechanisms of pregnancy on breast cancer risk from the above aspects, which is helpful to understand the complexity of female breast cancer occurrence.
Collapse
Affiliation(s)
- Shiting Fu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Hao Ke
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | | | - Huaimeng Xu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Wenyan Chen
- Department of Medical Oncology, The Third Hospital of Nanchang, Nanchang 330009, China
| | - Limin Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China.
| |
Collapse
|
3
|
Miyajima Y, Ochiai K, Fujii S. Design, Synthesis, and Evaluation of B-(Trifluoromethyl)phenyl Phosphine-Borane Derivatives as Novel Progesterone Receptor Antagonists. Molecules 2024; 29:1587. [PMID: 38611867 PMCID: PMC11013038 DOI: 10.3390/molecules29071587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
We previously revealed that phosphine-boranes can function as molecular frameworks for biofunctional molecules. In the present study, we exploited the diversity of available phosphines to design and synthesize a series of B-(trifluoromethyl)phenyl phosphine-borane derivatives as novel progesterone receptor (PR) antagonists. We revealed that the synthesized phosphine-borane derivatives exhibited LogP values in a predictable manner and that the P-H group in the phosphine-borane was almost nonpolar. Among the synthesized phosphine-boranes, which exhibited PR antagonistic activity, B-(4-trifluoromethyl)phenyl tricyclopropylphosphine-borane was the most potent with an IC50 value of 0.54 μM. A docking simulation indicated that the tricyclopropylphosphine moiety plays an important role in ligand-receptor interactions. These results support the idea that phosphine-boranes are versatile structural options in drug discovery, and the developed compounds are promising lead compounds for further structural development of next-generation PR antagonists.
Collapse
Affiliation(s)
| | | | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| |
Collapse
|
4
|
Gao Z, Shao D, Zhao C, Liu H, Zhao X, Wei Q, Ma B. The High Level of RANKL Improves IκB/p65/Cyclin D1 Expression and Decreases p-Stat5 Expression in Firm Udder of Dairy Goats. Int J Mol Sci 2023; 24:ijms24108841. [PMID: 37240191 DOI: 10.3390/ijms24108841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/05/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
Udder traits, influencing udder health and function, are positively correlated with lactation performance. Among them, breast texture influences heritability and impacts on the milk yield of cattle; however, there is a lack of systematic research on its underlying mechanism in dairy goats in particular. Here, we showed the structure of firm udders with developed connective tissue and smaller acini per lobule during lactation and confirmed that there were lower serum levels of estradiol (E2) and progesterone (PROG), and higher mammary expression of estrogen nuclear receptor (ER) α and progesterone receptor (PR), in dairy goats with firm udders. The results of transcriptome sequencing of the mammary gland revealed that the downstream pathway of PR, the receptor activator of nuclear factor-kappa B (NF-κB) ligand (RANKL) signal, participated in the formation of firm mammary glands. During the culture of goat mammary epithelial cells (GMECs), high RANKL level additions promote the Inhibitor kappaB (IκB)/p65/Cyclin D1 expression related to cell proliferation and decrease the phosphorylated signal transduction and transcription activator 5 (Stat5) expression related to milk-protein synthesis of GMECs, which is consistent with electron microscope results showing that there are fewer lactoprotein particles in the acinar cavity of a firm mammary. Furthermore, co-culturing with adipocyte-like cells for 7 d is beneficial for the acinar structure formation of GMECs, while there is a slightly negative effect of high RANKL level on it. In conclusion, the results of this study revealed the structure of firm udders structure and confirmed the serum hormone levels and their receptor expression in the mammary glands of dairy goats with firm udders. The underlying mechanism leading to firm udders and a decrease in milk yield were explored preliminarily, which provided an important foundation for the prevention and amelioration of firm udders and improving udder health and milk yield.
Collapse
Affiliation(s)
- Zhen Gao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Xianyang 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Dan Shao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Xianyang 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Chunrui Zhao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Xianyang 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Haokun Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Xianyang 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Xiaoe Zhao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Xianyang 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Qiang Wei
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Xianyang 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Baohua Ma
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Xianyang 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| |
Collapse
|
5
|
Lee SH, Yap YHY, Lim CL, Woo ARE, Lin VCL. Activation function 1 of progesterone receptor is required for mammary development and regulation of RANKL during pregnancy. Sci Rep 2022; 12:12286. [PMID: 35854046 PMCID: PMC9296660 DOI: 10.1038/s41598-022-16289-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Progesterone receptor (PGR) is a member of the nuclear receptor superfamily of transcription factors. It is critical for mammary stem cells expansion, mammary ductal branching and alveologenesis. The transcriptional activity of PGR is mainly mediated by activation functions AF1 and AF2. Although the discovery of AF1 and AF2 propelled the understanding of the mechanism of gene regulation by nuclear receptors, their physiological roles are still poorly understood. This is largely due to the lack of suitable genetic models. The present study reports gain or loss of AF1 function mutant mouse models in the study of mammary development. The gain of function mutant AF1_QQQ exhibits hyperactivity while the loss of function mutant AF1_FFF shows hypoactivity on mammary development. However, the involvement of AF1 is context dependent. Whereas the AF1_FFF mutation causes significant impairment in mammary development during pregnancy or in response to estrogen and progesterone, it has no effect on mammary development in nulliparous mice. Furthermore, Rankl, but not Wnt4 and Areg is a major target gene of AF1. In conclusion, PGR AF1 is a pivotal ligand-dependent activation domain critical for mammary development during pregnancy and it exerts gene specific effect on PGR regulated genes.
Collapse
Affiliation(s)
- Shi Hao Lee
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Yeannie H Y Yap
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.,Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Bandar Saujana Putra, 42610, Jenjarom, Selangor, Malaysia
| | - Chew Leng Lim
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Amanda Rui En Woo
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Valerie C L Lin
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
6
|
Rusidzé M, Adlanmérini M, Chantalat E, Raymond-Letron I, Cayre S, Arnal JF, Deugnier MA, Lenfant F. Estrogen receptor-α signaling in post-natal mammary development and breast cancers. Cell Mol Life Sci 2021; 78:5681-5705. [PMID: 34156490 PMCID: PMC8316234 DOI: 10.1007/s00018-021-03860-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022]
Abstract
17β-estradiol controls post-natal mammary gland development and exerts its effects through Estrogen Receptor ERα, a member of the nuclear receptor family. ERα is also critical for breast cancer progression and remains a central therapeutic target for hormone-dependent breast cancers. In this review, we summarize the current understanding of the complex ERα signaling pathways that involve either classical nuclear “genomic” or membrane “non-genomic” actions and regulate in concert with other hormones the different stages of mammary development. We describe the cellular and molecular features of the luminal cell lineage expressing ERα and provide an overview of the transgenic mouse models impacting ERα signaling, highlighting the pivotal role of ERα in mammary gland morphogenesis and function and its implication in the tumorigenic processes. Finally, we describe the main features of the ERα-positive luminal breast cancers and their modeling in mice.
Collapse
Affiliation(s)
- Mariam Rusidzé
- INSERM U1297, Institut Des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, CHU, Toulouse, France
| | - Marine Adlanmérini
- INSERM U1297, Institut Des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, CHU, Toulouse, France
| | - Elodie Chantalat
- INSERM U1297, Institut Des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, CHU, Toulouse, France
| | - I Raymond-Letron
- LabHPEC et Institut RESTORE, Université de Toulouse, CNRS U-5070, EFS, ENVT, Inserm U1301, Toulouse, France
| | - Surya Cayre
- Department of Cell Biology and Cancer, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR144, Paris, France
| | - Jean-François Arnal
- INSERM U1297, Institut Des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, CHU, Toulouse, France
| | - Marie-Ange Deugnier
- Department of Cell Biology and Cancer, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR144, Paris, France
| | - Françoise Lenfant
- INSERM U1297, Institut Des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, CHU, Toulouse, France.
| |
Collapse
|
7
|
Progesterone receptors in normal breast development and breast cancer. Essays Biochem 2021; 65:951-969. [PMID: 34061163 DOI: 10.1042/ebc20200163] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023]
Abstract
Progesterone receptors (PR) play a pivotal role in many female reproductive tissues such as the uterus, the ovary, and the mammary gland (MG). Moreover, PR play a key role in breast cancer growth and progression. This has led to the development and study of different progestins and antiprogestins, many of which are currently being tested in clinical trials for cancer treatment. Recent reviews have addressed the role of PR in MG development, carcinogenesis, and breast cancer growth. Thus, in this review, in addition to making an overview on PR action in normal and tumor breast, the focus has been put on highlighting the still unresolved topics on hormone treatment involving PR isoforms and breast cancer prognosis.
Collapse
|
8
|
Altamirano GA, Gomez AL, Schierano-Marotti G, Muñoz-de-Toro M, Rodriguez HA, Kass L. Bisphenol A and benzophenone-3 exposure alters milk protein expression and its transcriptional regulation during functional differentiation of the mammary gland in vitro. ENVIRONMENTAL RESEARCH 2020; 191:110185. [PMID: 32946892 DOI: 10.1016/j.envres.2020.110185] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/03/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
The plastic monomer and plasticizer bisphenol A (BPA), and the UV-filter benzophenone-3 (BP3) have been shown to have estrogenic activities that could alter mammary gland development. Our aim was to analyze whether BPA or BP3 direct exposure affects the functional differentiation of the mammary gland using an in vitro model. Mammary organoids were obtained and isolated from 8 week-old virgin female C57BL/6 mice and were differentiated on Matrigel with medium containing lactogenic hormones and exposed to: a) vehicle (0.01% ethanol); b) 1 × 10-9 M or 1 × 10-6 M BPA; or c) 1 × 10-12 M, 1 × 10-9 M or 1 × 10-6 M BP3 for 72 h. The mRNA and protein expression of estrogen receptor alpha (ESR1) and progesterone receptor (PR) were assessed. In addition, mRNA levels of PR-B isoform, glucocorticoid receptor (GR), prolactin receptor (PRLR) and Stat5a, and protein expression of pStat5a/b were evaluated at 72 h. The mRNA and protein expression of milk proteins and their DNA methylation status were also analyzed. Although mRNA level of PRLR and GR was similar between treatments, mRNA expression of ESR1, total PR, PR-B and Stat5a was increased in organoids exposed to 1 × 10-9 M BPA and 1 × 10-12 M BP3. Total PR expression was also increased with 1 × 10-6 M BPA. Nuclear ESR1 and PR expression was observed in all treated organoids; whereas nuclear pStat5a/b alveolar cells was observed only in organoids exposed to 1 × 10-9 M BPA and 1 × 10-12 M BP3. The beta-casein mRNA level was increased in both BPA concentrations and 1 × 10-12 M BP3, which was associated with hypomethylation of its promoter. The beta-casein protein expression was only increased with 1 × 10-9 M BPA or 1 × 10-12 M BP3. In contrast, BPA exposure decreased alpha-lactalbumin mRNA expression and increased DNA methylation level in different methylation-sensitive sites of the gene. Also, 1 × 10-9 M BPA decreased alpha-lactalbumin protein expression. Our results demonstrate that BPA or BP3 exposure alters milk protein synthesis and its transcriptional regulation during mammary gland differentiation in vitro.
Collapse
Affiliation(s)
- Gabriela A Altamirano
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ayelen L Gomez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gonzalo Schierano-Marotti
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Horacio A Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
9
|
Lamb CA, Fabris VT, Lanari C. Progesterone and breast. Best Pract Res Clin Obstet Gynaecol 2020; 69:85-94. [DOI: 10.1016/j.bpobgyn.2020.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/20/2020] [Accepted: 04/03/2020] [Indexed: 12/16/2022]
|
10
|
Islam MS, Afrin S, Jones SI, Segars J. Selective Progesterone Receptor Modulators-Mechanisms and Therapeutic Utility. Endocr Rev 2020; 41:bnaa012. [PMID: 32365199 PMCID: PMC8659360 DOI: 10.1210/endrev/bnaa012] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/30/2020] [Indexed: 02/07/2023]
Abstract
Selective progesterone receptor modulators (SPRMs) are a new class of compounds developed to target the progesterone receptor (PR) with a mix of agonist and antagonist properties. These compounds have been introduced for the treatment of several gynecological conditions based on the critical role of progesterone in reproduction and reproductive tissues. In patients with uterine fibroids, mifepristone and ulipristal acetate have consistently demonstrated efficacy, and vilaprisan is currently under investigation, while studies of asoprisnil and telapristone were halted for safety concerns. Mifepristone demonstrated utility for the management of endometriosis, while data are limited regarding the efficacy of asoprisnil, ulipristal acetate, telapristone, and vilaprisan for this condition. Currently, none of the SPRMs have shown therapeutic success in treating endometrial cancer. Multiple SPRMs have been assessed for efficacy in treating PR-positive recurrent breast cancer, with in vivo studies suggesting a benefit of mifepristone, and multiple in vitro models suggesting the efficacy of ulipristal acetate and telapristone. Mifepristone, ulipristal acetate, vilaprisan, and asoprisnil effectively treated heavy menstrual bleeding (HBM) in patients with uterine fibroids, but limited data exist regarding the efficacy of SPRMs for HMB outside this context. A notable class effect of SPRMs are benign, PR modulator-associated endometrial changes (PAECs) due to the actions of the compounds on the endometrium. Both mifepristone and ulipristal acetate are effective for emergency contraception, and mifepristone was approved by the US Food and Drug Administration (FDA) in 2012 for the treatment of Cushing's syndrome due to its additional antiglucocorticoid effect. Based on current evidence, SPRMs show considerable promise for treatment of several gynecologic conditions.
Collapse
Affiliation(s)
- Md Soriful Islam
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, Maryland
| | - Sadia Afrin
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, Maryland
| | - Sara Isabel Jones
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, Maryland
| | - James Segars
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Recouvreux MS, Diaz Bessone MI, Taruselli A, Todaro L, Lago Huvelle MA, Sampayo RG, Bissell MJ, Simian M. Alterations in Progesterone Receptor Isoform Balance in Normal and Neoplastic Breast Cells Modulates the Stem Cell Population. Cells 2020; 9:cells9092074. [PMID: 32932770 PMCID: PMC7564437 DOI: 10.3390/cells9092074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
To investigate the role of PR isoforms on the homeostasis of stem cells in the normal and neoplastic mammary gland, we used PRA and PRB transgenic mice and the T47D human breast cancer cell line and its derivatives, T47D YA and YB (manipulated to express only PRA or PRB, respectively). Flow cytometry and mammosphere assays revealed that in murine breast, overexpression of PRB leads to an increase in luminal and basal progenitor/stem cells. Ovariectomy had a negative impact on the luminal compartment and induced an increase in mammosphere-forming capacity in cells derived from WT and PRA mice only. Treatment with ICI 182,780 augmented the mammosphere-forming capacity of cells isolated from WT and PRA mice, whilst those from PRB remained unaltered. T47D YB cells showed an increase in the CD44+/CD24Low/- subpopulation; however, the number of tumorspheres did not vary relative to T47D and YA, even though they were larger, more irregular, and had increased clonogenic capacity. T47D and YA tumorspheres were modulated by estrogen/antiestrogens, whereas YB spheres remained unchanged in size and number. Our results show that alterations in PR isoform balance have an impact on normal and tumorigenic breast progenitor/stem cells and suggest a key role for the B isoform, with implications in response to antiestrogens.
Collapse
Affiliation(s)
- María Sol Recouvreux
- Área de Investigaciones, Instituto de Oncología, “Ángel H. Roffo”, Av. San Martín 5481, Buenos Aires C1417DTB, Argentina; (M.S.R.); (M.I.D.B.); (A.T.); (L.T.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - María Inés Diaz Bessone
- Área de Investigaciones, Instituto de Oncología, “Ángel H. Roffo”, Av. San Martín 5481, Buenos Aires C1417DTB, Argentina; (M.S.R.); (M.I.D.B.); (A.T.); (L.T.)
- Instituto de Nanosistemas, Universidad Nacional de San Martín, 25 de Mayo 1021, San Martín, Buenos Aires 1650, Argentina;
| | - Agustina Taruselli
- Área de Investigaciones, Instituto de Oncología, “Ángel H. Roffo”, Av. San Martín 5481, Buenos Aires C1417DTB, Argentina; (M.S.R.); (M.I.D.B.); (A.T.); (L.T.)
| | - Laura Todaro
- Área de Investigaciones, Instituto de Oncología, “Ángel H. Roffo”, Av. San Martín 5481, Buenos Aires C1417DTB, Argentina; (M.S.R.); (M.I.D.B.); (A.T.); (L.T.)
| | - María Amparo Lago Huvelle
- Instituto de Nanosistemas, Universidad Nacional de San Martín, 25 de Mayo 1021, San Martín, Buenos Aires 1650, Argentina;
| | - Rocío G. Sampayo
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA;
| | - Mina J. Bissell
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA;
| | - Marina Simian
- Área de Investigaciones, Instituto de Oncología, “Ángel H. Roffo”, Av. San Martín 5481, Buenos Aires C1417DTB, Argentina; (M.S.R.); (M.I.D.B.); (A.T.); (L.T.)
- Instituto de Nanosistemas, Universidad Nacional de San Martín, 25 de Mayo 1021, San Martín, Buenos Aires 1650, Argentina;
- Correspondence:
| |
Collapse
|
12
|
Dwyer AR, Truong TH, Ostrander JH, Lange CA. 90 YEARS OF PROGESTERONE: Steroid receptors as MAPK signaling sensors in breast cancer: let the fates decide. J Mol Endocrinol 2020; 65:T35-T48. [PMID: 32209723 PMCID: PMC7329584 DOI: 10.1530/jme-19-0274] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
Steroid hormone receptors (SRs) are classically defined as ligand-activated transcription factors that function as master regulators of gene programs important for a wide range of processes governing adult physiology, development, and cell or tissue homeostasis. A second function of SRs includes the ability to activate cytoplasmic signaling pathways. Estrogen (ER), androgen (AR), and progesterone (PR) receptors bind directly to membrane-associated signaling molecules including mitogenic protein kinases (i.e. c-SRC and AKT), G-proteins, and ion channels to mediate context-dependent actions via rapid activation of downstream signaling pathways. In addition to making direct contact with diverse signaling molecules, SRs are further fully integrated with signaling pathways by virtue of their N-terminal phosphorylation sites that act as regulatory hot-spots capable of sensing the signaling milieu. In particular, ER, AR, PR, and closely related glucocorticoid receptors (GR) share the property of accepting (i.e. sensing) ligand-independent phosphorylation events by proline-directed kinases in the MAPK and CDK families. These signaling inputs act as a 'second ligand' that dramatically impacts cell fate. In the face of drugs that reliably target SR ligand-binding domains to block uncontrolled cancer growth, ligand-independent post-translational modifications guide changes in cell fate that confer increased survival, EMT, migration/invasion, stemness properties, and therapy resistance of non-proliferating SR+ cancer cell subpopulations. The focus of this review is on MAPK pathways in the regulation of SR+ cancer cell fate. MAPK-dependent phosphorylation of PR (Ser294) and GR (Ser134) will primarily be discussed in light of the need to target changes in breast cancer cell fate as part of modernized combination therapies.
Collapse
Affiliation(s)
- Amy R. Dwyer
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455
| | - Thu H. Truong
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455
| | - Julie H. Ostrander
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis MN 55455
| | - Carol A. Lange
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis MN 55455
- Department of Pharmacology, University of Minnesota, Minneapolis MN 55455
- Corresponding author: Carol A Lange, Professor, ; 612-626-0621 (phone), University of Minnesota Masonic Cancer Center, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN 55455, USA
| |
Collapse
|
13
|
Mori S, Tsuemoto N, Kasagawa T, Nakano E, Fujii S, Kagechika H. Development of Boron-Cluster-Based Progesterone Receptor Antagonists Bearing a Pentafluorosulfanyl (SF 5) Group. Chem Pharm Bull (Tokyo) 2019; 67:1278-1283. [DOI: 10.1248/cpb.c19-00522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shuichi Mori
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU)
| | - Nozomi Tsuemoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU)
| | - Tomoya Kasagawa
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU)
| | - Eiichi Nakano
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU)
| | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU)
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU)
| |
Collapse
|
14
|
Dodda BR, Bondi CD, Hasan M, Clafshenkel WP, Gallagher KM, Kotlarczyk MP, Sethi S, Buszko E, Latimer JJ, Cline JM, Witt-Enderby PA, Davis VL. Co-administering Melatonin With an Estradiol-Progesterone Menopausal Hormone Therapy Represses Mammary Cancer Development in a Mouse Model of HER2-Positive Breast Cancer. Front Oncol 2019; 9:525. [PMID: 31355130 PMCID: PMC6636553 DOI: 10.3389/fonc.2019.00525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022] Open
Abstract
Melatonin has numerous anti-cancer properties reported to influence cancer initiation, promotion, and metastasis. With the need for effective hormone therapies (HT) to treat menopausal symptoms without increasing breast cancer risk, co-administration of nocturnal melatonin with a natural, low-dose HT was evaluated in mice that develop primary and metastatic mammary cancer. Individually, melatonin (MEL) and estradiol-progesterone therapy (EPT) did not significantly affect mammary cancer development through age 14 months, but, when combined, the melatonin-estradiol-progesterone therapy (MEPT) significantly repressed tumor formation. This repression was due to effects on tumor incidence, but not latency. These results demonstrate that melatonin and the HT cooperate to decrease the mammary cancer risk. Melatonin and EPT also cooperate to alter the balance of the progesterone receptor (PR) isoforms by significantly increasing PRA protein expression only in MEPT mammary glands. Melatonin significantly suppressed amphiregulin transcripts in MEL and MEPT mammary glands, suggesting that amphiregulin together with the higher PRA:PRB balance and other factors may contribute to reducing cancer development in MEPT mice. Melatonin supplementation influenced mammary morphology by increasing tertiary branching in the mouse mammary glands and differentiation in human mammary epithelial cell cultures. Uterine weight in the luteal phase was elevated after long-term exposure to EPT, but not to MEPT, indicating that melatonin supplementation may reduce estrogen-induced uterine stimulation. Melatonin supplementation significantly decreased the incidence of grossly-detected lung metastases in MEL mice, suggesting that melatonin delays the formation of metastatic lesions and/or decreases aggressiveness in this model of HER2+ breast cancer. Mammary tumor development was similar in EPT and MEPT mice until age 8.6 months, but after 8.6 months, only MEPT continued to suppress cancer development. These data suggest that melatonin supplementation has a negligible effect in young MEPT mice, but is required in older mice to inhibit tumor formation. Since melatonin binding was significantly decreased in older mammary glands, irrespective of treatment, melatonin supplementation may overcome reduced melatonin responsiveness in the aged MEPT mice. Since melatonin levels are known to decline near menopause, nocturnal melatonin supplementation may also be needed in aging women to cooperate with HT to decrease breast cancer risk.
Collapse
Affiliation(s)
- Balasunder R Dodda
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Corry D Bondi
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Mahmud Hasan
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - William P Clafshenkel
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Katie M Gallagher
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Mary P Kotlarczyk
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Shalini Sethi
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Ethan Buszko
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Jean J Latimer
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - J Mark Cline
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Paula A Witt-Enderby
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States.,UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Vicki L Davis
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Kaitoh K, Nakatsu A, Mori S, Kagechika H, Hashimoto Y, Fujii S. Design, Synthesis and Biological Evaluation of Novel Nonsteroidal Progesterone Receptor Antagonists Based on Phenylamino-1,3,5-triazine Scaffold. Chem Pharm Bull (Tokyo) 2019; 67:566-575. [PMID: 31155562 DOI: 10.1248/cpb.c19-00094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We report here the development of phenylamino-1,3,5-triazine derivatives as novel nonsteroidal progesterone receptor (PR) antagonists. PR plays key roles in various physiological systems, including the female reproductive system, and PR antagonists are promising candidates for clinical treatment of multiple diseases. By using the phenylamino-1,3,5-triazine scaffold as a template structure, we designed and synthesized a series of 4-cyanophenylamino-1,3,5-triazine derivatives. The synthesized compounds exhibited PR antagonistic activity, and among them, compound 12n was the most potent (IC50 = 0.30 µM); it also showed significant binding affinity to the PR ligand-binding domain. Docking simulation supported the design rationale of the compounds. Our results suggest that the phenylamino-1,3,5-triazine scaffold is a versatile template for development of nonsteroidal PR antagonists and that the developed compounds are promising lead compounds for further structural development of nonsteroidal PR antagonists.
Collapse
Affiliation(s)
- Kazuma Kaitoh
- Institute for Quantitative Biosciences, The University of Tokyo
| | - Aki Nakatsu
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University
| | - Shuichi Mori
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University
| | | | - Shinya Fujii
- Institute for Quantitative Biosciences, The University of Tokyo.,Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University
| |
Collapse
|
16
|
Tan S, Bajalovic N, Wong ESP, Lin VCL. Ligand-activated progesterone receptor B activates transcription factor EB to promote autophagy in human breast cancer cells. Exp Cell Res 2019; 382:111433. [PMID: 31100306 DOI: 10.1016/j.yexcr.2019.05.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/30/2019] [Accepted: 05/10/2019] [Indexed: 02/08/2023]
Abstract
Autophagy is an evolutionary conserved, self-eating process that targets cellular constituents for lysosomal degradation. Transcription factor EB (TFEB) is a master regulator of autophagy by inducing the expression of genes involved in autophagic and lysosomal degradation. In breast cancer, ligand-activated progesterone receptor has been reported to influence cancer development by manipulating the autophagy pathway. However, understanding of the mechanism that underlies this autophagic response remains limited. Herein, we report that prolonged treatment with progestin R5020 upregulates autophagy in MCF-7 human breast cancer cells via a novel interplay between progesterone receptor B (PRB) and TFEB. R5020 upregulates TFEB gene expression and protein levels in a PRB-dependent manner. Additionally, R5020 enhances the co-recruitment of PRB and TFEB to each other to facilitate TFEB nuclear localization. Once in the nucleus, TFEB induces the expression of autophagy and lysosomal genes to potentiate autophagy. Together, our findings highlight a novel functional connection between ligand-activated PRB and TFEB to modulate autophagy in MCF-7 breast cancer cells. As breast cancer development is controlled by autophagy, the progestin-PRB-TFEB transduction pathway warrants future attention as a potential therapeutic target in cancer therapy.
Collapse
Affiliation(s)
- Sijie Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Natasa Bajalovic
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Esther S P Wong
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore; Centre for Healthy Ageing, National University Health System, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Valerie C L Lin
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
17
|
Wang W, Do HN, Aupperlee MD, Durairaj S, Flynn EE, Miksicek RJ, Haslam SZ, Schwartz RC. C/EBPβ LIP and c-Jun synergize to regulate expression of the murine progesterone receptor. Mol Cell Endocrinol 2018; 477:57-69. [PMID: 29870755 PMCID: PMC6153074 DOI: 10.1016/j.mce.2018.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/11/2023]
Abstract
CCAAT/enhancer binding protein β (C/EBPβ) is required for murine mammary ductal morphogenesis and alveologenesis. Progesterone is critical for proliferation and alveologenesis in adult mammary glands, and there is a similar requirement for progesterone receptor isoform B (PRB) in alveologenesis. We examined C/EBPβ regulation of PR expression. All three C/EBPβ isoforms, including typically inhibitory LIP, transactivated the PR promoter. LIP, particularly, strongly synergized with c-Jun to drive PR transcription. Endogenous C/EBPβ and c-Jun stimulated a PR promoter-reporter and these two factors showed promoter occupancy on the endogenous PR gene. Additionally, LIP overexpression elevated endogenous PR protein expression. In pregnancy, both PRB and the relative abundance of LIP among C/EBPβ isoforms increase. Consistent with a role in PRB expression, in vivo C/EBPβ and PR isoform A expression showed mutually exclusive localization in mammary epithelium, while C/EBPβ and PRB largely co-localized. We suggest a critical role for C/EBPβ, particularly LIP, in PRB expression.
Collapse
Affiliation(s)
- Weizhong Wang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Han Ngoc Do
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Mark D Aupperlee
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA; Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Srinivasan Durairaj
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Emily E Flynn
- Genetics Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Richard J Miksicek
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Sandra Z Haslam
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA; Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Richard C Schwartz
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA; Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
18
|
Wahlbuhl M, Schuepbach-Mallepell S, Kowalczyk-Quintas C, Dick A, Fahlbusch FB, Schneider P, Schneider H. Attenuation of Mammary Gland Dysplasia and Feeding Difficulties in Tabby Mice by Fetal Therapy. J Mammary Gland Biol Neoplasia 2018; 23:125-138. [PMID: 29855766 DOI: 10.1007/s10911-018-9399-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Hypohidrotic ectodermal dysplasias (HED) are hereditary differentiation disorders of multiple ectodermal structures including the mammary gland. The X-linked form of HED (XLHED) is caused by a lack of the secreted signaling molecule ectodysplasin A1 (EDA1) which is encoded by the gene EDA and belongs to the tumor necrosis factor (TNF) superfamily. Although male patients (hemizygous) are usually more severely affected by XLHED, heterozygous female carriers of an EDA mutation may also suffer from a variety of symptoms, in particular from abnormal development of their breasts. In Tabby mice, a well-studied animal model of XLHED, EDA1 is absent. We investigated the effects of prenatal administration of Fc-EDA, a recombinant EDA1 replacement protein, on mammary gland development in female Tabby mice. Intra-amniotic delivery of Fc-EDA to fetal animals resulted later in improved breastfeeding and thus promoted the growth of their offspring. In detail, such treatment led to a normalization of the nipple shape (protrusion, tapering) that facilitated sucking. Mammary glands of treated female Tabby mice also showed internal changes, including enhanced branching morphogenesis and ductal elongation. Our findings indicate that EDA receptor stimulation during development has a stable impact on later stages of mammary gland differentiation, including lactation, but also show that intra-amniotic administration of an EDA1 replacement protein to fetal Tabby mice partially corrects the mammary gland phenotype in female adult animals.
Collapse
Affiliation(s)
- Mandy Wahlbuhl
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany.
| | | | | | - Angela Dick
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany
| | - Fabian B Fahlbusch
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Holm Schneider
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany
| |
Collapse
|
19
|
Hai L, Szwarc MM, Wetendorf M, Wu SP, Peavey MC, Grimm SL, Edwards DP, DeMayo FJ, Lydon JP. A mouse model engineered to conditionally express the progesterone receptor-B isoform. Genesis 2018; 56:e23223. [PMID: 30004627 PMCID: PMC6141023 DOI: 10.1002/dvg.23223] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/30/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023]
Abstract
Using a Rosa26 gene targeting strategy in mouse embryonic stem cells, we have generated a new transgenic mouse (Pgr-B LSL ), which is designed to conditionally express the epitope-tagged mouse progesterone receptor-B (PGR-B) isoform when crossed with a specific cre driver mouse. To functionally validate this transgenic mouse, we crossed the Pgr-B LSL mouse with the MMTV-CREA transgenic mouse to create the MMTV-CREA/Pgr-B LSL bigenic (termed PR-B:OE to denote PGR-B overexpressor). As expected, transgene-derived PGR-B protein was specifically targeted to the virgin mammary gland epithelium. At a functional level, the PR-B:OE bigenic exhibited abnormal mammary morphogenesis-dilated epithelial ducts, precocious alveologenesis and lateral side-branching, along with a prominent proliferative signature-that resulted in pregnant PR-B:OE mice unable to exhibit mammary gland terminal differentiation at parturition. Because of this developmental failure, the PR-B:OE mammary gland was incapable of producing milk resulting in early neonatal death of otherwise healthy litters. This first line of analysis demonstrates the utility of the Pgr-B LSL mouse to examine the role of the PGR-B isoform in different physiologic and pathophysiologic systems that are responsive to progesterone.
Collapse
Affiliation(s)
- Lan Hai
- Department of Molecular & Cellular Biology, Baylor
College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Maria M. Szwarc
- Department of Molecular & Cellular Biology, Baylor
College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | | | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina,
27709
| | | | - Sandra L. Grimm
- Department of Molecular & Cellular Biology, Baylor
College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Dean P. Edwards
- Department of Molecular & Cellular Biology, Baylor
College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Francesco J. DeMayo
- Reproductive and Developmental Biology Laboratory, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina,
27709
| | - John P. Lydon
- Department of Molecular & Cellular Biology, Baylor
College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| |
Collapse
|
20
|
Nishiyama Y, Mori S, Makishima M, Fujii S, Kagechika H, Hashimoto Y, Ishikawa M. Novel Nonsteroidal Progesterone Receptor (PR) Antagonists with a Phenanthridinone Skeleton. ACS Med Chem Lett 2018; 9:641-645. [PMID: 30034593 DOI: 10.1021/acsmedchemlett.8b00058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 06/23/2018] [Indexed: 12/15/2022] Open
Abstract
The progesterone receptor (PR) plays an important role in various physiological systems, including female reproduction and the central nervous system, and PR antagonists are thought to be effective not only as contraceptive agents and abortifacients but also in the treatment of various diseases, including hormone-dependent cancers and endometriosis. Here, we identified phenanthridin-6(5H)-one derivatives as a new class of PR antagonists and investigated their structure-activity relationships. Among the synthesized compounds, 37, 40, and 46 exhibited very potent PR antagonistic activity with high selectivity for PR over other nuclear receptors. These compounds are structurally distinct from other nonsteroidal PR antagonists, including cyanoaryl derivatives, and should be useful for further studies of the clinical utility of PR antagonists.
Collapse
Affiliation(s)
- Yuko Nishiyama
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Shuichi Mori
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Makoto Makishima
- Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Shinya Fujii
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Yuichi Hashimoto
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Minoru Ishikawa
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| |
Collapse
|
21
|
Lamb CA, Fabris VT, Jacobsen B, Molinolo AA, Lanari C. Biological and clinical impact of imbalanced progesterone receptor isoform ratios in breast cancer. Endocr Relat Cancer 2018; 25:ERC-18-0179. [PMID: 29991638 DOI: 10.1530/erc-18-0179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/17/2022]
Abstract
There is a consensus that progestins and thus their cognate receptor molecules, the progesterone receptors (PR), are essential in the development of the adult mammary gland and regulators of proliferation and lactation. However, a role for natural progestins in breast carcinogenesis remains poorly understood. A hint to that possible role came from studies in which the synthetic progestin medroxyprogesterone acetate was associated with an increased breast cancer risk in women under hormone replacement therapy. However, progestins have been also used for breast cancer treatment and to inhibit the growth of several experimental breast cancer models. More recently, PR have been shown to be regulators of estrogen receptor signaling. With all this information, the question is how can we target PR, and if so, which patients may benefit from such an approach? PR are not single unique molecules. Two main PR isoforms have been characterized, PRA and PRB, that exert different functions and the relative abundance of one isoform respect to the other determines the response of PR agonists and antagonists. Immunohistochemistry with standard antibodies against PR do not discriminate between isoforms. In this review, we summarize the current knowledge on the expression of both PR isoforms in mammary glands, in experimental models of breast cancer and in breast cancer patients, to better understand how the PRA/PRB ratio can be exploited therapeutically to design personalized therapeutic strategies.
Collapse
Affiliation(s)
- Caroline A Lamb
- C Lamb, Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Victoria T Fabris
- V Fabris, Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Britta Jacobsen
- B Jacobsen, Department of Pathology, University of Colorado at Denver - Anschutz Medical Campus, Aurora, United States
| | - Alfredo A Molinolo
- A Molinolo, Biorepository and Tissue Technology Shared Resource, University of California San Diego Moores Cancer Center, La Jolla, United States
| | - Claudia Lanari
- C Lanari, Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| |
Collapse
|
22
|
Aupperlee MD, Kariagina A, Zaremba N, Basson MD, Schwartz RC, Haslam SZ. The Proliferative Response to p27 Down-Regulation in Estrogen Plus Progestin Hormonal Therapy is Lost in Breast Tumors. Transl Oncol 2018; 11:518-527. [PMID: 29524829 PMCID: PMC5884216 DOI: 10.1016/j.tranon.2018.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/08/2018] [Accepted: 02/14/2018] [Indexed: 11/23/2022] Open
Abstract
Increased proliferation and breast cancer risk has been observed in postmenopausal women receiving estrogen (E) + progestin hormone replacement therapy (HRT). Progestin action is mediated through two progesterone receptor (PR) isoforms, PRA and PRB, with unique transcriptional activity and function. The current study examines hormonal regulation of PR isoforms in the normal postmenopausal human breast and the mechanism by which progestins increase proliferation and breast cancer risk. Archival benign breast biopsies from postmenopausal and premenopausal women, and luminal breast tumor biopsies from postmenopausal women, were analyzed for regulation of PRA and PRB expression by E and E+medroxyprogesterone acetate (MPA). In the postmenopausal breast without HRT, PRA and PRB expression was decreased compared to the premenopausal breast. Both E (n = 12) and E+MPA (n = 13) HRT in the postmenopausal breast were associated with increased PRA and PRB expression, increased nuclear cyclin E expression, and decreased nuclear p27 expression compared to no HRT (n = 16). With E+MPA HRT, there was a further decrease in nuclear p27 and increased Receptor Activator of NF-kappa B Ligand (RANKL) expression compared to E-alone HRT. In luminal breast cancers, E+MPA HRT (n = 6) was also associated with decreased nuclear expression of the cell cycle inhibitor p27 compared to E HRT (n = 6), but was not associated with increased proliferation. These results suggest that p27 mediates progestin-induced proliferation in the normal human breast and that regulation of this proliferative response by E+MPA is lost in breast tumors.
Collapse
Affiliation(s)
- Mark D Aupperlee
- Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI; Department of Physiology, Michigan State University, East Lansing, MI.
| | - Anastasia Kariagina
- Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI
| | - Nicole Zaremba
- Department of Surgery, University of Southern California, Los Angeles, CA
| | - Marc D Basson
- Department of Surgery, University of North Dakota, Grand Forks, ND
| | - Richard C Schwartz
- Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI
| | - Sandra Z Haslam
- Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI; Department of Physiology, Michigan State University, East Lansing, MI
| |
Collapse
|
23
|
Fabris V, Abascal MF, Giulianelli S, May M, Sequeira GR, Jacobsen B, Lombès M, Han J, Tran L, Molinolo A, Lanari C. Isoform specificity of progesterone receptor antibodies. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2017; 3:227-233. [PMID: 29085663 PMCID: PMC5653926 DOI: 10.1002/cjp2.83] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/19/2022]
Abstract
Progesterone receptors (PR) are prognostic and predictive biomarkers in hormone‐dependent cancers. Two main PR isoforms have been described, PRB and PRA, that differ only in that PRB has 164 extra N‐terminal amino acids. It has been reported that several antibodies empirically exclusively recognize PRA in formalin‐fixed paraffin‐embedded (FFPE) tissues. To confirm these findings, we used human breast cancer xenograft models, T47D‐YA and ‐YB cells expressing PRA or PRB, respectively, MDA‐MB‐231 cells modified to synthesize PRB, and MDA‐MB‐231/iPRAB cells which can bi‐inducibly express either PRA or PRB. Cells were injected into immunocompromised mice to generate tumours exclusively expressing PRA or PRB. PR isoform expression was verified using immunoblots. FFPE samples from the same tumours were studied by immunohistochemistry using H‐190, clone 636, clone 16, and Ab‐6 anti‐PR antibodies, the latter exclusively recognizing PRB. Except for Ab‐6, all antibodies displayed a similar staining pattern. Our results indicate that clones 16, 636, and the H‐190 antibody recognize both PR isoforms. They point to the need for more stringency in evaluating the true specificity of purported PRA‐specific antibodies as the PRA/PRB ratio may have prognostic and predictive value in breast cancer.
Collapse
Affiliation(s)
- Victoria Fabris
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICETBuenos AiresArgentina
| | - María F Abascal
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICETBuenos AiresArgentina
| | - Sebastián Giulianelli
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICETBuenos AiresArgentina.,Laboratorio de Reproducción y Biología Integrativa de Invertebrados Marinos, Instituto de Biología de Organismos Marinos (IBIOMAR), CONICETArgentina
| | - María May
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICETBuenos AiresArgentina
| | - Gonzalo R Sequeira
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICETBuenos AiresArgentina
| | | | - Marc Lombès
- Unité Mixte de Recherche, INSERM U 1185, Fac Med Paris SudUniversité Paris SaclayFrance
| | - Julie Han
- Department of Pathology, Moore's Cancer Center, UCSDLa JollaCAUSA
| | - Luan Tran
- Department of Pathology, Moore's Cancer Center, UCSDLa JollaCAUSA
| | - Alfredo Molinolo
- Department of Pathology, Moore's Cancer Center, UCSDLa JollaCAUSA
| | - Claudia Lanari
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICETBuenos AiresArgentina
| |
Collapse
|
24
|
Zhu Y, Aupperlee MD, Haslam SZ, Schwartz RC. Pubertally Initiated High-Fat Diet Promotes Mammary Tumorigenesis in Obesity-Prone FVB Mice Similarly to Obesity-Resistant BALB/c Mice. Transl Oncol 2017; 10:928-935. [PMID: 29024822 PMCID: PMC5704096 DOI: 10.1016/j.tranon.2017.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/14/2017] [Indexed: 01/27/2023] Open
Abstract
Premenopausal breast cancer is associated with increased animal fat consumption among normal-weight but not overweight women. Our previous findings in obesity-resistant BALB/c mice showed that a diet high in saturated animal fat (HFD) promotes mammary tumorigenesis in both DMBA carcinogenesis and Trp53-null transplant models. Having made these observations in BALB/c mice, which have very modest HFD weight gain, we determined the effects of HFD in FVB mice, which gain significant weight on HFD. Three-week-old FVB mice fed a low-fat diet or HFD were subjected to 7,12-dimethylbenz[a]anthracene-induced carcinogenesis. Like BALB/c mice, HFD promoted mammary tumorigenesis. Development of tumors largely occurred prior to mice becoming obese, indicating the role of animal-derived HFD rather than resulting obesity in tumor promotion. Also similar to BALB/c mice, early-occurring adenosquamous mammary tumors were abundant among HFD-fed FVB mice. Tumors from HFD mice also had increased intra-tumor M2 macrophages. Prior to tumor development, HFD accelerated normal mammary gland development and increased mammary M2 macrophages, similarly to BALB/c mice. The promotional effects of puberty-initiated HFD on carcinogen-induced mammary cancer are thus largely weight gain-independent. Like BALB/c mice, HFD promoted adenosquamous tumors, suggesting a role for early age HFD in promoting this subtype of triple negative mammary cancer. M2 macrophage recruitment was common to both mouse strains. We speculate that a similar effect of HFD on immune function may contribute to epidemiological findings of increased breast cancer risk in young, premenopausal, normal-weight women who consume a diet high in saturated animal fat.
Collapse
Affiliation(s)
- Yirong Zhu
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI; Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI
| | - Mark D Aupperlee
- Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI; Department of Physiology, Michigan State University, East Lansing, MI
| | - Sandra Z Haslam
- Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI; Department of Physiology, Michigan State University, East Lansing, MI.
| | - Richard C Schwartz
- Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI.
| |
Collapse
|
25
|
O'Leary KA, Shea MP, Salituro S, Blohm CE, Schuler LA. Prolactin Alters the Mammary Epithelial Hierarchy, Increasing Progenitors and Facilitating Ovarian Steroid Action. Stem Cell Reports 2017; 9:1167-1179. [PMID: 28919264 PMCID: PMC5639259 DOI: 10.1016/j.stemcr.2017.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 01/01/2023] Open
Abstract
Hormones drive mammary development and function and play critical roles in breast cancer. Epidemiologic studies link prolactin (PRL) to increased risk for aggressive cancers that express estrogen receptor α (ERα). However, in contrast to ovarian steroids, PRL actions on the mammary gland outside of pregnancy are poorly understood. We employed the transgenic NRL-PRL model to examine the effects of PRL alone and with defined estrogen/progesterone exposure on stem/progenitor activity and regulatory networks that drive epithelial differentiation. PRL increased progenitors and modulated transcriptional programs, even without ovarian steroids, and with steroids further raised stem cell activity associated with elevated canonical Wnt signaling. However, despite facilitating some steroid actions, PRL opposed steroid-driven luminal maturation and increased CD61+ luminal cells. Our findings demonstrate that PRL can powerfully influence the epithelial hierarchy alone and temper the actions of ovarian steroids, which may underlie its role in the development of breast cancer.
Collapse
Affiliation(s)
- Kathleen A O'Leary
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael P Shea
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Molecular and Environmental Toxicology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Stephanie Salituro
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Courtney E Blohm
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; UW Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53792, USA; Molecular and Environmental Toxicology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
26
|
Subramani R, Lakshmanaswamy R. Pregnancy and Breast Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 151:81-111. [PMID: 29096898 DOI: 10.1016/bs.pmbts.2017.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer is the most commonly diagnosed type of cancer among women worldwide. The majority of breast cancers are sporadic and the etiology is not well understood. Several factors have been attributed to altering the risk of breast cancer. A full-term pregnancy is a crucial factor in altering the risk. Early full-term pregnancy has been shown to reduce the lifetime risk of breast cancer, while a later first full-term pregnancy increases breast cancer risk. Epidemiological and experimental data demonstrate that spontaneous or induced abortions do not significantly alter the risk of breast cancer. In this study, we briefly discuss the different types and stages of breast cancer, various risk factors, and potential mechanisms involved in early full-term pregnancy-induced protection against breast cancer. Understanding how early full-term pregnancy induces protection against breast cancer will help design innovative preventive and therapeutic strategies. This understanding can also help in the development of molecular biomarkers that can be of tremendous help in predicting the risk of breast cancer in the general population.
Collapse
Affiliation(s)
- Ramadevi Subramani
- Center of Emphasis in Cancer Research, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Rajkumar Lakshmanaswamy
- Center of Emphasis in Cancer Research, Paul L. Foster School of Medicine, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States.
| |
Collapse
|
27
|
Rojas PA, May M, Sequeira GR, Elia A, Alvarez M, Martínez P, Gonzalez P, Hewitt S, He X, Perou CM, Molinolo A, Gibbons L, Abba MC, Gass H, Lanari C. Progesterone Receptor Isoform Ratio: A Breast Cancer Prognostic and Predictive Factor for Antiprogestin Responsiveness. J Natl Cancer Inst 2017; 109:3064537. [PMID: 28376177 DOI: 10.1093/jnci/djw317] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/01/2016] [Indexed: 12/12/2022] Open
Abstract
Background Compelling evidence shows that progestins regulate breast cancer growth. Using preclinical models, we demonstrated that antiprogestins are inhibitory when the level of progesterone receptor isoform A (PR-A) is higher than that of isoform B (PR-B) and that they might stimulate growth when PR-B is predominant. The aims of this study were to investigate ex vivo responses to mifepristone (MFP) in breast carcinomas with different PR isoform ratios and to examine their clinical and molecular characteristics. Methods We performed human breast cancer tissue culture assays (n = 36) to evaluate the effect of MFP on cell proliferation. PR isoform expression was determined by immunoblotting (n = 282). Tumors were categorized as PRA-H (PR-A/PR-B ≥ 1.2) or PRB-H (PR-A/PR-B ≤ 0.83). RNA was extracted for Ribo-Zero-Seq sequencing to evaluate differentially expressed genes. Subtypes and risk scores were predicted using the PAM50 gene set, the data analyzed using The Cancer Genome Atlas RNA-seq gene analysis and other publicly available gene expression data. Tissue microarrays were performed using paraffin-embedded tissues (PRA-H n = 53, PRB-H n = 24), and protein expression analyzed by immunohistochemistry. All statistical tests were two-sided. Results One hundred sixteen out of 222 (52.3%) PR+ tumors were PRA-H, and 64 (28.8%) PRB-H. Cell proliferation was inhibited by MFP in 19 of 19 tissue cultures from PRA-H tumors. A total of 139 transcripts related to proliferative pathways were differentially expressed in nine PRA-H and seven PRB-H tumors. PRB-H and PRA-H tumors were either luminal B or A phenotypes, respectively ( P = .03). PRB-H cases were associated with shorter relapse-free survival (hazard ratio [HR] = 2.70, 95% confidence interval [CI] = 1.71 to 6.20, P = .02) and distant metastasis-free survival (HR = 4.17, 95% CI = 2.18 to 7.97, P < .001). PRB-H tumors showed increased tumor size ( P < .001), Ki-67 levels ( P < .001), human epidermal growth factor receptor 2 expression ( P = .04), high grades ( P = .03), and decreased total PR ( P = .004) compared with PRA-H tumors. MUC-2 ( P < .001) and KRT6A ( P = .02) were also overexpressed in PRB-H tumors. Conclusion The PRA/PRB ratio is a prognostic and predictive factor for antiprogestin responsiveness in breast cancer.
Collapse
Affiliation(s)
- Paola A Rojas
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María May
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gonzalo R Sequeira
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Andrés Elia
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Michelle Alvarez
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula Martínez
- Hospital de Agudos Magdalena V de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Pedro Gonzalez
- Hospital de Agudos Magdalena V de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Stephen Hewitt
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xiaping He
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Charles M Perou
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Alfredo Molinolo
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Luz Gibbons
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Martin C Abba
- CINIBA-CONICET, Escuela de Ciencias Médicas, UNLP, La Plata, Argentina
| | - Hugo Gass
- Hospital de Agudos Magdalena V de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Claudia Lanari
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
28
|
Hosseini H, Obradović MMS, Hoffmann M, Harper KL, Sosa MS, Werner-Klein M, Nanduri LK, Werno C, Ehrl C, Maneck M, Patwary N, Haunschild G, Gužvić M, Reimelt C, Grauvogl M, Eichner N, Weber F, Hartkopf AD, Taran FA, Brucker SY, Fehm T, Rack B, Buchholz S, Spang R, Meister G, Aguirre-Ghiso JA, Klein CA. Early dissemination seeds metastasis in breast cancer. Nature 2016; 540:552-558. [PMID: 27974799 PMCID: PMC5390864 DOI: 10.1038/nature20785] [Citation(s) in RCA: 512] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 11/11/2016] [Indexed: 12/14/2022]
Abstract
Accumulating data suggest that metastatic dissemination often occurs early during tumour formation but the mechanisms of early metastatic spread have not yet been addressed. Here, we studied metastasis in a HER2-driven mouse breast cancer model and found that progesterone-induced signalling triggered migration of cancer cells from early lesions shortly after HER2 activation, but promoted proliferation in advanced primary tumour cells. The switch from migration to proliferation was regulated by elevated HER2 expression and increased tumour cell density involving miRNA-mediated progesterone receptor (PGR) down-regulation and was reversible. Cells from early, low-density lesions displayed more stemness features than cells from dense, advanced tumours, migrated more and founded more metastases. Strikingly, we found that at least 80% of metastases were derived from early disseminated cancer cells (DCC). Karyotypic and phenotypic analysis of human disseminated cancer cells and primary tumours corroborated the relevance of these findings for human metastatic dissemination.
Collapse
Affiliation(s)
- Hedayatollah Hosseini
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Milan M S Obradović
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Martin Hoffmann
- Project group 'Personalized Tumour Therapy', Fraunhofer Institute for Toxicology und Experimental Medicine, 93053 Regensburg, Germany
| | - Kathryn L Harper
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Maria Soledad Sosa
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | | | - Lahiri Kanth Nanduri
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Christian Werno
- Project group 'Personalized Tumour Therapy', Fraunhofer Institute for Toxicology und Experimental Medicine, 93053 Regensburg, Germany
| | - Carolin Ehrl
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Matthias Maneck
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Nina Patwary
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Gundula Haunschild
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Miodrag Gužvić
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Christian Reimelt
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Michael Grauvogl
- Department of Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Norbert Eichner
- Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Florian Weber
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Andreas D Hartkopf
- Department of Gynecology and Obstetrics, University of Tübingen, 72076 Tübingen, Germany
| | - Florin-Andrei Taran
- Department of Gynecology and Obstetrics, University of Tübingen, 72076 Tübingen, Germany
| | - Sara Y Brucker
- Department of Gynecology and Obstetrics, University of Tübingen, 72076 Tübingen, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Brigitte Rack
- Department of Gynecology and Obstetrics, University Munich, 80337 Munich, Germany
| | - Stefan Buchholz
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Rainer Spang
- Department of Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Gunter Meister
- Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Julio A Aguirre-Ghiso
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Christoph A Klein
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany.,Project group 'Personalized Tumour Therapy', Fraunhofer Institute for Toxicology und Experimental Medicine, 93053 Regensburg, Germany
| |
Collapse
|
29
|
Leehy KA, Regan Anderson TM, Daniel AR, Lange CA, Ostrander JH. Modifications to glucocorticoid and progesterone receptors alter cell fate in breast cancer. J Mol Endocrinol 2016; 56:R99-R114. [PMID: 26831511 PMCID: PMC7256961 DOI: 10.1530/jme-15-0322] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/16/2015] [Indexed: 12/21/2022]
Abstract
Steroid hormone receptors (SRs) are heavily posttranslationally modified by the reversible addition of a variety of molecular moieties, including phosphorylation, acetylation, methylation, SUMOylation, and ubiquitination. These rapid and dynamic modifications may be combinatorial and interact (i.e. may be sequential, complement, or oppose each other), creating a vast array of uniquely modified receptor subspecies that allow for diverse receptor behaviors that enable highly sensitive and context-dependent hormone action. For example, in response to hormone or growth factor membrane-initiated signaling events, posttranslational modifications (PTMs) to SRs alter protein-protein interactions that govern the complex process of promoter or gene-set selection coupled to transcriptional repression or activation. Unique phosphorylation events allow SRs to associate or disassociate with specific cofactors that may include pioneer factors and other tethering partners, which specify the resulting transcriptome and ultimately change cell fate. The impact of PTMs on SR action is particularly profound in the context of breast tumorigenesis, in which frequent alterations in growth factor-initiated signaling pathways occur early and act as drivers of breast cancer progression toward endocrine resistance. In this article, with primary focus on breast cancer relevance, we review the mechanisms by which PTMs, including reversible phosphorylation events, regulate the closely related SRs, glucocorticoid receptor and progesterone receptor, allowing for precise biological responses to ever-changing hormonal stimuli.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Female
- Gene Expression
- Gene Expression Regulation, Neoplastic
- Humans
- Prognosis
- Protein Isoforms
- Protein Processing, Post-Translational
- Receptors, Estrogen/metabolism
- Receptors, Glucocorticoid/chemistry
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Receptors, Progesterone/chemistry
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Signal Transduction
- Stress, Physiological
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Katherine A Leehy
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Tarah M Regan Anderson
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Andrea R Daniel
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Carol A Lange
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Julie H Ostrander
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| |
Collapse
|
30
|
Aupperlee MD, Zhao Y, Tan YS, Zhu Y, Langohr IM, Kirk EL, Pirone JR, Troester MA, Schwartz RC, Haslam SZ. Puberty-specific promotion of mammary tumorigenesis by a high animal fat diet. Breast Cancer Res 2015; 17:138. [PMID: 26526858 PMCID: PMC4630903 DOI: 10.1186/s13058-015-0646-4] [Citation(s) in RCA: 20] [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: 07/01/2015] [Accepted: 10/15/2015] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Increased animal fat consumption is associated with increased premenopausal breast cancer risk in normal weight, but not overweight, women. This agrees with our previous findings in obesity-resistant BALB/c mice, in which exposure to a high saturated animal fat diet (HFD) from peripuberty through adulthood promoted mammary tumorigenesis. Epidemiologic and animal studies support the importance of puberty as a life stage when diet and environmental exposures affect adult breast cancer risk. In this study, we identified the effects of peripubertal exposure to HFD and investigated its mechanism of enhancing tumorigenesis. METHODS Three-week-old BALB/c mice fed a low-fat diet (LFD) or HFD were subjected to 7,12-dimethylbenz[a]anthracene (DMBA)-induced carcinogenesis. At 9 weeks of age, half the mice on LFD were switched to HFD (LFD-HFD group) and half the mice on HFD were switched to LFD (HFD-LFD group). Tumor gene expression was evaluated in association with diet and tumor latency. RESULTS The peripubertal HFD reduced the latency of DMBA-induced mammary tumors and was associated with tumor characteristics similar to those in mice fed a continuous HFD. Notably, short-latency tumors in both groups shared gene expression characteristics and were more likely to have adenosquamous histology. Both HFD-LFD and continuous HFD tumors showed similar gene expression patterns and early latency. Adult switch from HFD to LFD did not reverse peripubertal HFD tumor promotion. Increased proliferation, hyperplasia, and macrophages were present in mammary glands before tumor development, implicating these as possible effectors of tumor promotion. Despite a significant interaction between pubertal diet and carcinogens in tumor promotion, peripubertal HFD by itself produced persistent macrophage recruitment to mammary glands. CONCLUSIONS In obesity-resistant mice, peripubertal HFD is sufficient to irreversibly promote carcinogen-induced tumorigenesis. Increased macrophage recruitment is likely a contributing factor. These results underscore the importance of early life exposures to increased adult cancer risk and are consistent with findings that an HFD in normal weight premenopausal women leads to increased breast cancer risk. Notably, short-latency tumors occurring after peripubertal HFD had characteristics similar to human basal-like breast cancers that predominantly develop in younger women.
Collapse
Affiliation(s)
- Mark D Aupperlee
- Breast Cancer and the Environment Research Program, Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building, Room 2201, 567 Wilson Road, East Lansing, MI, 48824, USA.
| | - Yong Zhao
- Breast Cancer and the Environment Research Program, Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building, Room 2201, 567 Wilson Road, East Lansing, MI, 48824, USA.
- Present address: College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China.
| | - Ying Siow Tan
- Breast Cancer and the Environment Research Program, Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building, Room 2201, 567 Wilson Road, East Lansing, MI, 48824, USA.
- Present address: Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| | - Yirong Zhu
- Breast Cancer and the Environment Research Program, Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building, Room 2201, 567 Wilson Road, East Lansing, MI, 48824, USA.
| | - Ingeborg M Langohr
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA.
- Present address: Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.
| | - Erin L Kirk
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jason R Pirone
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Melissa A Troester
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Richard C Schwartz
- Breast Cancer and the Environment Research Program, Department of Microbiology and Molecular Genetics, Michigan State University, Biomedical and Physical Sciences Building, Room 2201, 567 Wilson Road, East Lansing, MI, 48824, USA.
| | - Sandra Z Haslam
- Breast Cancer and the Environment Research Program, Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building, Room 2201, 567 Wilson Road, East Lansing, MI, 48824, USA.
| |
Collapse
|
31
|
Campo Verde Arboccó F, Sasso CV, Nasif DL, Hapon MB, Jahn GA. Effect of hypothyroidism on the expression of nuclear receptors and their co-regulators in mammary gland during lactation in the rat. Mol Cell Endocrinol 2015; 412:26-35. [PMID: 26027918 DOI: 10.1016/j.mce.2015.05.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/14/2015] [Accepted: 05/05/2015] [Indexed: 02/07/2023]
Abstract
Thyroid hormones (TH) regulate mammary function. Hypothyroidism (HypoT) has deleterious effects on lactation, litter growth and survival. We analyzed the effect of chronic 6-propyl-2-thiouracil (PTU)-induced HypoT in the expression of nuclear receptors, co-regulators and oxytocin receptor (OTR) on lactation (L) days 2, 7 and 14. TH receptors (TRs) were increased on L7 at mRNA and protein levels, except TRα protein, that fell on L14. HypoT decreased TRα2 mRNA on L7 and TRα1 protein on L2, while TRβ1 protein increased on L14. HypoT increased estrogen receptor β (ERβ) mRNA on L7 but decreased its protein levels on L14. Progesterone receptor A (PRA) mRNA decreased from L2 to L14 while PRB increased, and at protein levels PRA levels showed a nadir on L7, while PRB peaked. HypoT decreased PRA mRNA and protein and increased PRB mRNA at L14. Nuclear receptor co-activator (NCOA) 1 and RXRα mRNA showed an opposite pattern to the TRs, while NCOA2 increased at L14; HypoT blocked the variations in NCOA1 and NCOA2. HypoT increased NCOR1 on L2 and decreased OTR at L2 and circulating estradiol and NCOR2 at L14. In controls the most notable changes occurred on L7, suggesting it is a key inflection point in mammary metabolism. The low levels of TRα1, NCOA1 and OTR, and increased NCOR1 produced by HypoT on L2 may hinder the mammary ability to achieve normal milk synthesis and ejection, leading to defective lactation. Later on, altered ER and PR expression may impair further mammary function.
Collapse
MESH Headings
- Animals
- Female
- Gene Expression
- Hypothyroidism/chemically induced
- Hypothyroidism/metabolism
- Lactation
- Mammary Glands, Animal/metabolism
- Nuclear Receptor Co-Repressor 1/genetics
- Nuclear Receptor Co-Repressor 1/metabolism
- Nuclear Receptor Coactivator 1/genetics
- Nuclear Receptor Coactivator 1/metabolism
- Nuclear Receptor Coactivator 2/genetics
- Nuclear Receptor Coactivator 2/metabolism
- Propylthiouracil
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Rats, Wistar
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, Oxytocin/genetics
- Receptors, Oxytocin/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- Retinoid X Receptor alpha/genetics
- Retinoid X Receptor alpha/metabolism
Collapse
Affiliation(s)
- Fiorella Campo Verde Arboccó
- Laboratorio de Reproducción y Lactancia, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, 5500 Mendoza, Argentina
| | - Corina V Sasso
- Laboratorio de Hormonas y Biología del Cáncer, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Mendoza, Argentina
| | - Daniela L Nasif
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Belén Hapon
- Laboratorio de Reproducción y Lactancia, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, 5500 Mendoza, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Graciela A Jahn
- Laboratorio de Reproducción y Lactancia, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, 5500 Mendoza, Argentina.
| |
Collapse
|
32
|
Hilton HN, Graham JD, Clarke CL. Minireview: Progesterone Regulation of Proliferation in the Normal Human Breast and in Breast Cancer: A Tale of Two Scenarios? Mol Endocrinol 2015; 29:1230-42. [PMID: 26266959 DOI: 10.1210/me.2015-1152] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Progesterone (P), which signals through the P receptor (PR), is critical in normal development of the breast, but its signaling axis is also a major driver of breast cancer risk. Here we review recent advances in the understanding of P signaling in the normal human breast, with a focus on the importance of the balance between autocrine and paracrine signaling. To date, most data (which derive largely from mouse models or human breast cancer cell line studies) have demonstrated that the vast majority of PR+ cells appear to act as "sensor" cells, which respond to P stimulation by translating these hormonal cues into paracrine signals. However, growing evidence suggests that, dependent on the cellular context, P may also signal in an autocrine manner in a subset of cells in the normal mouse mammary gland and human breast. It has been suggested that it may be dysregulation of this autocrine signaling, resulting in a "switch" from a predominance of paracrine signaling to autocrine signaling in PR+ cells, which is an early event during breast tumorigenesis. This review summarizes current evidence in the literature that demonstrates the mechanisms through which P acts in the normal human breast, as well as highlighting the important questions that remain unanswered.
Collapse
Affiliation(s)
- Heidi N Hilton
- Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| | - J Dinny Graham
- Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| | - Christine L Clarke
- Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| |
Collapse
|
33
|
Tornillo G, Smalley MJ. ERrrr…where are the progenitors? Hormone receptors and mammary cell heterogeneity. J Mammary Gland Biol Neoplasia 2015; 20:63-73. [PMID: 26193872 PMCID: PMC4595529 DOI: 10.1007/s10911-015-9336-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 07/08/2015] [Indexed: 02/07/2023] Open
Abstract
The mammary epithelium is a highly heterogenous and dynamic tissue that includes a range of cell types with varying levels of proliferative capacity and differentiation potential, from stem to committed progenitor and mature cells. Generation of mature cells through expansion and specification of immature precursors is driven by hormonal and local stimuli. Intriguingly, although circulating hormones can be directly sensed only by a subset of mammary cells, they also regulate the behaviour of cells lacking their cognate receptors through paracrine mechanisms. Thus, mapping the hormonal signalling network on to the emerging mammary cell hierarchy appears to be a difficult task. Nevertheless, a first step towards a better understanding is the characterization of the hormone receptor expression pattern across individual cell types in the mammary epithelium. Here we review the most relevant findings on the cellular distribution of hormone receptors in the mammary gland, taking into account differences between mice and humans, the methods employed to assess receptor expression as well as the variety of approaches used to resolve the mammary cell heterogeneity.
Collapse
Affiliation(s)
- Giusy Tornillo
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff, CF24 4HQ, UK.
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff, CF24 4HQ, UK
| |
Collapse
|
34
|
Brisken C, Ataca D. Endocrine hormones and local signals during the development of the mouse mammary gland. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:181-95. [PMID: 25645332 DOI: 10.1002/wdev.172] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/02/2014] [Accepted: 12/08/2014] [Indexed: 01/03/2023]
Abstract
Most of mammary gland development occurs postnatally under the control of female reproductive hormones, which in turn interact with other endocrine factors. While hormones impinge on many tissues and trigger very complex biological responses, tissue recombination experiments with hormone receptor-deficient mammary epithelia revealed eminent roles for estrogens, progesterone, and prolactin receptor (PrlR) signaling that are intrinsic to the mammary epithelium. A subset of the luminal mammary epithelial cells expresses the estrogen receptor α (ERα), the progesterone receptor (PR), and the PrlR and act as sensor cells. These cells convert the detected systemic signals into local signals that are developmental stage-dependent and may be direct, juxtacrine, or paracrine. This setup ensures that the original input is amplified and that the biological responses of multiple cell types can be coordinated. Some key mediators of hormone action have been identified such as Wnt, EGFR, IGFR, and RANK signaling. Multiple signaling pathways such as FGF, Hedgehog, and Notch signaling participate in driving different aspects of mammary gland development locally but how they link to the hormonal control remains to be elucidated. An increasing number of endocrine factors are appearing to have a role in mammary gland development, the adipose tissue is increasingly recognized to play a role in endocrine regulation, and a complex role of the immune system with multiple different cell types is being revealed. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Cathrin Brisken
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | |
Collapse
|
35
|
Wargon V, Riggio M, Giulianelli S, Sequeira GR, Rojas P, May M, Polo ML, Gorostiaga MA, Jacobsen B, Molinolo A, Novaro V, Lanari C. Progestin and antiprogestin responsiveness in breast cancer is driven by the PRA/PRB ratio via AIB1 or SMRT recruitment to the CCND1 and MYC promoters. Int J Cancer 2014; 136:2680-92. [PMID: 25363551 DOI: 10.1002/ijc.29304] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 10/16/2014] [Indexed: 01/01/2023]
Abstract
There is emerging interest in understanding the role of progesterone receptors (PRs) in breast cancer. The aim of this study was to investigate the proliferative effect of progestins and antiprogestins depending on the relative expression of the A (PRA) and B (PRB) isoforms of PR. In mifepristone (MFP)-resistant murine carcinomas antiprogestin responsiveness was restored by re-expressing PRA using demethylating agents and histone deacetylase inhibitors. Consistently, in two human breast cancer xenograft models, one manipulated to overexpress PRA or PRB (IBH-6 cells), and the other expressing only PRA (T47D-YA) or PRB (T47D-YB), MFP selectively inhibited the growth of PRA-overexpressing tumors and stimulated IBH-6-PRB xenograft growth. Furthermore, in cells with high or equimolar PRA/PRB ratios, which are stimulated to proliferate in vitro by progestins, and are inhibited by MFP, MPA increased the interaction between PR and the coactivator AIB1, and MFP favored the interaction between PR and the corepressor SMRT. In a PRB-dominant context in which MFP stimulates and MPA inhibits cell proliferation, the opposite interactions were observed. Chromatin immunoprecipitation assays in T47D cells in the presence of MPA or MFP confirmed the interactions between PR and the coregulators at the CCND1 and MYC promoters. SMRT downregulation by siRNA abolished the inhibitory effect of MFP on MYC expression and cell proliferation. Our results indicate that antiprogestins are therapeutic tools that selectively inhibit PRA-overexpressing tumors by increasing the SMRT/AIB1 balance at the CCND1 and MYC promoters.
Collapse
Affiliation(s)
- Victoria Wargon
- Laboratory of Hormonal Carcinogenesis, Institute of Experimental Biology and Medicine (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhao Y, Tan YS, Aupperlee MD, Langohr IM, Kirk EL, Troester MA, Schwartz RC, Haslam SZ. Pubertal high fat diet: effects on mammary cancer development. Breast Cancer Res 2014; 15:R100. [PMID: 24156623 PMCID: PMC3978633 DOI: 10.1186/bcr3561] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 10/11/2013] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Epidemiological studies linking dietary fat intake and obesity to breast cancer risk have produced inconsistent results. This may be due to the difficulty of dissociating fat intake from obesity, and/or the lack of defined periods of exposure in these studies. The pubertal mammary gland is highly sensitive to cancer-causing agents. We assessed how high fat diet (HFD) affects inflammation, proliferative, and developmental events in the pubertal gland, since dysregulation of these can promote mammary tumorigenesis. To test the effect of HFD initiated during puberty on tumorigenesis, we utilized BALB/c mice, for which HFD neither induces obesity nor metabolic syndrome, allowing dissociation of HFD effects from other conditions associated with HFD. METHODS Pubertal BALB/c mice were fed a low fat diet (12% kcal fat) or a HFD (60% kcal fat), and subjected to carcinogen 7,12-dimethylbenz[a]anthracene (DMBA)-induced tumorigenesis. RESULTS HFD elevated mammary gland expression of inflammatory and growth factor genes at 3 and 4 weeks of diet. Receptor activator of nuclear factor kappa-B ligand (RANKL), robustly induced at 4 weeks, has direct mitogenic activity in mammary epithelial cells and, as a potent inducer of NF-κB activity, may induce inflammatory genes. Three weeks of HFD induced a transient influx of eosinophils into the mammary gland, consistent with elevated inflammatory factors. At 10 weeks, prior to the appearance of palpable tumors, there were increased numbers of abnormal mammary epithelial lesions, enhanced cellular proliferation, increased growth factors, chemokines associated with immune-suppressive regulatory T cells, increased vascularization, and elevated M2 macrophages. HFD dramatically reduced tumor latency. Early developing tumors were more proliferative and were associated with increased levels of tumor-related growth factors, including increased plasma levels of HGF in tumor-bearing animals. Early HFD tumors also had increased vascularization, and more intra-tumor and stromal M2 macrophages. CONCLUSIONS Taken together in this non-obesogenic context, HFD promotion of inflammatory processes, as well as local and systemically increased growth factor expression, are likely responsible for the enhanced tumorigenesis. It is noteworthy that although DMBA mutagenesis is virtually random in its targeting of genes in tumorigenesis, the short latency tumors arising in animals on HFD showed a unique gene expression profile, highlighting the potent overarching influence of HFD.
Collapse
|
37
|
Aupperlee MD, Zhao Y, Tan YS, Leipprandt JR, Bennett J, Haslam SZ, Schwartz RC. Epidermal growth factor receptor (EGFR) signaling is a key mediator of hormone-induced leukocyte infiltration in the pubertal female mammary gland. Endocrinology 2014; 155:2301-13. [PMID: 24693965 PMCID: PMC4020926 DOI: 10.1210/en.2013-1933] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well documented that macrophages and eosinophils play important roles in normal murine pubertal mammary gland development. Although it is accepted that estrogen (E) and progesterone (P) are key players in mammary gland development, the roles these hormones might play in regulating the actions of leukocytes in that process is an understudied area. We show here that P and E, respectively, induce unique, but overlapping, sets of proinflammatory and angiogenic cytokines and chemokines, in the pubertal female BALB/c mammary gland, as well as induce infiltration of macrophages and eosinophils to the mammary periepithelium. This extends earlier studies showing P induction of proinflammatory products in pubertal and adult mammary epithelial organoids and P-induced in vivo infiltration of leukocytes to the adult mammary periepithelium. Importantly, epidermal growth factor receptor-signaling, which is likely mediated by amphiregulin (Areg), a downstream mediator of E and P, is both necessary and sufficient for both E- and P-induced recruitment of macrophages and eosinophils to the pubertal mammary periepithelium. We further show that receptor activator of nuclear factor κB ligand (RANKL), although not sufficient of itself to cause macrophage and eosinophil recruitment, contributes to an optimal response to P. The potency of Areg is highlighted by the fact that it is sufficient to induce macrophage and eosinophil recruitment at levels equivalent to that induced by either E or P. Our finding of a dominant role for Areg in hormonally induced leukocyte recruitment to the pubertal mammary gland parallels its dominance in regulating ductal outgrowth and its role in P-induced proliferation in the pubertal gland.
Collapse
Affiliation(s)
- Mark D Aupperlee
- Breast Cancer and the Environment Research Program, Departments of Physiology (M.D.A., Y.Z., Y.S.T., J.R.L., J.B., S.Z.H.) and Microbiology and Molecular Genetics (R.C.S.), Michigan State University, East Lansing, Michigan 48824
| | | | | | | | | | | | | |
Collapse
|
38
|
Knutson TP, Lange CA. Tracking progesterone receptor-mediated actions in breast cancer. Pharmacol Ther 2014; 142:114-25. [PMID: 24291072 PMCID: PMC3943696 DOI: 10.1016/j.pharmthera.2013.11.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 11/15/2013] [Indexed: 12/13/2022]
Abstract
Ovarian steroid hormones contribute to breast cancer initiation and progression primarily through the actions of their nuclear transcription factors, the estrogen receptor alpha (ERα) and progesterone receptors (PRs). These receptors are important drivers of the luminal A and B subtypes of breast cancer, where estrogen-blocking drugs have been effective endocrine therapies for patients with these tumors. However, many patients do not respond, or become resistant to treatment. When endocrine therapies fail, the luminal subtypes of breast cancer are more difficult to treat because these subtypes are among the most heterogeneous in terms of mutation diversity and gene expression profiles. Recent evidence suggests that progestin and PR actions may be important drivers of luminal breast cancers. Clinical trial data has demonstrated that hormone replacement therapy with progestins drives invasive breast cancer and results in greater mortality. PR transcriptional activity is dependent upon cross-talk with growth factor signaling pathways that alter PR phosphorylation, acetylation, or SUMOylation as mechanisms for regulating PR target gene selection required for increased cell proliferation and survival. Site-specific PR phosphorylation is the primary driver of gene-selective PR transcriptional activity. However, PR phosphorylation and heightened transcriptional activity is coupled to rapid PR protein degradation; the range of active PR detected in tumors is likely to be dynamic. Thus, PR target gene signatures may provide a more accurate means of tracking PR's contribution to tumor progression rather than standard clinical protein-based (IHC) assays. Further development of antiprogestin therapies should be considered alongside antiestrogens and aromatase inhibitors.
Collapse
Affiliation(s)
- Todd P Knutson
- Departments of Medicine, Division of Hematology, Oncology, and Transplantation and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carol A Lange
- Departments of Medicine, Division of Hematology, Oncology, and Transplantation and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
39
|
Kariagina A, Xie J, Langohr IM, Opreanu RC, Basson MD, Haslam SZ. Progesterone stimulates proliferation and promotes cytoplasmic localization of the cell cycle inhibitor p27 in steroid receptor positive breast cancers. HORMONES & CANCER 2013; 4:381-390. [PMID: 23996077 PMCID: PMC10358121 DOI: 10.1007/s12672-013-0159-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/19/2013] [Indexed: 11/24/2022]
Abstract
Progestins are reported to increase the risk of more aggressive estrogen receptor positive, progesterone receptor positive (ER+ PR+) breast cancers in postmenopausal women. Using an in vivo rat model of ER+ PR + mammary cancer, we show that tumors arising in the presence of estrogen and progesterone exhibit increased proliferation and decreased nuclear expression of the cell cycle inhibitor p27 compared with tumors growing in the presence of estrogen alone. In human T47D breast cancer cells, progestin increased proliferation and decreased nuclear p27 expression. The decrease of nuclear p27 protein was dependent on activation of Src and PI3K by progesterone receptor isoforms PRA or PRB. Importantly, increased proliferation and decreased nuclear p27 expression were observed in invasive breast carcinoma compared with carcinoma in situ. These results suggest that progesterone specifically regulates intracellular localization of p27 protein and proliferation. Therefore, progesterone-activated pathways can provide useful therapeutic targets for treatment of more aggressive ER+ PR+ breast cancers.
Collapse
Affiliation(s)
- Anastasia Kariagina
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA,
| | | | | | | | | | | |
Collapse
|
40
|
Obr AE, Grimm SL, Bishop KA, Pike JW, Lydon JP, Edwards DP. Progesterone receptor and Stat5 signaling cross talk through RANKL in mammary epithelial cells. Mol Endocrinol 2013; 27:1808-24. [PMID: 24014651 PMCID: PMC3805851 DOI: 10.1210/me.2013-1077] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 08/21/2013] [Indexed: 02/08/2023] Open
Abstract
Progesterone (P4) stimulates proliferation of the mammary epithelium by a mechanism that involves paracrine signaling mediated from progesterone receptor (PR)-positive to neighboring PR-negative cells. Here we used a primary mouse mammary epithelial cell (MEC) culture system to define the molecular mechanism by which P4 regulates the expression of target gene effectors of proliferation including the paracrine factor receptor and activator of nuclear factor κB ligand (RANKL). MECs from adult virgin mice grown and embedded in three-dimensional basement-membrane medium resemble mammary ducts in vivo structurally and with respect to other properties including a heterogeneous pattern of PR expression, P4 induction of RANKL and other target genes in a PR-dependent manner, and a proliferative response to progestin. RANKL was demonstrated to have multiple functional P4-responsive enhancers that bind PR in a hormone-dependent manner as detected by chromatin immunoprecipitation assay. P4 also stimulated recruitment of signal transducer and activator of transcription (Stat)5a to RANKL enhancers through an apparent tethering with PR. Analysis of primary MECs from Stat5a knockout mice revealed that P4 induction of RANKL and a broad range of other PR target genes required Stat5a, as did P4-stimulated cell proliferation. In the absence of Stat5a, PR binding was lost at selective RANKL enhancers but was retained with others, suggesting that Stat5a acts to facilitate PR DNA binding at selective sites and to function as a coactivator with DNA-bound PR at others. These results show that RANKL is a direct PR target gene and that Stat5a has a novel role as a cofactor in PR-mediated transcriptional signaling in the mammary gland.
Collapse
Affiliation(s)
- Alison E Obr
- PhD, Department of Molecular & Cellular Biology, Baylor College of Medicine, BCM Box 130, One Baylor Plaza, Houston, Texas 77030.
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Many organs respond to physiological challenges by changing tissue size or composition. Such changes may originate from tissue-specific stem cells and their supportive environment (niche). The endocrine system is a major effector and conveyor of physiological changes and as such could alter stem cell behavior in various ways. In this review, we examine how hormones affect stem cell biology in four different organs: the ovary, intestine, hematopoietic system, and mammary gland. Hormones control every stage of stem cell life, including establishment, expansion, maintenance, and differentiation. The effects can be cell autonomous or non-cell autonomous through the niche. Moreover, a single hormone can affect different stem cells in different ways or affect the same stem cell differently at various developmental times. The vast complexity and diversity of stem cell responses to hormonal cues allow hormones to coordinate the body's reaction to physiological challenges.
Collapse
Affiliation(s)
- Dana Gancz
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100 Israel; ,
| | | |
Collapse
|
42
|
Abstract
Understanding the biology of the breast and how ovarian hormones impinge on it is key to rational new approaches in breast cancer prevention and therapy. Because of the success of selective oestrogen receptor modulators (SERMs), such as tamoxifen, and aromatase inhibitors in breast cancer treatment, oestrogens have long received the most attention. Early progesterone receptor (PR) antagonists, however, were dismissed because of severe side effects, but awareness is now increasing that progesterone is an important hormone in breast cancer. Oestrogen receptor-α (ERα) signalling and PR signalling have distinct roles in normal mammary gland biology in mice; both ERα and PR delegate many of their biological functions to distinct paracrine mediators. If the findings in the mouse model translate to humans, new preventive and therapeutic perspectives might open up.
Collapse
Affiliation(s)
- Cathrin Brisken
- ISREC - Swiss Institute for Experimental Cancer Research, National Center of Competence for Molecular Oncology, School of Life Sciences, Ecole polytechnique fédérale de Lausanne (EPFL), SV2.832 Station 19, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
43
|
Aupperlee MD, Leipprandt JR, Bennett JM, Schwartz RC, Haslam SZ. Amphiregulin mediates progesterone-induced mammary ductal development during puberty. Breast Cancer Res 2013; 15:R44. [PMID: 23705924 PMCID: PMC3738150 DOI: 10.1186/bcr3431] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 05/25/2013] [Indexed: 01/11/2023] Open
Abstract
Introduction Puberty is a period of increased susceptibility to factors that cause increased
breast cancer risk in adulthood. Mammary end buds (EBs) that develop during
puberty are believed to be the targets of breast cancer initiation. Whereas the
role of estrogen (E) has been extensively studied in pubertal mammary gland
development, the role of progesterone (P) during puberty is less defined. Methods Pubertal and prepubertal ovariectomized mice were treated with vehicle control
(C), E, P, or E+P. Mammary glands from these mice were analyzed for changes in
morphology, proliferation, and expression of the downstream targets amphiregulin
(AREG) and receptor activator of NF-κB ligand (RANKL). Results P, acting specifically through the progesterone receptor, induced increases in
mammary gland proliferation and EB formation that were associated with increased
AREG expression in ducts and EBs. E, acting specifically through the estrogen
receptor, produced similar responses also mediated by AREG. Blocking AREG action
by treatment with an EGFR inhibitor completely abrogated the effect of P on EB
formation and proliferation and significantly reduced proliferation within ducts.
P also increased expression of RANKL, primarily in ducts. Treatment with RANK-Fc,
an inhibitor of RANKL, reduced P-dependent proliferation in ducts and to a lesser
extent in EB, but did not cause EB regression. Conclusions These results demonstrate a novel P-specific effect through AREG to cause EB
formation and proliferation in the developing mammary gland both before and during
puberty. Thus, hormones and/or factors in addition to E that upregulate AREG can
promote mammary gland development and have the potential to affect breast cancer
risk associated with pubertal mammary gland development.
Collapse
|
44
|
Kim JJ, Kurita T, Bulun SE. Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr Rev 2013; 34:130-62. [PMID: 23303565 PMCID: PMC3565104 DOI: 10.1210/er.2012-1043] [Citation(s) in RCA: 350] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/17/2012] [Indexed: 12/19/2022]
Abstract
Progesterone receptor (PR) mediates the actions of the ovarian steroid progesterone, which together with estradiol regulates gonadotropin secretion, prepares the endometrium for implantation, maintains pregnancy, and differentiates breast tissue. Separation of estrogen and progesterone actions in hormone-responsive tissues remains a challenge. Pathologies of the uterus and breast, including endometrial cancer, endometriosis, uterine fibroids, and breast cancer, are highly associated with estrogen, considered to be the mitogenic factor. Emerging evidence supports distinct roles of progesterone and its influence on the pathogenesis of these diseases. Progesterone antagonizes estrogen-driven growth in the endometrium, and insufficient progesterone action strikingly increases the risk of endometrial cancer. In endometriosis, eutopic and ectopic tissues do not respond sufficiently to progesterone and are considered to be progesterone-resistant, which contributes to proliferation and survival. In uterine fibroids, progesterone promotes growth by increasing proliferation, cellular hypertrophy, and deposition of extracellular matrix. In normal mammary tissue and breast cancer, progesterone is pro-proliferative and carcinogenic. A key difference between these tissues that could explain the diverse effects of progesterone is the paracrine interactions of PR-expressing stroma and epithelium. Normal endometrium is a mucosa containing large quantities of distinct stromal cells with abundant PR, which influences epithelial cell proliferation and differentiation and protects against carcinogenic transformation. In contrast, the primary target cells of progesterone in the breast and fibroids are the mammary epithelial cells and the leiomyoma cells, which lack specifically organized stromal components with significant PR expression. This review provides a unifying perspective for the diverse effects of progesterone across human tissues and diseases.
Collapse
Affiliation(s)
- J Julie Kim
- Division of Reproductive Biology Research, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | |
Collapse
|
45
|
Sampayo R, Recouvreux S, Simian M. The hyperplastic phenotype in PR-A and PR-B transgenic mice: lessons on the role of estrogen and progesterone receptors in the mouse mammary gland and breast cancer. VITAMINS AND HORMONES 2013; 93:185-201. [PMID: 23810007 DOI: 10.1016/b978-0-12-416673-8.00012-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Progesterone receptor (PR) belongs to the superfamily of steroid receptors and mediates the action of progesterone in its target tissues. In the mammary gland, in particular, PR expression is restricted to the luminal epithelial cell compartment. The generation of estrogen receptor-α (ER) and PR knockout mice allowed the specific characterization of the roles of each of these in mammary gland development: ER is critical for ductal morphogenesis, whereas PR has a key role in lobuloalveolar differentiation. To further study the role PR isoforms have in mammary gland biology, transgenic mice overexpressing either the "A" (PR-A) or the "B" (PR-B) isoforms of PR were generated. Overexpression of the A isoform of PR led to increased side branching, multilayered ducts, loss of basement membrane integrity, and alterations in matrix metalloproteinase activation in the mammary gland. Moreover, levels of TGFβ1 and p21 were diminished and those of cyclin D1 increased. Interestingly, the phenotype was counteracted by antiestrogens, suggesting that ER is essential for the manifestation of the hyperplasias. Mice overexpressing the B isoform of PR had limited ductal growth but retained the ability to differentiate during pregnancy. Levels of latent and active TGFβ1 were increased compared to PR-A transgenics. The phenotypes of these transgenic mice are further discussed in the context of the impact of progesterone on mammary stem cells and breast cancer. We conclude that an adequate balance between the A and B isoforms of PR is critical for tissue homeostasis. Future work to further understand the biology of PR in breast biology will hopefully lead to new and effective preventive and therapeutic alternatives for patients.
Collapse
Affiliation(s)
- Rocio Sampayo
- Área Investigación, Instituto de Oncología "Angel H. Roffo", Avda. San Martin 5481, Buenos Aires, Argentina
| | | | | |
Collapse
|
46
|
Meyer G, Leipprandt J, Xie J, Aupperlee MD, Haslam SZ. A potential role of progestin-induced laminin-5/α6-integrin signaling in the formation of side branches in the mammary gland. Endocrinology 2012; 153:4990-5001. [PMID: 22910029 PMCID: PMC3512027 DOI: 10.1210/en.2012-1518] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/31/2012] [Indexed: 11/19/2022]
Abstract
Mammary organoids from adult mice produce tubules, analogous to mammary ducts in vivo, in response to hepatocyte growth factor (HGF) when cultured in collagen gels. The combination of HGF plus progestin (R5020) causes reduced tubule number and length. We hypothesized that the inhibitory effect on tubulogenesis was due to progestin-mediated alteration of HGF/c-Met signaling. Using molecular inhibitors and short hairpin RNA, it was determined that HGF activation of Ras-related C3 botulinum toxin substrate (Rac1) was required for the formation of cytoplasmic extensions, the first step of tubulogenesis, and that Rac1 activity was Src kinase (Src) and focal adhesion kinase (FAK) dependent. The highly novel finding was that R5020 reduced tubulogenesis by up-regulating and increasing extracellular laminin and α6-integrin ligation to reduce activation of the Src, focal adhesion kinase, and Rac1 pathway. Receptor activator of nuclear factor-κB ligand, another progesterone-induced paracrine factor, did not replicate this effect of R5020. The inhibitory effect of R5020 on tubulogenesis was likely mediated through progesterone receptor (PR) isoform A (PRA), because PRA is the predominant PR isoform expressed in the organoids, and the progestin-induced effect was prevented by the PR antagonist RU486. These results provide a plausible mechanism that explains progestin/PRA-mediated blunting of HGF-induced tubulogenesis in vitro and is proposed to be relevant to progesterone/PRA-induced side-branching in vivo during pregnancy.
Collapse
Affiliation(s)
- Gabriele Meyer
- Department of Physiology and Breast Cancer and the Environment Research Center, Michigan State University, East Lansing, Michigan 48824, USA
| | | | | | | | | |
Collapse
|
47
|
Obr A, Edwards DP. The biology of progesterone receptor in the normal mammary gland and in breast cancer. Mol Cell Endocrinol 2012; 357:4-17. [PMID: 22193050 PMCID: PMC3318965 DOI: 10.1016/j.mce.2011.10.030] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 09/23/2011] [Accepted: 10/26/2011] [Indexed: 11/21/2022]
Abstract
This paper reviews work on progesterone and the progesterone receptor (PR) in the mouse mammary gland that has been used extensively as an experimental model. Studies have led to the concept that progesterone controls proliferation and morphogenesis of the luminal epithelium in a tightly orchestrated manner at distinct stages of development by paracrine signaling pathways, including receptor activator of nuclear factor κB ligand (RANKL) as a major paracrine factor. Progesterone also drives expansion of stem cells by paracrine signals to generate progenitors required for alveologenesis. During mid-to-late pregnancy, progesterone has another role to suppress secretory activation until parturition mediated in part by crosstalk between PR and prolactin/Stat5 signaling to inhibit induction of milk protein gene expression, and by inhibiting tight junction closure. In models of hormone-dependent mouse mammary tumors, the progesterone/PR signaling axis enhances pre-neoplastic progression by a switch from a paracrine to an autocrine mode of proliferation and dysregulation of the RANKL signaling pathway. Limited experiments with normal human breast show that progesterone/PR signaling also stimulates epithelial cell proliferation by a paracrine mechanism; however, the signaling pathways and whether RANKL is a major mediator remains unknown. Work with human breast cancer cell lines, patient tumor samples and clinical studies indicates that progesterone is a risk factor for breast cancer and that alteration in progesterone/PR signaling pathways contributes to early stage human breast cancer progression. However, loss of PR expression in primary tumors is associated with a less differentiated more invasive phenotype and worse prognosis, suggesting that PR may limit later stages of tumor progression.
Collapse
Affiliation(s)
- Alison Obr
- Departments of Molecular & Cellular Biology and Pathology and Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Dean P. Edwards
- Departments of Molecular & Cellular Biology and Pathology and Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA
| |
Collapse
|
48
|
Jacobsen BM, Horwitz KB. Progesterone receptors, their isoforms and progesterone regulated transcription. Mol Cell Endocrinol 2012; 357:18-29. [PMID: 21952082 PMCID: PMC3272316 DOI: 10.1016/j.mce.2011.09.016] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 08/11/2011] [Accepted: 09/11/2011] [Indexed: 01/16/2023]
Abstract
This review discusses mechanisms by which progesterone receptors (PR) regulate transcription. We examine available data in different species and tissues regarding: (1) regulation of PR levels; and (2) expression profiling of progestin-regulated genes by total PRs, or their PRA and PRB isoforms. (3) We address current views about the composition of progesterone response elements, and postulate that PR monomers acting through "half-site" elements are common, entailing cooperativity with neighboring DNA-bound transcription factors. (4) We summarize transcription data for multiple progestin-regulated promoters as directed by total PR, or PRA vs. PRB. We conclude that current models and methods used to study PR function are problematical, and recommend that future work employ cells and receptors appropriate to the species, focusing on analyses of the effects of endogenous receptors targeting endogenous genes in native chromatin.
Collapse
Affiliation(s)
- Britta M Jacobsen
- Department of Medicine/Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States.
| | | |
Collapse
|
49
|
Joshi PA, Di Grappa MA, Khokha R. Active allies: hormones, stem cells and the niche in adult mammopoiesis. Trends Endocrinol Metab 2012; 23:299-309. [PMID: 22613704 DOI: 10.1016/j.tem.2012.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 01/17/2023]
Abstract
Adult stem cells are recruited in response to specific physiological demands to regenerate, repair or maintain essential cellular components of tissues, while preserving self-renewal capacity. Signals that activate adult stem cells are not simply cell autonomous and stem cells are part of a larger dynamic framework, the stem cell 'niche', which integrates systemic and local cues to sustain stem cell functionality. The mammary stem cell niche responds readily to hormonal stimuli, generating pertinent signals that activate stem cells, culminating in stem cell expansion and tissue growth. We review here current knowledge of the mammary stem cell niche with attention to the potent stimulation rendered by ovarian hormones, relevant cellular and molecular players, and the implication of a deregulated niche, for breast cancer risk.
Collapse
Affiliation(s)
- Purna A Joshi
- Ontario Cancer Insitute, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | | | | |
Collapse
|
50
|
López-Fontana CM, Maselli ME, Salicioni AM, Carón RW. The inhibitory effect of progesterone on lactogenesis during pregnancy is already evident by mid- to late gestation in rodents. Reprod Fertil Dev 2012; 24:704-14. [DOI: 10.1071/rd11160] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 10/18/2011] [Indexed: 11/23/2022] Open
Abstract
Lactogenesis is a very complex process highly dependent on hormonal regulation. In the present study the time-course of the inhibitory actions of progesterone on prolactin secretion, mammary gland morphology and lactogenesis from mid- to late gestation in rodents was investigated. Groups of pregnant rats were luteectomised or administered with mifepristone on Day 10, 13, 15 or 17 of gestation and decapitated 28 or 48 h later. Whole-blood samples and the inguinal mammary glands were taken for determinations of hormone levels and for measurement of mammary content of casein and lactose and for tissue morphology analyses, respectively. Luteectomy or mifepristone evoked prolactin increases only after Day 17 of gestation. Mammary content of casein was increased by both treatments regardless of timing or duration. Mifepristone was less effective than luteectomy in inducing lactose production and the effect was only observed after Day 15 of gestation. Analysis of mammary gland morphology confirmed the observed effect of progesterone on lactogenesis. Both treatments triggered remarkable secretory activity in the mammary gland, even without a parallel epithelial proliferation, demonstrating that the mammary epithelium is able to synthesise milk compounds long before its full lobulo–alveolar development is achieved, provided that progesterone action is abolished. Thus, the present study demonstrates that progesterone is a potent hormonal switch for the prolactin and prolactin-like effects on mammary gland development and its milk-synthesising capacity during pregnancy, and that its inhibitory action is already evident by mid-pregnancy in rodents.
Collapse
|