1
|
Sun G, Zhao C, Han J, Wu S, Chen Y, Yao J, Li L. Regulatory mechanisms of steroid hormone receptors on gene transcription through chromatin interaction and enhancer reprogramming. Cell Oncol (Dordr) 2024; 47:2073-2090. [PMID: 39543064 DOI: 10.1007/s13402-024-01011-y] [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] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
Regulation of steroid hormone receptors (SHRs) on transcriptional reprogramming is crucial for breast cancer progression. SHRs, including estrogen receptor (ER), androgen receptor (AR), progesterone receptor (PR), and glucocorticoid receptor (GR) play key roles in remodeling the transcriptome of breast cancer cells. However, the molecular mechanisms by which SHRs regulate chromatin landscape in enhancer regions and transcription factor interactions remain largely unknown. In this review, we summarized the regulatory effects of 3 types of SHRs (AR, PR, and GR) on gene transcription through chromatin interactions and enhancer reprogramming. Specifically, AR and PR exhibit bi-directional regulatory effects (both inhibitory and promoting) on ER-mediated gene transcription, while GR modulates the transcription of pro-proliferation genes in ER-positive breast cancer cells. In addition, we have presented four enhancer reprogramming mechanisms (transcription factor cooperation, pioneer factor binding, dynamic assisted loading, and tethering) and the multiple enhancer-promoter contact models. Based on these mechanisms and models, this review proposes that the combination of multiple therapy strategies such as agonists/antagonists of SHRs plus endocrine therapy and the adoption of the latest sequencing technologies are expected to improve the efficacy of ER positive breast cancer treatment.
Collapse
Affiliation(s)
- Ge Sun
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Chunguang Zhao
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Jing Han
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P.R. China
| | - Shaoya Wu
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Yan Chen
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Jing Yao
- Cancer Center, Institute of Radiation Oncology, Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
| | - Li Li
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China.
| |
Collapse
|
2
|
Hilgart E, Zhou W, Martinez-Montes E, Idrizi A, Tryggvadottir R, Gondek LP, Majeti R, Ji H, Koldobskiy MA, Feinberg AP. DNA methylation stochasticity is linked to transcriptional variability and identifies convergent epigenetic disruption across genetically-defined subtypes of AML. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.26.620422. [PMID: 39554147 PMCID: PMC11565875 DOI: 10.1101/2024.10.26.620422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Disruption of the epigenetic landscape is of particular interest in acute myeloid leukemia (AML) due to its relatively low mutational burden and frequent occurrence of mutations in epigenetic regulators. Here, we applied an information-theoretic analysis of methylation potential energy landscapes, capturing changes in mean methylation level and methylation entropy, to comprehensively analyze DNA methylation stochasticity in subtypes of AML defined by mutually exclusive genetic mutations. We identified AML subtypes with CEBPA double mutation and those with IDH mutations as distinctly high-entropy subtypes, marked by methylation disruption over a convergent set of genes. We found a core program of epigenetic landscape disruption across all AML subtypes, with discordant methylation stochasticity and transcriptional dysregulation converging on functionally important leukemic signatures, suggesting a genotype-independent role of stochastic disruption of the epigenetic landscape in mediating leukemogenesis. We further established a relationship between methylation entropy and gene expression variability, connecting the disruption of the epigenetic landscape to transcription in AML. This approach identified a convergent program of epigenetic dysregulation in leukemia, clarifying the contribution of specific genetic mutations to stochastic disruption of the epigenetic and transcriptional landscapes of AML.
Collapse
|
3
|
Pavlik T, Konchekov E, Shimanovskii N. Antitumor progestins activity: Cytostatic effect and immune response. Steroids 2024; 210:109474. [PMID: 39048056 DOI: 10.1016/j.steroids.2024.109474] [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: 04/25/2024] [Revised: 07/13/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Progestins are used to treat some hormone-sensitive tumors. This review discusses the mechanisms of progestins' effects on tumor cells, the differences in the effects of progesterone and its analogs on different tumor types, and the influence of progestins on the antitumor immune response. Progestins cause a cytostatic effect, but at the same time they can suppress the antitumor immune response, and this can promote the proliferation and metastasis of tumor cells. Such progestins as dienogest, megestrol acetate and levonorgestrel increase the activity of NK-cells, which play a major role in the body's fight against tumor cells. The use of existing progestins and the development of new drugs with gestagenic activity may hold promise in oncotherapy.
Collapse
Affiliation(s)
- T Pavlik
- Pirogov Russian National Research Medical University, Russia; Prokhorov General Physics Institute of the Russian Academy of Sciences, Russia.
| | - E Konchekov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Russia; Peoples Friendship University of Russia (RUDN University), Russia
| | - N Shimanovskii
- Pirogov Russian National Research Medical University, Russia
| |
Collapse
|
4
|
Liao Z, Tang S, Jiang P, Geng T, Cope DI, Dunn TN, Guner J, Radilla LA, Guan X, Monsivais D. Impaired bone morphogenetic protein (BMP) signaling pathways disrupt decidualization in endometriosis. Commun Biol 2024; 7:227. [PMID: 38402336 PMCID: PMC10894266 DOI: 10.1038/s42003-024-05898-z] [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: 10/20/2023] [Accepted: 02/07/2024] [Indexed: 02/26/2024] Open
Abstract
Endometriosis is linked to increased infertility and pregnancy complications due to defective endometrial decidualization. We hypothesized that identification of altered signaling pathways during decidualization could identify the underlying cause of infertility and pregnancy complications. Our study reveals that transforming growth factor β (TGFβ) pathways are impaired in the endometrium of individuals with endometriosis, leading to defective decidualization. Through detailed transcriptomic analyses, we discovered abnormalities in TGFβ signaling pathways and key regulators, such as SMAD4, in the endometrium of affected individuals. We also observed compromised activity of bone morphogenetic proteins (BMP), a subset of the TGFβ family, that control endometrial receptivity. Using 3-dimensional models of endometrial stromal and epithelial assembloids, we showed that exogenous BMP2 improved decidual marker expression in individuals with endometriosis. Our findings reveal dysfunction of BMP/SMAD signaling in the endometrium of individuals with endometriosis, explaining decidualization defects and subsequent pregnancy complications in these individuals.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Peixin Jiang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ting Geng
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dominique I Cope
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Timothy N Dunn
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
- Division of Reproductive Endocrinology & Infertility, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Joie Guner
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, 90033, USA
| | - Linda Alpuing Radilla
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaoming Guan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Monsivais D, Liao Z, Tang S, Jiang P, Geng T, Cope D, Dunn T, Guner J, Radilla LA, Guan X. Impaired bone morphogenetic protein (BMP) signaling pathways disrupt decidualization in endometriosis. RESEARCH SQUARE 2023:rs.3.rs-3471243. [PMID: 37986901 PMCID: PMC10659538 DOI: 10.21203/rs.3.rs-3471243/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Endometriosis is linked to increased infertility and pregnancy complications due to defective endometrial decidualization. We hypothesized that identification of altered signaling pathways during decidualization could identify the underlying cause of infertility and pregnancy complications. Our study reveals that transforming growth factor β (TGFβ) pathways are impaired in the endometrium of individuals with endometriosis, leading to defective decidualization. Through detailed transcriptomic analyses, we discovered abnormalities in TGFβ signaling pathways and key regulators, such as SMAD4, in the endometrium of affected individuals. We also observed compromised activity of bone morphogenetic proteins (BMP), a subset of the TGFβ family, that control endometrial receptivity. Using 3-dimensional models of endometrial stromal and epithelial assembloids, we showed that exogenous BMP2 improved decidual marker expression in individuals with endometriosis. Our findings unveil a previously unidentified dysfunction in BMP/SMAD signaling in the endometrium of individuals with endometriosis, explaining decidualization defects and subsequent pregnancy complications in these individuals.
Collapse
|
6
|
Liao Z, Tang S, Jiang P, Geng T, Cope DI, Dunn TN, Guner J, Radilla LA, Guan X, Monsivais D. Impaired bone morphogenetic protein signaling pathways disrupt decidualization in endometriosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558268. [PMID: 37790548 PMCID: PMC10542516 DOI: 10.1101/2023.09.21.558268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
It is hypothesized that impaired endometrial decidualization contributes to decreased fertility in individuals with endometriosis. To identify the molecular defects that underpin defective decidualization in endometriosis, we subjected endometrial stromal cells from individuals with or without endometriosis to time course in vitro decidualization with estradiol, progesterone, and 8-bromo-cyclic-AMP (EPC) for 2, 4, 6, or 8 days. Transcriptomic profiling identified differences in key pathways between the two groups, including defective bone morphogenetic protein (BMP)/SMAD4 signaling (ID2, ID3, FST), oxidate stress response (NFE2L2, ALOX15, SLC40A1), and retinoic acid signaling pathways (RARRES, RARB, ALDH1B1). Genome-wide binding analyses identified an altered genomic distribution of SMAD4 and H3K27Ac in the decidualized stromal cells from individuals without endometriosis relative to those with endometriosis, with target genes enriched in pathways related to signaling by transforming growth factor β (TGFβ), neurotrophic tyrosine kinase receptors (NTRK), and nerve growth factor (NGF)-stimulated transcription. We found that direct SMAD1/5/4 target genes control FOXO, PI3K/AKT, and progesterone-mediated signaling in decidualizing cells and that BMP2 supplementation in endometriosis patient-derived assembloids elevated the expression of decidualization markers. In summary, transcriptomic and genome-wide binding analyses of patient-derived endometrial cells and assembloids identified that a functional BMP/SMAD1/5/4 signaling program is crucial for engaging decidualization.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Peixin Jiang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ting Geng
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dominique I. Cope
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Timothy N. Dunn
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
- Division of Reproductive Endocrinology & Infertility, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Joie Guner
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, 90033, USA
| | - Linda Alpuing Radilla
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaoming Guan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
7
|
Hasnawati H, Wahyuono S, Susidarti RA, Santosa D, Arfan A. A New Diterpenoid of Indonesian Scoparia dulcis Linn: Isolation and Cytotoxic Activity against MCF-7 and T47D Cell Lines. Molecules 2023; 28:5960. [PMID: 37630212 PMCID: PMC10459870 DOI: 10.3390/molecules28165960] [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: 06/29/2023] [Revised: 07/30/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Scoparia dulcis Linn plays an important role in treatment because it contains active compounds that are proven to have a variety of activities, including cytotoxicity on various cancer cells. The objective of this study is to isolate and identify the cytotoxic compounds in the ethyl acetate fraction of Scoparia dulcis, observe cell cycle inhibition and induction of apoptosis in vitro, and carry out molecular studies using in silico studies. A new diterpene compound was isolated from the ethyl acetate fraction of Scoparia dulcis L. of Indonesian origin. Chromatographic methods were used to isolate the compound, spectroscopic methods were used to elucidate its structure, and these data were compared with those reported in the literature. The compound was tested for its cytotoxic activity against two breast cancer cells (MCF-7 and T47D). The results of the isolated compound showed a cytotoxic effect on MCF-7 and T47D breast cancer cells at IC50 70.56 ± 1.54 and <3.125 ± 0.43 µg/mL, respectively. The compound inhibited the growth of MCF-7 and T47D breast cancer cells and the accumulation of cells in the G1 phases, and it induced apoptosis. Based on a spectroscopic analysis, the isolated compound was identified as 2α-hydroxyscopadiol, which is a new diterpenoid. A docking study revealed that the isolate's hydroxyl groups are essential for interacting with crucial residues on the active sites of the ER and PR and caspase-9. The isolate inhibits ER and PR activity with binding energies of -8.2 kcal/mol and -7.3 kcal/mol, respectively. In addition, the isolate was also able to induce apoptosis through the activation of the caspase-9 pathway with an affinity of -9.0 kcal/mol. In conclusion, the isolated compound from S. dulcis demonstrated anticancer activity based on in vitro and in silico studies.
Collapse
Affiliation(s)
- Hasnawati Hasnawati
- Faculty of Pharmacy, Universitas Halu Oleo, Kendari 93232, Indonesia;
- Doctoral Program in Pharmaceutical Science, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Subagus Wahyuono
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | - Ratna Asmah Susidarti
- Department of Chemistry Pharmaceutical, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | - Djoko Santosa
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | - Arfan Arfan
- Faculty of Pharmacy, Universitas Halu Oleo, Kendari 93232, Indonesia;
| |
Collapse
|
8
|
Lin W, Chen L, Meng W, Yang K, Wei S, Wei W, Chen J, Zhang L. C/EBPα promotes porcine pre-adipocyte proliferation and differentiation via mediating MSTRG.12568.2/FOXO3 trans-activation for STYX. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159206. [PMID: 35870701 DOI: 10.1016/j.bbalip.2022.159206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 10/17/2022]
Abstract
As a key adipogenic marker, C/EBPα (CCAAT/enhancer binding protein α) is also an important factor in regulating targets containing CCAAT element for transcription, whose products include coding and non-coding RNAs (ncRNAs). However, knowledge of the mechanism of C/EBPα affecting pre-adipocyte proliferation and adipogenesis through regulating ncRNA is still limited. In this study, we firstly conducted an investigation concerning the impact of C/EBPα knockdown on porcine pre-adipocytes by using RNA sequencing (RNA-Seq) to identify the role of key ncRNAs, especially lncRNAs and their correlated mRNAs in regulating proliferation and differentiation of porcine pre-adipocytes. 97 differentially expressed (DE) mRNAs and 4 DE lncRNAs were identified in si-C/EBPα groups compared with the si-NC groups. Meanwhile, we found C/EBPα directly target the promoter of a novel lncRNA, namely MSTRG.12568.2, which was trans-correlated with STYX (serine/threonine/tyrosine interacting protein), an important candidate gene for regulating cell proliferation. Moreover, FOXO3 (forkhead box O3) was identified as a co-regulator with MSTR.12568.2 for STYX. Overexpression and knockdown of any of the MSTRG.12568.2, STYX, and FOXO3 increased and decreased the levels of pre-adipocyte proliferation and differentiation, respectively, which demonstrated that they played a positive role in adipogenesis of pre-adipocytes. Moreover, our results revealed that FOXO3 was necessary for MSTRG.12568.2 to trans-activate STYX. We revealed that C/EBPα regulated pre-adipocyte proliferation and differentiation through mediating trans-activation of MSTRG.12568.2-FOXO3 to STYX. These results provide a novel regulation signal for C/EBPα to influence porcine pre-adipocyte proliferation and differentiation and greatly benefit to our understanding of molecular mechanism regulating subcutaneous adipogenesis.
Collapse
Affiliation(s)
- Weimin Lin
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; Sanya Institute of Nanjing Agricultural University, Sanya, Hainan 572024, China; College of Animal Science, Fujian Agriculture & Forestry University, Fuzhou, Fujian 350002, China
| | - Lei Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Wenjing Meng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Kai Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Shengjuan Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; Sanya Institute of Nanjing Agricultural University, Sanya, Hainan 572024, China.
| |
Collapse
|
9
|
La Greca A, Bellora N, Le Dily F, Jara R, Nacht AS, Quilez Oliete J, Villanueva JL, Vidal E, Merino G, Fresno C, Tarifa Reischle I, Vallejo G, Vicent GP, Fernández E, Beato M, Saragüeta P. Chromatin topology defines estradiol-primed progesterone receptor and PAX2 binding in endometrial cancer cells. eLife 2022; 11:66034. [PMID: 35018885 PMCID: PMC8887898 DOI: 10.7554/elife.66034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/11/2022] [Indexed: 11/15/2022] Open
Abstract
Estrogen (E2) and Progesterone (Pg), via their specific receptors (ERalpha and PR), are major determinants in the development and progression of endometrial carcinomas, However, their precise mechanism of action and the role of other transcription factors involved are not entirely clear. Using Ishikawa endometrial cancer cells, we report that E2 treatment exposes a set of progestin-dependent PR binding sites which include both E2 and progestin target genes. ChIP-seq results from hormone-treated cells revealed a non-random distribution of PAX2 binding in the vicinity of these estrogen-promoted PR sites. Altered expression of hormone regulated genes in PAX2 knockdown cells suggests a role for PAX2 in fine-tuning ERalpha and PR interplay in transcriptional regulation. Analysis of long-range interactions by Hi-C coupled with ATAC-seq data showed that these regions, that we call ‘progestin control regions’ (PgCRs), exhibited an open chromatin state even before hormone exposure and were non-randomly associated with regulated genes. Nearly 20% of genes potentially influenced by PgCRs were found to be altered during progression of endometrial cancer. Our findings suggest that endometrial response to progestins in differentiated endometrial tumor cells results in part from binding of PR together with PAX2 to accessible chromatin regions. What maintains these regions open remains to be studied.
Collapse
Affiliation(s)
| | - Nicolás Bellora
- National Scientific and Technical Research Council (CONICET), Institute of Nuclear Technologies for Health, Bariloche, Argentina
| | - François Le Dily
- Gene Regulation, Centre for Genomic Regulation, Barcelona, Spain
| | - Rodrigo Jara
- Biology and Experimental Medicine Institute, Buenos Aires, Argentina
| | | | | | | | - Enrique Vidal
- Gene Regulation, Centre for Genomic Regulation, Barcelona, Spain
| | - Gabriela Merino
- Bioscience Data Mining Group, Córdoba University, Córdoba, Argentina
| | - Cristóbal Fresno
- Bioscience Data Mining Group, Córdoba University, Córdoba, Argentina
| | | | - Griselda Vallejo
- Biology and Experimental Medicine Institute, Buenos Aires, Argentina
| | | | - Elmer Fernández
- Bioscience Data Mining Group, Córdoba University, Córdoba, Argentina
| | - Miguel Beato
- Gene Regulation, Centre for Genomic Regulation, Barcelona, Spain
| | | |
Collapse
|
10
|
Zaurin R, Ferrari R, Nacht AS, Carbonell J, Le Dily F, Font-Mateu J, de Llobet Cucalon LI, Vidal E, Lioutas A, Beato M, Vicent GP. A set of accessible enhancers enables the initial response of breast cancer cells to physiological progestin concentrations. Nucleic Acids Res 2021; 49:12716-12731. [PMID: 34850111 PMCID: PMC8682742 DOI: 10.1093/nar/gkab1125] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 11/14/2022] Open
Abstract
Here, we report that in T47D breast cancer cells 50 pM progestin is sufficient to activate cell cycle entry and the progesterone gene expression program. At this concentration, equivalent to the progesterone blood levels found around the menopause, progesterone receptor (PR) binds only to 2800 genomic sites, which are accessible to ATAC cleavage prior to hormone exposure. These highly accessible sites (HAs) are surrounded by well-organized nucleosomes and exhibit breast enhancer features, including estrogen receptor alpha (ERα), higher FOXA1 and BRD4 (bromodomain containing 4) occupancy. Although HAs are enriched in RAD21 and CTCF, PR binding is the driving force for the most robust interactions with hormone-regulated genes. HAs show higher frequency of 3D contacts among themselves than with other PR binding sites, indicating colocalization in similar compartments. Gene regulation via HAs is independent of classical coregulators and ATP-activated remodelers, relying mainly on MAP kinase activation that enables PR nuclear engagement. HAs are also preferentially occupied by PR and ERα in breast cancer xenografts derived from MCF-7 cells as well as from patients, indicating their potential usefulness as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Roser Zaurin
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Roberto Ferrari
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Ana Silvina Nacht
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Jose Carbonell
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Francois Le Dily
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Jofre Font-Mateu
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Lara Isabel de Llobet Cucalon
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Enrique Vidal
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Antonios Lioutas
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain
| | - Miguel Beato
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, 08003, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Guillermo P Vicent
- Center for Genomic Regulation (CRG), Barcelona, 08003, Spain.,Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, 08003, Spain
| |
Collapse
|
11
|
Feng Y, Liu X, Pauklin S. 3D chromatin architecture and epigenetic regulation in cancer stem cells. Protein Cell 2021; 12:440-454. [PMID: 33453053 PMCID: PMC8160035 DOI: 10.1007/s13238-020-00819-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/05/2020] [Indexed: 12/29/2022] Open
Abstract
Dedifferentiation of cell identity to a progenitor-like or stem cell-like state with increased cellular plasticity is frequently observed in cancer formation. During this process, a subpopulation of cells in tumours acquires a stem cell-like state partially resembling to naturally occurring pluripotent stem cells that are temporarily present during early embryogenesis. Such characteristics allow these cancer stem cells (CSCs) to give rise to the whole tumour with its entire cellular heterogeneity and thereby support metastases formation while being resistant to current cancer therapeutics. Cancer development and progression are demarcated by transcriptional dysregulation. In this article, we explore the epigenetic mechanisms shaping gene expression during tumorigenesis and cancer stem cell formation, with an emphasis on 3D chromatin architecture. Comparing the pluripotent stem cell state and epigenetic reprogramming to dedifferentiation in cellular transformation provides intriguing insight to chromatin dynamics. We suggest that the 3D chromatin architecture could be used as a target for re-sensitizing cancer stem cells to therapeutics.
Collapse
Affiliation(s)
- Yuliang Feng
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences Old Road, University of Oxford, Oxford, OX3 7LD, UK
| | - Xingguo Liu
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences Old Road, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
12
|
Huang CCF, Lingadahalli S, Morova T, Ozturan D, Hu E, Yu IPL, Linder S, Hoogstraat M, Stelloo S, Sar F, van der Poel H, Altintas UB, Saffarzadeh M, Le Bihan S, McConeghy B, Gokbayrak B, Feng FY, Gleave ME, Bergman AM, Collins C, Hach F, Zwart W, Emberly E, Lack NA. Functional mapping of androgen receptor enhancer activity. Genome Biol 2021; 22:149. [PMID: 33975627 PMCID: PMC8112059 DOI: 10.1186/s13059-021-02339-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 04/02/2021] [Indexed: 01/22/2023] Open
Abstract
Background Androgen receptor (AR) is critical to the initiation, growth, and progression of prostate cancer. Once activated, the AR binds to cis-regulatory enhancer elements on DNA that drive gene expression. Yet, there are 10–100× more binding sites than differentially expressed genes. It is unclear how or if these excess binding sites impact gene transcription. Results To characterize the regulatory logic of AR-mediated transcription, we generated a locus-specific map of enhancer activity by functionally testing all common clinical AR binding sites with Self-Transcribing Active Regulatory Regions sequencing (STARRseq). Only 7% of AR binding sites displayed androgen-dependent enhancer activity. Instead, the vast majority of AR binding sites were either inactive or constitutively active enhancers. These annotations strongly correlated with enhancer-associated features of both in vitro cell lines and clinical prostate cancer samples. Evaluating the effect of each enhancer class on transcription, we found that AR-regulated enhancers frequently interact with promoters and form central chromosomal loops that are required for transcription. Somatic mutations of these critical AR-regulated enhancers often impact enhancer activity. Conclusions Using a functional map of AR enhancer activity, we demonstrated that AR-regulated enhancers act as a regulatory hub that increases interactions with other AR binding sites and gene promoters.
Collapse
Affiliation(s)
- Chia-Chi Flora Huang
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Shreyas Lingadahalli
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Tunc Morova
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Dogancan Ozturan
- School of Medicine, Koç University, Istanbul, Turkey.,Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Eugene Hu
- Department of Physics, Simon Fraser University, Burnaby, Canada
| | - Ivan Pak Lok Yu
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Simon Linder
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marlous Hoogstraat
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Suzan Stelloo
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Funda Sar
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Henk van der Poel
- Division of Urology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Umut Berkay Altintas
- School of Medicine, Koç University, Istanbul, Turkey.,Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Mohammadali Saffarzadeh
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Stephane Le Bihan
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Brian McConeghy
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Bengul Gokbayrak
- School of Medicine, Koç University, Istanbul, Turkey.,Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Felix Y Feng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, USA
| | - Martin E Gleave
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Andries M Bergman
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Colin Collins
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Faraz Hach
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| | - Eldon Emberly
- Department of Physics, Simon Fraser University, Burnaby, Canada
| | - Nathan A Lack
- Vancouver Prostate Centre, Department of Urologic Science, University of British Columbia, Vancouver, Canada. .,School of Medicine, Koç University, Istanbul, Turkey. .,Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.
| |
Collapse
|
13
|
Guan Y, Zhang C, Lyu G, Huang X, Zhang X, Zhuang T, Jia L, Zhang L, Zhang C, Li C, Tao W. Senescence-activated enhancer landscape orchestrates the senescence-associated secretory phenotype in murine fibroblasts. Nucleic Acids Res 2020; 48:10909-10923. [PMID: 33045748 PMCID: PMC7641768 DOI: 10.1093/nar/gkaa858] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
The three-dimensional configuration of the chromatin architecture is known to be crucial for alterations in the transcriptional network; however, the underlying mechanisms of epigenetic control of senescence-related gene expression by modulating the chromatin architecture remain unknown. Here, we demonstrate frequent chromosomal compartment switching during mouse embryonic fibroblasts (MEFs) replicative senescence as characterized by senescence-inactivated (SIAEs) and -activated enhancers (SAEs) in topologically associated domains (TADs). Mechanistically, SAEs are closely correlated with senescence-associated secretory phenotype (SASP) genes, which are a key transcriptional feature of an aging microenvironment that contributes to tumor progression, aging acceleration, and immunoinflammatory responses. Moreover, SAEs can positively regulate robust changes in SASP expression. The transcription factor CCAAT/enhancer binding protein α (C/EBPα) is capable of enhancing SAE activity, which accelerates the emergence of SAEs flanking SASPs and the secretion of downstream factors, contributing to the progression of senescence. Our results provide novel insight into the TAD-related control of SASP gene expression by revealing hierarchical roles of the chromatin architecture, transcription factors, and enhancer activity in the regulation of cellular senescence.
Collapse
Affiliation(s)
- Yiting Guan
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China.,Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524000, China
| | - Chao Zhang
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China.,Center for Bioinformatics, School of Life Sciences and Center for Statistical Science, Peking University, Beijing 100871, China
| | - Guoliang Lyu
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Xiaoke Huang
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Xuebin Zhang
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Tenghan Zhuang
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Lumeng Jia
- Center for Bioinformatics, School of Life Sciences and Center for Statistical Science, Peking University, Beijing 100871, China
| | - Lijun Zhang
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Chen Zhang
- China Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Cheng Li
- Center for Bioinformatics, School of Life Sciences and Center for Statistical Science, Peking University, Beijing 100871, China
| | - Wei Tao
- Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
14
|
Kothari C, Diorio C, Durocher F. The Importance of Breast Adipose Tissue in Breast Cancer. Int J Mol Sci 2020; 21:ijms21165760. [PMID: 32796696 PMCID: PMC7460846 DOI: 10.3390/ijms21165760] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is a complex endocrine organ, with a role in obesity and cancer. Adipose tissue is generally linked to excessive body fat, and it is well known that the female breast is rich in adipose tissue. Hence, one can wonder: what is the role of adipose tissue in the breast and why is it required? Adipose tissue as an organ consists of adipocytes, an extracellular matrix (ECM) and immune cells, with a significant role in the dynamics of breast changes throughout the life span of a female breast from puberty, pregnancy, lactation and involution. In this review, we will discuss the importance of breast adipose tissue in breast development and its involvement in breast changes happening during pregnancy, lactation and involution. We will focus on understanding the biology of breast adipose tissue, with an overview on its involvement in the various steps of breast cancer development and progression. The interaction between the breast adipose tissue surrounding cancer cells and vice-versa modifies the tumor microenvironment in favor of cancer. Understanding this mutual interaction and the role of breast adipose tissue in the tumor microenvironment could potentially raise the possibility of overcoming breast adipose tissue mediated resistance to therapies and finding novel candidates to target breast cancer.
Collapse
Affiliation(s)
- Charu Kothari
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1T 1C2, Canada;
- Cancer Research Centre, CHU de Quebec Research Centre, Quebec, QC G1V 4G2, Canada;
| | - Caroline Diorio
- Cancer Research Centre, CHU de Quebec Research Centre, Quebec, QC G1V 4G2, Canada;
- Department of Preventive and Social Medicine, Faculty of Medicine, Laval University, Quebec, QC G1T 1C2, Canada
| | - Francine Durocher
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1T 1C2, Canada;
- Cancer Research Centre, CHU de Quebec Research Centre, Quebec, QC G1V 4G2, Canada;
- Correspondence: ; Tel.: +1-(418)-525-4444 (ext. 48508)
| |
Collapse
|
15
|
Beato M, Wright RHG, Dily FL. 90 YEARS OF PROGESTERONE: Molecular mechanisms of progesterone receptor action on the breast cancer genome. J Mol Endocrinol 2020; 65:T65-T79. [PMID: 32485671 PMCID: PMC7354705 DOI: 10.1530/jme-19-0266] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022]
Abstract
Gene regulation by steroid hormones has been at the forefront in elucidating the intricacies of transcriptional regulation in eukaryotes ever since the discovery by Karlson and Clever that the insect steroid hormone ecdysone induces chromatin puffs in giant chromosomes. After the successful cloning of the hormone receptors toward the end of the past century, detailed mechanistic insight emerged in some model systems, in particular the MMTV provirus. With the arrival of next generation DNA sequencing and the omics techniques, we have gained even further insight into the global cellular response to steroid hormones that in the past decades also extended to the function of the 3D genome topology. More recently, advances in high resolution microcopy, single cell genomics and the new vision of liquid-liquid phase transitions in the context of nuclear space bring us closer than ever to unravelling the logic of gene regulation and its complex integration of global cellular signaling networks. Using the function of progesterone and its cellular receptor in breast cancer cells, we will briefly summarize the history and describe the present extent of our knowledge on how regulatory proteins deal with the chromatin structure to gain access to DNA sequences and interpret the genomic instructions that enable cells to respond selectively to external signals by reshaping their gene regulatory networks.
Collapse
Affiliation(s)
- Miguel Beato
- Centre de Regulació Genomica (CRG), Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Roni H G Wright
- Centre de Regulació Genomica (CRG), Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, Spain
| | - François Le Dily
- Centre de Regulació Genomica (CRG), Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, Spain
| |
Collapse
|
16
|
Konan HP, Kassem L, Omarjee S, Surmieliova-Garnès A, Jacquemetton J, Cascales E, Rezza A, Trédan O, Treilleux I, Poulard C, Le Romancer M. ERα-36 regulates progesterone receptor activity in breast cancer. Breast Cancer Res 2020; 22:50. [PMID: 32429997 PMCID: PMC7238515 DOI: 10.1186/s13058-020-01278-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/13/2020] [Indexed: 01/12/2023] Open
Abstract
Background Alterations in estrogen and progesterone signaling, via their respective receptors, estrogen receptor alpha (ERα) and progesterone receptor (PR), respectively, are largely involved in the development of breast cancer (BC). The recent identification of ERα-36, a splice variant of ERα, has uncovered a new facet of this pathology. Although ERα-36 expression is associated with poor prognosis, metastasis development, and resistance to treatment, its predictive value has so far not been associated with a BC subtype and its mechanisms of action remain understudied. Methods To study ERα-36 expression in BC specimens, we performed immunochemical experiments. Next, the role of ERα-36 in progesterone signaling was investigated by generating KO clones using the CRISPR/CAS9 technology. PR signaling was also assessed by proximity ligation assay, Western blotting, RT-QPCR, and ChIP experiments. Finally, proliferation assays were performed with the IncuCyte technology and migration experiments using scratch assays. Results Here, we demonstrate that ERα-36 expression at the plasma membrane is correlated with a reduced disease-free survival in a cohort of 160 BC patients, particularly in PR-positive tumors, suggesting a crosstalk between ERα-36 and PR. Indeed, we show that ERα-36 interacts constitutively with PR in the nucleus of tumor cells. Moreover, it regulates PR expression and phosphorylation on key residues, impacting the biological effects of progesterone. Conclusions ERα-36 is thus a regulator of PR signaling, interfering with its transcriptional activity and progesterone-induced anti-proliferative effects as well as migratory capacity. Hence, ERα-36 may constitute a new prognostic marker as well as a potential target in PR-positive BC.
Collapse
Affiliation(s)
- Henri-Philippe Konan
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Loay Kassem
- Clinical Oncology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Soleilmane Omarjee
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Ausra Surmieliova-Garnès
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Julien Jacquemetton
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | | | | | - Olivier Trédan
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, 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
| | - Isabelle Treilleux
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,Pathology 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, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, 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, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373 Lyon Cedex 08, F-69000, Lyon, France. .,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.
| |
Collapse
|
17
|
Nacht AS, Ferrari R, Zaurin R, Scabia V, Carbonell-Caballero J, Le Dily F, Quilez J, Leopoldi A, Brisken C, Beato M, Vicent GP. C/EBPα mediates the growth inhibitory effect of progestins on breast cancer cells. EMBO J 2019; 38:e101426. [PMID: 31373033 DOI: 10.15252/embj.2018101426] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 01/19/2023] Open
Abstract
Steroid hormones are key gene regulators in breast cancer cells. While estrogens stimulate cell proliferation, progestins activate a single cell cycle followed by proliferation arrest. Here, we use biochemical and genome-wide approaches to show that progestins achieve this effect via a functional crosstalk with C/EBPα. Using ChIP-seq, we identify around 1,000 sites where C/EBPα binding precedes and helps binding of progesterone receptor (PR) in response to hormone. These regions exhibit epigenetic marks of active enhancers, and C/EBPα maintains an open chromatin conformation that facilitates loading of ligand-activated PR. Prior to hormone exposure, C/EBPα favors promoter-enhancer contacts that assure hormonal regulation of key genes involved in cell proliferation by facilitating binding of RAD21, YY1, and the Mediator complex. Knockdown of C/EBPα disrupts enhancer-promoter contacts and decreases the presence of these architectural proteins, highlighting its key role in 3D chromatin looping. Thus, C/EBPα fulfills a previously unknown function as a potential growth modulator in hormone-dependent breast cancer.
Collapse
Affiliation(s)
- A Silvina Nacht
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Roberto Ferrari
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Roser Zaurin
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Valentina Scabia
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - José Carbonell-Caballero
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Francois Le Dily
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Javier Quilez
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Alexandra Leopoldi
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Miguel Beato
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Guillermo P Vicent
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| |
Collapse
|