1
|
Hamilton AM, Srivastava VK, Hiney JK, Dees WL, Dearth RK. Manganese-induced Precocious Puberty Alters Mammary Epithelial Cell Proliferation in Female Rats. Endocrinology 2025; 166:bqaf052. [PMID: 40105700 PMCID: PMC12006721 DOI: 10.1210/endocr/bqaf052] [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: 09/11/2024] [Revised: 01/02/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
Precocious puberty (PP) is an established breast cancer risk factor. In the normal mammary gland, hormone receptor-positive (HR+) cells rarely proliferate. In breast cancer, proliferating epithelial cells are often HR+. It is not known if PP can modify this population of proliferating HR+ cells. Previously, we established a manganese-induced precocious puberty (MnPP) model to study the effects of PP on mammary gland development in female rats. Here, we characterized the distribution of HR+ proliferating mammary epithelial cells in prepubertal and adult rodents, in association with precocious puberty. Female rats were exposed daily to 10 mg/kg manganese chloride or saline (control) from postnatal day (PND) 12 to PND 30. Mammary glands were collected on PNDs 30 and 120, processed for western blot analysis and double immunofluorescence staining for proliferating cell nuclear antigen and progesterone receptor or estrogen receptor. MnPP increased the percentage of HR+ mammary epithelial cells coexpressing proliferating cell nuclear antigen relative to normally developed controls at PND 30. This correlated with increased expression of estrogen receptor-regulated proteins in MnPP mammary glands relative to controls at PND 30, including FOXA1, AREG, and c-Myc. Conversely, at PND 120 relative to PND 30, proliferating HR+ cells remained chronically elevated in MnPP mammary glands at PND 120, which coincided with decreased expression of cell-cycle regulator, p27, and increased expression of progesterone receptor-regulated markers, EREG and sp1. Collectively, these results suggest early puberty alters steroidal regulation of classic proliferative mechanisms in the prepubertal gland with increased prevalence of high-risk proliferating HR+ cells.
Collapse
MESH Headings
- Animals
- Female
- Cell Proliferation/drug effects
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/pathology
- Rats
- Puberty, Precocious/chemically induced
- Puberty, Precocious/metabolism
- Puberty, Precocious/pathology
- Rats, Sprague-Dawley
- Receptors, Progesterone/metabolism
- Proliferating Cell Nuclear Antigen/metabolism
- Receptors, Estrogen/metabolism
- Sexual Maturation/drug effects
- Manganese
- Chlorides
- Manganese Compounds
Collapse
Affiliation(s)
- Alina M Hamilton
- School of Integrative Biological and Chemical Sciences, College of Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27606, USA (Current)
| | - Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - Robert K Dearth
- School of Integrative Biological and Chemical Sciences, College of Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
2
|
Xie J, Wang J, Cui X. Research progress on estrogen and estrogen receptors in the occurrence and progression of autoimmune thyroid diseases. Autoimmun Rev 2025; 24:103803. [PMID: 40089093 DOI: 10.1016/j.autrev.2025.103803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Autoimmune thyroid disease (AITD) is a category of disease related to sex differences, with a significantly higher incidence in women than in men. In addition to X chromosome inactivation abnormalities, Estrogen and estrogen receptors may lead to the sex differences in AITD. Estrogen, estrogen receptors and estrogen receptor-mediated signaling pathways can affect the number and function of immune cells and the function of the thyroid to promote the development of AITD. This article describes the role of estrogen in regulating the composition ratio and the function of immune cells and the role of estrogen in promoting thyroid cell proliferation and thyroxine-binding protein and thyroid antibody production; the role of estrogen in stimulating the hypothalamus-pituitary-thyroid gland axis; and the role of estrogen and the estrogen receptor in the progression of AITD. These roles offer a new perspective for understanding the pathological mechanism of AITD and provide new targets for future therapeutic strategies.
Collapse
Affiliation(s)
- Jiewen Xie
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, PR China
| | - Jie Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, PR China
| | - Xuejiao Cui
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
3
|
Das GM, Oturkar CC, Menon V. Interaction between Estrogen Receptors and p53: A Broader Role for Tamoxifen? Endocrinology 2025; 166:bqaf020. [PMID: 39891710 PMCID: PMC11837209 DOI: 10.1210/endocr/bqaf020] [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: 09/29/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Tamoxifen is one of the most widely used anticancer drugs in the world. It is a safe drug with generally well-tolerated side effects and has been prescribed for the treatment of early-stage and advanced-stage or metastatic estrogen receptor α (ERα/ESR1)-positive breast cancer. Tamoxifen therapy also provides a 38% reduction of the risk of developing breast cancer in women at high risk. With the advent of newer medications targeting ERα-positive breast cancer, tamoxifen is now mainly used as adjuvant therapy for lower-risk premenopausal breast cancer and cancer prevention. It is widely accepted that tamoxifen as a selective estrogen receptor modulator exerts its therapeutic effect by competitively binding to ERα, leading to the recruitment of corepressors and inhibition of transcription of genes involved in the proliferation of breast cancer epithelium. As such, expression of ERα in breast tumors has been considered necessary for tumors to be responsive to tamoxifen therapy. However, ERα-independent effects of tamoxifen in various in vitro and in vivo contexts have been reported over the years. Importantly, the recent discovery that ERα and estrogen receptor β (ERβ/ESR2) can bind tumor suppressor protein p53 with functional consequences has provided new insights into the mechanisms underlying response to tamoxifen therapy and resistance. Furthermore, these findings have paved the way for broadening the use of tamoxifen by potentially repurposing it to treat triple negative (negative for ERα, human epidermal growth factor receptor 2, and progesterone receptor) breast cancer. Herein, we summarize these developments and discuss their mechanistic underpinnings and clinical implications.
Collapse
Affiliation(s)
- Gokul M Das
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Chetan C Oturkar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Vishnu Menon
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
4
|
Long BY, Liao X, Liang X. The Hypothalamus and Pituitary Gland Regulate Reproduction and Are Involved in the Development of Polycystic Ovary Syndrome. Neuroendocrinology 2025; 115:315-334. [PMID: 39894018 DOI: 10.1159/000543877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex condition with unclear mechanisms, posing a challenge for prevention and treatment of PCOS. The role of the hypothalamus and pituitary gland in regulating female reproduction is critical. Abnormalities in the hypothalamus and pituitary can impair reproductive function. It is important to study hypothalamic and pituitary changes in patients with PCOS. SUMMARY This article reviews articles on the hypothalamus and PCOS with the goal of summarizing what abnormalities of the hypothalamic-pituitary-ovarian axis are present in patients with PCOS and to clarify the pathogenesis of PCOS. We find that the mechanisms by which the hypothalamus and pituitary regulate reproduction in girls are complex and are associated with altered sex hormone levels, obesity, and insulin resistance. Different animal models of PCOS are characterized by different alterations in the hypothalamus and pituitary and respond differently to different treatments, which correspond to the complex pathogenesis of patients with PCOS. KEY MESSAGES Arcuate nucleus (ARC) is associated with luteinizing hormone (LH) surges. Suprachiasmatic nucleus, ARC, and RP3V are associated with LH surges. Animal models of PCOS have different characteristics.
Collapse
Affiliation(s)
- Bin-Yang Long
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xipeng Liao
- Tianjin University of Technology, Tianjin, China
| | - Xin Liang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
5
|
Park CJ, Oh JE, Lin P, Zhou S, Bunnell M, Bikorimana E, Spinella MJ, Lim HJ, Ko CJ. A Dynamic Shift in Estrogen Receptor Expression During Granulosa Cell Differentiation in the Ovary. Endocrinology 2025; 166:bqaf006. [PMID: 39834231 PMCID: PMC12054734 DOI: 10.1210/endocr/bqaf006] [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: 10/06/2024] [Revised: 12/29/2024] [Accepted: 01/18/2025] [Indexed: 01/22/2025]
Abstract
This study uncovers a dynamic shift in estrogen receptor expression during granulosa cell (GC) differentiation in the ovary, highlighting a transition from estrogen receptor alpha (ESR1) to estrogen receptor beta (ESR2). Using a transgenic mouse model with Esr1-iCre-mediated Esr2 deletion, we demonstrate that ESR2 expression is absent in GCs derived from ESR1-expressing ovarian surface epithelium (OSE) cells. Single-cell analysis of the OSE-GC lineage reveals a developmental trajectory from Esr1-expressing OSE cells to Foxl2-expressing pre-GCs, culminating in GCs exclusively expressing Esr2. Transcriptome analyses identified vasculature-derived TGFβ1 ligands as key regulators of this transition. Supporting this, TGFβ1 treatment of cultured embryonic ovaries reduced Esr1 expression while promoting Esr2 expression. This study underscores the capability of GCs to switch from ESR1 to ESR2 expression as a fundamental aspect of normal differentiation.
Collapse
Affiliation(s)
- Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
- Epivara, Inc., Research Park, Champaign, IL 61820, USA
| | - Ji-Eun Oh
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - PoChing Lin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Sherry Zhou
- Epivara, Inc., Research Park, Champaign, IL 61820, USA
| | - Mary Bunnell
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Emmanuel Bikorimana
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Michael J Spinella
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Hyunjung Jade Lim
- Department of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea
| | - CheMyong J Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| |
Collapse
|
6
|
İpçak HH, Denli M, Yokuş B, Bademkıran S. The Impact of Dietary Encapsulated Fennel Seed (Foeniculum vulgare Mill.) Essential Oil Inclusion Levels on Performance, Serum Hormone Profiles, and Expression of Reproductive Axis-Related Genes in the Early and Late Laying Phases of Hens. Vet Med Sci 2025; 11:e70150. [PMID: 39655356 PMCID: PMC11629027 DOI: 10.1002/vms3.70150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/15/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Fennel seed (Foeniculum vulgare Mill.) essential oil (FEO), which is rich in the phytoestrogenic compound trans-anethole, interacts with oestrogen receptors and influences molecular targets within cells and hormonal responses. This study examined the effect of dietary encapsulated FEO inclusion levels on performance, reproductive hormone profiles, and gene expression in laying hens during the early and late phases. The study was conducted in two independent trials, each involving 210 Atak-S laying hens that were randomly distributed into 3 experimental groups, each having 10 replicates with 7 hens. The dietary treatments included a basal diet without FEO (Control) and a basal diet supplemented with 175 (FEO175) or 350 mg (FEO350) of encapsulated FEO/kg for 12 weeks. The results showed that FEO350 treatment improved egg production, egg mass, and feed conversion ratio during both early and late phases (p < 0.05). Moreover, increasing FEO inclusion levels enhanced oestradiol, follicle-stimulating hormone, luteinizing hormone and progesterone concentrations in both early and late laying hens, reaching peak levels at FEO350 (p < 0.05). FEO supplementation upregulated the expression of oestrogen receptor 2 (ESR2) and follicle-stimulating hormone receptor (FSHR) (p < 0.05). Furthermore, FEO350 increased prolactin receptor (PRLR) expression during the early phase but decreased it during the late laying phase (p < 0.05). Positive correlations were observed between egg production and FSHR, ESR2 and steroidogenic acute regulatory protein (STAR) expression, with a negative correlation for PRLR (p < 0.05). In conclusion, 350 mg FEO/kg was found to be the most effective level for enhancing layer performance.
Collapse
Affiliation(s)
- Hasan Hüseyin İpçak
- Department of Animal Science, Faculty of AgricultureDicle UniversityDiyarbakırTurkey
| | - Muzaffer Denli
- Department of Animal Science, Faculty of AgricultureDicle UniversityDiyarbakırTurkey
| | - Beran Yokuş
- Department of Biochemistry, Faculty of Veterinary MedicineDicle UniversityDiyarbakırTurkey
| | - Servet Bademkıran
- Department of Obstetrics and Gynaecology, Faculty of Veterinary MedicineDicle UniversityDiyarbakirTurkey
| |
Collapse
|
7
|
González-Flores O, Garcia-Juárez M, Tecamachaltzi-Silvarán MB, Lucio RA, Ordoñez RD, Pfaus JG. Cellular and molecular mechanisms of action of ovarian steroid hormones. I: Regulation of central nervous system function. Neurosci Biobehav Rev 2024; 167:105937. [PMID: 39510217 DOI: 10.1016/j.neubiorev.2024.105937] [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: 08/27/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
The conventional way steroid hormones work through receptors inside cells is widely acknowledged. There are unanswered questions about what happens to the hormone in the end and why there isn't always a strong connection between how much tissue takes up and its biological effects through receptor binding. Steroid hormones can also have non-traditional effects that happen quickly but don't involve entering the cell. Several possible mechanisms for these non-traditional actions include (a) changes in membrane fluidity, (b) steroid hormones acting on receptors on the outer surface of cells, (c) steroid hormones regulating GABAA receptors on cell membranes, and (d) activation of steroid receptors by factors like EGF, IGF-1, and dopamine. Data also suggests that steroid hormones may be inserted into DNA through receptors, acting as transcription factors. These proposed new mechanisms of action should not be seen as challenging the conventional mechanism. Instead, they contribute to a more comprehensive understanding of how hormones work, allowing for rapid, short-term, and prolonged effects to meet the body's physiological needs.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico.
| | - Marcos Garcia-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | | | - Rosa Angélica Lucio
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Raymundo Domínguez Ordoñez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico; Licenciatura en Ingeniería Agronómica y Zootecnia, Complejo Regional Centro, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany, Czech Republic; Department of Psychology and Life Sciences, Faculty of Humanities, Charles University, Prague, Czech Republic
| |
Collapse
|
8
|
Almuntashiri S, Dutta S, Zhu Y, Gamare S, Ramírez G, Irineo‐Moreno V, Camarena A, Regino N, Campero P, Hernández‐Cardenas CM, Rodriguez‐ Reyna TS, Zuñiga J, Owen CA, Wang X, Zhang D. Estrogen-dependent gene regulation: Molecular basis of TIMP-1 as a sex-specific biomarker for acute lung injury. Physiol Rep 2024; 12:e70047. [PMID: 39267201 PMCID: PMC11392656 DOI: 10.14814/phy2.70047] [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: 04/25/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024] Open
Abstract
Increased circulating tissue inhibitor of metalloproteinases-1 (TIMP-1) levels have been observed in patients with acute lung injury (ALI). However, the sex-specific regulation of TIMP-1 and the underlying molecular mechanisms have not been well elucidated. In this study, we found that plasma TIMP-1 levels were significantly higher in COVID-19 and H1N1 patients compared with those in healthy subjects (n = 25). TIMP-1 concentrations were significantly different between males and females in each disease group. Among female but not male patients, TIMP-1 levels significantly correlated with the PaO2/FiO2 ratio and hospital length of stay. Using the mouse model of ALI induced by the H1N1 virus, we found that TIMP-1 is strikingly induced in PDGFRα-positive cells in the murine lungs. Moreover, female mice showed a higher Timp-1 expression in the lungs on day 3 postinfection. Mechanistically, we observed that estrogen can upregulate TIMP-1 expression in lung fibroblasts, not epithelial cells. In addition, overexpression of estrogen receptor α (ERα) increased the TIMP-1 promoter activity. In summary, TIMP-1 is an estrogen-responsive gene, and its promoter activity is regulated by ERα. Circulating TIMP-1 may serve as a sex-specific marker, reflecting the severity and worst outcomes in female patients with SARS-CoV2- and IAV-related ALI.
Collapse
Affiliation(s)
- Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
- Department of Clinical Pharmacy, College of PharmacyUniversity of HailHailSaudi Arabia
| | - Saugata Dutta
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Siddhika Gamare
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Gustavo Ramírez
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Valeria Irineo‐Moreno
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
- Tecnologico de Monterrey, School of Medicine and Health SciencesMexico CityMexico
| | - Angel Camarena
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Nora Regino
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
- Tecnologico de Monterrey, School of Medicine and Health SciencesMexico CityMexico
| | - Paloma Campero
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | | | - Tatiana S. Rodriguez‐ Reyna
- Department of Immunology and RheumatologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Joaquin Zuñiga
- Laboratory of Immunobiology and Genetics and Intensive Care UnitInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
- Tecnologico de Monterrey, School of Medicine and Health SciencesMexico CityMexico
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care MedicineBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGeorgiaUSA
- Department of Medicine, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
9
|
Dong H, Zeng X, Xu J, He C, Sun Z, Liu L, Huang Y, Sun Z, Cao Y, Peng Z, Qiu YA, Yu T. Advances in immune regulation of the G protein-coupled estrogen receptor. Int Immunopharmacol 2024; 136:112369. [PMID: 38824903 DOI: 10.1016/j.intimp.2024.112369] [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: 02/16/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
Estrogen and related receptors have been shown to have a significant impact on human development, reproduction, metabolism and immune regulation and to play a critical role in tumor development and treatment. Traditionally, the nuclear estrogen receptors (nERs) ERα and ERβ have been thought to be involved in mediating the estrogenic effects. However, our group and others have previously demonstrated that the G protein-coupled estrogen receptor (GPER) is the third independent ER, and estrogen signaling mediated by GPER is known to play an important role in normal physiology and a variety of abnormal diseases. Interestingly, recent studies have progressively revealed GPER involvement in the maintenance of the normal immune system, abnormal immune diseases, and inflammatory lesions, which may be of significant clinical value primarily in the immunotherapy of tumors. In this article, we review current advances in GPER-related immunomodulators and provide a theoretical basis and potential clinical targets to ameliorate immune-related diseases and immunotherapy for tumors.
Collapse
Affiliation(s)
- Hanzhi Dong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Xiaoqiang Zeng
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Jiawei Xu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Zhengkui Sun
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Liyan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Yanxiao Huang
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhe Sun
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Yuan Cao
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhiqiang Peng
- Department of Lymphohematology, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Yu-An Qiu
- Department of Critical Care Medicine, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| |
Collapse
|
10
|
Hancock GR, Gertz J, Jeselsohn R, Fanning SW. Estrogen Receptor Alpha Mutations, Truncations, Heterodimers, and Therapies. Endocrinology 2024; 165:bqae051. [PMID: 38643482 PMCID: PMC11075793 DOI: 10.1210/endocr/bqae051] [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: 01/05/2024] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024]
Abstract
Annual breast cancer (BCa) deaths have declined since its apex in 1989 concomitant with widespread adoption of hormone therapies that target estrogen receptor alpha (ERα), the prominent nuclear receptor expressed in ∼80% of BCa. However, up to ∼50% of patients who are ER+ with high-risk disease experience post endocrine therapy relapse and metastasis to distant organs. The vast majority of BCa mortality occurs in this setting, highlighting the inadequacy of current therapies. Genomic abnormalities to ESR1, the gene encoding ERα, emerge under prolonged selective pressure to enable endocrine therapy resistance. These genetic lesions include focal gene amplifications, hotspot missense mutations in the ligand binding domain, truncations, fusions, and complex interactions with other nuclear receptors. Tumor cells utilize aberrant ERα activity to proliferate, spread, and evade therapy in BCa as well as other cancers. Cutting edge studies on ERα structural and transcriptional relationships are being harnessed to produce new therapies that have shown benefits in patients with ESR1 hotspot mutations. In this review we discuss the history of ERα, current research unlocking unknown aspects of ERα signaling including the structural basis for receptor antagonism, and future directions of ESR1 investigation. In addition, we discuss the development of endocrine therapies from their inception to present day and survey new avenues of drug development to improve pharmaceutical profiles, targeting, and efficacy.
Collapse
Affiliation(s)
- Govinda R Hancock
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60513, USA
| | - Jason Gertz
- Department of Oncological Sciences, Huntsman Cancer Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Rinath Jeselsohn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Sean W Fanning
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60513, USA
| |
Collapse
|
11
|
Kranjčević JK, Čonkaš J, Ozretić P. The Role of Estrogen and Estrogen Receptors in Head and Neck Tumors. Cancers (Basel) 2024; 16:1575. [PMID: 38672656 PMCID: PMC11049451 DOI: 10.3390/cancers16081575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 04/21/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the most common histological form of head and neck tumors (HNTs), which originate from the epithelium of the lips and oral cavity, pharynx, larynx, salivary glands, nasal cavity, and sinuses. The main risk factors include consumption of tobacco in all forms and alcohol, as well as infections with high-risk human papillomaviruses or the Epstein-Barr virus. Regardless of the etiological agent, the risk of developing different types of HNTs is from two to more than six times higher in males than in females. The reason for such disparities probably lies in a combination of both biological and psychosocial factors. Therefore, it is hypothesized that exposure to female sex hormones, primarily estrogen, provides women with protection against the formation and metastasis of HNTs. In this review, we synthesized available knowledge on the role of estrogen and estrogen receptors (ERs) in the development and progression of HNTs, with special emphasis on membrane ERs, which are much less studied. We can summarize that in addition to epidemiologic studies unequivocally pointing to the protective effect of estrogen in women, an increased expression of both nuclear ERs, ERα, and ERβ, and membrane ERs, ERα36, GPER1, and NaV1.2, was present in different types of HNSCC, for which anti-estrogens could be used as an effective therapeutic approach.
Collapse
Affiliation(s)
| | | | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia (J.Č.)
| |
Collapse
|
12
|
Rymbai E, Sugumar D, Chakkittukandiyil A, Kothandan R, Selvaraj D. Molecular insights into the potential effects of selective estrogen receptor β agonists in Alzheimer's and Parkinson's diseases. Cell Biochem Funct 2024; 42:e4014. [PMID: 38616346 DOI: 10.1002/cbf.4014] [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: 12/20/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative disorders. Pathologically, AD and PD are characterized by the accumulation of misfolded proteins. Hence, they are also called as proteinopathy diseases. Gender is considered as one of the risk factors in both diseases. Estrogens are widely accepted to be neuroprotective in several neurodegenerative disorders. Estrogens can be produced in the central nervous system, where they are called as neurosteroids. Estrogens mediate their neuroprotective action mainly through their actions on estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). However, ERα is mainly involved in the growth and development of the primary and secondary sexual organs in females. Hence, the activation of ERα is associated with undesired side effects such as gynecomastia and increase in the risk of breast cancer, thromboembolism, and feminization. Therefore, selective activation of ERβ is often considered to be safer. In this review, we explore the role of ERβ in regulating the expression and functions of AD- and PD-associated genes. Additionally, we discuss the association of these genes with the amyloid-beta peptide (Aβ) and α-synuclein mediated toxicity. Ultimately, we established a correlation between the importance of ERβ activation and the process underlying ERβ's neuroprotective mechanisms in AD and PD.
Collapse
Affiliation(s)
- Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Deepa Sugumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Ram Kothandan
- Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| |
Collapse
|
13
|
Rawłuszko-Wieczorek AA, Lipowicz J, Nowacka M, Ostrowska K, Pietras P, Blatkiewicz M, Ruciński M, Jagodziński PP, Nowicki M. Estrogen receptor β affects hypoxia response in colorectal cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166894. [PMID: 37748565 DOI: 10.1016/j.bbadis.2023.166894] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023]
Abstract
The occurrence of colorectal cancer (CRC) is inversely correlated with estrogen receptor beta (ERβ) presence. Additionally, multiple studies associate low ERβ expression with poorer overall survival of CRC patients. Molecular pathways involved in ERβ - related reduced tumorigenesis include enhanced apoptosis, decreased proliferation, or repression of oncogenes. Moreover, the development of solid tumors, such as CRC, is often associated with an increased tumor mass that results in decreased oxygen partial tension, known as hypoxia, clinically associated with decreased prognosis and therapeutic resistance. Our high-throughput study suggests that ERβ also represses a hypoxic response in CRC cells. We observed a significantly altered transcriptional profile in HCT116 ERβ overexpressing cells that was further stimulated by E2 treatment under hypoxic conditions. The achieved data for downregulation of VEGFA, PDGFA and ANGPTL4 were validated in a time course experiment in DLD-1 cells. In addition, using an ERβ construct with a mutated DNA binding domain we observed that the downregulation of selected genes is dependent on the direct binding of this receptor to regulatory region genes. In addition, we observed that ERβ may affect the expression of the main hypoxia regulator, HIF1A, at the transcriptional and translational levels. In summary, ERβ alters the hypoxic outcome in CRC cells.
Collapse
Affiliation(s)
| | - Julia Lipowicz
- Department of Histology, Poznań University of Medical Sciences, Poland
| | - Marta Nowacka
- Department of Histology, Poznań University of Medical Sciences, Poland
| | - Kamila Ostrowska
- Department of Histology, Poznań University of Medical Sciences, Poland; Department of Head and Neck Oncology, Poznań University of Medical Sciences, The Greater Poland Cancer Centre, Poland
| | - Paulina Pietras
- Department of Histology, Poznań University of Medical Sciences, Poland
| | | | - Marcin Ruciński
- Department of Histology, Poznań University of Medical Sciences, Poland
| | - Paweł Piotr Jagodziński
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Poland
| | - Michał Nowicki
- Department of Histology, Poznań University of Medical Sciences, Poland
| |
Collapse
|
14
|
Colón-Caraballo M, Flores-Caldera I. Translational aspects of the endometriosis epigenome. EPIGENETICS IN HUMAN DISEASE 2024:883-929. [DOI: 10.1016/b978-0-443-21863-7.00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
15
|
Hoffmann JP, Liu JA, Seddu K, Klein SL. Sex hormone signaling and regulation of immune function. Immunity 2023; 56:2472-2491. [PMID: 37967530 DOI: 10.1016/j.immuni.2023.10.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/25/2023] [Accepted: 10/14/2023] [Indexed: 11/17/2023]
Abstract
Immune responses to antigens, including innocuous, self, tumor, microbial, and vaccine antigens, differ between males and females. The quest to uncover the mechanisms for biological sex differences in the immune system has intensified, with considerable literature pointing toward sex hormonal influences on immune cell function. Sex steroids, including estrogens, androgens, and progestins, have profound effects on immune function. As such, drastic changes in sex steroid concentrations that occur with aging (e.g., after puberty or during the menopause transition) or pregnancy impact immune responses and the pathogenesis of immune-related diseases. The effect of sex steroids on immunity involves both the concentration of the ligand and the density and distribution of genomic and nongenomic receptors that serve as transcriptional regulators of immune cellular responses to affect autoimmunity, allergy, infectious diseases, cancers, and responses to vaccines. The next frontier will be harnessing these effects of sex steroids to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Joseph P Hoffmann
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jennifer A Liu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kumba Seddu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
16
|
Tsoi H, Lok J, Man EP, Cheng CN, Leung MH, You CP, Chan SY, Chan WL, Khoo US. Overexpression of BQ323636.1 contributes to anastrozole resistance in AR+ve/ER+ve breast cancer. J Pathol 2023; 261:156-168. [PMID: 37555303 DOI: 10.1002/path.6157] [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: 11/07/2022] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 08/10/2023]
Abstract
Aromatase inhibitors (Ais) are used as adjuvant endocrine therapy for oestrogen receptor-positive (ER+ve) post-menopausal breast cancer patients. Ais, by inhibiting the enzyme aromatase, block the conversion of androgen to oestrogen, reducing oestrogen levels. Resistance to Ais limits their clinical utilisation. Here, we show that overexpression of BQ323636.1 (BQ), a novel splice variant of nuclear co-repressor NCOR2, is associated with resistance to the non-steroidal aromatase inhibitor anastrozole in ER+ve post-menopausal breast cancer. Mechanistic study indicates that BQ overexpression enhances androgen receptor (AR) activity and in the presence of anastrozole, causes hyper-activation of AR signalling, which unexpectedly enhanced cell proliferation, through increased expression of CDK2, CDK4, and CCNE1. BQ overexpression reverses the effect of anastrozole in ER+ve breast cancer in an AR-dependent manner, whilst co-treatment with the AR antagonist bicalutamide recovered its therapeutic effect both in vitro and in vivo. Thus, for BQ-overexpressing breast cancer, targeting AR can combat anastrozole resistance. Clinical study of 268 primary breast cancer samples of ER+ve patients who had been treated with non-steroidal Ais showed 32.5% (38/117) of cases with combined high nuclear expression of BQ and AR, which were found to be significantly associated with Ai resistance. Non-steroidal Ai-treated patients with high nuclear expression of both BQ and AR had poorer overall, disease-specific, and disease-free survival. These findings suggest the importance of assessing BQ and AR expression status in the primary ER+ve breast tumour prior to Ai treatment. This may save patients from inappropriate treatment and enable effective therapy to be given at an early stage. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ho Tsoi
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Johann Lok
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Ellen Ps Man
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Cheuk-Nam Cheng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Man-Hong Leung
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Chan-Ping You
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Sum-Yin Chan
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, SAR, PR China
| | - Wing-Lok Chan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Ui-Soon Khoo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| |
Collapse
|
17
|
Li M, Duan J, Liu Y, Zou J, Yang X, Zeng H. The habitual characteristic of smart phone use under relevant cues among Chinese college students. Front Psychol 2023; 14:1218886. [PMID: 37799513 PMCID: PMC10548464 DOI: 10.3389/fpsyg.2023.1218886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023] Open
Abstract
Excessive smartphone use may be habitual behavior induced by cues associated with the phone. Habitual behavior occurs outside of awareness and is characterized by lack of control. It is not like problematic smartphone use (PSU) (Brand et al., 2016), which is used to either limit behavior or produce pleasure and relieve feelings of pain, stress, and failure despite significant harmful consequences. 62 college students participated in experiments to test the effects of visual cues and self-control, which are the important characteristic of habitual behavior in smartphone-related behavior. The results showed that a significantly larger amount of cue-related phone use behavior occurred in the setting where participants (a) had their smartphones in sight and (b) were given no instructions to exert self-control, compared to when neither of the two conditions was imposed. The habitual model is a useful framework for understanding PSU and can help people avoid it with less stress. The results provide substantial implications for reducing the frequency and duration of smartphone use among college populations.
Collapse
Affiliation(s)
- Ming Li
- Department of Psychology, Guangzhou University, Guangzhou, China
- College of Science and Engineering, Jinan University, Guangzhou, China
| | - Jieyue Duan
- Zhuhai Center for Chronic Disease Control, Zhuhai, China
| | - Yuning Liu
- School of Philosophy, Psychology and Language sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jingxin Zou
- Department of Psychology, Guangzhou University, Guangzhou, China
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China
| | - Hong Zeng
- Clinical and Counseling Psychology Research Center, Guangzhou University, Guangzhou, China
| |
Collapse
|
18
|
Lindblad A, Wu R, Persson K, Demirel I. The Role of NLRP3 in Regulation of Antimicrobial Peptides and Estrogen Signaling in UPEC-Infected Bladder Epithelial Cells. Cells 2023; 12:2298. [PMID: 37759520 PMCID: PMC10526908 DOI: 10.3390/cells12182298] [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: 08/09/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The NLRP3 inflammasome, estrogen and antimicrobial peptides have all been found to have a vital role in the protection of the bladder urothelium. However, the interdependence between these protective factors during a bladder infection is currently unknown. Our aim was to investigate the role of NLRP3 in the regulation of antimicrobial peptides and estrogen signaling in bladder epithelial cells during a UPEC infection. Human bladder epithelial cells and CRISPR/Cas9-generated NLRP3-deficient cells were stimulated with the UPEC strain CFT073 and estradiol. The gene and protein expression were evaluated with microarray, qRT-PCR, western blot and ELISA. Microarray results showed that the expression of most antimicrobial peptides was reduced in CFT073-infected NLRP3-deficient cells compared to Cas9 control cells. Conditioned medium from NLRP3-deficient cells also lost the ability to suppress CFT073 growth. Moreover, NLRP3-deficient cells had lower basal release of Beta-defensin-1, Beta-defensin-2 and RNase7. The ability of estradiol to induce an increased expression of antimicrobial peptides was also abrogated in NLRP3-deficient cells. The decreased antimicrobial peptide expression might be linked to the observed reduced expression and activity of estradiol receptor beta in NLRP3-deficient cells. This study suggests that NLRP3 may regulate the release and expression of antimicrobial peptides and affect estrogen signaling in bladder epithelial cells.
Collapse
Affiliation(s)
| | | | | | - Isak Demirel
- School of Medical Sciences, Örebro University, 701 82 Örebro, Sweden; (A.L.); (R.W.); (K.P.)
| |
Collapse
|
19
|
Balcazar Lopez CE, Albrecht J, Hafstað V, Börjesson Freitag C, Vallon‐Christersson J, Bellodi C, Persson H. Alternative promoters and splicing create multiple functionally distinct isoforms of oestrogen receptor alpha in breast cancer and healthy tissues. Cancer Med 2023; 12:18931-18945. [PMID: 37676103 PMCID: PMC10557849 DOI: 10.1002/cam4.6508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Oestrogen receptor alpha (ER) is involved in cell growth and proliferation and functions as a transcription factor, a transcriptional coregulator, and in cytoplasmic signalling. It affects, for example, bone, endometrium, ovaries and mammary epithelium. It is a key biomarker in clinical management of breast cancer, where it is used as a prognostic and treatment-predictive factor, and a therapeutical target. Several ER isoforms have been described, but transcript annotation in public databases is incomplete and inconsistent, and functional differences are not well understood. METHODS We have analysed short- and long-read RNA sequencing data from breast tumours, breast cancer cell lines, and normal tissues to create a comprehensive annotation of ER transcripts and combined it with experimental studies of full-length protein and six alternative isoforms. RESULTS The isoforms have varying transcription factor activity, subcellular localisation, and response to the ER-targeting drugs tamoxifen and fulvestrant. Antibodies differ in ability to detect alternative isoforms, which raises concerns for the interpretation of ER-status in routine pathology. CONCLUSIONS Future work should investigate the effects of alternative isoforms on patient survival and therapy response. An accurate annotation of ER isoforms will aid in interpretation of clinical data and inform functional studies to improve our understanding of the ER in health and disease.
Collapse
Affiliation(s)
| | - Juliane Albrecht
- Department of Clinical Sciences Lund, OncologyLund University Cancer CentreLundSweden
| | - Völundur Hafstað
- Department of Clinical Sciences Lund, OncologyLund University Cancer CentreLundSweden
| | | | | | - Cristian Bellodi
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of MedicineLund UniversityLundSweden
| | - Helena Persson
- Department of Clinical Sciences Lund, OncologyLund University Cancer CentreLundSweden
| |
Collapse
|
20
|
Jang SH, Paek SH, Kim JK, Seong JK, Lim W. A New Culture Model for Enhancing Estrogen Responsiveness in HR+ Breast Cancer Cells through Medium Replacement: Presumed Involvement of Autocrine Factors in Estrogen Resistance. Int J Mol Sci 2023; 24:ijms24119474. [PMID: 37298425 DOI: 10.3390/ijms24119474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/20/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Hormone receptor-positive breast cancer (HR+ BC) cells depend on estrogen and its receptor, ER. Due to this dependence, endocrine therapy (ET) such as aromatase inhibitor (AI) treatment is now possible. However, ET resistance (ET-R) occurs frequently and is a priority in HR+ BC research. The effects of estrogen have typically been determined under a special culture condition, i.e., phenol red-free media supplemented with dextran-coated charcoal-stripped fetal bovine serum (CS-FBS). However, CS-FBS has some limitations, such as not being fully defined or ordinary. Therefore, we attempted to find new experimental conditions and related mechanisms to improve cellular estrogen responsiveness based on the standard culture medium supplemented with normal FBS and phenol red. The hypothesis of pleiotropic estrogen effects led to the discovery that T47D cells respond well to estrogen under low cell density and medium replacement. These conditions made ET less effective there. The fact that several BC cell culture supernatants reversed these findings implies that housekeeping autocrine factors regulate estrogen and ET responsiveness. Results reproduced in T47D subclone and MCF-7 cells highlight that these phenomena are general among HR+ BC cells. Our findings offer not only new insights into ET-R but also a new experimental model for future ET-R studies.
Collapse
Affiliation(s)
- Seok-Hoon Jang
- Department of Surgery, Ewha Womans University Mokdong Hospital, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Republic of Korea
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Se Hyun Paek
- Department of Surgery, Ewha Womans University Seoul Hospital, 260, Gonghang-daero, Gangseo-gu, Seoul 07804, Republic of Korea
| | - Jong-Kyu Kim
- Department of Surgery, Ewha Womans University Seoul Hospital, 260, Gonghang-daero, Gangseo-gu, Seoul 07804, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Woosung Lim
- Department of Surgery, Ewha Womans University Mokdong Hospital, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Republic of Korea
| |
Collapse
|
21
|
Amran NH, Zaid SSM, Meng GY, Salleh A, Mokhtar MH. Protective Role of Kelulut Honey against Toxicity Effects of Polystyrene Microplastics on Morphology, Hormones, and Sex Steroid Receptor Expression in the Uterus of Rats. TOXICS 2023; 11:324. [PMID: 37112551 PMCID: PMC10141738 DOI: 10.3390/toxics11040324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/15/2023] [Accepted: 03/23/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Microplastics (MPs) are an emerging global pollutant. Previous studies have revealed that chronic exposure to MPs can affect animal and human reproductive health, particularly by impairing the reproductive system's normal functions, which may increase the risk of infertility in both males and females. Kelulut honey (KH), an excellent source of antioxidants, has been used to counteract the disruptive effects of Polystyrene microplastics (PS-MPs) in the rat uterus. Thus, this study aimed to investigate the potential protective effects of Kelulut honey against PS-MPs-induced uterine toxicity in pubertal rats. METHODS Prepubertal female Sprague Dawley rats were divided into four groups (n = 8): (i) normal control group (NC: treated with deionized water), MPs-exposed group (M: exposed to PS-MPs at 2.5 mg/kg), (iii) Kelulut honey group (DM: pretreated with 1200 mg/kg of KH 30 minutes before they were administered with PS-MPs at 2.5 mg/kg), and (iv) Kelulut honey control group (DC: only treated with KH at 2.5 mg/kg). The rats were treated orally once daily for six consecutive weeks. RESULTS Uterine abnormalities in PS-MPs-exposed rats were significantly improved after concurrent treatment with Kelulut honey. Morphology improvement was observed and luminal epithelial cells seemed thicker with more goblet cells, glandular cells had a more regular and circular shape, stromal cell increased in size, interstitial gaps between stromal cells expanded, and the myometrium layer was thicker. Kelulut honey treatment also effectively normalized the suppressive effect of PS-MPs on the expression and distribution of sex steroid receptors (ERα and ERβ), as well as the level of serum gonadotropin (LH and FSH) and sex steroid (estradiol and progesterone) hormones. CONCLUSION Kelulut honey can protect the female reproductive system against the disruptive effects of PS-MPs. The phytochemical properties of Kelulut honey might be responsible for these beneficial benefits. However, future studies are warranted to identify the mechanisms involved.
Collapse
Affiliation(s)
- Nur Hanisah Amran
- Department of Environment, Faculty of Forestry and Environment, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Siti Sarah Mohamad Zaid
- Department of Environment, Faculty of Forestry and Environment, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Goh Yong Meng
- Department of Veterinary Pre-Clinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Annas Salleh
- Department of Veterinary Laboratory Diagnostic, Faculty of Veterinary Medicine, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
22
|
Choi Y, Pollack S. Significant Association of Estrogen Receptor-β Isoforms and Coactivators in Breast Cancer Subtypes. Curr Issues Mol Biol 2023; 45:2533-2548. [PMID: 36975536 PMCID: PMC10047005 DOI: 10.3390/cimb45030166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/22/2023] Open
Abstract
Nuclear receptor coregulators are the principal regulators of Estrogen Receptor (ER)-mediated transcription. ERβ, an ER subtype first identified in 1996, is associated with poor outcomes in breast cancer (BCa) subtypes, and the coexpression of the ERβ1 isoform and AIB-1 and TIF-2 coactivators in BCa-associated myofibroblasts is associated with high-grade BCa. We aimed to identify the specific coactivators that are involved in the progression of ERβ-expressing BCa. ERβ isoforms, coactivators, and prognostic markers were tested using standard immunohistochemistry. AIB-1, TIF-2, NF-kB, p-c-Jun, and/or cyclin D1 were differentially correlated with ERβ isoform expression in the BCa subtypes and subgroups. The coexpression of the ERβ5 and/or ERβ1 isoforms and the coactivators were found to be correlated with a high expression of P53, Ki-67, and Her2/neu and large-sized and/or high-grade tumors in BCa. Our study supports the notion that ERβ isoforms and coactivators seemingly coregulate the proliferation and progression of BCa and may provide insight into the potential therapeutic uses of the coactivators in BCa.
Collapse
Affiliation(s)
- Young Choi
- Department of Pathology, Yale School of Medicine, 434 Pine Grove Lane, Hartsdale, NY 10530, USA
- Correspondence:
| | - Simcha Pollack
- Department of Statistics, St. John’s University, New York, NY 11423, USA
| |
Collapse
|
23
|
Chakraborty B, Byemerwa J, Krebs T, Lim F, Chang CY, McDonnell DP. Estrogen Receptor Signaling in the Immune System. Endocr Rev 2023; 44:117-141. [PMID: 35709009 DOI: 10.1210/endrev/bnac017] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Indexed: 01/14/2023]
Abstract
The immune system functions in a sexually dimorphic manner, with females exhibiting more robust immune responses than males. However, how female sex hormones affect immune function in normal homeostasis and in autoimmunity is poorly understood. In this review, we discuss how estrogens affect innate and adaptive immune cell activity and how dysregulation of estrogen signaling underlies the pathobiology of some autoimmune diseases and cancers. The potential roles of the major circulating estrogens, and each of the 3 estrogen receptors (ERα, ERβ, and G-protein coupled receptor) in the regulation of the activity of different immune cells are considered. This provides the framework for a discussion of the impact of ER modulators (aromatase inhibitors, selective estrogen receptor modulators, and selective estrogen receptor downregulators) on immunity. Synthesis of this information is timely given the considerable interest of late in defining the mechanistic basis of sex-biased responses/outcomes in patients with different cancers treated with immune checkpoint blockade. It will also be instructive with respect to the further development of ER modulators that modulate immunity in a therapeutically useful manner.
Collapse
Affiliation(s)
- Binita Chakraborty
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jovita Byemerwa
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Taylor Krebs
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.,Known Medicine, Salt Lake City, UT 84108, USA
| | - Felicia Lim
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
24
|
Michishita S, Gibble C, Tubbs C, Felton R, Gjeltema J, Lang J, Finkelstein M. Microplastic in northern anchovies (Engraulis mordax) and common murres (Uria aalge) from the Monterey Bay, California USA - Insights into prevalence, composition, and estrogenic activity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120548. [PMID: 36336179 DOI: 10.1016/j.envpol.2022.120548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/08/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
Microplastic (particle size <5 mm) is considered an emerging threat to the marine environment, yet data are limited for coastal ecosystems. To provide information related to microplastic in a coastal system, we used alkaline tissue digestion and Raman spectroscopy to quantify the prevalence and composition (e.g. fiber, fragment, foam, etc.) of anthropogenic microparticles in the digestive tracts of northern anchovies (Engraulis mordax, anchovy, n = 24), and common murres (Uria aalge, murre, n = 19) from the Monterey Bay, California USA. We also determined microplastic prevalence and composition in seawater (n = 12 17-h sampling periods representing ∼46,000 L sampled) from two Monterey Bay intake systems (Moss Landing, CA and Santa Cruz, CA USA). Microparticles recovered from murre digestive tracts were assessed for estrogenic activity using an in-vitro estrogen receptor activation assay. Suspected anthropogenic microparticles based on visual characteristics were recovered from all sample types with ∼2 particles per 1000 L from the seawater sampling periods, 58% prevalence in anchovies, and 100% prevalence in murres. Across samples of seawater, anchovies, and murres, the most abundant microparticle type found were fibers (78%), followed by fragments (13%), foam (6%), film (2%), and beads (1%). Raman spectroscopy identified 57% of microparticles (excluding dye-prominent and unknown) as plastic (synthetic, semi-synthetic, or blends). Almost one quarter (23%) of the murre digestive tracts contained microparticles that exhibited estrogenic activity. Our study describes the widespread occurrence, composition, and potential estrogenic activity of microplastic in the Monterey Bay and provides important information to aid in the understanding of microplastic contamination in coastal systems.
Collapse
Affiliation(s)
- Sami Michishita
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Corinne Gibble
- California Department of Fish and Wildlife, Office of Spill Prevention and Response, Marine Wildlife Veterinary Care and Research Center, 151 McAllister Way, Santa Cruz, CA, 95060, USA
| | - Christopher Tubbs
- Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, 15600 San Pasqual Valley Road, Escondido, CA, 92027, USA
| | - Rachel Felton
- Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, 15600 San Pasqual Valley Road, Escondido, CA, 92027, USA
| | - Jenessa Gjeltema
- Department of Medicine and Epidemiology and the Karen C. Drayer Wildlife Health Center, University of California Davis School of Veterinary Medicine, One Shields Drive, Davis, CA, 95616, USA
| | - Jackelyn Lang
- Department of Medicine and Epidemiology and the Karen C. Drayer Wildlife Health Center, University of California Davis School of Veterinary Medicine, One Shields Drive, Davis, CA, 95616, USA
| | - Myra Finkelstein
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
25
|
Ng ASN, Zhang S, Mak VCY, Zhou Y, Yuen Y, Sharma R, Lu Y, Zhuang G, Zhao W, Pang HH, Cheung LWT. AKTIP loss is enriched in ERα-positive breast cancer for tumorigenesis and confers endocrine resistance. Cell Rep 2022; 41:111821. [PMID: 36516775 PMCID: PMC9837615 DOI: 10.1016/j.celrep.2022.111821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
Recurrent deletion of 16q12.2 is observed in luminal breast cancer, yet the causal genomic alterations in this region are largely unknown. In this study, we identify that loss of AKTIP, which is located on 16q12.2, drives tumorigenesis of estrogen receptor alpha (ERα)-positive, but not ERα-negative, breast cancer cells and is associated with poor prognosis of patients with ERα-positive breast cancer. Intriguingly, AKTIP-depleted tumors have increased ERα protein level and activity. Cullin-associated and neddylation-dissociated protein 1 (CAND1), which regulates the cullin-RING E3 ubiquitin ligases, protects ERα from cullin 2-dependent proteasomal degradation. Apart from ERα signaling, AKTIP loss triggers JAK2-STAT3 activation, which provides an alternative survival signal when ERα is inhibited. AKTIP-depleted MCF7 cells and ERα-positive patient-derived organoids are more resistant to ERα antagonists. Importantly, the resistance can be overcome by co-inhibition of JAK2/STAT3. Together, our results highlight the subtype-specific functional consequences of AKTIP loss and provide a mechanistic explanation for the enriched AKTIP copy-number loss in ERα-positive breast cancer.
Collapse
Affiliation(s)
- Angel S N Ng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shibo Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Victor C Y Mak
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuan Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yin Yuen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Rakesh Sharma
- Proteomics and Metabolomics Core, Center for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yiling Lu
- Department of Genomic Medicine, Division of Cancer Medicine, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guanglei Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China; Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Wei Zhao
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Herbert H Pang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lydia W T Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
26
|
Kehmeier MN, Bedell BR, Cullen AE, Khurana A, D'Amico HJ, Henson GD, Walker AE. In vivo arterial stiffness, but not isolated artery endothelial function, varies with the mouse estrous cycle. Am J Physiol Heart Circ Physiol 2022; 323:H1057-H1067. [PMID: 36240435 PMCID: PMC9678414 DOI: 10.1152/ajpheart.00369.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 12/14/2022]
Abstract
With the increasing appreciation for sex as a biological variable and the inclusion of female mice in research, it is important to understand the influence of the estrous cycle on physiological function. Sex hormones are known to modulate vascular function, but the effects of the mouse estrous cycle phase on arterial stiffness, endothelial function, and arterial estrogen receptor expression remain unknown. In 23 female C57BL/6 mice (6 mo of age), we determined the estrous cycle stage via vaginal cytology and plasma hormone concentrations. Aortic stiffness, assessed by pulse wave velocity, was lower during the estrus phase compared with diestrus. In ex vivo assessment of isolated pressurized mesenteric and posterior cerebral arteries, the responses to acetylcholine, insulin, and sodium nitroprusside, as well as nitric oxide-mediated dilation, were not different between estrous cycle phases. In the aorta, expression of phosphorylated estrogen receptor-α was higher for mice in estrus compared with mice in proestrus. In the cerebral arteries, gene expression for estrogen receptor-β (Esr2) was lowest for mice in estrus compared with diestrus and proestrus. These results demonstrate that the estrus phase is associated with lower in vivo large artery stiffness in mice. In contrast, ex vivo resistance artery endothelial function is not different between estrous cycle phases. Estrogen receptor expression is modulated by the estrus cycle in an artery-dependent manner. These results suggest that the estrous cycle phase should be considered when measuring in vivo arterial stiffness in young female mice.NEW & NOTEWORTHY To design rigorous vascular research studies using young female rodents, the influence of the estrous cycle on vascular function must be known. We found that in vivo aortic stiffness was lower during estrus compared with the diestrus phase in female mice. In contrast, ex vivo mesenteric and cerebral artery endothelial function did not differ between estrous cycle stages. These results suggest that the estrous cycle stage should be accounted for when measuring in vivo arterial stiffness.
Collapse
Affiliation(s)
| | - Bradley R Bedell
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Abigail E Cullen
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Aleena Khurana
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Holly J D'Amico
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Grant D Henson
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| |
Collapse
|
27
|
Vallet A, El Ezzy M, Diennet M, Haidar S, Bouvier M, Mader S. The AF-2 cofactor binding region is key for the selective SUMOylation of estrogen receptor alpha by antiestrogens. J Biol Chem 2022; 299:102757. [PMID: 36460099 PMCID: PMC9823126 DOI: 10.1016/j.jbc.2022.102757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Antiestrogens (AEs) are used to treat all stages of estrogen receptor (ER)-positive breast cancer. Selective estrogen receptor modulators such as tamoxifen have tissue-specific partial agonist activity, while selective estrogen receptor downregulators such as fulvestrant (ICI182,780) display a more complete antiestrogenic profile. We have previously observed that fulvestrant-induced ERα SUMOylation contributes to transcriptional suppression, but whether this effect is seen with other AEs and is specific to ERα is unclear. Here we show that several AEs induce SUMOylation of ERα, but not ERβ, at different levels. Swapping domains between ERα and ERβ indicates that the ERα identity of the ligand-binding domain helices 3 and 4 (H3-H4 region), which contribute to the static part of the activation function-2 (AF-2) cofactor binding groove, is sufficient to confer fulvestrant-induced SUMOylation to ERβ. This region does not contain lysine residues unique to ERα, suggesting that ERα-specific residues in H3-H4 determine the capacity of the AE-bound ERα ligand-binding domain to recruit the SUMOylation machinery. We also show that the SUMO E3 ligase protein inhibitor of activated STAT 1 increases SUMOylation of ERα and of ERβ containing the H3-H4 region of ERα, but not of ERβ. Together, these results shed new light on the molecular basis for the differential capacity of selective estrogen receptor modulators and selective estrogen receptor downregulators to suppress transcription by ERα.
Collapse
Affiliation(s)
- Amandine Vallet
- Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, Québec, Canada
| | - Mohamed El Ezzy
- Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, Québec, Canada
| | - Marine Diennet
- Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, Québec, Canada
| | - Salwa Haidar
- Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, Québec, Canada
| | - Michel Bouvier
- Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, Québec, Canada,Département de Biochimie et Biologie Moléculaire, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Mader
- Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, Québec, Canada,Département de Biochimie et Biologie Moléculaire, Université de Montréal, Montréal, Québec, Canada,For correspondence: Sylvie Mader
| |
Collapse
|
28
|
Uzar I, Bogacz A, Sowińska-Przepiera E, Kotrych K, Wolek M, Sulikowski T, Kamiński A. The influence of ESR1 polymorphisms on selected hormonal, metabolic and mineral balance markers in women with hyperandrogenism. Sci Rep 2022; 12:19712. [PMID: 36385124 PMCID: PMC9668905 DOI: 10.1038/s41598-022-17383-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Hyperandrogenism is the most common endocrine disorder in women, characterized by an imbalance in normal estrogen and androgen levels in the blood. Androgens influence bone mineral density, body mass composition, muscle mass, mental state, and the regulation of sexual function.. The aim of the study was to assess the effect of estrogen receptor α gene (ESR1) polymorphisms on selected markers of bone metabolism and hormonal parameters in women with hyperandrogenism. The study group included 80 young women with hyperandrogenism who underwent measurements of bone mineral density (BMD), and determination of hormonal and metabolic parameters. Enzyme immunoassays were used to measure leptin, sRANKL (soluble receptor activator of nuclear factor-kB ligand), osteoprotegerin and 25-OH vitamin D total levels. An analysis of ESR1 gene polymorphisms was performed using the real-time PCR method. A relationship was demonstrated between the concentration of free estradiol (FEI) and the concentration of 17-OH-progesterone, and the ESR1 gene polymorphisms: rs3020314 (p = 0.031, p = 0.026 respectively) and rs1884051 (p = 0.033, p = 0.026 respectively). In conclusion, the ESR gene polymorphisms may be associated with hormonal disturbances in the concentration of estrogens and androgens, in hyperandrogenism in young women which may indirectly affect bone mineral density. However, no statistically significant relationships between the studied polymorphisms and the selected parameters of mineral metabolism have been demonstrated..
Collapse
Affiliation(s)
- Izabela Uzar
- Department of Pharmacology and Pharmacoeconomics, Pomeranian Medical University in Szczecin, 71-230, Szczecin, Poland
| | - Anna Bogacz
- Department of Stem Cells and Regenerative Medicine, Institute of Natural Fibers and Medicinal Plants, 62-064, Plewiska, Poland.
| | - Elżbieta Sowińska-Przepiera
- Department of Endocrinology, Metabolic Diseases, and Internal Diseases, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland
| | - Katarzyna Kotrych
- Department of General and Dental Radiology, Pomeranian Medical University in Szczecin, 70-111, Szczecin, Poland
| | - Marlena Wolek
- Department of Stem Cells and Regenerative Medicine, Institute of Natural Fibers and Medicinal Plants, 62-064, Plewiska, Poland
| | - Tadeusz Sulikowski
- General, Mini-Invasive and Gastroenterogical Surgery Clinic, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland
| | - Adam Kamiński
- Department of Orthopedics and Traumatology, Independent Public Clinical Hospital No. 1, Pomeranian Medical University, 71-252, Szczecin, Poland
| |
Collapse
|
29
|
Hirao-Suzuki M. Mechanisms of Cancer Malignancy Elicited by Environmental Chemicals: Analysis Focusing on Cadmium and Bisphenol A. YAKUGAKU ZASSHI 2022; 142:1161-1168. [DOI: 10.1248/yakushi.22-00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
30
|
Ewert J, Heintze L, Jordà-Redondo M, von Glasenapp JS, Nonell S, Bucher G, Peifer C, Herges R. Photoswitchable Diazocine-Based Estrogen Receptor Agonists: Stabilization of the Active Form inside the Receptor. J Am Chem Soc 2022; 144:15059-15071. [PMID: 35952371 DOI: 10.1021/jacs.2c03649] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Photopharmacology is an emerging approach in drug design and pharmacological therapy. Light is used to switch a pharmacophore between a biologically inactive and an active isomer with high spatiotemporal resolution at the site of illness, thus potentially avoiding side effects in neighboring healthy tissue. The most frequently used strategy to design a photoswitchable drug is to replace a suitable functional group in a known bioactive molecule with azobenzene. Our strategy is different in that the photoswitch moiety is closer to the drug's scaffold. Docking studies reveal a very high structural similarity of natural 17β-estradiol and the E isomers of dihydroxy diazocines, but not their Z isomers, respectively. Seven dihydroxy diazocines were synthesized and subjected to a biological estrogen reporter gene assay. Four derivatives exhibit distinct estrogenic activity after irradiation with violet light, which can be shut off with green light. Most remarkably, the photogenerated, active E form of one of the active compounds isomerizes back to the inactive Z form with a half-life of merely several milliseconds in water, but nevertheless is active for more than 3 h in the presence of the estrogen receptor. The results suggest a significant local impact of the ligand-receptor complex toward back-isomerization. Thus, drugs that are active when bound but lose their activity immediately after leaving the receptor could be of great pharmacological value because they strongly increase target specificity. Moreover, the drugs are released into the environment in their inactive form. The latter argument is particularly important for drugs that act as endocrine disruptors.
Collapse
Affiliation(s)
- Julia Ewert
- Otto-Diels-Institute of Organic Chemistry, Christian-Albrechts-University of Kiel, 24098 Kiel, Germany
| | - Linda Heintze
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany
| | | | - Jan-Simon von Glasenapp
- Otto-Diels-Institute of Organic Chemistry, Christian-Albrechts-University of Kiel, 24098 Kiel, Germany
| | - Santi Nonell
- Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain
| | - Götz Bucher
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, U. K
| | - Christian Peifer
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany
| | - Rainer Herges
- Otto-Diels-Institute of Organic Chemistry, Christian-Albrechts-University of Kiel, 24098 Kiel, Germany
| |
Collapse
|
31
|
Bonfim Neto AP, Cardoso APMM, Silva RDS, Sousa LMMDC, Giometti IC, Binelli M, Bauersachs S, Kowalewski MP, Papa PDC. An approach to uncover the relationship between 17b-estradiol and ESR1/ESR2 ratio in the regulation of canine corpus luteum. Front Vet Sci 2022; 9:885257. [PMID: 35982918 PMCID: PMC9378837 DOI: 10.3389/fvets.2022.885257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022] Open
Abstract
The canine corpus luteum (CL) is able to synthetise, activate and deactivate 17b-estradiol (E2) and also expresses nuclear estrogen receptors in a time-dependent manner during diestrus. Nevertheless, we are still missing a better comprehension of E2 functions in the canine CL, especially regarding the specific roles of estrogen receptor alpha (ERa) and ERb, encoded by ESR1 and 2, respectively. For that purpose, we analyzed transcriptomic data of canine non-pregnant CL collected on days 10, 20, 30, 40, 50 and 60 of diestrus and searched for differentially expressed genes (DEG) containing predicted transcription factor binding sites (TFBS) for ESR1 or ESR2. Based on biological functions of DEG presenting TFBS, expression of select transcripts and corresponding proteins was assessed. Additionally, luteal cells were collected across specific time points during diestrus and specificity of E2 responses was tested using ERa and/or ERb inhibitors. Bioinformatic analyses revealed 517 DEGs containing TFBS, from which 67 for both receptors. In general, abundance of predicted ESR1 targets was greater in the beginning, while abundance of ESR2 targets was greater in the end of diestrus. ESR1/ESR2 ratio shifted from an increasing to a decreasing pattern from day 30 to 40 post ovulation. Specific receptor inhibition suggested an ERa-mediated positive regulation of CL function at the beginning of diestrus and an ERb-mediated effect contributing to luteal regression. In conclusion, our data points toward a broad spectrum of action of E2 and its nuclear receptors, which can also act as transcription factors for other genes regulating canine CL function.
Collapse
Affiliation(s)
| | | | - Renata dos Santos Silva
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Ines Cristina Giometti
- Faculty of Veterinary Medicine, University of Western São Paulo, Presidente Prudente, Brazil
| | - Mario Binelli
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Stefan Bauersachs
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | - Paula de Carvalho Papa
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- *Correspondence: Paula de Carvalho Papa
| |
Collapse
|
32
|
Davey MG, Davey CM, Bouz L, Kerin E, McFeetors C, Lowery AJ, Kerin MJ. Relevance of the 21-gene expression assay in male breast cancer: A systematic review and meta-analysis. Breast 2022; 64:41-46. [PMID: 35512428 PMCID: PMC9079225 DOI: 10.1016/j.breast.2022.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/22/2022] [Accepted: 04/26/2022] [Indexed: 12/19/2022] Open
Affiliation(s)
- Matthew G Davey
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, H91YR71, Ireland.
| | - Ciara M Davey
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, H91YR71, Ireland
| | - Luis Bouz
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, H91YR71, Ireland
| | - Eoin Kerin
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, H91YR71, Ireland
| | - Carson McFeetors
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, H91YR71, Ireland
| | - Aoife J Lowery
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, H91YR71, Ireland
| | - Michael J Kerin
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, H91YR71, Ireland
| |
Collapse
|
33
|
Henriques PC, Aquino NSS, Campideli-Santana AC, Silva JF, Araujo-Lopes R, Franci CR, Coimbra CC, Szawka RE. Hypothalamic Expression of Estrogen Receptor Isoforms Underlies Estradiol Control of Luteinizing Hormone in Female Rats. Endocrinology 2022; 163:6631316. [PMID: 35789268 DOI: 10.1210/endocr/bqac101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Indexed: 11/19/2022]
Abstract
Luteinizing hormone (LH) secretion during the ovarian cycle is governed by fluctuations in circulating estradiol (E2) that oppositely regulate kisspeptin neurons in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC) of the hypothalamus. However, how these effects are orchestrated to achieve fertility is unknown. Here, we have tested the hypothesis that AVPV and ARC neurons have different sensitivities to E2 to coordinate changes in LH secretion. Cycling and ovariectomized rats with low and high E2 levels were used. As an index of E2 responsiveness, progesterone receptor (PR) was expressed only in the AVPV of rats with high E2, showing the preovulatory LH surge. On the other hand, kisspeptin neurons in the ARC responded to low E2 levels sufficient to suppress LH release. Notably, the Esr1/Esr2 ratio of gene expression was higher in the ARC than AVPV, regardless of E2 levels. Accordingly, the selective pharmacological activation of estrogen receptor α (ERα) required lower doses to induce PR in the ARC. The activation of ERβ, in turn, amplified E2-induced PR expression in the AVPV and the LH surge. Thus, ARC and AVPV neurons are differently responsive to E2. Lower E2 levels activate ERα in the ARC, whereas ERβ potentiates the E2 positive feedback in the AVPV, which appears related to the differential Esr1/Esr2 ratio in these 2 brain areas. Our findings provide evidence that the distinct expression of ER isoforms in the AVPV and ARC plays a key role in the control of periodic secretion of LH required for fertility in females.
Collapse
Affiliation(s)
- Patricia C Henriques
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Nayara S S Aquino
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Ana C Campideli-Santana
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Juneo F Silva
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Roberta Araujo-Lopes
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Celso R Franci
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirao Preto, 14049-900, Universidade de São Paulo, Ribeirao Preto, Brazil
| | - Candido C Coimbra
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Raphael E Szawka
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| |
Collapse
|
34
|
Molecular Mechanism of Equine Endometrosis: The NF-κB-Dependent Pathway Underlies the Ovarian Steroid Receptors’ Dysfunction. Int J Mol Sci 2022; 23:ijms23137360. [PMID: 35806363 PMCID: PMC9266418 DOI: 10.3390/ijms23137360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 12/10/2022] Open
Abstract
Endometrosis is a frequently occurring disease decreasing mares’ fertility. Thus, it is an important disease of the endometrium associated with epithelial and stromal cell alterations, endometrium gland degeneration and periglandular fibrosis. Multiple degenerative changes are found in uterine mucosa, the endometrium. However, their pathogenesis is not well known. It is thought that nuclear factor-κB (NF-κB), a cell metabolism regulator, and its activation pathways take part in it. The transcription of the profibrotic pathway genes of the NF-κB in fibrotic endometria differed between the follicular (FLP) and mid-luteal (MLP) phases of the estrous cycle, as well as with fibrosis progression. This study aimed to investigate the transcription of genes of estrogen (ESR1, ESR2) and progesterone receptors (PGR) in equine endometria to find relationships between the endocrine environment, NF-κB-pathway, and fibrosis. Endometrial samples (n = 100), collected in FLP or MLP, were classified histologically, and examined using quantitative PCR. The phase of the cycle was determined through the evaluation of ovarian structures and hormone levels (estradiol, progesterone) in serum. The transcription of ESR1, ESR2, and PGR decreased with the severity of endometrial fibrosis and degeneration of the endometrium. Moreover, differences in the transcription of ESR1, ESR2, and PGR were noted between FLP and MLP in the specific categories and histopathological type of equine endometrosis. In FLP and MLP, specific moderate and strong correlations between ESR1, ESR2, PGR and genes of the NF-κB pathway were evidenced. The transcription of endometrial steroid receptors can be subjected to dysregulation with the degree of equine endometrosis, especially in both destructive types of endometrosis, and mediated by the canonical NF-κB pathway depending on the estrous cycle phase.
Collapse
|
35
|
Stack ME, Cossaboon JM, Tubbs CW, Vilchis LI, Felton RG, Johnson JL, Danil K, Heckel G, Hoh E, Dodder NG. Assessing Marine Endocrine-Disrupting Chemicals in the Critically Endangered California Condor: Implications for Reintroduction to Coastal Environments. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:7800-7809. [PMID: 35579339 DOI: 10.1021/acs.est.1c07302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Coastal reintroduction sites for California condors (Gymnogyps californianus) can lead to elevated halogenated organic compound (HOC) exposure and potential health impacts due to the consumption of scavenged marine mammals. Using nontargeted analysis based on comprehensive two-dimensional gas chromatography coupled to time-of-flight mass spectrometry (GC×GC/TOF-MS), we compared HOC profiles of plasma from inland and coastal scavenging California condors from the state of California (CA), and marine mammal blubber from CA and the Gulf of California off Baja California (BC), Mexico. We detected more HOCs in coastal condors (32 ± 5, mean number of HOCs ± SD, n = 7) than in inland condors (8 ± 1, n = 10) and in CA marine mammals (136 ± 87, n = 25) than in BC marine mammals (55 ± 46, n = 8). ∑DDT-related compounds, ∑PCBs, and total tris(chlorophenyl)methane (∑TCPM) were, respectively, ∼7, ∼3.5, and ∼148 times more abundant in CA than in BC marine mammals. The endocrine-disrupting potential of selected polychlorinated biphenyls (PCB) congeners, TCPM, and TCPMOH was determined by in vitro California condor estrogen receptor (ER) activation. The higher levels of HOCs in coastal condors compared to those in inland condors and lower levels of HOC contamination in Baja California marine mammals compared to those from the state of California are factors to consider in condor reintroduction efforts.
Collapse
Affiliation(s)
- Margaret E Stack
- San Diego State University Research Foundation, San Diego, California 92182, United States
| | - Jennifer M Cossaboon
- School of Public Health, San Diego State University, San Diego, California 92182, United States
| | - Christopher W Tubbs
- Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, California 92027, United States
| | - L Ignacio Vilchis
- Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, California 92027, United States
| | - Rachel G Felton
- Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, California 92027, United States
| | - Jade L Johnson
- School of Public Health, San Diego State University, San Diego, California 92182, United States
| | - Kerri Danil
- Southwest Fisheries Science Center, National Marine Fisheries Service, National Oceanographic and Atmospheric Administration, La Jolla, California 92037, United States
| | - Gisela Heckel
- Centro de Investigacion Cientifica y de Educacion Superior de Ensenada, 22860 Ensenada, Baja California, Mexico
| | - Eunha Hoh
- School of Public Health, San Diego State University, San Diego, California 92182, United States
| | - Nathan G Dodder
- San Diego State University Research Foundation, San Diego, California 92182, United States
- School of Public Health, San Diego State University, San Diego, California 92182, United States
| |
Collapse
|
36
|
Bhave S, Ho WLN, Cheng K, Omer M, Bousquet N, Guyer RA, Hotta R, Goldstein AM. Tamoxifen administration alters gastrointestinal motility in mice. Neurogastroenterol Motil 2022; 34:e14357. [PMID: 35279902 DOI: 10.1111/nmo.14357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 12/13/2021] [Accepted: 01/28/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Tamoxifen is widely used for Cre-estrogen receptor-mediated genomic recombination in transgenic mouse models to mark cells for lineage tracing and to study gene function. However, recent studies have highlighted off-target effects of tamoxifen in various tissues and cell types when used for induction of Cre recombination. Despite the widespread use of these transgenic Cre models to assess gastrointestinal (GI) function, the effect of tamoxifen exposure on GI motility has not been described. METHODS We examined the effects of tamoxifen on GI motility by measuring total GI transit, gastric emptying, small intestinal transit, and colonic contractility in wild-type adult mice. KEY RESULTS We observed a significant delay in total GI transit in tamoxifen-treated mice, with unaltered gastric emptying, accelerated small intestinal transit, and abnormal colonic motility. CONCLUSION Our findings highlight the importance of considering GI motility alterations induced by tamoxifen when designing protocols that utilize tamoxifen as a Cre-driver for studying GI function.
Collapse
Affiliation(s)
- Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wing Lam N Ho
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Katarina Cheng
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meredith Omer
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicole Bousquet
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard A Guyer
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
ERβ Isoforms Have Differential Clinical Significance in Breast Cancer Subtypes and Subgroups. Curr Issues Mol Biol 2022; 44:1564-1586. [PMID: 35723365 PMCID: PMC9164084 DOI: 10.3390/cimb44040107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
ERβ, an ER subtype first identified in 1996, is highly expressed in different types of BCa including ERα-negative BCa and TNBC. Many studies on ERβ expression investigated mostly on ERβ1 protein expression in ERα-positive and ERα-negative BCa combined. The results are conflicting. This may be due to the complexity of ERβ isoforms, subject heterogeneity, and various study designs targeting different ERβ isoforms and either ERβ protein or mRNA expression, as well as to the lack of a standardized testing protocol. Herein, we simultaneously investigated both mRNA and protein expression of ERβ isoforms 1, 2, and 5 in different BCa subtypes and clinical characteristics. Patient samples (138) and breast cancer cell lines (BCC) reflecting different types of BCa were tested for ERα and ERβ mRNA expression using quantitative real-time PCR, as well as for protein expression of ERα, ERβ1, ERβ2, and ERβ5 isoforms, PR, HER2/neu, Ki-67, CK 5/6, and p53 using immunohistochemistry. Associations of ERβ isoform expression with clinical characteristics and overall survival (OS) were analyzed. ERβ1, 2, and 5 isoforms are differentially expressed in different BCa subtypes including ERα-negative and TNBC. Each ERβ isoform seemingly plays a distinct role and is associated with clinical tumor characteristics and patient outcomes. ERβ isoform expression is significantly associated with >15% Ki-67 positivity and poor prognostic markers, and it predicts poorer OS, mostly in the subgroups. High ERβ2 and 5 isoform expression in ERα-negative BCa and TNBC is predictive of poor OS. Further investigation of ERβ isoforms in a larger cohort of BCa subgroups is needed to evaluate the role of ERβ for the potential usefulness of ERβ as a prognostic and predictive marker and for therapeutic use. The inconsistent outcomes of ERβ isoform mRNA or protein expression in many studies suggest that the standardization of ERβ testing would facilitate the use of ERβ in a clinical setting.
Collapse
|
38
|
Jehle S, Kunowska N, Benlasfer N, Woodsmith J, Weber G, Wahl MC, Stelzl U. A human kinase yeast array for the identification of kinases modulating phosphorylation-dependent protein-protein interactions. Mol Syst Biol 2022; 18:e10820. [PMID: 35225431 PMCID: PMC8883442 DOI: 10.15252/msb.202110820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Protein kinases play an important role in cellular signaling pathways and their dysregulation leads to multiple diseases, making kinases prime drug targets. While more than 500 human protein kinases are known to collectively mediate phosphorylation of over 290,000 S/T/Y sites, the activities have been characterized only for a minor, intensively studied subset. To systematically address this discrepancy, we developed a human kinase array in Saccharomyces cerevisiae as a simple readout tool to systematically assess kinase activities. For this array, we expressed 266 human kinases in four different S. cerevisiae strains and profiled ectopic growth as a proxy for kinase activity across 33 conditions. More than half of the kinases showed an activity-dependent phenotype across many conditions and in more than one strain. We then employed the kinase array to identify the kinase(s) that can modulate protein-protein interactions (PPIs). Two characterized, phosphorylation-dependent PPIs with unknown kinase-substrate relationships were analyzed in a phospho-yeast two-hybrid assay. CK2α1 and SGK2 kinases can abrogate the interaction between the spliceosomal proteins AAR2 and PRPF8, and NEK6 kinase was found to mediate the estrogen receptor (ERα) interaction with 14-3-3 proteins. The human kinase yeast array can thus be used for a variety of kinase activity-dependent readouts.
Collapse
Affiliation(s)
- Stefanie Jehle
- Otto-Warburg-Laboratory, Max-Planck-Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Natalia Kunowska
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Nouhad Benlasfer
- Otto-Warburg-Laboratory, Max-Planck-Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Jonathan Woodsmith
- Otto-Warburg-Laboratory, Max-Planck-Institute for Molecular Genetics (MPIMG), Berlin, Germany
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Gert Weber
- Institut für Chemie und Biochemie, Freie Universität, Berlin, Germany
- Helmholtz-Zentrum Berlin für Materialien und Energie, Macromolecular Crystallography, Berlin, Germany
| | - Markus C Wahl
- Institut für Chemie und Biochemie, Freie Universität, Berlin, Germany
| | - Ulrich Stelzl
- Otto-Warburg-Laboratory, Max-Planck-Institute for Molecular Genetics (MPIMG), Berlin, Germany
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz and BioTechMed-Graz, Graz, Austria
| |
Collapse
|
39
|
Chimento A, De Luca A, Avena P, De Amicis F, Casaburi I, Sirianni R, Pezzi V. Estrogen Receptors-Mediated Apoptosis in Hormone-Dependent Cancers. Int J Mol Sci 2022; 23:1242. [PMID: 35163166 PMCID: PMC8835409 DOI: 10.3390/ijms23031242] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
It is known that estrogen stimulates growth and inhibits apoptosis through estrogen receptor(ER)-mediated mechanisms in many cancer cell types. Interestingly, there is strong evidence that estrogens can also induce apoptosis, activating different ER isoforms in cancer cells. It has been observed that E2/ERα complex activates multiple pathways involved in both cell cycle progression and apoptotic cascade prevention, while E2/ERβ complex in many cases directs the cells to apoptosis. However, the exact mechanism of estrogen-induced tumor regression is not completely known. Nevertheless, ERs expression levels of specific splice variants and their cellular localization differentially affect outcome of estrogen-dependent tumors. The goal of this review is to provide a general overview of current knowledge on ERs-mediated apoptosis that occurs in main hormone dependent-cancers. Understanding the molecular mechanisms underlying the induction of ER-mediated cell death will be useful for the development of specific ligands capable of triggering apoptosis to counteract estrogen-dependent tumor growth.
Collapse
Affiliation(s)
- Adele Chimento
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Arianna De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Paola Avena
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Ivan Casaburi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rosa Sirianni
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| |
Collapse
|
40
|
Tsoi H, Shi L, Leung MH, Man EPS, So ZQ, Chan WL, Khoo US. Overexpression of BQ323636.1 Modulated AR/IL-8/CXCR1 Axis to Confer Tamoxifen Resistance in ER-Positive Breast Cancer. Life (Basel) 2022; 12:93. [PMID: 35054486 PMCID: PMC8778777 DOI: 10.3390/life12010093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 02/07/2023] Open
Abstract
NCOR2 is a co-repressor for estrogen receptor (ER) and androgen receptor (AR). Our group previously identified a novel splice variant of NCOR2, BQ323636.1 (BQ), that mediates tamoxifen resistance via interference of NCOR2 repression on ER. Luciferase reporter assay showed BQ overexpression could enhance the transcriptional activity of androgen response element (ARE). We proposed that BQ employs both AR and ER to confer tamoxifen resistance. Through in silico analysis, we identified interleukin-8 (IL-8) as the sole ERE and ARE containing gene responsiveness to ER and AR activation. We confirmed that BQ overexpression enhanced the expression of IL-8 in ER+ve breast cancer cells, and AR inhibition reduced IL-8 expression in the BQ overexpressing cell lines, suggesting that AR was involved in the modulation of IL-8 expression by BQ. Moreover, we demonstrated that IL-8 could activate both AKT and ERK1/2 via CXCR1 to confer tamoxifen resistance. Targeting CXCR1/2 by a small inhibitor repertaxin reversed tamoxifen resistance of BQ overexpressing breast cancer cells in vitro and in vivo. In conclusion, BQ overexpression in ER+ve breast cancer can enhance IL-8 mediated signaling to modulate tamoxifen resistance. Targeting IL-8 signaling is a promising approach to overcome tamoxifen resistance in ER+ve breast cancer.
Collapse
Affiliation(s)
- Ho Tsoi
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.T.); (L.S.); (M.-H.L.); (E.P.S.M.); (Z.-Q.S.)
| | - Ling Shi
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.T.); (L.S.); (M.-H.L.); (E.P.S.M.); (Z.-Q.S.)
| | - Man-Hong Leung
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.T.); (L.S.); (M.-H.L.); (E.P.S.M.); (Z.-Q.S.)
| | - Ellen P. S. Man
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.T.); (L.S.); (M.-H.L.); (E.P.S.M.); (Z.-Q.S.)
| | - Zi-Qing So
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.T.); (L.S.); (M.-H.L.); (E.P.S.M.); (Z.-Q.S.)
| | - Wing-Lok Chan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Ui-Soon Khoo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.T.); (L.S.); (M.-H.L.); (E.P.S.M.); (Z.-Q.S.)
| |
Collapse
|
41
|
Chauvin S, Cohen-Tannoudji J, Guigon CJ. Estradiol Signaling at the Heart of Folliculogenesis: Its Potential Deregulation in Human Ovarian Pathologies. Int J Mol Sci 2022; 23:ijms23010512. [PMID: 35008938 PMCID: PMC8745567 DOI: 10.3390/ijms23010512] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/26/2023] Open
Abstract
Estradiol (E2) is a major hormone controlling women fertility, in particular folliculogenesis. This steroid, which is locally produced by granulosa cells (GC) within ovarian follicles, controls the development and selection of dominant preovulatory follicles. E2 effects rely on a complex set of nuclear and extra-nuclear signal transduction pathways principally triggered by its nuclear receptors, ERα and ERβ. These transcription factors are differentially expressed within follicles, with ERβ being the predominant ER in GC. Several ERβ splice isoforms have been identified and display specific structural features, which greatly complicates the nature of ERβ-mediated E2 signaling. This review aims at providing a concise overview of the main actions of E2 during follicular growth, maturation, and selection in human. It also describes the current understanding of the various roles of ERβ splice isoforms, especially their influence on cell fate. We finally discuss how E2 signaling deregulation could participate in two ovarian pathogeneses characterized by either a follicular arrest, as in polycystic ovary syndrome, or an excess of GC survival and proliferation, leading to granulosa cell tumors. This review emphasizes the need for further research to better understand the molecular basis of E2 signaling throughout folliculogenesis and to improve the efficiency of ovarian-related disease therapies.
Collapse
|
42
|
Xia S, Lin Q. Estrogen Receptor Bio-Activities Determine Clinical Endocrine Treatment Options in Estrogen Receptor-Positive Breast Cancer. Technol Cancer Res Treat 2022; 21:15330338221090351. [PMID: 35450488 PMCID: PMC9036337 DOI: 10.1177/15330338221090351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In estrogen receptor positive (ER+) breast cancer therapy, estrogen receptors (ERs) are the major targeting molecules. ER-targeted therapy has provided clinical benefits for approximately 70% of all breast cancer patients through targeting the ERα subtype. In recent years, mechanisms underlying breast cancer occurrence and progression have been extensively studied and largely clarified. The PI3K/AKT/mTOR pathway, microRNA regulation, and other ER downstream signaling pathways are found to be the effective therapeutic targets in ER+ BC therapy. A number of the ER+ (ER+) breast cancer biomarkers have been established for diagnosis and prognosis. The ESR1 gene mutations that lead to endocrine therapy resistance in ER+ breast cancer had been identified. Mutations in the ligand-binding domain of ERα which encoded by ESR1 gene occur in most cases. The targeted drugs combined with endocrine therapy have been developed to improve the therapeutic efficacy of ER+ breast cancer, particularly the endocrine therapy resistance ER+ breast cancer. The combination therapy has been demonstrated to be superior to monotherapy in overall clinical evaluation. In this review, we focus on recent progress in studies on ERs and related clinical applications for targeted therapy and provide a perspective view for therapy of ER+ breast cancer.
Collapse
Affiliation(s)
- Song Xia
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang, China
- Qiong Lin, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China.
| |
Collapse
|
43
|
Choi Y. Estrogen Receptor β Expression and Its Clinical Implication in Breast Cancers: Favorable or Unfavorable? J Breast Cancer 2022; 25:75-93. [PMID: 35380018 PMCID: PMC9065353 DOI: 10.4048/jbc.2022.25.e9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 02/03/2022] [Indexed: 11/30/2022] Open
Abstract
There are two estrogen receptor (ER) genes (ESR1/ERα and ESR2/ERβ) in humans. Of those. ERβ, the second ER isotype identified in 1996, is differentially expressed in different phenotypes and molecular subtypes of breast cancer (BCa), and is highly expressed in ERα-negative BCa and triple-negative BCa (TNBC). This review summarizes the potential clinical relevance of ERβ in BCa and the challenges associated with studies on the role of ERβ in BCa. The experimental and clinical studies evaluating clinical outcomes and associations with clinical characteristics and responses to endocrine therapy on targeting ERβ reviewed herein indicate that ERβ is a clinically important biomarker in BCa. The reviewed studies also suggest that each ERβ isoform has a distinct role in BCa subtypes and the potential of novel- targeted therapies in BCa, especially ERα-negative BCa and TNBC. However, the findings of many studies on ERβ are inconsistent, and the exact role of ERβ in BCa remains elusive; this may potentially be attributed to the complexity of ERβ isoforms, but also to the lack of standardized testing protocol. Thus, successful clinical application of ERβ requires the development of standardized, reproducible, and objective measurement methods for ERβ that can be widely and routinely applied in clinical setting.
Collapse
Affiliation(s)
- Young Choi
- Department of Pathology, Yale School of Medicine, Hartsdale, NY, USA
| |
Collapse
|
44
|
Maharjan CK, Mo J, Wang L, Kim MC, Wang S, Borcherding N, Vikas P, Zhang W. Natural and Synthetic Estrogens in Chronic Inflammation and Breast Cancer. Cancers (Basel) 2021; 14:cancers14010206. [PMID: 35008370 PMCID: PMC8744660 DOI: 10.3390/cancers14010206] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022] Open
Abstract
The oncogenic role of estrogen receptor (ER) signaling in breast cancer has long been established. Interaction of estrogen with estrogen receptor (ER) in the nucleus activates genomic pathways of estrogen signaling. In contrast, estrogen interaction with the cell membrane-bound G-protein-coupled estrogen receptor (GPER) activates the rapid receptor-mediated signaling transduction cascades. Aberrant estrogen signaling enhances mammary epithelial cell proliferation, survival, and angiogenesis, hence is an important step towards breast cancer initiation and progression. Meanwhile, a growing number of studies also provide evidence for estrogen's pro- or anti-inflammatory roles. As other articles in this issue cover classic ER and GPER signaling mediated by estrogen, this review will discuss the crucial mechanisms by which estrogen signaling influences chronic inflammation and how that is involved in breast cancer. Xenoestrogens acquired from plant diet or exposure to industrial products constantly interact with and alter innate estrogen signaling at various levels. As such, they can modulate chronic inflammation and breast cancer development. Natural xenoestrogens generally have anti-inflammatory properties, which is consistent with their chemoprotective role in breast cancer. In contrast, synthetic xenoestrogens are proinflammatory and carcinogenic compounds that can increase the risk of breast cancer. This article also highlights important xenoestrogens with a particular focus on their role in inflammation and breast cancer. Improved understanding of the complex relationship between estrogens, inflammation, and breast cancer will guide clinical research on agents that could advance breast cancer prevention and therapy.
Collapse
Affiliation(s)
- Chandra K. Maharjan
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (C.K.M.); (J.M.); (L.W.); (M.-C.K.)
| | - Jiao Mo
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (C.K.M.); (J.M.); (L.W.); (M.-C.K.)
| | - Lei Wang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (C.K.M.); (J.M.); (L.W.); (M.-C.K.)
| | - Myung-Chul Kim
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (C.K.M.); (J.M.); (L.W.); (M.-C.K.)
| | - Sameul Wang
- Canyonoak Consulting LLC, San Diego, CA 92127, USA;
| | - Nicholas Borcherding
- Department of Pathology and Immunology, School of Medicine, Washington University, St. Louis, MO 63110, USA;
| | - Praveen Vikas
- Department of Internal Medicine, Carver College of Medicine, Iowa City, IA 52242, USA;
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (C.K.M.); (J.M.); (L.W.); (M.-C.K.)
- Mechanism of Oncogenesis Program, University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Correspondence: to: ; Tel.: +1-352-273-6748
| |
Collapse
|
45
|
Asiamah CA, Liu Y, Ye R, Pan Y, Lu LL, Zou K, Zhao Z, Jiang P, Su Y. Polymorphism analysis and expression profile of the estrogen receptor 2 gene in Leizhou black duck. Poult Sci 2021; 101:101630. [PMID: 35033905 PMCID: PMC8762077 DOI: 10.1016/j.psj.2021.101630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/13/2021] [Accepted: 11/25/2021] [Indexed: 12/30/2022] Open
Abstract
Our previous study on the ovarian transcriptomic analysis in Leizhou black duck revealed that the ESR2 gene was involved in hormone regulation in reproduction and the estrogen signaling pathway related to reproductive performance was enriched. This suggested that ESR2 may have a functional role in the reproductive performance of the Leizhou black duck. Thus, this study aimed at evaluating the polymorphism of the ESR2 gene and its association with egg-laying traits and the distribution pattern of ESR2 mRNA in laying and non-laying Leizhou black ducks. In this study, genomic DNA was extracted from blood samples of 101 Leizhou black ducks to identify single nucleotide polymorphisms (SNPs) of the ESR2 gene to elucidate molecular markers highly associated with egg-laying traits. Four each of laying and non-laying Leizhou black ducks were selected to collect different tissues to analyze the ESR2 gene expression. A total of 23 SNPs were identified and association analysis of the single SNP sites showed that SNPs g.56805646 T>C and exon 3-20G>A were significantly (P < 0.05) associated with egg weight. Ducks with CT and AG genotypes had significantly higher (P < 0.05) egg weights than their respective other genotypes. Haplotype association analysis of g.56805646 T>C and exon 3-20G>A showed that the haplotypes were significantly associated with egg weight. Higher egg weight was seen in individuals with H3H4 haplotypes. In the hypothalamus-pituitary-gonadal (HPG) axis, the results of qRT/PCR showed that ESR2 mRNA was significantly (P < 0.05) expressed in the ovaries of both duck groups than in the hypothalamus and pituitary. In the oviduct, ESR2 was significantly (P < 0.05) higher in the infundibulum and magnum of laying and non-laying ducks respectively. This study provides a molecular marker for selecting Leizhou black ducks for egg production. In addition, it offers theoretical knowledge for studying the related biological functions of the ESR2 gene at the cellular level.
Collapse
Affiliation(s)
- Collins Amponsah Asiamah
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Yuanbo Liu
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Rungen Ye
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Yiting Pan
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Li-Li Lu
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Kun Zou
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Zhihui Zhao
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Ping Jiang
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China
| | - Ying Su
- College of Coastal Agriculture, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, PR China.
| |
Collapse
|
46
|
The Effects of Osteoporotic and Non-osteoporotic Medications on Fracture Risk and Bone Mineral Density. Drugs 2021; 81:1831-1858. [PMID: 34724173 PMCID: PMC8578161 DOI: 10.1007/s40265-021-01625-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/26/2022]
Abstract
Osteoporosis is a highly prevalent bone disease affecting more than 37.5 million individuals in the European Union (EU) and the United States of America (USA). It is characterized by low bone mineral density (BMD), impaired bone quality, and loss of structural and biomechanical properties, resulting in reduced bone strength. An increase in morbidity and mortality is seen in patients with osteoporosis, caused by the approximately 3.5 million new osteoporotic fractures occurring every year in the EU. Currently, different medications are available for the treatment of osteoporosis, including anti-resorptive and osteoanabolic medications. Bisphosphonates, which belong to the anti-resorptive medications, are the standard treatment for osteoporosis based on their positive effects on bone, long-term experience, and low costs. However, not only medications used for the treatment of osteoporosis can affect bone: several other medications are suggested to have an effect on bone as well, especially on fracture risk and BMD. Knowledge about the positive and negative effects of different medications on both fracture risk and BMD is important, as it can contribute to an improvement in osteoporosis prevention and treatment in general, and, even more importantly, to the individual's health. In this review, we therefore discuss the effects of both osteoporotic and non-osteoporotic medications on fracture risk and BMD. In addition, we discuss the underlying mechanisms of action.
Collapse
|
47
|
Hirao-Suzuki M. Estrogen Receptor β as a Possible Double-Edged Sword Molecule in Breast Cancer: A Mechanism of Alteration of Its Role by Exposure to Endocrine-Disrupting Chemicals. Biol Pharm Bull 2021; 44:1594-1597. [PMID: 34719637 DOI: 10.1248/bpb.b21-00468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Estrogen is essential for the growth and development of mammary glands and its signaling is associated with breast cancer growth. Estrogen can exert physiological actions via estrogen receptors α/β (ERα/β). There is experimental evidence suggesting that in ERα/β-positive breast cancer, ERα promotes tumor cell proliferation and ERβ inhibits ERα-mediated transcriptional activity, resulting in abrogation of cell growth. Therefore, ERβ is attracting attention as a potential tumor suppressor, and as a biomarker and therapeutic target in the ERα/β-positive breast cancer. Based on this information, we have hypothesized that some endocrine-disrupting chemicals (EDCs) that can perturb the balance between ERα and ERβ expression levels in breast cancer cells might have effects on the breast cancer proliferation (i.e., down-regulation of the α-type of ER). We have recently reported that 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), an active metabolite of bisphenol A, in ERα/β-positive human breast cancer significantly down-regulates ERα expression, yet stimulates cell proliferation through the activation of ERβ-mediated transcription. These results support our hypothesis by demonstrating that exposure to MBP altered the functional role of ERβ in breast cancer cells from suppressor to promoter. In contrast, some EDCs, such as Δ9-tetrahydrocannabinol and bisphenol AF, can exhibit anti-estrogenic effects through up-regulation of ERβ expression without affecting the ERα expression levels. However, there is no consensus on the correlation between ERβ expression levels and clinical prognosis, which might be due to differences in exposed chemicals. Therefore, elucidating the exposure effects of EDCs can reveal the reason for inconsistent functional role of ERβ in ERα/β-positive breast cancer.
Collapse
Affiliation(s)
- Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University
| |
Collapse
|
48
|
Role of Polyphenols in the Metabolism of the Skeletal System in Humans and Animals – A Review. ANNALS OF ANIMAL SCIENCE 2021. [DOI: 10.2478/aoas-2021-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abstract
Polyphenols are a group of compounds arousing enormous interest due to their multiple effects on both human and animal health and omnipresence in plants. A number of in vitro and animal model studies have shown that all polyphenols exhibit anti-inflammatory and antioxidant activities, and play a significant role against oxidative stress-related pathologies. They also exert gut promotory effects and prevent chronic degenerative diseases. However, less attention has been paid to the potential influence of polyphenols on bone properties and metabolism. It is well known that proper growth and functioning of the organism depend largely on bone growth and health. Therefore, understanding the action of substances (including polyphenols) that may improve the health and functioning of the skeletal system and bone metabolism is extremely important for the health of the present and future generations of both humans and farm animals. This review provides a comprehensive summary of literature related to causes of bone loss during ageing of the organism (in both humans and animals) and possible effects of dietary polyphenols preventing bone loss and diseases. In particular, the underlying cellular and molecular mechanisms that can modulate skeletal homeostasis and influence the bone modeling and remodeling processes are presented.
Collapse
|
49
|
Mandujano-Lázaro G, Galaviz-Hernández C, Reyes-López CA, Almanza-Pérez JC, Giacoman-Martínez A, López-Camarillo C, Huang F, Marchat LA. A Short S-Equol Exposure Has a Long-Term Inhibitory Effect on Adipogenesis in Mouse 3T3-L1 Cells. APPLIED SCIENCES 2021; 11:9657. [DOI: 10.3390/app11209657] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
Abstract
In the search for new drugs against obesity, the chronic disease that threatens human health worldwide, several works have focused on the study of estrogen homologs because of the role of estrogen receptors (ERs) in adipocyte growth. The isoflavone equol, an ERβ agonist, has shown beneficial metabolic effects in in vivo and in vitro assays; however, additional studies are required to better characterize its potential for body weight control. Here, we showed that the treatment of 3T3-L1 cells with 10 μM of S-equol for the first three days of the adipocyte differentiation protocol was able to prevent cells becoming semi-rounded and having a lipid droplet formation until the seventh day of culture; moreover, lipid accumulation was reduced by about 50%. Congruently, S-equol induced a reduction in mRNA expression of the adipogenic markers C/EBPα and PPARγ, and adipokines secretion, mainly Adiponectin, Leptin, Resistin, and MCP-1, while the release of PAI-1 was augmented. Moreover, it also reduced the expression of ERα and attenuated the subexpression of ERβ associated with adipogenesis. Altogether, our data suggested that S-equol binding to ERβ affects the transcriptional program that regulates adipogenesis and alters adipocyte functions. Future efforts will focus on studying the impact of S-equol on ER signaling pathways.
Collapse
Affiliation(s)
- Gilberto Mandujano-Lázaro
- Laboratorio 2 de Biomedicina Molecular, ENMH, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico
| | | | - César A. Reyes-López
- Laboratorio de Bioquímica Estructural, ENMH, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico
| | - Julio C. Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Abraham Giacoman-Martínez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Sede Sur, Ciudad de México 14330, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México 03100, Mexico
| | - Fengyang Huang
- Laboratorio de Farmacología y Toxicología, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico
| | - Laurence A. Marchat
- Laboratorio 2 de Biomedicina Molecular, ENMH, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico
| |
Collapse
|
50
|
Sun Y, Sangam S, Guo Q, Wang J, Tang H, Black SM, Desai AA. Sex Differences, Estrogen Metabolism and Signaling in the Development of Pulmonary Arterial Hypertension. Front Cardiovasc Med 2021; 8:719058. [PMID: 34568460 PMCID: PMC8460911 DOI: 10.3389/fcvm.2021.719058] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex and devastating disease with a poor long-term prognosis. While women are at increased risk for developing PAH, they exhibit superior right heart function and higher survival rates than men. Susceptibility to disease risk in PAH has been attributed, in part, to estrogen signaling. In contrast to potential pathological influences of estrogen in patients, studies of animal models reveal estrogen demonstrates protective effects in PAH. Consistent with this latter observation, an ovariectomy in female rats appears to aggravate the condition. This discrepancy between observations from patients and animal models is often called the "estrogen paradox." Further, the tissue-specific interactions between estrogen, its metabolites and receptors in PAH and right heart function remain complex; nonetheless, these relationships are essential to characterize to better understand PAH pathophysiology and to potentially develop novel therapeutic and curative targets. In this review, we explore estrogen-mediated mechanisms that may further explain this paradox by summarizing published literature related to: (1) the synthesis and catabolism of estrogen; (2) activity and functions of the various estrogen receptors; (3) the multiple modalities of estrogen signaling in cells; and (4) the role of estrogen and its diverse metabolites on the susceptibility to, and progression of, PAH as well as their impact on right heart function.
Collapse
Affiliation(s)
- Yanan Sun
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shreya Sangam
- Department of Medicine, Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, United States
| | - Qiang Guo
- Department of Critical Care Medicine, Suzhou Dushu Lake Hospital, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haiyang Tang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Stephen M. Black
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Miami, FL, United States
- Center for Translational Science and Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Port St. Lucie, FL, United States
| | - Ankit A. Desai
- Department of Medicine, Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, United States
| |
Collapse
|