1
|
Ramadan WS, Alseksek RK, Mouffak S, Talaat IM, Saber-Ayad MM, Menon V, Ilce BY, El-Awady R. Impact of HDAC6-mediated progesterone receptor expression on the response of breast cancer cells to hormonal therapy. Eur J Pharmacol 2024; 983:177001. [PMID: 39284403 DOI: 10.1016/j.ejphar.2024.177001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Modulation of estrogen receptor (ER) and progesterone receptor (PR) expression, as well as their emerging functional crosstalk, remains a potential approach for enhancing the response to hormonal therapy in breast cancer. Aberrant epigenetic alterations induced by histone deacetylases (HDACs) were massively implicated in dysregulating the function of hormone receptors in breast cancer. Although much is known about the regulation of ER signaling by HDAC, the precise role of HDAC in modulating the expression of PR and its impact on the outcomes of hormonal therapy is poorly defined. Here, we demonstrate the involvement of HDAC6 in regulating PR expression in breast cancer cells. The correlation between HDAC6 and hormone receptors was investigated in patients' tissues by immunohistochemistry (n = 80) and publicly available data (n = 3260) from breast cancer patients. We explored the effect of modulating the expression of HDAC6 as well as its catalytic inhibition on the level of hormone receptors by a variety of molecular analyses, including Western blot, immunofluorescence, Real-time PCR, RNA-seq analysis and chromatin immunoprecipitation. Based on our in-silico and immunohistochemistry analyses, HDAC6 levels were negatively correlated with PR status in breast cancer tissues. The downregulation of HDAC6 enhanced the expression of PR-B in hormone receptor-positive and triple-negative breast cancer (TNBC) cells. The selective targeting of HDAC6 by tubacin resulted in the enrichment of the H3K9 acetylation mark at the PGR-B gene promoter region and enhanced the expression of PR-B. Additionally, transcriptomic analysis of tubacin-treated cells revealed enhanced activity of acetyltransferase and growth factor signaling pathways, along with the enrichment of transcription factors involved in the transcriptional activity of ER, underscoring the crucial role of HDAC6 in regulating hormone receptors. Notably, the addition of HDAC6 inhibitor potentiated the effects of anti-ER and anti-PR drugs mainly in TNBC cells. Together, these data highlight the role of HDAC6 in regulating PR expression and provide a promising therapeutic approach for boosting breast cancer sensitivity to hormonal therapy.
Collapse
Affiliation(s)
- Wafaa S Ramadan
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Rahma K Alseksek
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University City Road, Sharjah, 27272, United Arab Emirates
| | - Soraya Mouffak
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Iman M Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; Clinical Sciences Department, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, Champollion Street, Alexandria, 21131, Egypt
| | - Maha M Saber-Ayad
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; Clinical Sciences Department, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Varsha Menon
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Burcu Yener Ilce
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Raafat El-Awady
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University City Road, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
2
|
Quintero J, Saad NY, Pagnoni SM, Jacquelin DK, Gatica L, Harper SQ, Rosa AL. The DUX4 protein is a co-repressor of the progesterone and glucocorticoid nuclear receptors. FEBS Lett 2022; 596:2644-2658. [PMID: 35662006 DOI: 10.1002/1873-3468.14416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/28/2022] [Accepted: 05/28/2022] [Indexed: 11/09/2022]
Abstract
DUX4 is a transcription factor required during early embryonic development in placental mammals. In this work we provide evidence that DUX4 is a co-repressor of nuclear receptors (NRs) of progesterone (PR) and glucocorticoids (GR). The DUX4 C-ter and N-ter regions, including the nuclear localization signals and homeodomain motifs, contribute to the corepressor activity of DUX4 on PR and GR. Immunoprecipitation studies, using total protein extracts of cells expressing tagged versions of DUX4 and GR, support that these proteins are physically associated. Our studies suggest that DUX4 could modulate gene expression by coregulating the activity of hormone NRs. This is the first report highlighting a potential endocrine role for DUX4.
Collapse
Affiliation(s)
- Julieta Quintero
- Laboratorio de Genética y Biología Molecular, IRNASUS-CONICET, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Argentina
| | - Nizar Y Saad
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Sabrina M Pagnoni
- Laboratorio de Genética y Biología Molecular, IRNASUS-CONICET, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Argentina
| | - Daniela K Jacquelin
- Laboratorio de Genética y Biología Molecular, IRNASUS-CONICET, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Argentina.,INFIQC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Laura Gatica
- Laboratorio de Genética y Biología Molecular, IRNASUS-CONICET, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Argentina.,CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Scott Q Harper
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Alberto L Rosa
- Laboratorio de Genética y Biología Molecular, IRNASUS-CONICET, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Argentina.,Fundación Allende-CONICET, Córdoba, Argentina
| |
Collapse
|
3
|
Progesterone receptor expression contributes to gemcitabine resistance at higher ECM stiffness in breast cancer cell lines. PLoS One 2022; 17:e0268300. [PMID: 35617163 PMCID: PMC9135204 DOI: 10.1371/journal.pone.0268300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Chemoresistance poses a great barrier to breast cancer treatment and is thought to correlate with increased matrix stiffness. We developed two-dimensional (2D) polyacrylamide (PAA) and three-dimensional (3D) alginate in vitro models of tissue stiffness that mimic the stiffness of normal breast and breast cancer. We then used these to compare cell viability in response to chemotherapeutic treatment. In both 2D and 3D we observed that breast cancer cell growth and size was increased at a higher stiffness corresponding to tumours compared to normal tissue. When chemotherapeutic response was measured, a specific differential response in cell viability was observed for gemcitabine in 2 of the 7 breast cancer cell lines investigated. MCF7 and T-47D cell lines showed gemcitabine resistance at 4 kPa compared to 500 Pa. These cell lines share a common phenotype of progesterone receptor (PGR) expression and, indeed, pre-treatment with the selective progesterone receptor modulator (SPRM) mifepristone abolished resistance to gemcitabine at high stiffness. Our data reveals that combined treatment with SPRMs may therefore help in reducing resistance to gemcitabine in stiffer breast tumours which are PGR positive.
Collapse
|
4
|
Thiebaut C, Vlaeminck-Guillem V, Trédan O, Poulard C, Le Romancer M. Non-genomic signaling of steroid receptors in cancer. Mol Cell Endocrinol 2021; 538:111453. [PMID: 34520815 DOI: 10.1016/j.mce.2021.111453] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 12/21/2022]
Abstract
Steroid receptors (SRs) are members of the nuclear receptor family, which are ligand-activated transcription factors. SRs regulate many physiological functions including development and reproduction, though they can also be involved in several pathologies, especially cancer. Highly controlled cellular responses to steroids involve transcriptional regulation (genomic activity) combined with direct activation of signaling cascades (non-genomic activity). Non-genomic signaling has been extensively studied in cancer, mainly in breast cancer for ER and PR, and prostate cancer for AR. Even though most of the studies have been conducted in cells, some of them have been confirmed in vivo, highlighting the relevance of this pathway in cancer. This review provides an overview of the current and emerging knowledge on non-genomic signaling with a focus on breast and prostate cancers and its clinical relevance. A thorough understanding of ER, PR, AR and GR non-genomic pathways may open new perspectives for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Charlène Thiebaut
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Virginie Vlaeminck-Guillem
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; Service de Biochimie Biologie Moléculaire Sud, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, F-69495, Pierre-Bénite, France
| | - Olivier Trédan
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; Medical Oncology Department, Centre Léon Bérard, F-69000, Lyon, France
| | - Coralie Poulard
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Muriel Le Romancer
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.
| |
Collapse
|
5
|
Ando K, Hédou JJ, Feyaerts D, Han X, Ganio EA, Tsai ES, Peterson LS, Verdonk F, Tsai AS, Marić I, Wong RJ, Angst MS, Aghaeepour N, Stevenson DK, Blumenfeld YJ, Sultan P, Carvalho B, Stelzer IA, Gaudillière B. A Peripheral Immune Signature of Labor Induction. Front Immunol 2021; 12:725989. [PMID: 34566984 PMCID: PMC8458888 DOI: 10.3389/fimmu.2021.725989] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/20/2021] [Indexed: 11/23/2022] Open
Abstract
Approximately 1 in 4 pregnant women in the United States undergo labor induction. The onset and establishment of labor, particularly induced labor, is a complex and dynamic process influenced by multiple endocrine, inflammatory, and mechanical factors as well as obstetric and pharmacological interventions. The duration from labor induction to the onset of active labor remains unpredictable. Moreover, prolonged labor is associated with severe complications for the mother and her offspring, most importantly chorioamnionitis, uterine atony, and postpartum hemorrhage. While maternal immune system adaptations that are critical for the maintenance of a healthy pregnancy have been previously characterized, the role of the immune system during the establishment of labor is poorly understood. Understanding maternal immune adaptations during labor initiation can have important ramifications for predicting successful labor induction and labor complications in both induced and spontaneous types of labor. The aim of this study was to characterize labor-associated maternal immune system dynamics from labor induction to the start of active labor. Serial blood samples from fifteen participants were collected immediately prior to labor induction (baseline) and during the latent phase until the start of active labor. Using high-dimensional mass cytometry, a total of 1,059 single-cell immune features were extracted from each sample. A multivariate machine-learning method was employed to characterize the dynamic changes of the maternal immune system after labor induction until the establishment of active labor. A cross-validated linear sparse regression model (least absolute shrinkage and selection operator, LASSO) predicted the minutes since induction of labor with high accuracy (R = 0.86, p = 6.7e-15, RMSE = 277 min). Immune features most informative for the model included STAT5 signaling in central memory CD8+ T cells and pro-inflammatory STAT3 signaling responses across multiple adaptive and innate immune cell subsets. Our study reports a peripheral immune signature of labor induction, and provides important insights into biological mechanisms that may ultimately predict labor induction success as well as complications, thereby facilitating clinical decision-making to improve maternal and fetal well-being.
Collapse
Affiliation(s)
- Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Julien J Hédou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Department of Biomedical Sciences, University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA, United States
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Eileen S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Laura S Peterson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Franck Verdonk
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Amy S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ivana Marić
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Ronald J Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States.,Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, United States
| | - David K Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Yair J Blumenfeld
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, United States
| | - Pervez Sultan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Brendan Carvalho
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ina A Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
6
|
Moore NL, Hanson AR, Ebrahimie E, Hickey TE, Tilley WD. Anti-proliferative transcriptional effects of medroxyprogesterone acetate in estrogen receptor positive breast cancer cells are predominantly mediated by the progesterone receptor. J Steroid Biochem Mol Biol 2020; 199:105548. [PMID: 31805393 DOI: 10.1016/j.jsbmb.2019.105548] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/19/2019] [Indexed: 01/14/2023]
Abstract
Medroxyprogesterone acetate (MPA) is a first generation progestin that has been in clinical use for various hormonal conditions in women since the 1960s. Although developed as a progesterone receptor (PR) agonist, MPA also has strong binding affinity for other steroid receptors. This promiscuity confounds the mechanistic action of MPA in target cells that express multiple steroid receptors. This study is the first to assess the relative contribution of progesterone, androgen and glucocorticoid receptors in mediating the transcriptional activity of MPA on endogenous targets in breast cancer cells that endogenously express all three receptors at comparable levels. Gene expression profiling in estrogen receptor positive (ER+) ZR-75-1 breast cancer cells demonstrated that although the MPA-regulated transcriptome strongly overlapped with that of Progesterone (PROG), 5α-dihydrotestosterone (DHT) and Dexamethasone (DEX), it clustered most strongly with that of PROG, suggesting that MPA predominantly acts via the progesterone receptor (PR) rather than androgen receptor (AR) or glucocorticoid receptor (GR). Subsequent experiments manipulating levels of these receptors, either through specific culture conditions or with lentiviral shRNAs targeting individual receptors, also revealed a stronger contribution of PR compared to AR and GR on the expression of endogenous target genes that are either commonly regulated by all ligands or specifically regulated only by MPA. A predominant contribution of PR to MPA action in ER+ T-47D breast cancer cells was also observed, although a stronger role for AR was evident in T-47D compared to that observed in ZR-75-1 cells. Network analysis of ligand-specific and commonly regulated genes demonstrated that MPA utilises different transcription factors and signalling pathways to inhibit proliferation compared with PROG. This study reaffirms the importance of PR in mediating MPA action in an endogenous breast cancer context where multiple steroid receptors are co-expressed and has potential implications for PR-targeting therapeutic strategies in ER+ breast cancer.
Collapse
Affiliation(s)
- Nicole L Moore
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Adrienne R Hanson
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Esmaeil Ebrahimie
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
7
|
Cenciarini ME, Proietti CJ. Molecular mechanisms underlying progesterone receptor action in breast cancer: Insights into cell proliferation and stem cell regulation. Steroids 2019; 152:108503. [PMID: 31562879 DOI: 10.1016/j.steroids.2019.108503] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/13/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
The ovarian steroid hormone progesterone and its nuclear receptor, the Progesterone Receptor (PR), play an essential role in the regulation of cell proliferation and differentiation in the mammary gland. In addition, experimental and clinical evidence demonstrate their critical role in controlling mammary gland tumorigenesis and breast cancer development. When bound to its ligand, the main action of PR is as a transcription factor, which regulates the expression of target genes networks. PR also activates signal transduction pathways through a rapid or non-genomic mechanism in breast cancer cells, an event that is fully integrated with its genomic effects. This review summarizes the molecular mechanisms of the ligand-activated PR actions that drive epithelial cell proliferation and the regulation of the stem cell population in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Mauro E Cenciarini
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | - Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina.
| |
Collapse
|
8
|
Leehy KA, Truong TH, Mauro LJ, Lange CA. Progesterone receptors (PR) mediate STAT actions: PR and prolactin receptor signaling crosstalk in breast cancer models. J Steroid Biochem Mol Biol 2018; 176:88-93. [PMID: 28442393 PMCID: PMC5653461 DOI: 10.1016/j.jsbmb.2017.04.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/28/2017] [Accepted: 04/20/2017] [Indexed: 12/17/2022]
Abstract
Estrogen is the major mitogenic stimulus of mammary gland development during puberty wherein ER signaling acts to induce abundant PR expression. PR signaling, in contrast, is the primary driver of mammary epithelial cell proliferation in adulthood. The high circulating levels of progesterone during pregnancy signal through PR, inducing expression of the prolactin receptor (PRLR). Cooperation between PR and prolactin (PRL) signaling, via regulation of downstream components in the PRL signaling pathway including JAKs and STATs, facilitates the alveolar morphogenesis observed during pregnancy. Indeed, these pathways are fully integrated via activation of shared signaling pathways (i.e. JAKs, MAPKs) as well as by the convergence of PRs and STATs at target genes relevant to both mammary gland biology and breast cancer progression (i.e. proliferation, stem cell outgrowth, tissue cell type heterogeneity). Thus, rather than a single mediator such as ER, transcription factor cascades (ER>PR>STATs) are responsible for rapid proliferative and developmental programming in the normal mammary gland. It is not surprising that these same mediators typify uncontrolled proliferation in a majority of breast cancers, where ER and PR are most often co-expressed and may cooperate to drive malignant tumor progression. This review will primarily focus on the integration of PR and PRL signaling in breast cancer models and the importance of this cross-talk in cancer progression in the context of mammographic density. Components of these PR/PRL signaling pathways could offer alternative drug targets and logical complements to anti-ER or anti-estrogen-based endocrine therapies.
Collapse
Affiliation(s)
- Katherine A Leehy
- Departments of Medicine and Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States
| | - Thu H Truong
- Departments of Medicine and Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States
| | - Laura J Mauro
- Department of Animal Sciences, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States
| | - Carol A Lange
- Departments of Medicine and Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States.
| |
Collapse
|
9
|
Grimm SL, Hartig SM, Edwards DP. Progesterone Receptor Signaling Mechanisms. J Mol Biol 2016; 428:3831-49. [PMID: 27380738 DOI: 10.1016/j.jmb.2016.06.020] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/25/2016] [Accepted: 06/27/2016] [Indexed: 12/27/2022]
Abstract
Progesterone receptor (PR) is a master regulator in female reproductive tissues that controls developmental processes and proliferation and differentiation during the reproductive cycle and pregnancy. PR also plays a role in progression of endocrine-dependent breast cancer. As a member of the nuclear receptor family of ligand-dependent transcription factors, the main action of PR is to regulate networks of target gene expression in response to binding its cognate steroid hormone, progesterone. This paper summarizes recent advances in understanding the structure-function properties of the receptor protein and the tissue/cell-type-specific PR signaling pathways that contribute to the biological actions of progesterone in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Sandra L Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA.
| |
Collapse
|
10
|
RETRACTED: MiR-143 inhibits tumor cell proliferation and invasion by targeting STAT3 in esophageal squamous cell carcinoma. Cancer Lett 2016; 373:97-108. [PMID: 26806810 DOI: 10.1016/j.canlet.2016.01.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/29/2015] [Accepted: 01/13/2016] [Indexed: 11/27/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy).
This article has been retracted at the request of the Editor in Chief. The term miR-143 has been used instead of the term miR-143* in the introduction and discussion which is misleading. With the misuse of this term subsequent errors and misleading statements appear throughout the paper. The authors apologize for this mistake.
Collapse
|
11
|
Proietti CJ, Izzo F, Díaz Flaqué MC, Cordo Russo R, Venturutti L, Mercogliano MF, De Martino M, Pineda V, Muñoz S, Guzmán P, Roa JC, Schillaci R, Elizalde PV. Heregulin Co-opts PR Transcriptional Action Via Stat3 Role As a Coregulator to Drive Cancer Growth. Mol Endocrinol 2015; 29:1468-85. [PMID: 26340407 DOI: 10.1210/me.2015-1170] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Accumulated findings have demonstrated the presence of bidirectional interactions between progesterone receptor (PR) and the ErbB family of receptor tyrosine kinases signaling pathways in breast cancer. We previously revealed signal transducer and activator of transcription 3 (Stat3) as a nodal convergence point between said signaling pathways proving that Stat3 is activated by one of the ErbBs' ligands, heregulin (HRG)β1 via ErbB2 and through the co-option of PR as a signaling molecule. Here, we found that HRGβ1 induced Stat3 recruitment to the promoters of the progestin-regulated cell cycle modulators Bcl-XL and p21(CIP1) and also stimulated Stat3 binding to the mouse mammary tumor virus promoter, which carries consensus progesterone response elements. Interestingly, HRGβ1-activated Stat3 displayed differential functions on PR activity depending on the promoter bound. Indeed, Stat3 was required for PR binding in bcl-X, p21(CIP1), and c-myc promoters while exerting a PR coactivator function on the mouse mammary tumor virus promoter. Stat3 also proved to be necessary for HRGβ1-induced in vivo tumor growth. Our results endow Stat3 a novel function as a coregulator of HRGβ1-activated PR to promote breast cancer growth. These findings underscore the importance of understanding the complex interactions between PR and other regulatory factors, such as Stat3, that contribute to determine the context-dependent transcriptional actions of PR.
Collapse
Affiliation(s)
- Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Franco Izzo
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - María Celeste Díaz Flaqué
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Rosalía Cordo Russo
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Leandro Venturutti
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - María Florencia Mercogliano
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Mara De Martino
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Viviana Pineda
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Sergio Muñoz
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Pablo Guzmán
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Juan C Roa
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Patricia V Elizalde
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| |
Collapse
|
12
|
Bravo ML, Pinto MP, Gonzalez I, Oliva B, Kato S, Cuello MA, Lange CA, Owen GI. Progesterone regulation of tissue factor depends on MEK1/2 activation and requires the proline-rich site on progesterone receptor. Endocrine 2015; 48:309-20. [PMID: 24853881 DOI: 10.1007/s12020-014-0288-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/05/2014] [Indexed: 10/25/2022]
Abstract
To characterize the molecular mechanism and map the response element used by progesterone (P) to upregulate tissue factor (TF) in breast cancer cells. TF expression and mRNA levels were analyzed in breast cancer ZR-75 and T47D cells, using Western blot and real-time PCR, respectively. Mapping of the TF promoter was performed using luciferase vectors. Progesterone receptor (PR) and specificity protein 1 (Sp1) binding to the TF promoter were analyzed by chromatin immuno precipitation assay. Specific or selective inhibitors were used for the MEK1/2 and the c-Src pathways (UO126 and PP2, respectively). TF mRNA increase peaks at 18 h following P treatment in ZR-75 and T47D cells. P upregulation occurs via a transcriptional mechanism that depends on PR and MEK1/2 activation, PR and Sp1 transcription factors bind to a region in the TF promoter that contains three Sp1 sites. TF mRNA upregulation requires an intact PR proline-rich site (mPRO), but it is independent from c-Src. TF upregulation by P is mediated by Sp1 sites in the TF promoter region. Transcriptional upregulation in breast cancer cells occurs via a new mechanism that requires MEK1/2 activation and the mPRO site but independent of c-Src activity. PR Phosphorylation at serine 294 and 345 is not essential.
Collapse
Affiliation(s)
- Maria Loreto Bravo
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Izzo F, Mercogliano F, Venturutti L, Tkach M, Inurrigarro G, Schillaci R, Cerchietti L, Elizalde PV, Proietti CJ. Progesterone receptor activation downregulates GATA3 by transcriptional repression and increased protein turnover promoting breast tumor growth. Breast Cancer Res 2014; 16:491. [PMID: 25479686 PMCID: PMC4303201 DOI: 10.1186/s13058-014-0491-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/28/2014] [Indexed: 11/10/2022] Open
Abstract
Introduction The transcription factor GATA3 is involved in mammary gland development and is crucial for the maintenance of the differentiated status of luminal epithelial cells. The role of GATA3 in breast cancer as a tumor suppressor has been established, although insights into the mechanism of GATA3 expression loss are still required. Methods Chromatin immunoprecipitation assays were conducted to study progestin modulation of recruitment of transcription factors to GATA3 promoter. We performed western blot and reverse RT-qPCR experiments to explore progestin regulation of GATA3 protein and mRNA expression respectively. Confocal microscopy and in vitro phosphorylation studies were conducted to examine progestin capacity to induce GATA3 serine phosphorylation in its 308 residue. GATA3 participation in progestin-induced breast cancer growth was addressed in in vitro proliferation and in vivo tumor growth experiments. Results In this study, we demonstrate that progestin-activated progesterone receptor (PR) reduces GATA3 expression through regulation at the transcriptional and post-translational levels in breast cancer cells. In the former mechanism, the histone methyltransferase enhancer of zeste homolog 2 is co-recruited with activated PR to a putative progesterone response element in the GATA3 proximal promoter, increasing H3K27me3 levels and inducing chromatin compaction, resulting in decreased GATA3 mRNA levels. This transcriptional regulation is coupled with increased GATA3 protein turnover through progestin-induced GATA3 phosphorylation at serine 308 followed by 26S proteasome-mediated degradation. Both molecular mechanisms converge to accomplish decreased GATA3 expression levels in breast cancer cells upon PR activation. In addition, we demonstrated that decreased GATA3 levels are required for progestin-induced upregulation of cyclin A2, which mediates the G1 to S phase transition of the cell cycle and was reported to be associated with poor prognosis in breast cancer. Finally, we showed that downregulation of GATA3 is required for progestin stimulation of both in vitro cell proliferation and in vivo tumor growth. Conclusions In the present study, we reveal that progestin-induced PR activation leads to loss of GATA3 expression in breast cancer cells through transcriptional and post-translational regulation. Importantly, we demonstrate that GATA3 downregulation is required for progestin-induced upregulation of cyclin A2 and for progestin-induced in vitro and in vivo breast cancer cell growth. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0491-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Franco Izzo
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Florencia Mercogliano
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Leandro Venturutti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Mercedes Tkach
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | | | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | | | - Patricia V Elizalde
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| |
Collapse
|
14
|
Xuan X, Li S, Lou X, Zheng X, Li Y, Wang F, Gao Y, Zhang H, He H, Zeng Q. Stat3 promotes invasion of esophageal squamous cell carcinoma through up-regulation of MMP2. Mol Biol Rep 2014; 42:907-15. [DOI: 10.1007/s11033-014-3828-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 11/10/2014] [Indexed: 01/06/2023]
|
15
|
Moore NL, Edwards DP, Weigel NL. Cyclin A2 and its associated kinase activity are required for optimal induction of progesterone receptor target genes in breast cancer cells. J Steroid Biochem Mol Biol 2014; 144 Pt B:471-82. [PMID: 25220500 PMCID: PMC4201666 DOI: 10.1016/j.jsbmb.2014.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 08/09/2014] [Accepted: 09/05/2014] [Indexed: 12/30/2022]
Abstract
A role for the cell cycle protein cyclin A2 in regulating progesterone receptor (PR) activity is emerging. This study investigates the role of cyclin A2 in regulating endogenous PR activity in T47D breast cancer cells by depleting cyclin A2 expression and measuring PR target genes using q-RT-PCR. Targets examined included genes induced by the PR-B isoform more strongly than PR-A (SGK1, FKBP5), a gene induced predominantly by PR-A (HEF1), genes induced via PR tethering to other transcription factors (p21, p27), a gene induced in part via extra-nuclear PR signaling mechanisms (cyclin D1) and PR-repressed genes (DST, IL1R1). Progestin induction of target genes was reduced following cyclin A2 depletion. However, cyclin A2 depletion did not diminish progestin target gene repression. Furthermore, inhibition of the associated Cdk2 kinase activity of cyclin A2 also reduced progestin induction of target genes, while Cdk2 enhanced the interaction between PR and cyclin A2. These results demonstrate that cyclin A2 and its associated kinase activity are important for progestin-induced activation of endogenous PR target genes in breast cancer cells.
Collapse
Affiliation(s)
- Nicole L Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | - Nancy L Weigel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
16
|
Blumert C, Kalkhof S, Brocke-Heidrich K, Kohajda T, von Bergen M, Horn F. Analysis of the STAT3 interactome using in-situ biotinylation and SILAC. J Proteomics 2013; 94:370-86. [PMID: 24013128 DOI: 10.1016/j.jprot.2013.08.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/01/2013] [Accepted: 08/26/2013] [Indexed: 12/28/2022]
Abstract
UNLABELLED Signal transducer and activator of transcription 3 (STAT3) is activated by a variety of cytokines and growth factors. To generate a comprehensive data set of proteins interacting specifically with STAT3, we applied stable isotope labeling with amino acids in cell culture (SILAC). For high-affinity pull-down using streptavidin, we fused STAT3 with a short peptide tag allowing biotinylation in situ (bio-tag), which did not affect STAT3 functions. By this approach, 3642 coprecipitated proteins were detected in human embryonic kidney-293 cells. Filtering using statistical and functional criteria finally extracted 136 proteins as putative interaction partners of STAT3. Both, a physical interaction network analysis and the enrichment of known and predicted interaction partners suggested that our filtering criteria successfully enriched true STAT3 interactors. Our approach identified numerous novel interactors, including ones previously predicted to associate with STAT3. By reciprocal coprecipitation, we were able to verify the physical association between STAT3 and selected interactors, including the novel interaction with TOX4, a member of the TOX high mobility group box family. Applying the same method, we next investigated the activation-dependency of the STAT3 interactome. Again, we identified both known and novel interactions. Thus, our approach allows to study protein-protein interaction effectively and comprehensively. BIOLOGICAL SIGNIFICANCE The location, activity, function, degradation, and synthesis of proteins are significantly regulated by interactions of proteins with other proteins, biopolymers and small molecules. Thus, the comprehensive characterization of interactions of proteins in a given proteome is the next milestone on the path to understanding the biochemistry of the cell. In order to generate a comprehensive interactome dataset of proteins specifically interacting with a selected bait protein, we fused our bait protein STAT3 with a short peptide tag allowing biotinylation in situ (bio-tag). This bio-tag allows an affinity pull-down using streptavidin but affected neither the activation of STAT3 by tyrosine phosphorylation nor its transactivating potential. We combined SILAC for accurate relative protein quantification, subcellular fractionation to increase the coverage of interacting proteins, high-affinity pull-down and a stringent filtering method to successfully analyze the interactome of STAT3. With our approach we confirmed several already known and identified numerous novel STAT3 interactors. The approach applied provides a rapid and effective method, which is broadly applicable for studying protein-protein interactions and their dependency on post-translational modifications.
Collapse
Affiliation(s)
- Conny Blumert
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany; Fraunhofer Institute for Cell Therapy and Immunology, Perlickstrasse 1, 04103 Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Sun X, Bartos A, Whitsett JA, Dey SK. Uterine deletion of Gp130 or Stat3 shows implantation failure with increased estrogenic responses. Mol Endocrinol 2013; 27:1492-501. [PMID: 23885093 DOI: 10.1210/me.2013-1086] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Leukemia inhibitory factor (LIF), a downstream target of estrogen, is essential for implantation in mice. LIF function is thought to be mediated by its binding to LIF receptor (LIFR) and recruitment of coreceptor GP130 (glycoprotein 130), and this receptor complex then activates signal transducer and activator of transcription (STAT)1/3. However, the importance of LIFR and GP130 acting via STAT3 in implantation remains uncertain, because constitutive inactivation of Lifr, Gp130, or Stat3 shows embryonic lethality in mice. To address this issue, we generated mice with conditional deletion of uterine Gp130 or Stat3 and show that both GP130 and STAT3 are critical for uterine receptivity and implantation. Implantation failure in these deleted mice is associated with higher uterine estrogenic responses prior to the time of implantation. These heightened estrogenic responses are not due to changes in ovarian hormone levels or expression of their nuclear receptors. In the deleted mice, estrogen-responsive gene, Lactoferrin (Ltf), and Mucin 1 protein, were up-regulated in the uterus. In addition, progesterone-responsive genes, Hoxa10 and Indian hedgehog (Ihh), were markedly down-regulated in STAT3-inactivated uteri. These changes in uteri of deleted mice were reflected by the failure of differentiation of the luminal epithelium, which is essential for blastocyst attachment.
Collapse
Affiliation(s)
- Xiaofei Sun
- Division of Reproductive Sciences,Perinatal Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | |
Collapse
|
18
|
Diaz Flaqué MC, Vicario R, Proietti CJ, Izzo F, Schillaci R, Elizalde PV. Progestin drives breast cancer growth by inducing p21(CIP1) expression through the assembly of a transcriptional complex among Stat3, progesterone receptor and ErbB-2. Steroids 2013. [PMID: 23178160 DOI: 10.1016/j.steroids.2012.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cell cycle regulator p21(CIP1) has controversial biological effects in breast cancer since in spite of its role as cell cycle inhibitor and promoter of cellular senescence, it also induces cell proliferation and chemoteraphy resistance. We here explored the molecular mechanisms involved in progestin regulation of p21(CIP1) expression. We also investigated the biological effects of p21(CIP1) in breast cancer cells. We found that the synthetic progestin medroxyprogesterone acetate (MPA) upregulates p21(CIP1) protein expression via c-Src, signal transducer and activator of transcription 3 (Stat3) and ErbB-2 phosphorylation. Notably, we also found that ErbB-2 nuclear function plays a key role in MPA-induction of p21(CIP1) expression. Interestingly, we determined that progestin drives p21(CIP1) transcriptional activation via a novel nonclassical transcriptional mechanism in which progesterone receptor is recruited along with Stat3 and ErbB-2 to a Stat3 binding site at p21(CIP1) promoter. Our findings revealed that ErbB-2 functions as a coactivator of Stat3 in progestin induction of p21(CIP1) transcriptional activation. Furthermore, we demonstrated that blockage of p21(CIP1) expression strongly inhibited in vitro and in vivo progestin-induced breast cancer cell proliferation. These results further support the hypothesis that according to cell context and type of stimulus, p21(CIP1) is capable of inducing cell cycle progression. Moreover, we provided evidence that Stat3 and nuclear ErbB-2 are key players in progestin-induced p21(CIP1) regulation.
Collapse
Affiliation(s)
- María C Diaz Flaqué
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | | | | | | | | | | |
Collapse
|
19
|
Tanos T, Rojo L, Echeverria P, Brisken C. ER and PR signaling nodes during mammary gland development. Breast Cancer Res 2012; 14:210. [PMID: 22809143 PMCID: PMC3680919 DOI: 10.1186/bcr3166] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The ovarian hormones estrogen and progesterone orchestrate postnatal mammary gland development and are implicated in breast cancer. Most of our understanding of the molecular mechanisms of estrogen receptor (ER) and progesterone receptor (PR) signaling stems from in vitro studies with hormone receptor-positive cell lines. They have shown that ER and PR regulate gene transcription either by binding to DNA response elements directly or via other transcription factors and recruiting co-regulators. In addition they cross-talk with other signaling pathways through nongenomic mechanisms. Mouse genetics combined with tissue recombination techniques have provided insights about the action of these two hormones in vivo. It has emerged that hormones act on a subset of mammary epithelial cells and relegate biological functions to paracrine factors. With regards to hormonal signaling in breast carcinomas, global gene expression analyses have led to the identification of gene expression signatures that are characteristic of ERα-positive tumors that have stipulated functional studies of hitherto poorly understood transcription factors. Here, we highlight what has been learned about ER and PR signaling nodes in these different systems and attempt to lay out in which way the insights may converge.
Collapse
|
20
|
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: 157] [Impact Index Per Article: 12.1] [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
|
21
|
Hagan CR, Daniel AR, Dressing GE, Lange CA. Role of phosphorylation in progesterone receptor signaling and specificity. Mol Cell Endocrinol 2012; 357:43-9. [PMID: 21945472 PMCID: PMC3265648 DOI: 10.1016/j.mce.2011.09.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 08/08/2011] [Accepted: 09/11/2011] [Indexed: 10/17/2022]
Abstract
Progesterone receptors (PR), in concert with peptide growth factor-initiated signaling pathways, initiate massive expansion of the epithelial cell compartment associated with the process of alveologenesis in the developing mammary gland. PR-dependent signaling events also contribute to inappropriate proliferation observed in breast cancer. Notably, PR-B isoform-specific cross talk with growth factor-driven pathways is required for the proliferative actions of progesterone. Indeed, PRs act as heavily phosphorylated transcription factor "sensors" for mitogenic protein kinases that are often elevated and/or constitutively activated in invasive breast cancers. In addition, phospho-PR-target genes frequently include the components of mitogenic signaling pathways, revealing a mechanism for feed-forward signaling that confers increased responsiveness of, PR +mammary epithelial cells to these same mitogenic stimuli. Understanding the mechanisms and isoform selectivity of PR/kinase interactions may yield further insight into targeting altered signaling networks in breast and other hormonally responsive cancers (i.e. lung, uterine and ovarian) in the clinic. This review focuses on PR phosphorylation by mitogenic protein kinases and mechanisms of PR-target gene selection that lead to increased cell proliferation.
Collapse
Affiliation(s)
- Christy R Hagan
- University of Minnesota, Departments of Medicine and Pharmacology, Division of Hematology, Oncology, and Transplantation, Women's Cancer Program, Masonic Cancer Center, Minneapolis, MN 55455, United States
| | | | | | | |
Collapse
|
22
|
Rivas MA, Venturutti L, Huang YW, Schillaci R, Huang THM, Elizalde PV. Downregulation of the tumor-suppressor miR-16 via progestin-mediated oncogenic signaling contributes to breast cancer development. Breast Cancer Res 2012; 14:R77. [PMID: 22583478 PMCID: PMC3446340 DOI: 10.1186/bcr3187] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 04/29/2012] [Accepted: 05/14/2012] [Indexed: 12/19/2022] Open
Abstract
Introduction Experimental and clinical evidence points to a critical role of progesterone and the nuclear progesterone receptor (PR) in controlling mammary gland tumorigenesis. However, the molecular mechanisms of progesterone action in breast cancer still remain elusive. On the other hand, micro RNAs (miRNAs) are short ribonucleic acids which have also been found to play a pivotal role in cancer pathogenesis. The role of miRNA in progestin-induced breast cancer is poorly explored. In this study we explored progestin modulation of miRNA expression in mammary tumorigenesis. Methods We performed a genome-wide study to explore progestin-mediated regulation of miRNA expression in breast cancer. miR-16 expression was studied by RT-qPCR in cancer cell lines with silenced PR, signal transducer and activator of transcription 3 (Stat3) or c-Myc, treated or not with progestins. Breast cancer cells were transfected with the precursor of miR-16 and proliferation assays, Western blots or in vivo experiments were performed. Target genes of miR-16 were searched through a bioinformatical approach, and the study was focused on cyclin E. Reporter gene assays were performed to confirm that cyclin E 3'UTR is a direct target of miR-16. Results We found that nine miRNAs were upregulated and seven were downregulated by progestin in mammary tumor cells. miR-16, whose function as a tumor suppressor in leukemia has already been shown, was identified as one of the downregulated miRNAs in murine and human breast cancer cells. Progestin induced a decrease in miR-16 levels via the classical PR and through a hierarchical interplay between Stat3 and the oncogenic transcription factor c-Myc. A search for miR-16 targets showed that the CCNE1 gene, encoding the cell cycle regulator cyclin E, contains conserved putative miR-16 target sites in its mRNA 3' UTR region. We found that, similar to the molecular mechanism underlying progestin-modulated miR-16 expression, Stat3 and c-Myc participated in the induction of cyclin E expression by progestin. Moreover, overexpression of miR-16 abrogated the ability of progestin to induce cyclin E upregulation, revealing that cyclin E is a novel target of miR-16 in breast cancer. Overexpression of miR-16 also inhibited progestin-induced breast tumor growth in vitro and in vivo, demonstrating for the first time, a role for miR-16 as a tumor suppressor in mammary tumorigenesis. We also found that the ErbB ligand heregulin (HRG) downregulated the expression of miR-16, which then participates in the proliferative activity of HRG in breast tumor cells. Conclusions In this study, we reveal the first progestin-regulated miRNA expression profile and identify a novel role for miR-16 as a tumor suppressor in progestin- and growth factor-induced growth in breast cancer.
Collapse
Affiliation(s)
- Martin A Rivas
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
23
|
Cui W, Li Q, Feng L, Ding W. MiR-126-3p regulates progesterone receptors and involves development and lactation of mouse mammary gland. Mol Cell Biochem 2011; 355:17-25. [PMID: 21526342 DOI: 10.1007/s11010-011-0834-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Accepted: 04/15/2011] [Indexed: 01/19/2023]
Abstract
MicroRNAs (miRNAs) are small (18-22 nucleotide) non-coding, endogenous regulatory RNA molecules, and they regulate gene expression at the post-transcriptional level through binding to their target mRNAs by base-pairing and subsequently inducing either translational repression or mRNA destabilization by plants, animals, and some viruses. In this study, combining microarray techniques with qRT-PCR, we found that miR-126-3p expression showed significant difference in the mouse mammary cycle during pregnancy, particularly on transition from pregnancy to lactation. Bioinformatics were used to predict target gene of miR-126-3p, and luciferase activity assay to test it, it showed that the progesterone receptor (PGR) 3'UTR is directly targeted by miR-126-3p. In this study, mouse mammary epithelial cells as cell model in vitro were used to study the function of miR-126-3p. Using gene silencing and over-expression for miR-126-3p, the expression of PGR protein and the secretion of casein were detected by western blotting and HPLC, respectively. To determine whether miR-126-3p can affect mouse mammary epithelial cells viability, cells were analyzed by CASY-YY. In conclusion, PGR gene confirmed miR-126-3p target genes through luciferase activity and western blotting. And miR-126-3p could also inhibit proliferation of mouse mammary epithelial cells (P < 0.01) and expression of β-casein (P < 0.01), and down-regulate PGR protein (P < 0.05). Our results suggested that miR-126-3p inhibited expression of PGR protein level as well as the proliferation of mammary epithelial cells, therefore miR-126-3p could play an important role in the process of mammary gland development.
Collapse
Affiliation(s)
- Wei Cui
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| | | | | | | |
Collapse
|