1
|
Melone V, Palumbo D, Palo L, Brusco N, Salvati A, Tarallo A, Giurato G, Rizzo F, Nassa G, Weisz A, Tarallo R. LncRNA PVT1 links estrogen receptor alpha and the polycomb repressive complex 2 in suppression of pro-apoptotic genes in hormone-responsive breast cancer. Cell Death Dis 2025; 16:80. [PMID: 39922814 PMCID: PMC11807188 DOI: 10.1038/s41419-025-07423-4] [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: 06/27/2024] [Revised: 01/13/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
RNA-based therapeutics highlighted novel approaches to target either coding or noncoding molecules for multiple diseases treatment. In breast cancer (BC), a multitude of deregulated long noncoding RNAs (lncRNAs) have been identified as potential therapeutic targets also in the context of antiestrogen resistance, and the RNA binding activity of the estrogen receptor α (ERα) points additional potential candidates to interfere with estrogenic signaling. A set of lncRNAs was selected among ERα-associated RNAs in BC cell nuclei due to their roles in processes such as transcriptional regulation and epigenetic chromatin modifications. Native immunoprecipitation of nuclear ERα-interacting RNAs coupled to NGS (RIP-Seq) was performed in MCF-7 cells, leading to the identification of essential lncRNAs interacting with the receptor in multi-molecular regulatory complexes. Among these, PVT1, FGD5-AS1 and EPB41L4A-AS1 were selected for further investigation. Functional assays and transcriptome analysis following lncRNA knock-down indicated PVT1 as the master modulator of some of the most relevant BC hallmarks, such as cell proliferation, apoptosis, migration and response to hypoxia. In addition, targeted experiments identified PVT1 as a key factor in the composition of PRC2-ERα network involved in downregulation of tumor suppressor genes, including BTG2.
Collapse
Affiliation(s)
- Viola Melone
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
| | - Domenico Palumbo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
| | - Luigi Palo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
- Genome Research Center for Health, 84081, Baronissi, SA, Italy
| | - Noemi Brusco
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
| | - Antonietta Tarallo
- Department of Translational Medical Sciences, Federico II University, 80131, Naples, Italy
- Telethon Institute of Genetics and Medicine, 80078, Pozzuoli, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy.
- Genome Research Center for Health, 84081, Baronissi, SA, Italy.
- Medical Genomics Program and Division of Oncology, AOU 'S. Giovanni di Dio e Ruggi d'Aragona' University of Salerno, and Rete Oncologica Campana, 84131, Salerno, Italy.
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, SA, Italy.
| |
Collapse
|
2
|
Khambata K, Raut S, Parte P, Balasinor NH. Estrogen Receptor Signaling Alters Sperm DNA Methylation Landscape in Adult Male Rats. Endocrinology 2025; 166:bqaf017. [PMID: 39865879 DOI: 10.1210/endocr/bqaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/19/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Estrogen through its receptors, ERα and ERβ, regulate various aspects of spermatogenesis and male fertility. Because the sperm epigenome is an important contributing factor to male fertility, we evaluated the effects of estrogen signaling activation through the ERs on sperm DNA methylome in adult rats. Whole genome-bisulfite sequencing in caudal sperm DNA was performed. The differentially methylated CpG (DMC) sites were validated by pyrosequencing, and the expression of differentially methylated genes (DMGs) was evaluated in testis by quantitative RT-PCR. Activation of ERα signaling brought about large-scale changes in the sperm DNA methylome compared to ERβ. There were 28074 DMCs and 5189 DMGs obtained after ERα agonist 4,4',4''-(4-Propyl-[1H] pyrazole-1,3,5-triyl) (PPT) treatment, whereas 1492 DMCs and 336 DMGs for ERβ agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN). In genic regions, most of the DMCs were intronic, followed by promoter and upstream regions. DMCs were distributed around the transcription start site and in transcription factor-binding regions, implicating their plausible role in gene expression regulation. Genes important for spermatogenesis were identified and validated which showed a similar trend of differential methylation as obtained by whole genome-bisulfite sequencing. The expression of the DMGs was also found to be altered in the testis. There was a considerable overlap (14% to 50%) of PPT DMGs with the DMGs reported to be affected in clinical conditions of male infertility. This study highlights the role of ERs in shaping the sperm epigenome and that aberrant estrogen signaling could be a contributing factor in clinical conditions of male infertility.
Collapse
Affiliation(s)
- Kushaan Khambata
- Gamete Immunobiology Department, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Sanketa Raut
- Neuroendocrinology Department, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Priyanka Parte
- Gamete Immunobiology Department, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Nafisa H Balasinor
- Neuroendocrinology Department, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| |
Collapse
|
3
|
Skrzypczak M, Wolinska E, Adaszek Ł, Ortmann O, Treeck O. Epigenetic Modulation of Estrogen Receptor Signaling in Ovarian Cancer. Int J Mol Sci 2024; 26:166. [PMID: 39796024 PMCID: PMC11720219 DOI: 10.3390/ijms26010166] [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: 11/27/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Ovarian cancer remains one of the leading causes of cancer-related deaths in women. There are several processes that are described to have a causal relationship in ovarian cancer development, progression, and metastasis formation, that occur both at the genetic and epigenetic level. One of the mechanisms involved in its pathogenesis and progression is estrogen signaling. Estrogen receptors (ER) α, ERβ, and G-protein coupled estrogen receptor 1 (GPER1), in concert with various coregulators and pioneer transcription factors, mediate the effects of estrogens primarily by the transcriptional regulation of estrogen responsive genes, thereby exerting pleiotropic effects including the regulation of cellular proliferation and apoptosis. The expression and activity of estrogen receptors and their coregulators have been demonstrated to be regulated by epigenetic mechanisms like histone modifications and DNA methylation. Here, we intend to summarize and to provide an update on the current understanding of epigenetic mechanisms regulating estrogen signaling and their role in ovarian cancer. For this purpose, we reviewed publications on this topic listed in the PubMed database. Finally, we assess to which extent drugs acting on the epigenetic level might be suitable for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Maciej Skrzypczak
- Chair and Department of Gynecology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Ewa Wolinska
- Department of Pathology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Łukasz Adaszek
- Clinic of Infectious Diseases, University of Life Sciences Lublin, 20-950 Lublin, Poland;
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany;
| | - Oliver Treeck
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany;
| |
Collapse
|
4
|
Alanazi H, Zhang Y, Fatunbi J, Luu T, Kwak-Kim J. The impact of reproductive hormones on T cell immunity; normal and assisted reproductive cycles. J Reprod Immunol 2024; 165:104295. [PMID: 39053203 DOI: 10.1016/j.jri.2024.104295] [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: 04/24/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
During pregnancy, a unique immune milieu is established systemically and locally at the maternal-fetal interface. While preparing for embryonic implantation, endometrial effectors significantly change their proportions and function, which are synchronized with hormonal changes. During assisted reproductive technology cycles, various cytokines, chemokines, and immune factors dynamically change with the altered receptor expressions on the immune effectors. Thus, the hormonal regulation of immune effectors is critical to maintaining the immune milieu. In this review, hormonal effects on T cell subsets are reviewed. Sex hormones affect T cell ontogeny and development, consequently affecting their functions. Like other T cell subsets, CD4+ T helper (Th) cells are modulated by estrogen, where low estrogen concentration promotes Th1-driven cell-mediated immunity in the uterus and in vitro by enhancing IFN-γ production, while a high estrogen level decreases it. The abundance and differentiation of T regulatory (Treg) cells are controlled by estrogen, inducing Treg expansion. Conversely, progesterone maintains immune homeostasis by balancing Th1/Th2 and Th17/Treg immunity, leading to maternal-fetal tolerance. Therefore, the understanding of the hormonal impact on various T cell subsets during the reproductive cycles is critical to improving reproductive outcomes in women with recurrent pregnancy losses, repeated implantation failures, and undergoing assisted reproductive cycles.
Collapse
Affiliation(s)
- Hallah Alanazi
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA; IVF and Reproductive Endocrinology Department, Women's Health Hospital, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Yuan Zhang
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA; Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Joy Fatunbi
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA
| | - Than Luu
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA.
| |
Collapse
|
5
|
Kafle A, Suttiprapa S, Muhammad M, Tenorio JCB, Mahato RK, Sahimin N, Loong SK. Epigenetic Biomarkers and the Wnt/β-Catenin Pathway in Opisthorchis viverrini-associated Cholangiocarcinoma: A Scoping Review on Therapeutic Opportunities. PLoS Negl Trop Dis 2024; 18:e0012477. [PMID: 39236081 PMCID: PMC11407677 DOI: 10.1371/journal.pntd.0012477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/17/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Epigenetic modifications, such as DNA methylation and histone modifications, are pivotal in regulating gene expression pathways related to inflammation and cancer. While there is substantial research on epigenetic markers in cholangiocarcinoma (CCA), Opisthorchis viverrini-induced cholangiocarcinoma (Ov-CCA) is overlooked as a neglected tropical disease (NTD) with limited representation in the literature. Considering the distinct etiological agent, pathogenic mechanisms, and pathological manifestations, epigenetic research plays a pivotal role in uncovering markers and potential targets related to the cancer-promoting and morbidity-inducing liver fluke parasite prevalent in the Great Mekong Subregion (GMS). Emerging studies highlight a predominant hypermethylation phenotype in Opisthorchis viverrini (O. viverrini) tumor tissues, underscoring the significance of abnormal DNA methylation and histone modifications in genes and their promoters as reliable targets for Ov-CCA. PRINCIPAL FINDINGS Relevant published literature was identified by searching major electronic databases using targeted search queries. This process retrieved a total of 81 peer-reviewed research articles deemed eligible for inclusion, as they partially or fully met the pre-defined selection criteria. These eligible articles underwent a qualitative synthesis and were included in the scoping review. Within these, 11 studies specifically explored Ov-CCA tissues to investigate potential epigenetic biomarkers and therapeutic targets. This subset of 11 articles provided a foundation for exploring the applications of epigenetics-based therapies and biomarkers for Ov-CCA. These articles delved into various epigenetic modifications, including DNA methylation and histone modifications, and examined genes with aberrant epigenetic changes linked to deregulated signalling pathways in Ov-CCA progression. CONCLUSIONS This review identified epigenetic changes and Wnt/β-catenin pathway deregulation as key drivers in Ov-CCA pathogenesis. Promoter hypermethylation of specific genes suggests potential diagnostic biomarkers and dysregulation of Wnt/β-catenin-modulating genes contributes to pathway activation in Ov-CCA progression. Reversible epigenetic changes offer opportunities for dynamic disease monitoring and targeted interventions. Therefore, this study underscores the importance of these epigenetic modifications in Ov-CCA development, suggesting novel therapeutic targets within disrupted signalling networks. However, additional validation is crucial for translating these novel insights into clinically applicable strategies, enhancing personalised Ov-CCA management approaches.
Collapse
Affiliation(s)
- Alok Kafle
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Mubarak Muhammad
- Department of Physiology and Graduate School, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jan Clyden B. Tenorio
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | | | - Norhidayu Sahimin
- Tropical Infectious Diseases Research & Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Shih Keng Loong
- Tropical Infectious Diseases Research & Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Rubin JB, Abou-Antoun T, Ippolito JE, Llaci L, Marquez CT, Wong JP, Yang L. Epigenetic developmental mechanisms underlying sex differences in cancer. J Clin Invest 2024; 134:e180071. [PMID: 38949020 PMCID: PMC11213507 DOI: 10.1172/jci180071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Cancer risk is modulated by hereditary and somatic mutations, exposures, age, sex, and gender. The mechanisms by which sex and gender work alone and in combination with other cancer risk factors remain underexplored. In general, cancers that occur in both the male and female sexes occur more commonly in XY compared with XX individuals, regardless of genetic ancestry, geographic location, and age. Moreover, XY individuals are less frequently cured of their cancers, highlighting the need for a greater understanding of sex and gender effects in oncology. This will be necessary for optimal laboratory and clinical cancer investigations. To that end, we review the epigenetics of sexual differentiation and its effect on cancer hallmark pathways throughout life. Specifically, we will touch on how sex differences in metabolism, immunity, pluripotency, and tumor suppressor functions are patterned through the epigenetic effects of imprinting, sex chromosome complement, X inactivation, genes escaping X inactivation, sex hormones, and life history.
Collapse
Affiliation(s)
| | | | - Joseph E. Ippolito
- Department of Radiology
- Department of Biochemistry and Molecular Biophysics
| | - Lorida Llaci
- Deartment of Genetics Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
7
|
Miller RAJ, Williams AP, Kovats S. Sex chromosome complement and sex steroid signaling underlie sex differences in immunity to respiratory virus infection. Front Pharmacol 2023; 14:1150282. [PMID: 37063266 PMCID: PMC10097973 DOI: 10.3389/fphar.2023.1150282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/08/2023] [Indexed: 04/18/2023] Open
Abstract
Epidemiological studies have revealed sex differences in the incidence and morbidity of respiratory virus infection in the human population, and often these observations are correlated with sex differences in the quality or magnitude of the immune response. Sex differences in immunity and morbidity also are observed in animal models of respiratory virus infection, suggesting differential dominance of specific immune mechanisms. Emerging research shows intrinsic sex differences in immune cell transcriptomes, epigenomes, and proteomes that may regulate human immunity when challenged by viral infection. Here, we highlight recent research into the role(s) of sex steroids and X chromosome complement in immune cells and describe how these findings provide insight into immunity during respiratory virus infection. We focus on the regulation of innate and adaptive immune cells by receptors for androgen and estrogens, as well as genes with a propensity to escape X chromosome inactivation. A deeper mechanistic knowledge of these pathways will help us to understand the often significant sex differences in immunity to endemic or pandemic respiratory pathogens such as influenza viruses, respiratory syncytial viruses and pathogenic coronaviruses.
Collapse
Affiliation(s)
- Reegan A. J. Miller
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Abigael P. Williams
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Susan Kovats
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
8
|
Ryu DY, Pang WK, Adegoke EO, Rahman MS, Park YJ, Pang MG. Abnormal histone replacement following BPA exposure affects spermatogenesis and fertility sequentially. ENVIRONMENT INTERNATIONAL 2022; 170:107617. [PMID: 36347119 DOI: 10.1016/j.envint.2022.107617] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 06/16/2023]
Abstract
Bisphenol A (BPA) is an endocrine-disrupting chemical widely distributed in the environment. Its exposure has been linked to male infertility in animals and humans due to its ability to induce epigenetic modification. Despite extensive research confirming the impact of BPA on epigenetic regulation, fundamental concerns about how BPA causes epigenetic changes and the underlying mechanism of BPA on the male reproductive system remain unresolved. Therefore, we sought to investigate the effects of BPA on epigenetic regulation and the histone-to-protamine (PRM) transition, which is fundamental process for male fertility in testes and spermatozoa by exposing male mice to BPA for 6 weeks while giving the mice in the control group corn oil by oral gavage. Our results demonstrated that the mRNA levels of the histone family and PRMs were significantly altered by BPA exposure in testes and spermatozoa. Subsequently, core histone proteins, the PRM1/PRM2 ratio, directly linked to male fertility, and transition proteins were significantly reduced. Furthermore, we discovered that BPA significantly caused abnormal histone-to-protamine replacement during spermiogenesis by increased histone variants-related to histone-to-PRM transition. The levels of histone H3 modification in the testes and DNA methylation in spermatozoa were significantly increased. Consequently, sperm concentration/motility/hyperactivation, fertilization, and early embryonic development were adversely affected as a consequence of altered signaling proteins following BPA exposure. To our knowledge, this is the first study to indicate that BPA exposure influences the histone-to-PRM transition via altering epigenetic modification and eventually causing reduced male fertility.
Collapse
Affiliation(s)
- Do-Yeal Ryu
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Won-Ki Pang
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Elikanah Olusayo Adegoke
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Md Saidur Rahman
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Yoo-Jin Park
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Myung-Geol Pang
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea.
| |
Collapse
|
9
|
Messier TL, Boyd JR, Gordon JAR, Tye CE, Page NA, Toor RH, Zaidi SK, Komm BS, Frietze S, Stein JL, Lian JB, Stein GS. Epigenetic and transcriptome responsiveness to ER modulation by tissue selective estrogen complexes in breast epithelial and breast cancer cells. PLoS One 2022; 17:e0271725. [PMID: 35862394 PMCID: PMC9302754 DOI: 10.1371/journal.pone.0271725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 07/06/2022] [Indexed: 01/07/2023] Open
Abstract
Selective estrogen receptor modulators (SERMs), including the SERM/SERD bazedoxifene (BZA), are used to treat postmenopausal osteoporosis and may reduce breast cancer (BCa) risk. One of the most persistent unresolved questions regarding menopausal hormone therapy is compromised control of proliferation and phenotype because of short- or long-term administration of mixed-function estrogen receptor (ER) ligands. To gain insight into epigenetic effectors of the transcriptomes of hormone and BZA-treated BCa cells, we evaluated a panel of histone modifications. The impact of short-term hormone treatment and BZA on gene expression and genome-wide epigenetic profiles was examined in ERαneg mammary epithelial cells (MCF10A) and ERα+ luminal breast cancer cells (MCF7). We tested individual components and combinations of 17β-estradiol (E2), estrogen compounds (EC10) and BZA. RNA-seq for gene expression and ChIP-seq for active (H3K4me3, H3K4ac, H3K27ac) and repressive (H3K27me3) histone modifications were performed. Our results show that the combination of BZA with E2 or EC10 reduces estrogen-mediated patterns of histone modifications and gene expression in MCF-7ERα+ cells. In contrast, BZA has minimal effects on these parameters in MCF10A mammary epithelial cells. BZA-induced changes in histone modifications in MCF7 cells are characterized by altered H3K4ac patterns, with changes at distal enhancers of ERα-target genes and at promoters of non-ERα bound proliferation-related genes. Notably, the ERα target gene GREB1 is the most sensitive to BZA treatment. Our findings provide direct mechanistic-based evidence that BZA induces epigenetic changes in E2 and EC10 mediated control of ERα regulatory programs to target distinctive proliferation gene pathways that restrain the potential for breast cancer development.
Collapse
Affiliation(s)
- Terri L. Messier
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Joseph R. Boyd
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Jonathan A. R. Gordon
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Coralee E. Tye
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Natalie A. Page
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Rabail H. Toor
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Sayyed K. Zaidi
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Barry S. Komm
- Komm Pharma Consulting LLC, San Francisco, CA, United States of America
| | - Seth Frietze
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States of America
| | - Janet L. Stein
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Jane B. Lian
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
| | - Gary S. Stein
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States of America
- Department of Surgery, University of Vermont, Burlington, VT, United States of America
| |
Collapse
|
10
|
Sukocheva OA, Lukina E, Friedemann M, Menschikowski M, Hagelgans A, Aliev G. The crucial role of epigenetic regulation in breast cancer anti-estrogen resistance: Current findings and future perspectives. Semin Cancer Biol 2022; 82:35-59. [PMID: 33301860 DOI: 10.1016/j.semcancer.2020.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer (BC) cell de-sensitization to Tamoxifen (TAM) or other selective estrogen receptor (ER) modulators (SERM) is a complex process associated with BC heterogeneity and the transformation of ER signalling. The most influential resistance-related mechanisms include modifications in ER expression and gene regulation patterns. During TAM/SERM treatment, epigenetic mechanisms can effectively silence ER expression and facilitate the development of endocrine resistance. ER status is efficiently regulated by specific epigenetic tools including hypermethylation of CpG islands within ER promoters, increased histone deacetylase activity in the ER promoter, and/or translational repression by miRNAs. Over-methylation of the ER α gene (ESR1) promoter by DNA methyltransferases was associated with poor prognosis and indicated the development of resistance. Moreover, BC progression and spreading were marked by transformed chromatin remodelling, post-translational histone modifications, and expression of specific miRNAs and/or long non-coding RNAs. Therefore, targeted inhibition of histone acetyltransferases (e.g. MYST3), deacetylases (e.g. HDAC1), and/or demethylases (e.g. lysine-specific demethylase LSD1) was shown to recover and increase BC sensitivity to anti-estrogens. Indicated as a powerful molecular instrument, the administration of epigenetic drugs can regain ER expression along with the activation of tumour suppressor genes, which can in turn prevent selection of resistant cells and cancer stem cell survival. This review examines recent advances in the epigenetic regulation of endocrine drug resistance and evaluates novel anti-resistance strategies. Underlying molecular mechanisms of epigenetic regulation will be discussed, emphasising the utilization of epigenetic enzymes and their inhibitors to re-program irresponsive BCs.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia.
| | - Elena Lukina
- Discipline of Biology, College of Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Markus Friedemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Albert Hagelgans
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russia; Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation; GALLY International Research Institute, San Antonio, TX, 78229, USA.
| |
Collapse
|
11
|
Sex Differences in Psychostimulant Abuse: Implications for Estrogen Receptors and Histone Deacetylases. Genes (Basel) 2022; 13:genes13050892. [PMID: 35627277 PMCID: PMC9140379 DOI: 10.3390/genes13050892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Substance abuse is a chronic pathological disorder that negatively affects many health and neurological processes. A growing body of literature has revealed gender differences in substance use. Compared to men, women display distinct drug-use phenotypes accompanied by recovery and rehabilitation disparities. These observations have led to the notion that sex-dependent susceptibilities exist along the progression to addiction. Within this scope, neuroadaptations following psychostimulant exposure are thought to be distinct for each sex. This review summarizes clinical findings and animal research reporting sex differences in the subjective and behavioral responses to cocaine, methamphetamine, and nicotine. This discussion is followed by an examination of epigenetic and molecular alterations implicated in the addiction process. Special consideration is given to histone deacetylases and estrogen receptor-mediated gene expression.
Collapse
|
12
|
High Risk-Human Papillomavirus in HNSCC: Present and Future Challenges for Epigenetic Therapies. Int J Mol Sci 2022; 23:ijms23073483. [PMID: 35408843 PMCID: PMC8998945 DOI: 10.3390/ijms23073483] [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: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC) is a highly heterogeneous group of tumors characterized by an incidence of 650,000 new cases and 350,000 deaths per year worldwide and a male to female ratio of 3:1. The main risk factors are alcohol and tobacco consumption and Human Papillomavirus (HPV) infections. HNSCC cases are divided into two subgroups, the HPV-negative (HPV−) and the HPV-positive (HPV+) which have different clinicopathological and molecular profiles. However, patients are still treated with the same therapeutic regimens. It is thus of utmost importance to characterize the molecular mechanisms underlying these differences to find new biomarkers and novel therapeutic targets towards personalized therapies. Epigenetic alterations are a hallmark of cancer and can be exploited as both promising biomarkers and potential new targets. E6 and E7 HPV oncoviral proteins besides targeting p53 and pRb, impair the expression and the activity of several epigenetic regulators. While alterations in DNA methylation patterns have been well described in HPV+ and HPV− HNSCC, accurate histone post-translational modifications (hPTMs) characterization is still missing. Herein, we aim to provide an updated overview on the impact of HPV on the hPTMs landscape in HNSCC. Moreover, we will also discuss the sex and gender bias in HNSCC and how the epigenetic machinery could be involved in this process, and the importance of taking into account sex and/or gender also in this field.
Collapse
|
13
|
Vadakedath S, Kandi V, Mohapatra RK, Pinnelli VBK, Yegurla RR, Shahapur PR, Godishala V, Natesan S, Vora KS, Sharun K, Tiwari R, Bilal M, Dhama K. Immunological aspects and gender bias during respiratory viral infections including novel Coronavirus disease-19 (COVID-19): A scoping review. J Med Virol 2021; 93:5295-5309. [PMID: 33990972 PMCID: PMC8242919 DOI: 10.1002/jmv.27081] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/19/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023]
Abstract
The human immune system is not adequately equipped to eliminate new microbes and could result in serious damage on first exposure. This is primarily attributed to the exaggerated immune response (inflammatory disease), which may prove detrimental to the host, as evidenced by SARS-CoV-2 infection. From the experiences of Novel Coronavirus Disease-19 to date, male patients are likely to suffer from high-intensity inflammation and disease severity than the female population. Hormones are considered the significant pillars of sex differences responsible for the discrepancy in immune response exhibited by males and females. Females appear to be better equipped to counter invading respiratory viral pathogens, including the novel SARS-CoV-2, than males. It can be hypothesized that females are more shielded from disease severity, probably owing to the diverse action/influence of estrogen and other sex hormones on both cellular (thymus-derived T lymphocytes) and humoral immunity (antibodies).
Collapse
Affiliation(s)
- Sabitha Vadakedath
- Department of BiochemistryPrathima Institute of Medical SciencesKarimnagarTelanganaIndia
| | - Venkataramana Kandi
- Department of MicrobiologyPrathima Institute of Medical SciencesKarimnagarTelanganaIndia
| | | | - Venkata B. K. Pinnelli
- Department of BiochemistryVydehi Institute of Medical Sciences and Research CentreBangaloreKarnatakaIndia
| | | | | | - Vikram Godishala
- Department of BiotechnologyGanapthi Degree CollegeParakalTelanganaIndia
| | - Senthilkumar Natesan
- Department of Infectious DiseasesIndian Institute of Public Health GandhinagarGanghinagarGujaratIndia
| | - Kranti S. Vora
- Department of Infectious DiseasesIndian Institute of Public Health GandhinagarGanghinagarGujaratIndia
| | - Khan Sharun
- Division of SurgeryICAR‐Indian Veterinary Research InstituteBareillyUttar PradeshIndia
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and ImmunologyCollege of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU)MathuraIndia
| | - Muhammad Bilal
- School of Life Science and Food EngineeringHuaiyin Institute of TechnologyHuaianChina
| | - Kuldeep Dhama
- Division of PathologyICAR‐Indian Veterinary Research InstituteBareillyUttar PradeshIndia
| |
Collapse
|
14
|
Núñez-Murrieta MA, Noguez P, Coria-Avila GA, García-García F, Santiago-García J, Bolado-García VE, Corona-Morales AA. Maternal behavior, novelty confrontation, and subcortical c-Fos expression during lactation period are shaped by gestational environment. Behav Brain Res 2021; 412:113432. [PMID: 34186145 DOI: 10.1016/j.bbr.2021.113432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/29/2021] [Accepted: 06/21/2021] [Indexed: 11/26/2022]
Abstract
The environmental context during gestation may modulate the postpartum variations in maternal behaviors observed within different animal species. Most of our experimental knowledge on this phenomenon and its physiological effects have been gained by confronting the pregnant mother with stressful situations, with the consensual results indicating a reduced maternal behavior and a hyper reactivity of stress-related neural paths. Here, in contrast, by exposing nulliparous rats strictly during pregnancy to a standard laboratory environment (STD) or a highly stimulating sensory and social environment (EE), we investigated the hypothesis that subjects frequently exposed to social stimuli and novel situations during pregnancy will show postpartum changes in subcortical brain areas' activity related to the processing of social stimuli and novelty, such that there will be modifications in maternal behavior. We found that EE mothers doubled the levels of licking and grooming, and active hovering over pups during the first postpartum week than STD dams, without a difference in the time of contact with the pups. Associated with these behaviors, EE dams showed increased c-Fos immunoreaction in hypothalamic nuclei and distinct responses in amygdalar nuclei, than STD dams. In the maternal defensive test, EE dams tripled the levels of aggressive behaviors of the STD rats. Additionally, in two different tests, EE mothers showed lower levels of postpartum anxiety-like behaviors when confronted with novel situations. Our results demonstrate that the activity of brain areas related to social behavior is adaptable by environmental circumstances experienced during gestation, presumably to prepare the progeny for these particular conditions.
Collapse
Affiliation(s)
| | - Paula Noguez
- Doctorado en Ciencias Biomédicas, Instituto de Fisiología Celular, UNAM, Mexico.
| | - Genaro A Coria-Avila
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Ver., Mexico.
| | - Fabio García-García
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Ver., Mexico.
| | - Juan Santiago-García
- Instituto de Investigaciones Biológicas, Universidad Veracruzana, Xalapa, Ver., Mexico.
| | - Victoria E Bolado-García
- Laboratorio de Investigación Genómica y Fisiológica, Facultad de Nutrición, Universidad Veracruzana, Xalapa, Ver., Mexico.
| | - Aleph A Corona-Morales
- Laboratorio de Investigación Genómica y Fisiológica, Facultad de Nutrición, Universidad Veracruzana, Xalapa, Ver., Mexico.
| |
Collapse
|
15
|
Darbre PD. Endocrine disrupting chemicals and breast cancer cells. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 92:485-520. [PMID: 34452695 DOI: 10.1016/bs.apha.2021.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Many hundreds of endocrine disrupting chemicals (EDCs) have been measured as entering human breast tissue from a range of environmental sources, and this review focuses on discussion of mechanisms by which such EDCs may be contributing to the globally rising incidence of breast cancer. Many of the distinguishing features of breast cancer may be accounted for by EDC exposure, including, but not limited to, the fact that many EDCs possess estrogenic activity and exposure to estrogen is a main risk factor for breast cancer. Studies of the actions of EDCs in human breast cancer cells are aided by use of the conceptual framework of the hallmarks of cancer, and, acting by a variety of genomic and nongenomic mechanisms, EDCs have now been shown to enable all the hallmarks of cancer to develop in human breast cancer cells. Many studies report that hallmarks can develop at concentrations which are within the range of those measured in human breast tissues, especially when added as mixtures. The varied levels of different EDCs measured in individual breast tissue samples together with the overlapping and complementary mechanisms of action of the EDCs imply that thematic mechanisms will be driven inevitably by different chemical mixtures. Despite the complexity, EDCs do need to now be acknowledged as a risk factor for breast cancer in order for preventative strategies to include reduction in EDC exposure.
Collapse
Affiliation(s)
- Philippa D Darbre
- School of Biological Sciences, University of Reading, Reading, United Kingdom.
| |
Collapse
|
16
|
Zhang FL, Kong L, Zhao AH, Ge W, Yan ZH, Li L, De Felici M, Shen W. Inflammatory cytokines as key players of apoptosis induced by environmental estrogens in the ovary. ENVIRONMENTAL RESEARCH 2021; 198:111225. [PMID: 33971129 DOI: 10.1016/j.envres.2021.111225] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/02/2021] [Accepted: 04/22/2021] [Indexed: 06/12/2023]
Abstract
Natural and synthetic environmental estrogens (EEs), interfering with the physiological functions of the body's estrogens, are widespread and are rising much concern for their possible deleterious effects on human and animal health, in particular on reproduction. In fact, increasing evidence indicate that EEs can be responsible for a variety of disfunctions of the reproductive system especially in females such as premature ovarian insufficiency (POI). Because of their great structural diversity, the modes of action of EEs are controversial. One important way through which EEs exert their effects on reproduction is the induction of apoptosis in the ovary. In general, EEs can exert pro-and anti-apoptotic effects by agonizing or antagonizing numerous estrogen-dependent signaling pathways. In the present work, results concerning apoptotic pathways and diseases induced by representative EEs (such as zearalenone, bisphenol A and di-2-ethylhexyl phthalate), in ovaries throughout development are presented into an integrated network. By reviewing and elaborating these studies, we propose inflammatory factors, centered on the production of tumor necrosis factor (TNF), as a major cause of the induction of apoptosis by EEs in the mammalian ovary. As a consequence, potential strategies to prevent such EE effect are suggested.
Collapse
Affiliation(s)
- Fa-Li Zhang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Li Kong
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ai-Hong Zhao
- Qingdao Academy of Agricultural Sciences, Qingdao, 266100, China
| | - Wei Ge
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zi-Hui Yan
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Lan Li
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy.
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
17
|
Quatrini L, Ricci B, Ciancaglini C, Tumino N, Moretta L. Regulation of the Immune System Development by Glucocorticoids and Sex Hormones. Front Immunol 2021; 12:672853. [PMID: 34248954 PMCID: PMC8260976 DOI: 10.3389/fimmu.2021.672853] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
Through the release of hormones, the neuro-endocrine system regulates the immune system function promoting adaptation of the organism to the external environment and to intrinsic physiological changes. Glucocorticoids (GCs) and sex hormones not only regulate immune responses, but also control the hematopoietic stem cell (HSC) differentiation and subsequent maturation of immune cell subsets. During the development of an organism, this regulation has long-term consequences. Indeed, the effects of GC exposure during the perinatal period become evident in the adulthood. Analogously, in the context of HSC transplantation (HSCT), the immune system development starts de novo from the donor HSCs. In this review, we summarize the effects of GCs and sex hormones on the regulation of HSC, as well as of adaptive and innate immune cells. Moreover, we discuss the short and long-term implications on hematopoiesis of sex steroid ablation and synthetic GC administration upon HSCT.
Collapse
Affiliation(s)
- Linda Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Biancamaria Ricci
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Cecilia Ciancaglini
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicola Tumino
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
18
|
The G-Protein-Coupled Estrogen Receptor (GPER) Regulates Trimethylation of Histone H3 at Lysine 4 and Represses Migration and Proliferation of Ovarian Cancer Cells In Vitro. Cells 2021; 10:cells10030619. [PMID: 33799631 PMCID: PMC8001910 DOI: 10.3390/cells10030619] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Histone H3 lysine 4 trimethylation (H3K4me3) is one of the most recognized epigenetic regulators of transcriptional activity representing, an epigenetic modification of Histone H3. Previous reports have suggested that the broad H3K4me3 domain can be considered as an epigenetic signature for tumor-suppressor genes in human cells. G-protein-coupled estrogen receptor (GPER), a new membrane-bound estrogen receptor, acts as an inhibitor on cell growth via epigenetic regulation in breast and ovarian cancer cells. This study was conducted to evaluate the relationship of GPER and H3K4me3 in ovarian cancer tissue samples as well as in two different cell lines (Caov3 and Caov4). Silencing of GPER by a specific siRNA and two selective regulators with agonistic (G1) and antagonistic (G15) activity were applied for consecutive in vitro studies to investigate their impacts on tumor cell growth and the changes in phosphorylated ERK1/2 (p-ERK1/2) and H3K4me3. We found a positive correlation between GPER and H3K4me3 expression in ovarian cancer patients. Patients overexpressing GPER as well as H3K4me3 had significantly improved overall survival. Increased H3K4me3 and p-ERK1/2 levels and attenuated cell proliferation and migration were observed in Caov3 and Caov4 cells via activation of GPER by G1. Conversely, antagonizing GPER activity by G15 resulted in opposite effects in the Caov4 cell line. In conclusion, interaction of GPER and H3K4me3 appears to be of prognostic significance for ovarian cancer patients. The results of the in vitro analyses confirm the biological rationale for their interplay and identify GPER agonists, such as G1, as a potential therapeutic approach for future investigations.
Collapse
|
19
|
Shepherd R, Cheung AS, Pang K, Saffery R, Novakovic B. Sexual Dimorphism in Innate Immunity: The Role of Sex Hormones and Epigenetics. Front Immunol 2021; 11:604000. [PMID: 33584674 PMCID: PMC7873844 DOI: 10.3389/fimmu.2020.604000] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022] Open
Abstract
Sexual dimorphism refers to differences between biological sexes that extend beyond sexual characteristics. In humans, sexual dimorphism in the immune response has been well demonstrated, with females exhibiting lower infection rates than males for a variety of bacterial, viral, and parasitic pathogens. There is also a substantially increased incidence of autoimmune disease in females compared to males. Together, these trends indicate that females have a heightened immune reactogenicity to both self and non-self-molecular patterns. However, the molecular mechanisms driving the sexually dimorphic immune response are not fully understood. The female sex hormones estrogen and progesterone, as well as the male androgens, such as testosterone, elicit direct effects on the function and inflammatory capacity of immune cells. Several studies have identified a sex-specific transcriptome and methylome, independent of the well-described phenomenon of X-chromosome inactivation, suggesting that sexual dimorphism also occurs at the epigenetic level. Moreover, distinct alterations to the transcriptome and epigenetic landscape occur in synchrony with periods of hormonal change, such as puberty, pregnancy, menopause, and exogenous hormone therapy. These changes are also mirrored by changes in immune cell function. This review will outline the evidence for sex hormones and pregnancy-associated hormones as drivers of epigenetic change, and how this may contribute to the sexual dimorphism. Determining the effects of sex hormones on innate immune function is important for understanding sexually dimorphic autoimmune diseases, sex-specific responses to pathogens and vaccines, and how innate immunity is altered during periods of hormonal change (endogenous or exogenous).
Collapse
Affiliation(s)
- Rebecca Shepherd
- Epigenetics Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Ada S. Cheung
- Department of Medicine (Austin Health), The University of Melbourne, Parkville, VIC, Australia
- Department of Endocrinology, Austin Health, Heidelberg, VIC, Australia
| | - Ken Pang
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Brain and Mitochondrial Research, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Adolescent Medicine, Royal Children’s Hospital, Parkville, VIC, Australia
| | - Richard Saffery
- Epigenetics Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Boris Novakovic
- Epigenetics Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
20
|
Liu XR, Zhang RY, Gong H, Rugo HS, Chen LB, Fu Y, Che JW, Tie J, Shao B, Wan FL, Kong WY, Song GH, Jiang HF, Xu GB, Li HP. Methylome Variation Predicts Exemestane Resistance in Advanced ER + Breast Cancer. Technol Cancer Res Treat 2020; 19:1533033819896331. [PMID: 32129154 PMCID: PMC7057408 DOI: 10.1177/1533033819896331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: More than 30% of estrogen receptor-positive breast cancers are resistant to primary
hormone therapy, and about 40% that initially respond to hormone therapy eventually
acquire resistance. Although the mechanisms of hormone therapy resistance remain
unclear, aberrant DNA methylation has been implicated in oncogenesis and drug
resistance. Purpose: We investigated the relationship between methylome variations in circulating tumor DNA
and exemestane resistance, to track hormone therapy efficacy. Methods: We prospectively recruited 16 patients who were receiving first-line therapy in our
center. All patients received exemestane-based hormone therapy after enrollment. We
collected blood samples at baseline, first follow-up (after 2 therapeutic cycles) and at
detection of disease progression. Disease that progressed within 6 months under
exemestane treatment was considered exemestane resistance but was considered relatively
exemestane-sensitive otherwise. We obtained circulating tumor DNA-derived methylomes
using the whole-genome bisulfide sequencing method. Methylation calling was done by
BISMARK software; differentially methylated regions for exemestane resistance were
calculated afterward. Results: Median follow-up for the 16 patients was 19.0 months. We found 7 exemestane
resistance-related differentially methylated regions, located in different chromosomes,
with both significantly different methylation density and methylation ratio. Baseline
methylation density and methylation ratio of chromosome 6 [32400000-32599999] were both
high in exemestane resistance. High baseline methylation ratios of chromosome 3
[67800000-67999999] (P = .013), chromosome 3 [140200000-140399999]
(P = .037), and chromosome 12 [101200000-101399999]
(P = .026) could also predict exemestane resistance. During
exemestane treatment, synchronized changes in methylation density and methylation ratio
in chromosome 6 [32400000-32599999] could accurately stratify patients in terms of
progression-free survival (P = .000033). Cutoff values of methylation
density and methylation ratio for chromosome 6 [149600000-149799999] were 0.066 and
0.076, respectively. Conclusion: Methylation change in chromosome 6 [149600000-149799999] is an ideal predictor of
exemestane resistance with great clinical potential.
Collapse
Affiliation(s)
- Xiao-Ran Liu
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China.,The authors contributed eually to the article
| | - Ru-Yan Zhang
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China.,The authors contributed eually to the article
| | - Hao Gong
- M3 Genomics, Inc, Guangzhou, Guangdong, China.,The authors contributed eually to the article
| | - Hope S Rugo
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, CA, USA
| | | | - Yuan Fu
- M3 Genomics, Inc, Guangzhou, Guangdong, China
| | - Jian-Wei Che
- M3 Genomics, Inc, Guangzhou, Guangdong, China.,Life Healthcare Group Ltd, Beijing, China
| | - Jian Tie
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Bin Shao
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Feng-Ling Wan
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei-Yao Kong
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Guo-Hong Song
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Han-Fang Jiang
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Guo-Bing Xu
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Hui-Ping Li
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
21
|
Chen Z, Wang L, Ke J, Xiao D. Effects of Estrogen in Gender-dependent Fetal Programming of Adult Cardiovascular Dysfunction. Curr Vasc Pharmacol 2020; 17:147-152. [PMID: 29493455 DOI: 10.2174/1570161116666180301142453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/15/2018] [Accepted: 02/21/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Epidemiological studies and experimental studies have demonstrated that intrauterine adverse environment increases the risk of Cardiovascular Disease (CVD) in adulthood. However, whether an individual develops a cardiovascular dysfunctional phenotype may depend on genetic background, age, and sex. METHODS In this review, we summarize some of the recent experimental animal studies in the developmental programming of adult CVD with an emphasis on sex differences and the potential role of estrogen in mediating sexual dimorphism. RESULTS Few epidemiological studies report the effect of sex on the developmental programming of CVD. However, numerous experimental animal studies have shown a sex difference in fetal programming of adult cardiovascular dysfunction. Most of the animal studies indicate that male offspring develop cardiovascular dysfunction and CVD in adulthood, whereas adult females appear to be protected. Estrogen is one of the key factors that contributes to the sex difference of adult CVD. Estrogen/its Receptor (ER) may interact with the RAS system by changes of DNA methylation patterns at the target gene promoter, serve as an antioxidant to counteract the prenatal insults-induced heightened ROS, and function as an eNOS activator to increase vasodilation, resulting in the protection of female offspring from the development of hypertension and other CVDs. CONCLUSION These studies suggest that estrogen/ER may contribute to sex differences in cardiovascular response to an adverse intrauterine environment and play a significant role in modulating the cardiovascular response in adulthood.
Collapse
Affiliation(s)
- Zewen Chen
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States.,Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong, China
| | - Lei Wang
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States
| | - Jun Ke
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States.,Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong, China
| | - Daliao Xiao
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States
| |
Collapse
|
22
|
Dworatzek E, Mahmoodzadeh S, Schriever C, Kusumoto K, Kramer L, Santos G, Fliegner D, Leung YK, Ho SM, Zimmermann WH, Lutz S, Regitz-Zagrosek V. Sex-specific regulation of collagen I and III expression by 17β-Estradiol in cardiac fibroblasts: role of estrogen receptors. Cardiovasc Res 2020; 115:315-327. [PMID: 30016401 DOI: 10.1093/cvr/cvy185] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 07/12/2018] [Indexed: 12/23/2022] Open
Abstract
Aims Sex differences in cardiac fibrosis point to the regulatory role of 17β-Estradiol (E2) in cardiac fibroblasts (CF). We, therefore, asked whether male and female CF in rodent and human models are differentially susceptible to E2, and whether this is related to sex-specific activation of estrogen receptor alpha (ERα) and beta (ERβ). Methods and results In female rat CF (rCF), 24 h E2-treatment (10-8 M) led to a significant down-regulation of collagen I and III expression, whereas both collagens were up-regulated in male rCF. E2-induced sex-specific collagen regulation was also detected in human CF, indicating that this regulation is conserved across species. Using specific ERα- and ERβ-agonists (10-7 M) for 24 h, we identified ERα as repressive and ERβ as inducing factor in female and male rCF, respectively. In addition, E2-induced ERα phosphorylation at Ser118 only in female rCF, whereas Ser105 phosphorylation of ERβ was exclusively found in male rCF. Further, in female rCF we found both ER bound to the collagen I and III promoters using chromatin immunoprecipitation assays. In contrast, in male rCF only ERβ bound to both promoters. In engineered connective tissues (ECT) from rCF, collagen I and III mRNA were down-regulated in female ECT and up-regulated in male ECT by E2. This was accompanied by an impaired condensation of female ECT, whereas male ECT showed an increased condensation and stiffness upon E2-treatment, analysed by rheological measurements. Finally, we confirmed the E2-effect on both collagens in an in vivo mouse model with ovariectomy for E2 depletion, E2 substitution, and pressure overload by transverse aortic constriction. Conclusion The mechanism underlying the sex-specific regulation of collagen I and III in the heart appears to involve E2-mediated differential ERα and ERβ signaling in CFs.
Collapse
Affiliation(s)
- Elke Dworatzek
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Shokoufeh Mahmoodzadeh
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Cindy Schriever
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kana Kusumoto
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany
| | - Lisa Kramer
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Gabriela Santos
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK, partner site Göttingen, Göttingen, Germany
| | | | - Yuet-Kin Leung
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Shuk-Mei Ho
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK, partner site Göttingen, Göttingen, Germany
| | - Susanne Lutz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK, partner site Göttingen, Göttingen, Germany
| | - Vera Regitz-Zagrosek
- Charité-Universitätsmedizin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
23
|
Zhou Z, Xu B, Minn A, Zhang NR. DENDRO: genetic heterogeneity profiling and subclone detection by single-cell RNA sequencing. Genome Biol 2020; 21:10. [PMID: 31937348 PMCID: PMC6961311 DOI: 10.1186/s13059-019-1922-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/16/2019] [Indexed: 12/18/2022] Open
Abstract
Although scRNA-seq is now ubiquitously adopted in studies of intratumor heterogeneity, detection of somatic mutations and inference of clonal membership from scRNA-seq is currently unreliable. We propose DENDRO, an analysis method for scRNA-seq data that clusters single cells into genetically distinct subclones and reconstructs the phylogenetic tree relating the subclones. DENDRO utilizes transcribed point mutations and accounts for technical noise and expression stochasticity. We benchmark DENDRO and demonstrate its application on simulation data and real data from three cancer types. In particular, on a mouse melanoma model in response to immunotherapy, DENDRO delineates the role of neoantigens in treatment response.
Collapse
Affiliation(s)
- Zilu Zhou
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA USA
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA USA
| | - Bihui Xu
- Department of Radiation Oncology, Parker Institute for Cancer Immunotherapy, Abramson Family Cancer Research Institute, Graduate Group in Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Andy Minn
- Department of Radiation Oncology, Parker Institute for Cancer Immunotherapy, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Nancy R. Zhang
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
24
|
Brown MA, Su MA. An Inconvenient Variable: Sex Hormones and Their Impact on T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2020; 202:1927-1933. [PMID: 30885988 DOI: 10.4049/jimmunol.1801403] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/05/2018] [Indexed: 12/15/2022]
Abstract
Epidemiologic data demonstrate sex differences in autoimmune diseases, immune responses against infection, and antitumor immunity, and accumulating evidence suggests a major role for sex hormones in mediating these differences. In this study, we review recent advances in understanding how sex hormones regulate T cell responses to alter susceptibility to autoimmunity. Although sex hormones can directly alter gene transcriptional programs of T cells, we focus in this study on how sex hormones alter T cell development and function through their effects on thymic stromal cells and innate cell types. In addition to contributing to our understanding of sex differences, these findings also have implications for the therapeutic use of sex hormones and sex hormone modulators, which are now being prescribed to increasing numbers of patients for a wide variety of indications.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Maureen A Su
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, College of Life Sciences, University of California at Los Angeles, Los Angeles, CA 90095; and .,Department of Pediatrics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
25
|
Guo WY, Zeng SMZ, Deora GS, Li QS, Ruan BF. Estrogen Receptor α (ERα)-targeting Compounds and Derivatives: Recent Advances in Structural Modification and Bioactivity. Curr Top Med Chem 2019; 19:1318-1337. [PMID: 31215379 DOI: 10.2174/1568026619666190619142504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 04/29/2019] [Accepted: 05/05/2019] [Indexed: 12/17/2022]
Abstract
Breast cancer is the most common cancer suffered by female, and the second highest cause of cancer-related death among women worldwide. At present, hormone therapy is still the main treatment route and can be divided into three main categories: selective estrogen receptor modulators (SERMs), selective estrogen receptor downregulators (SERDs), and aromatase inhibitors (AIs). However, breast cancer is difficult to cure even after several rounds of anti-estrogen therapy and most drugs have serious side-effects. Here, we review the literature published over the past five years regarding the isolation and synthesis of analogs and their derivatives.
Collapse
Affiliation(s)
- Wei-Yun Guo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Shang-Ming-Zhu Zeng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Girdhar Singh Deora
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Qing-Shan Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Ban-Feng Ruan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| |
Collapse
|
26
|
Widelitz RB, Lin GW, Lai YC, Mayer JA, Tang PC, Cheng HC, Jiang TX, Chen CF, Chuong CM. Morpho-regulation in diverse chicken feather formation: Integrating branching modules and sex hormone-dependent morpho-regulatory modules. Dev Growth Differ 2018; 61:124-138. [PMID: 30569461 DOI: 10.1111/dgd.12584] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022]
Abstract
Many animals can change the size, shape, texture and color of their regenerated coats in response to different ages, sexes, or seasonal environmental changes. Here, we propose that the feather core branching morphogenesis module can be regulated by sex hormones or other environmental factors to change feather forms, textures or colors, thus generating a large spectrum of complexity for adaptation. We use sexual dimorphisms of the chicken to explore the role of hormones. A long-standing question is whether the sex-dependent feather morphologies are autonomously controlled by the male or female cell types, or extrinsically controlled and reversible. We have recently identified core feather branching molecular modules which control the anterior-posterior (bone morphogenetic orotein [BMP], Wnt gradient), medio-lateral (Retinoic signaling, Gremlin), and proximo-distal (Sprouty, BMP) patterning of feathers. We hypothesize that morpho-regulation, through quantitative modulation of existing parameters, can act on core branching modules to topologically tune the dimension of each parameter during morphogenesis and regeneration. Here, we explore the involvement of hormones in generating sexual dimorphisms using exogenously delivered hormones. Our strategy is to mimic male androgen levels by applying exogenous dihydrotestosterone and aromatase inhibitors to adult females and to mimic female estradiol levels by injecting exogenous estradiol to adult males. We also examine differentially expressed genes in the feathers of wildtype male and female chickens to identify potential downstream modifiers of feather morphogenesis. The data show male and female feather morphology and their color patterns can be modified extrinsically through molting and resetting the stem cell niche during regeneration.
Collapse
Affiliation(s)
- Randall B Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Gee-Way Lin
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Chih Lai
- Department of Pathology, University of Southern California, Los Angeles, California.,Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Julie A Mayer
- Department of Pathology, University of Southern California, Los Angeles, California.,Biocept Inc., San Diego, California
| | - Pin-Chi Tang
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Hsu-Chen Cheng
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Ting-Xin Jiang
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Chih-Feng Chen
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
27
|
Yang GJ, Lei PM, Wong SY, Ma DL, Leung CH. Pharmacological Inhibition of LSD1 for Cancer Treatment. Molecules 2018; 23:E3194. [PMID: 30518104 PMCID: PMC6320820 DOI: 10.3390/molecules23123194] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Lysine-specific demethylase 1A (LSD1, also named KDM1A) is a demethylase that can remove methyl groups from histones H3K4me1/2 and H3K9me1/2. It is aberrantly expressed in many cancers, where it impedes differentiation and contributes to cancer cell proliferation, cell metastasis and invasiveness, and is associated with inferior prognosis. Pharmacological inhibition of LSD1 has been reported to significantly attenuate tumor progression in vitro and in vivo in a range of solid tumors and acute myeloid leukemia. This review will present the structural aspects of LSD1, its role in carcinogenesis, a comparison of currently available approaches for screening LSD1 inhibitors, a classification of LSD1 inhibitors, and its potential as a drug target in cancer therapy.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| | - Pui-Man Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| | - Suk-Yu Wong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China.
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
28
|
Sellers KJ, Watson IA, Gresz RE, Raval P, Srivastava DP. Cyto-nuclear shuttling of afadin is required for rapid estradiol-mediated modifications of histone H3. Neuropharmacology 2018; 143:153-162. [PMID: 30268521 PMCID: PMC6277849 DOI: 10.1016/j.neuropharm.2018.09.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/18/2018] [Accepted: 09/25/2018] [Indexed: 12/19/2022]
Abstract
Estrogens have been shown to rapidly regulate local signalling at synapses and within the nucleus. The result of these signalling events is to rapidly modulate synapse structure and function, as well as epigenetic mechanisms including histone modifications. Ultimately these mechanisms are thought to contribute to long-lasting changes in neural circuitry, and thus influence cognitive functions such as learning and memory. However, the mechanisms by which estrogen-mediated local synaptic and nuclear signalling events are coordinated are not well understood. In this study we have found that the scaffold protein afadin, (also known as AF-6), undergoes a bi-directional trafficking to both synaptic and nuclear compartment in response to acute 17β-estradiol (estradiol) treatment, in mixed sex neuronal cultures derived from fetal cortex. Interestingly, nuclear accumulation of afadin was coincidental with an increase in the phosphorylation of histone H3 at serine 10 (H3S10p). This epigenetic modification is associated with the remodeling of chromatin into an open euchromatin state, allowing for transcriptional activation and related learning and memory processes. Critically, the cyto-nuclear trafficking of afadin was required for estradiol-dependent H3S10p. We further determined that nuclear accumulation of afadin is sufficient to induce phosphorylation of the mitogentic kinases ERK1/2 (pERK1/2) within the nucleus. Moreover, nuclear pERK1/2 was required for estradiol-dependent H3S10p. Taken together, we propose a model whereby estradiol induces the bi-directional trafficking of afadin to synaptic and nuclear sub-compartments. Within the nucleus, afadin is required for increased pERK1/2 which in turn is required for H3S10p. Therefore this represents a mechanism through which estrogens may be able to coordinate both synaptic and nucleosomal events within the same neuronal population. 17β-estradiol targets afadin to membrane and nuclear subcompartments. Histone H3 is rapidly phosphorylated by 17β-estradiol. Histone H3 phosphorylation by 17β-estradiol requires afadin nuclear accumulation. 17β-estradiol-mediated ERK1/2 activation is required for histone H3 phosphorylation.
Collapse
Affiliation(s)
- Katherine J Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Iain A Watson
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Rahel E Gresz
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK.
| |
Collapse
|
29
|
Ghosh K, Chatterjee B, Jayaprasad AG, Kanade SR. The persistent organochlorine pesticide endosulfan modulates multiple epigenetic regulators with oncogenic potential in MCF-7 cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 624:1612-1622. [PMID: 29054638 DOI: 10.1016/j.scitotenv.2017.10.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/28/2017] [Accepted: 10/07/2017] [Indexed: 06/07/2023]
Abstract
Environmental cues and chemicals can potentially modulate the phenotypic expression of genome through alterations in the epigenetic mechanisms. Endosulfan is one of the extensively used organochlorine pesticides around the world which is known for its endocrine, neuro- and reproductive toxicity. This study was aimed to investigate the potential of α-endosulfan in modulation of multiple epigenetic enzymes in MCF-7 cells. The cells were treated with DMSO (control) or α-endosulfan (1 and 10μM) and the expression of various epigenetic enzymes was assayed by real-time PCR and immunoblotting, in addition to their activity assays. The results shows α-endosulfan, at 1 and 10μM concentration, significantly promoted viability of MCF-7 cells compared to untreated cells after 24h. The expression of DNA methyltransferases (DNMTs) was upregulated while the global DNA methylation status was initially affected, but later recovered. Total intracellular histone deacetylase (HDAC) activity was found to be significantly increased which was correlated with upregulation of class I HDACs (HDAC 1 and 3) while no significant alteration in the other HDAC classes was observed. The expression and activity of arginine and lysine methylation enzymes, protein arginine methyltransferase 5 (PRMT5) and Enhancer of Zeste homolog 2 (EZH2), respectively, were also found to be modulated by α-endosulfan. We found increased expression of histones H3 and H4, trimethylated H3K27 (product of EZH2), symmetric dimethylation of H4R3 (product of PRMT5) and five different (unidentified) proteins whose arginine residues are symmetrically dimethylated (by increased level of PRMT5) were enhanced in response to 10μM α-endosulfan after 24h exposure window. Moreover, overexpression of basal level of estrogen receptor alpha (ERα), suggests estrogenicity of α-endosulfan. In summary, our results shows modulatory impact of α-endosulfan on multiple cellular epigenetic regulators, known to possess oncogenic potential which might contribute to mechanistic insight of its action in future.
Collapse
Affiliation(s)
- Krishna Ghosh
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod 671314, Kerala, India
| | - Biji Chatterjee
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod 671314, Kerala, India
| | - Aparna Geetha Jayaprasad
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod 671314, Kerala, India
| | - Santosh R Kanade
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod 671314, Kerala, India.
| |
Collapse
|
30
|
Michalczyk AA, Janus ED, Judge A, Ebeling PR, Best JD, Ackland MJ, Asproloupos D, Dunbar JA, Ackland ML. Transient epigenomic changes during pregnancy and early postpartum in women with and without type 2 diabetes. Epigenomics 2018; 10:419-431. [PMID: 29561170 DOI: 10.2217/epi-2017-0129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
AIM To investigate epigenomic changes in pregnancy and early postpartum in women with and without type 2 diabetes. METHODS Dimethylation of histones H3K4, H3K9, H3K27, H3K36 and H3K79 was measured in white blood cells of women at 30 weeks pregnancy, at 8-10 and 20 weeks postpartum and in never-pregnant women. RESULTS Dimethylation levels of all five histones were different between women in pregnancy and early postpartum compared with never-pregnant women and were different between women with and without type 2 diabetes. CONCLUSION Histone methylation changes are transient in pregnancy and early postpartum and may represent normal physiological responses to hormones. Different epigenomic profiles in women with type 2 diabetes mellitus may correlate with hormonal responses, leading to high risk pregnancy outcomes.
Collapse
Affiliation(s)
- Agnes A Michalczyk
- Centre for Cellular & Molecular Biology, School of Life & Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia
| | - Edward D Janus
- University of Melbourne, Western Centre for Health Research & Education, Western Health, St Albans VIC 3021, Australia.,General Internal Medicine Unit, Western Health, Sunshine Hospital, St Albans, VIC 3021, Australia
| | - Alisha Judge
- Centre for Cellular & Molecular Biology, School of Life & Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3168, Australia
| | - James D Best
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
| | - Michael J Ackland
- The Alfred Centre, Monash University, Melbourne, Victoria 3004, Australia
| | - Dino Asproloupos
- Centre for Population Health Research, Faculty of Health, Deakin University, Burwood, Victoria 3125, Australia
| | - James A Dunbar
- Centre for Population Health Research, Faculty of Health, Deakin University, Burwood, Victoria 3125, Australia
| | - M Leigh Ackland
- Centre for Cellular & Molecular Biology, School of Life & Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia
| |
Collapse
|
31
|
Singla H, Ludhiadch A, Kaur RP, Chander H, Kumar V, Munshi A. Recent advances in HER2 positive breast cancer epigenetics: Susceptibility and therapeutic strategies. Eur J Med Chem 2017; 142:316-327. [DOI: 10.1016/j.ejmech.2017.07.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/27/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
32
|
Abstract
Multiple mechanisms of epigenetic control that include DNA methylation, histone modification, noncoding RNAs, and mitotic gene bookmarking play pivotal roles in stringent gene regulation during lineage commitment and maintenance. Experimental evidence indicates that bivalent chromatin domains, i.e., genome regions that are marked by both H3K4me3 (activating) and H3K27me3 (repressive) histone modifications, are a key property of pluripotent stem cells. Bivalency of developmental genes during the G1 phase of the pluripotent stem cell cycle contributes to cell fate decisions. Recently, some cancer types have been shown to exhibit partial recapitulation of bivalent chromatin modifications that are lost along with pluripotency, suggesting a mechanism by which cancer cells reacquire properties that are characteristic of undifferentiated, multipotent cells. This bivalent epigenetic control of oncofetal gene expression in cancer cells may offer novel insights into the onset and progression of cancer and may provide specific and selective options for diagnosis as well as for therapeutic intervention.
Collapse
|
33
|
Abdel-Hafiz HA. Epigenetic Mechanisms of Tamoxifen Resistance in Luminal Breast Cancer. Diseases 2017; 5:E16. [PMID: 28933369 PMCID: PMC5622332 DOI: 10.3390/diseases5030016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/28/2017] [Accepted: 06/30/2017] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is one of the most common cancers and the second leading cause of cancer death in the United States. Estrogen receptor (ER)-positive cancer is the most frequent subtype representing more than 70% of breast cancers. These tumors respond to endocrine therapy targeting the ER pathway including selective ER modulators (SERMs), selective ER downregulators (SERDs) and aromatase inhibitors (AIs). However, resistance to endocrine therapy associated with disease progression remains a significant therapeutic challenge. The precise mechanisms of endocrine resistance remain unclear. This is partly due to the complexity of the signaling pathways that influence the estrogen-mediated regulation in breast cancer. Mechanisms include ER modifications, alteration of coregulatory function and modification of growth factor signaling pathways. In this review, we provide an overview of epigenetic mechanisms of tamoxifen resistance in ER-positive luminal breast cancer. We highlight the effect of epigenetic changes on some of the key mechanisms involved in tamoxifen resistance, such as tumor-cell heterogeneity, ER signaling pathway and cancer stem cells (CSCs). It became increasingly recognized that CSCs are playing an important role in driving metastasis and tamoxifen resistance. Understanding the mechanism of tamoxifen resistance will provide insight into the design of novel strategies to overcome the resistance and make further improvements in breast cancer therapeutics.
Collapse
Affiliation(s)
- Hany A Abdel-Hafiz
- Department of Medicine/Endocrinology, School of Medicine, University of Colorado, Ms 8106 PO Box 6511, 12801 E 17th Avenue, Aurora, Denver, CO 80010, USA; Tel.: +1-303-724-1013; Fax: +1-303-724-3920.
| |
Collapse
|
34
|
Abstract
Background: Features of malignant melanoma (MM) vary in the different geographic regions of the world. This may be attributable to environmental, ethnic, and genetic factors. The aim of this study was to determine the expression of estrogen receptor alpha (ER-α) in MM in Isfahan, Iran. Materials and Methods: This study was planned as a descriptive, analytical, cross-sectional investigation. During this study, paraffin-embedded tissue blocks of patients with a histopathologic diagnosis of MM was studied for ER-α using immunohistochemistry (IHC). Results: In this study, 38 patients (female/male; 20/18) with a definite diagnosis of malignant cutaneous melanoma and mean age of 52.4 ± 11.2 years were investigated. Using envision IHC staining, there were not any cases with ER-α expression. Conclusion: In confirmation to the most previous studies, expression of ER-α was negative in MM. It is recommended to investigate the expression of estrogen receptor beta and other markers in MM.
Collapse
Affiliation(s)
- Parvin Rajabi
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Bagheri
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Hani
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
35
|
Laffont S, Seillet C, Guéry JC. Estrogen Receptor-Dependent Regulation of Dendritic Cell Development and Function. Front Immunol 2017; 8:108. [PMID: 28239379 PMCID: PMC5300975 DOI: 10.3389/fimmu.2017.00108] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/23/2017] [Indexed: 12/23/2022] Open
Abstract
Autoimmunity, infectious diseases and cancer affect women and men differently. Because they tend to develop more vigorous adaptive immune responses than men, women are less susceptible to some infectious diseases but also at higher risk of autoimmunity. The regulation of immune responses by sex-dependent factors probably involves several non-redundant mechanisms. A privileged area of study, however, concerns the role of sex steroid hormones in the biology of innate immune cells, especially dendritic cells (DCs). In recent years, our understanding of the lineage origin of DC populations has expanded, and the lineage-committing transcription factors shaping peripheral DC subsets have been identified. Both progenitor cells and mature DC subsets express estrogen receptors (ERs), which are ligand-dependent transcription factors. This suggests that estrogens may contribute to the reported sex differences in immunity by regulating DC biology. Here, we review the recent literature and highlight evidence that estrogen-dependent activation of ERα regulates the development or the functional responses of particular DC subsets. The in vitro model of GM-CSF-induced DC differentiation shows that CD11c+ CD11bint Ly6cneg cells depend on ERα activation by estrogen for their development, and for the acquisition of competence to activate naive CD4+ T lymphocytes and mount a robust pro-inflammatory cytokine response to CD40 stimulation. In this model, estrogen signaling in conjunction with GM-CSF is necessary to promote early interferon regulatory factor (Irf)-4 expression in macrophage-DC progenitors and their subsequent differentiation into IRF-4hi CD11c+ CD11bint Ly6cneg cells, closely related to the cDC2 subset. The Flt3L-induced model of DC differentiation in turn shows that ERα signaling promotes the development of conventional DC (cDC) and plasmacytoid DC (pDC) with higher capability of pro-inflammatory cytokine production in response to TLR stimulation. Likewise, cell-intrinsic ER signaling positively regulates the TLR-driven production of type I interferons (IFNs) in mouse pDCs in vivo. This effect of estrogens likely contributes to the greater proficiency of women's pDCs than men's as regards the production of type I IFNs elicited by TLR7 ligands. In summary, evidence is emerging in support of the notion that estrogen signaling regulates important aspects of cDC and pDC development and/or effector functions, in both mice and humans.
Collapse
Affiliation(s)
- Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| | - Cyril Seillet
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Jean-Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS , Toulouse , France
| |
Collapse
|
36
|
Ariazi EA, Taylor JC, Black MA, Nicolas E, Slifker MJ, Azzam DJ, Boyd J. A New Role for ERα: Silencing via DNA Methylation of Basal, Stem Cell, and EMT Genes. Mol Cancer Res 2016; 15:152-164. [PMID: 28108626 DOI: 10.1158/1541-7786.mcr-16-0283] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/10/2016] [Accepted: 10/18/2016] [Indexed: 12/30/2022]
Abstract
Resistance to hormonal therapies is a major clinical problem in the treatment of estrogen receptor α-positive (ERα+) breast cancers. Epigenetic marks, namely DNA methylation of cytosine at specific CpG sites (5mCpG), are frequently associated with ERα+ status in human breast cancers. Therefore, ERα may regulate gene expression in part via DNA methylation. This hypothesis was evaluated using a panel of breast cancer cell line models of antiestrogen resistance. Microarray gene expression profiling was used to identify genes normally silenced in ERα+ cells but derepressed upon exposure to the demethylating agent decitabine, derepressed upon long-term loss of ERα expression, and resuppressed by gain of ERα activity/expression. ERα-dependent DNA methylation targets (n = 39) were enriched for ERα-binding sites, basal-up/luminal-down markers, cancer stem cell, epithelial-mesenchymal transition, and inflammatory and tumor suppressor genes. Kaplan-Meier survival curve and Cox proportional hazards regression analyses indicated that these targets predicted poor distant metastasis-free survival among a large cohort of breast cancer patients. The basal breast cancer subtype markers LCN2 and IFI27 showed the greatest inverse relationship with ERα expression/activity and contain ERα-binding sites. Thus, genes that are methylated in an ERα-dependent manner may serve as predictive biomarkers in breast cancer. IMPLICATIONS ERα directs DNA methylation-mediated silencing of specific genes that have biomarker potential in breast cancer subtypes. Mol Cancer Res; 15(2); 152-64. ©2016 AACR.
Collapse
Affiliation(s)
- Eric A Ariazi
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania.
| | - John C Taylor
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Michael A Black
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Emmanuelle Nicolas
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Michael J Slifker
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Diana J Azzam
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Jeff Boyd
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania.
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
37
|
Zhang T, Shen W, De Felici M, Zhang XF. Di(2-ethylhexyl)phthalate: Adverse effects on folliculogenesis that cannot be neglected. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:579-588. [PMID: 27530864 DOI: 10.1002/em.22037] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/30/2016] [Accepted: 07/05/2016] [Indexed: 06/06/2023]
Abstract
Primordial follicle formation and the subsequent transition of follicles through primary and secondary stages constitute crucial events of oogenesis. In particular, in mammals, defects in the processes that precede and accompany the formation of the primordial follicle pool can affect the size of this population significantly, while alterations in follicle activation, growth and maturation can result in premature depletion of the follicle reserve or cause follicle arrest at immature stages. Over the last decade, in vitro and in vivo approaches have been used to provide evidence that exposure to di(2-ethylhexyl)phthalate(DEHP), the most widely used plasticizer, has a deleterious effect on various stages of folliculogenesis in rodents. There is growing concern, supported by epidemiological and experimental data, that DEHP may have similar effects in women. This article reviews the evidence, with particular reference to our own findings, that DEHP may actually exert a variety of adverse effects on mammalian folliculogenesis from early to final stages of oogenesis, including altered development of the primordial germ cells, impaired fetal oocyte survival and meiotic progression, reduced oocyte nest breakdown, acceleration of primordial follicle activation, altered follicle steroidogenesis and increased follicle atresia. These effects can cause serious complications for reproductive and nonreproductive women's health. In addition, emerging data indicate that phthalates, including DEHP, may cause subtle epigenetic changes in germ cells that can be transmitted to subsequent generations, with potential negative effects on human health. Environ. Mol. Mutagen. 57:589-604, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Teng Zhang
- College of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Wei Shen
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata,", Rome, 00133, Italy.
| | - Xi-Feng Zhang
- College of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
38
|
Kavoosi F, Dastjerdi MN, Valiani A, Esfandiari E, Sanaei M, Hakemi MG. Genistein potentiates the effect of 17-beta estradiol on human hepatocellular carcinoma cell line. Adv Biomed Res 2016; 5:133. [PMID: 27656602 PMCID: PMC5025906 DOI: 10.4103/2277-9175.187395] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/11/2015] [Indexed: 11/16/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. This cancer may be due to a multistep process with an accumulation of epigenetic alterations in tumor suppressor genes (TSGs), leading to hypermethylation of the genes. Hypermethylation of TSGs is associated with silencing and inactivation of them. It is well-known that DNA hypomethylation is the initial epigenetic abnormality recognized in human tumors. Estrogen receptor alpha (ERα) is one of the TSGs which modulates gene transcription and its hypermethylation is because of overactivity of DNA methyltransferases. Fortunately, epigenetic changes especially hypermethylation can be reversed by pharmacological compounds such as genistein (GE) and 17-beta estradiol (E2) which involve in preventing the development of certain cancers by maintaining a protective DNA methylation. The aim of the present study was to analyze the effects of GE on ERα and DNMT1 genes expression and also apoptotic and antiproliferative effects of GE and E2 on HCC. Materials and Methods: Cells were treated with various concentrations of GE and E2 and the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay was used. Furthermore, cells were treated with single dose of GE and E2 (25 μM) and flow cytometry assay was performed. The expression level of the genes was determined by quantitative real-time reverse transcription polymerase chain reaction. Results: GE increased ERα and decreased DNMT1 genes expression, GE and E2 inhibited cell viability and induced apoptosis significantly. Conclusion: GE can epigenetically increase ERα expression by inhibition of DNMT1 expression which in turn increases apoptotic effect of E2. Furthermore, a combination of GE and E2 can induce apoptosis more significantly.
Collapse
Affiliation(s)
- Fraidoon Kavoosi
- Department of Anatomical Sciences, Medical School, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mehdi Nikbakht Dastjerdi
- Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Valiani
- Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masumeh Sanaei
- Department of Anatomical Sciences, Medical School, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mazdak Ganjalikhani Hakemi
- Cellular and Molecular Immunology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
39
|
Abbas A, Hall JA, Patterson WL, Ho E, Hsu A, Al-Mulla F, Georgel PT. Sulforaphane modulates telomerase activity via epigenetic regulation in prostate cancer cell lines. Biochem Cell Biol 2016; 94:71-81. [DOI: 10.1139/bcb-2015-0038] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Epidemiologic studies have revealed that diets rich in sulforaphane (SFN), an isothiocyanate present in cruciferous vegetables, are associated with a marked decrease in prostate cancer incidence. The chemo-preventive role of SFN is associated with its histone de-acetylase inhibitor activity. However, the effect of SFN on chromatin composition and dynamic folding, especially in relation to HDAC inhibitor activity, remains poorly understood. In this study, we found that SFN can inhibit the expression and activity of human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, in 2 prostate cancer cell lines. This decrease in gene expression is correlated with SFN-induced changes in chromatin structure and composition. The SFN-mediated changes in levels of histone post-translational modifications, more specifically acetylation of histone H3 lysine 18 and di-methylation of histone H3 lysine 4, 2 modifications linked with high risk of prostate cancer recurrence, were associated with regulatory elements within the hTERT promoter region. Chromatin condensation may also play a role in SFN-mediated hTERT repression, since expression and recruitment of MeCP2, a known chromatin compactor, were altered in SFN treated prostate cancer cells. Chromatin immuno-precipitation (ChIP) of MeCP2 showed enrichment over regions of the hTERT promoter with increased nucleosome density. These combined results strongly support a role for SFN in the mediation of epigenetic events leading to the repression of hTERT in prostate cancer cells. This ability of SFN to modify chromatin composition and structure associated with target gene expression provides a new model by which dietary phytochemicals may exert their chemoprevention activity.
Collapse
Affiliation(s)
- Ata Abbas
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
| | - J. Adam Hall
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| | - William L. Patterson
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| | - Emily Ho
- Oregon State University, School of Biological and Population Health Sciences, Linus Pauling Institute, Corvallis, OR, USA
| | - Anna Hsu
- Oregon State University, School of Biological and Population Health Sciences, Linus Pauling Institute, Corvallis, OR, USA
| | - Fahd Al-Mulla
- Kuwait University, Health Sciences Center, Faculty of Medicine, Molecular Pathology Unit, Kuwait City, Kuwait
| | - Philippe T. Georgel
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| |
Collapse
|
40
|
Stolzenberg DS, Champagne FA. Hormonal and non-hormonal bases of maternal behavior: The role of experience and epigenetic mechanisms. Horm Behav 2016; 77:204-10. [PMID: 26172856 DOI: 10.1016/j.yhbeh.2015.07.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 11/24/2022]
Abstract
This article is part of a Special Issue "Parental Care". Though hormonal changes occurring throughout pregnancy and at the time of parturition have been demonstrated to prime the maternal brain and trigger the onset of mother-infant interactions, extended experience with neonates can induce similar behavioral interactions. Sensitization, a phenomenon in which rodents engage in parental responses to young following constant cohabitation with donor pups, was elegantly demonstrated by Rosenblatt (1967) to occur in females and males, independent of hormonal status. Study of the non-hormonal basis of maternal behavior has contributed significantly to our understanding of hormonal influences on the maternal brain and the cellular and molecular mechanisms that mediate maternal behavior. Here, we highlight our current understanding regarding both hormone-induced and experience-induced maternal responsivity and the mechanisms that may serve as a common pathway through which increases in maternal behavior are achieved. In particular, we describe the epigenetic changes that contribute to chromatin remodeling and how these molecular mechanisms may influence the neural substrates of the maternal brain. We also consider how individual differences in these systems emerge during development in response to maternal care. This research has broad implications for our understanding of the parental brain and the role of experience in the induction of neurobiological and behavior changes.
Collapse
|
41
|
Reproductive experiential regulation of cognitive and emotional resilience. Neurosci Biobehav Rev 2015; 58:92-106. [DOI: 10.1016/j.neubiorev.2015.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 04/16/2015] [Accepted: 05/21/2015] [Indexed: 11/17/2022]
|
42
|
Kocatürk B, Tieken C, Vreeken D, Ünlü B, Engels CC, de Kruijf EM, Kuppen PJ, Reitsma PH, Bogdanov VY, Versteeg HH. Alternatively spliced tissue factor synergizes with the estrogen receptor pathway in promoting breast cancer progression. J Thromb Haemost 2015; 13:1683-93. [PMID: 26179105 PMCID: PMC4560996 DOI: 10.1111/jth.13049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/18/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Procoagulant full-length tissue factor (flTF) and its minimally coagulant alternatively spliced isoform (asTF), promote breast cancer (BrCa) progression via different mechanisms. We previously showed that flTF and asTF are expressed by BrCa cells, resulting in autoregulation in a cancer milieu. BrCa cells often express hormone receptors such as the estrogen receptor (ER), leading to the formation of hormone-regulated cell populations. OBJECTIVE To investigate whether TF isoform-specific and ER-dependent pathways interact in BrCa. METHODS Tissue factor isoform-regulated gene sets were assessed using ingenuity pathway analysis. Tissues from a cohort of BrCa patients were divided into ER-positive and ER-negative groups. Associations between TF isoform levels and tumor characteristics were analyzed in these groups. BrCa cells expressing TF isoforms were assessed for proliferation, migration and in vivo growth in the presence or absence of estradiol. RESULTS Ingenuity pathway analysis pointed to similarities between ER- and TF-induced gene expression profiles. In BrCa tissue specimens, asTF expression was associated with grade and stage in ER-positive but not in ER-negative tumors. flTF was only associated with grade in ER-positive tumors. In MCF-7 cells, asTF accelerated proliferation in the presence of estradiol in a β1 integrin-dependent manner. No synergy between asTF and the ER pathway was observed in a migration assay. Estradiol accelerated the growth of asTF-expressing tumors but not control tumors in vivo in an orthotopic setting. CONCLUSION Tissue factor isoform and estrogen signaling share downstream targets in BrCa; the concomitant presence of asTF and estrogen signaling is required to promote BrCa cell proliferation.
Collapse
Affiliation(s)
- B Kocatürk
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - C Tieken
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - D Vreeken
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - B Ünlü
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - C C Engels
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - E M de Kruijf
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - P J Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - P H Reitsma
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - V Y Bogdanov
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - H H Versteeg
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
43
|
Alam H, Gu B, Lee MG. Histone methylation modifiers in cellular signaling pathways. Cell Mol Life Sci 2015; 72:4577-92. [PMID: 26305020 DOI: 10.1007/s00018-015-2023-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/02/2015] [Accepted: 08/14/2015] [Indexed: 02/06/2023]
Abstract
Histone methyltransferases and demethylases epigenetically regulate gene expression by modifying histone methylation status in numerous cellular processes, including cell differentiation and proliferation. These modifiers also control methylation levels of various non-histone proteins, such as effector proteins that play critical roles in cellular signaling networks. Dysregulated histone methylation modifiers alter expression of oncogenes and tumor suppressor genes and change methylation states of effector proteins, frequently resulting in aberrant cellular signaling cascades and cellular transformation. In this review, we summarize the role of histone methylation modifiers in regulating the following signaling pathways: NF-κB, RAS/RAF/MEK/MAPK, PI3K/Akt, Wnt/β-catenin, p53, and ERα.
Collapse
Affiliation(s)
- Hunain Alam
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Bingnan Gu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Min Gyu Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
- Cancer Biology Program, Graduate School of Biomedical Sciences, The University of Texas Health Science Center, Houston, TX, 77030, USA.
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
44
|
Estrogens Correlate with PELP1 Expression in ER Positive Breast Cancer. PLoS One 2015; 10:e0134351. [PMID: 26247365 PMCID: PMC4527840 DOI: 10.1371/journal.pone.0134351] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/09/2015] [Indexed: 11/19/2022] Open
Abstract
The Proline-, glutamic acid- and leucine-rich protein 1 (PELP1) is an estrogen receptor (ER) coactivator and a proto-oncogene known to be deregulated in endocrine cancers. In breast cancer, PELP1 overexpression has been associated with endocrine therapy resistance. Although PELP1 is known to be regulated by estrogens in vitro, its association with estrogen levels within the tissue of breast cancer patients has not previously been assessed. Here, we determined PELP1 mRNA expression levels in paired samples of normal and malignant breast tissue obtained from 32 postmenopausal and 11 premenopausal women. In the total sample set, PELP1 levels were higher in tumors compared to normal breast tissue (P = 0.041). Among postmenopausal women, PELP1 tumor levels correlated positively with estrone (E1) and estradiol (E2) levels in both normal tissue (r = 0.543, P = 0.003 and r = 0.601, P = 0.001, respectively) and plasma (r = 0.392, P = 0.053 and r = 0.403, P = 0.046, respectively). Analyzing all ER+ tumors (n = 26), PELP1 correlated positively with E1 and E2 in tumor tissue (r = 0.562, P = 0.003 and r = 0.411, P = 0.037, respectively) and normal tissue (r = 0.461, P = 0.018 and r = 0.427, P = 0.030, respectively) in addition to plasma E1, E2 and estrone sulphate (E1S) concentrations (r = 0.576, P = 0.003, r = 0.456, P = 0.025 and r = 0.406, P = 0.049, respectively). Finally, PELP1 correlated positively with ER mRNA (ESR1) (r = 0.553, P = 0.026) in ER+ tumors, whereas a negative association between PELP1 and ESR1 (r = -0.733, P = 0.010) was observed in ER- breast tumors. Taken together, tumor PELP1 mRNA expression is associated with estrogen levels in breast cancer, suggesting a potentially important role of PELP1 in ER+ breast cancer growth in vivo.
Collapse
|
45
|
Krishnan SR, Nair BC, Sareddy GR, Roy SS, Natarajan M, Suzuki T, Peng Y, Raj G, Vadlamudi RK. Novel role of PELP1 in regulating chemotherapy response in mutant p53-expressing triple negative breast cancer cells. Breast Cancer Res Treat 2015; 150:487-99. [PMID: 25788226 DOI: 10.1007/s10549-015-3339-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/10/2015] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC), the most aggressive breast cancer subtype, occurs in younger women and is associated with poor prognosis. Gain-of-function mutations in TP53 are a frequent occurrence in TNBC and have been demonstrated to repress apoptosis and up-regulate cell cycle progression. Even though TNBC responds to initial chemotherapy, resistance to chemotherapy develops and is a major clinical problem. Tumor recurrence eventually occurs and most patients die from their disease. An urgent need exists to identify molecular-targeted therapies that can enhance chemotherapy response. In the present study, we report that targeting PELP1, an oncogenic co-regulator molecule, could enhance the chemotherapeutic response of TNBC through the inhibition of cell cycle progression and activation of apoptosis. We demonstrate that PELP1 interacts with MTp53, regulates its recruitment, and alters epigenetic marks at the target gene promoters. PELP1 knockdown reduced MTp53 target gene expression, resulting in decreased cell survival and increased apoptosis upon genotoxic stress. Mechanistic studies revealed that PELP1 depletion contributes to increased stability of E2F1, a transcription factor that regulates both cell cycle and apoptosis in a context-dependent manner. Further, PELP1 regulates E2F1 stability in a KDM1A-dependent manner, and PELP1 phosphorylation at the S1033 residue plays an important role in mediating its oncogenic functions in TNBC cells. Accordingly, depletion of PELP1 increased the expression of E2F1 target genes and reduced TNBC cell survival in response to genotoxic agents. PELP1 phosphorylation was significantly greater in the TNBC tumors than in the other subtypes of breast cancer and in the normal tissues. These findings suggest that PELP1 is an important molecular target in TNBC, and that PELP1-targeted therapies may enhance response to chemotherapies.
Collapse
Affiliation(s)
- Samaya R Krishnan
- Department of Cellular and Structural Biology, UTHSCSA, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Chen RY, Fan YM, Zhang Q, Liu S, Li Q, Ke GL, Li C, You Z. Estradiol inhibits Th17 cell differentiation through inhibition of RORγT transcription by recruiting the ERα/REA complex to estrogen response elements of the RORγT promoter. THE JOURNAL OF IMMUNOLOGY 2015; 194:4019-28. [PMID: 25769926 DOI: 10.4049/jimmunol.1400806] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 02/09/2015] [Indexed: 12/24/2022]
Abstract
The symptoms of vaginal candidiasis exacerbate in the second half of the menstrual cycle in premenopausal women when the serum estradiol level is elevated. Estradiol has been shown to inhibit Th17 differentiation and production of antifungal IL-17 cytokines. However, little is known about the mechanisms. In the present study, we used mouse splenocytes and found that estradiol inhibited Th17 differentiation through downregulation of Rorγt mRNA and protein expression. Estradiol activated estrogen receptor (ER)α to recruit repressor of estrogen receptor activity (REA) and form the ERα/REA complex. This complex bound to three estrogen response element (ERE) half-sites on the Rorγt promoter region to suppress Rorγt expression. Estradiol induced Rea mRNA and protein expression in mouse splenocytes. Using Rea small interfering RNA to knock down Rea expression enhanced Rorγt expression and Th17 differentiation. Alternatively, histone deacetylase 1 and 2 bound to the three ERE half-sites, independent of estradiol. Histone deacetylase inhibitor MS-275 dose- and time-dependently increased Rorγt expression and subsequently enhanced Th17 differentiation. In 15 healthy premenopausal women, high serum estradiol levels are correlated with low RORγT mRNA levels and high REA mRNA levels in the vaginal lavage. These results demonstrate that estradiol upregulates REA expression and recruits REA via ERα to the EREs on the RORγT promoter region, thus inhibiting RORγT expression and Th17 differentiation. This study suggests that the estradiol/ERα/REA axis may be a feasible target in the management of recurrent vaginal candidiasis.
Collapse
Affiliation(s)
- Rong-Yi Chen
- Department of Dermatology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China; Department of Structural and Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA 70112; and
| | - Yi-Ming Fan
- Department of Dermatology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China;
| | - Qiuyang Zhang
- Department of Structural and Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA 70112; and
| | - Sen Liu
- Department of Structural and Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA 70112; and
| | - Qingli Li
- Department of Structural and Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA 70112; and Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Guo-Lin Ke
- Department of Dermatology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Chen Li
- Department of Dermatology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA 70112; and Department of Orthopaedic Surgery, Tulane University Health Sciences Center, New Orleans, LA 70112; Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, LA 70112; Tulane Center for Aging, Tulane University Health Sciences Center, New Orleans, LA 70112; and Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112
| |
Collapse
|
47
|
Kovats S. Estrogen receptors regulate innate immune cells and signaling pathways. Cell Immunol 2015; 294:63-9. [PMID: 25682174 PMCID: PMC4380804 DOI: 10.1016/j.cellimm.2015.01.018] [Citation(s) in RCA: 694] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/26/2015] [Accepted: 01/29/2015] [Indexed: 02/07/2023]
Abstract
Humans show strong sex differences in immunity to infection and autoimmunity,
suggesting sex hormones modulate immune responses. Indeed, receptors for estrogens (ER)
regulate cells and pathways in the innate and adaptive immune system, as well as immune
cell development. ERs are ligand-dependent transcription factors that mediate long-range
chromatin interactions and form complexes at gene regulatory elements, thus promoting
epigenetic changes and transcription. ERs also participate in membrane-initiated steroid
signaling to generate rapid responses. Estradiol and ER activity show profound dose- and
context-dependent effects on innate immune signaling pathways and myeloid cell
development. While estradiol most often promotes the production of type I interferon, innate pathways
leading to pro-inflammatory cytokine production may be enhanced or dampened by ER
activity. Regulation of innate immune cells and signaling by ERs may contribute to the
reported sex differences in innate immune pathways. Here we review the recent literature
and highlight several molecular mechanisms by which ERs regulate the development or
functional responses of innate immune cells.
Collapse
Affiliation(s)
- Susan Kovats
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, United States.
| |
Collapse
|
48
|
Kawaratani Y, Matsuoka T, Hirata Y, Fukata N, Nagaoka Y, Uesato S. Influence of the carbamate fungicide benomyl on the gene expression and activity of aromatase in the human breast carcinoma cell line MCF-7. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:292-299. [PMID: 25543211 DOI: 10.1016/j.etap.2014.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 11/07/2014] [Indexed: 06/04/2023]
Abstract
The carbamate fungicide benomyl reportedly inhibited the growth of the human breast cancer cell line MCF-7 by inducing apoptosis. However, influence of benomyl on the expression and activity of aromatase of MCF-7 cells remains to be examined, since benomyl was identified as an endocrine disruptor. We here confirmed through cell cycle analysis and immunofluorescence staining that benomyl damaged microtubules and caused apoptosis. We also found that benomyl inhibited histone deacetylase (HDAC) 1 and accumulated acetylated histone H3 in MCF-7 cells. Additionally, benomyl enhanced the levels of aromatase protein and mRNA, albeit at high concentrations. It is thus likely that benomyl enhanced the promoter activity of the aromatase gene via acetylation of histone H3 as does the HDAC inhibitor Vorinostat. In conclusion, benomyl remains to be a risk factor as an endocrine disruptor for breast cancer.
Collapse
Affiliation(s)
- Yasuyuki Kawaratani
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka 564-8680, Japan.
| | - Takeshi Matsuoka
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka 564-8680, Japan.
| | - Yoshiyuki Hirata
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka 564-8680, Japan.
| | - Naofumi Fukata
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka 564-8680, Japan.
| | - Yasuo Nagaoka
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka 564-8680, Japan.
| | - Shinichi Uesato
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka 564-8680, Japan.
| |
Collapse
|
49
|
Mau T, Yung R. Potential of epigenetic therapies in non-cancerous conditions. Front Genet 2014; 5:438. [PMID: 25566322 PMCID: PMC4271720 DOI: 10.3389/fgene.2014.00438] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 11/25/2014] [Indexed: 12/20/2022] Open
Abstract
There has been an explosion of knowledge in the epigenetics field in the past 20 years. The first epigenetic therapies have arrived in the clinic for cancer treatments. In contrast, much of the promise of epigenetic therapies for non-cancerous conditions remains in the laboratories. The current review will focus on the recent progress that has been made in understanding the pathogenic role of epigenetics in immune and inflammatory conditions, and how the knowledge may provide much needed new therapeutic targets for many autoimmune diseases. Dietary factors are increasingly recognized as potential modifiers of epigenetic marks that can influence health and diseases across generations. The current epigenomics revolution will almost certainly complement the explosion of personal genetics medicine to help guide treatment decisions and disease risk stratification.
Collapse
Affiliation(s)
- Theresa Mau
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan Ann Arbor, MI, USA
| | - Raymond Yung
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan Ann Arbor, MI, USA ; Department of Veterans Affairs Ann Arbor Health System, Geriatric Research, Education and Clinical Care Center Ann Arbor, MI, USA
| |
Collapse
|
50
|
Remely M, Lovrecic L, de la Garza AL, Migliore L, Peterlin B, Milagro FI, Martinez AJ, Haslberger AG. Therapeutic perspectives of epigenetically active nutrients. Br J Pharmacol 2014; 172:2756-68. [PMID: 25046997 DOI: 10.1111/bph.12854] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/24/2014] [Accepted: 07/10/2014] [Indexed: 12/17/2022] Open
Abstract
Many nutrients are known for a wide range of activities in prevention and alleviation of various diseases. Recently, their potential role in regulating human health through effects on epigenetics has become evident, although specific mechanisms are still unclear. Thus, nutriepigenetics/nutriepigenomics has emerged as a new and promising field in current epigenetics research in the past few years. In particular, polyphenols, as part of the central dynamic interaction between the genome and the environment with specificity at physiological concentrations, are well known to affect mechanisms underlying human health. This review summarizes the effects of dietary compounds on epigenetic mechanisms in the regulation of gene expression including expression of enzymes and other molecules responsible for drug absorption, distribution, metabolism and excretion in cancer, metabolic syndrome, neurodegenerative disorders and hormonal dysfunction.
Collapse
Affiliation(s)
- M Remely
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - L Lovrecic
- Clinical Institute of Medical Genetics, Department of Gynecology and Obstetrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - A L de la Garza
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, Pamplona, Spain
| | - L Migliore
- Department of Translational Research and New Technologies in Medicine and Surgery, Division of Medical Genetics, University of Pisa, Pisa, Italy.,Research Center Nutraceuticals and Food for Health - Nutrafood, University of Pisa, Pisa, Italy
| | - B Peterlin
- Clinical Institute of Medical Genetics, Department of Gynecology and Obstetrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - F I Milagro
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, Pamplona, Spain
| | - A J Martinez
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, Pamplona, Spain.,Physiopathology of Obesity and Nutrition, CIBERobn, Carlos III Health Research Institute, Madrid, Spain
| | - A G Haslberger
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| |
Collapse
|