1
|
Logsdon DM, Churchwell A, Schoolcraft WB, Krisher RL, Yuan Y. Estrogen signaling encourages blastocyst development and implantation potential. J Assist Reprod Genet 2023; 40:1003-1014. [PMID: 37017886 PMCID: PMC10239412 DOI: 10.1007/s10815-023-02783-2] [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: 01/19/2023] [Accepted: 03/21/2023] [Indexed: 04/06/2023] Open
Abstract
PURPOSE Estrogen is well-known for preparing uterine receptivity. However, its roles in regulating embryo development and implantation are unclear. Our objective was to characterize estrogen receptor 1 (ESR1) in human and mouse embryos and determine the effect of estradiol (E2) supplementation on pre- and peri-implantation blastocyst development. METHODS Mouse embryos, 8-cell through hatched blastocyst stages, and human embryonic days 5-7 blastocysts were stained for ESR1 and imaged using confocal microscopy. We then treated 8-cell mouse embryos with 8 nM E2 during in vitro culture (IVC) and examined embryo morphokinetics, blastocyst development, and cell allocation into the inner cell mass (ICM) and trophectoderm (TE). Finally, we disrupted ESR1, using ICI 182,780, and evaluated peri-implantation development. RESULTS ESR1 exhibits nuclear localization in early blastocysts followed by aggregation, predominantly in the TE of hatching and hatched blastocysts, in human and mouse embryos. During IVC, most E2 was absorbed by the mineral oil, and no effect on embryo development was found. When IVC was performed without an oil overlay, embryos treated with E2 exhibited increased blastocyst development and ICM:TE ratio. Additionally, embryos treated with ICI 182,780 had significantly decreased trophoblast outgrowth during extended embryo culture. CONCLUSION Similar ESR1 localization in mouse and human blastocysts suggests a conserved role in blastocyst development. These mechanisms may be underappreciated due to the use of mineral oil during conventional IVC. This work provides important context for how estrogenic toxicants may impact reproductive health and offers an avenue to further optimize human-assisted reproductive technology (ART) to treat infertility.
Collapse
Affiliation(s)
- Deirdre M. Logsdon
- Colorado Center for Reproductive Medicine, 10290 RidgeGate Circle, Lone Tree, CO 80124 USA
| | - Ashlyn Churchwell
- Colorado Center for Reproductive Medicine, 10290 RidgeGate Circle, Lone Tree, CO 80124 USA
| | - William B. Schoolcraft
- Colorado Center for Reproductive Medicine, 10290 RidgeGate Circle, Lone Tree, CO 80124 USA
| | | | - Ye Yuan
- Colorado Center for Reproductive Medicine, 10290 RidgeGate Circle, Lone Tree, CO 80124 USA
| |
Collapse
|
2
|
Aspros KGM, Emch MJ, Wang X, Subramaniam M, Hinkle ML, Rodman EPB, Goetz MP, Hawse JR. Disruption of estrogen receptor beta's DNA binding domain impairs its tumor suppressive effects in triple negative breast cancer. Front Med (Lausanne) 2023; 10:1047166. [PMID: 36926316 PMCID: PMC10011152 DOI: 10.3389/fmed.2023.1047166] [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: 09/17/2022] [Accepted: 02/09/2023] [Indexed: 03/08/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive sub-type of the disease which accounts for a disproportionately high percentage of breast cancer morbidities and mortalities. For these reasons, a better understanding of TNBC biology is required and the development of novel therapeutic approaches are critically needed. Estrogen receptor beta (ERβ) is a reported tumor suppressor that is expressed in approximately 20% of primary TNBC tumors, where it is associated with favorable prognostic features and patient outcomes. Previous studies have shown that ERβ mediates the assembly of co-repressor complexes on DNA to inhibit the expression of multiple growth promoting genes and to suppress the ability of oncogenic transcription factors to drive cancer progression. To further elucidate the molecular mechanisms by which ERβ elicits its anti-cancer effects, we developed MDA-MB-231 cells that inducibly express a mutant form of ERβ incapable of directly binding DNA. We demonstrate that disruption of ERβ's direct interaction with DNA abolishes its ability to regulate the expression of well characterized immediate response genes and renders it unable to suppress TNBC cell proliferation. Loss of DNA binding also diminishes the ability of ERβ to suppress oncogenic NFκB signaling even though it still physically associates with NFκB and other critical co-factors. These findings enhance our understanding of how ERβ functions in this disease and provide a model system that can be utilized to further investigate the mechanistic processes by which ERβ elicits its anti-cancer effects.
Collapse
Affiliation(s)
- Kirsten G. M. Aspros
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Michael J. Emch
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Xiyin Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Malayannan Subramaniam
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Megan L. Hinkle
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Esther P. B. Rodman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Matthew P. Goetz
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - John R. Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
3
|
Kuryłowicz A. Estrogens in Adipose Tissue Physiology and Obesity-Related Dysfunction. Biomedicines 2023; 11:biomedicines11030690. [PMID: 36979669 PMCID: PMC10045924 DOI: 10.3390/biomedicines11030690] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/30/2023] Open
Abstract
Menopause-related decline in estrogen levels is accompanied by a change in adipose tissue distribution from a gynoid to an android and an increased prevalence of obesity in women. These unfavorable phenomena can be partially restored by hormone replacement therapy, suggesting a significant role for estrogen in the regulation of adipocytes' function. Indeed, preclinical studies proved the involvement of these hormones in adipose tissue development, metabolism, and inflammatory activity. However, the relationship between estrogen and obesity is bidirectional. On the one hand-their deficiency leads to excessive fat accumulation and impairs adipocyte function, on the other-adipose tissue of obese individuals is characterized by altered expression of estrogen receptors and key enzymes involved in their synthesis. This narrative review aims to summarize the role of estrogen in adipose tissue development, physiology, and in obesity-related dysfunction. Firstly, the estrogen classification, synthesis, and modes of action are presented. Next, their role in regulating adipogenesis and adipose tissue activity in health and the course of obesity is described. Finally, the potential therapeutic applications of estrogen and its derivates in obesity treatment are discussed.
Collapse
Affiliation(s)
- Alina Kuryłowicz
- Department of Human Epigenetics, Mossakowski Medical Research Centre PAS, 02-106 Warsaw, Poland
- Department of General Medicine and Geriatric Cardiology, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland
| |
Collapse
|
4
|
Kim DY, Ko SH. Common Regulators of Lipid Metabolism and Bone Marrow Adiposity in Postmenopausal Women. Pharmaceuticals (Basel) 2023; 16:322. [PMID: 37259464 PMCID: PMC9967016 DOI: 10.3390/ph16020322] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 09/13/2024] Open
Abstract
A variety of metabolic disorders are associated with a decrease in estradiol (E2) during natural or surgical menopause. Postmenopausal women are prone to excessive fat accumulation in skeletal muscle and adipose tissue due to the loss of E2 via abnormalities in lipid metabolism and serum lipid levels. In skeletal muscle and adipose tissue, genes related to energy metabolism and fatty acid oxidation, such as those encoding peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) and estrogen-related receptor alpha (ERRα), are downregulated, leading to increased fat synthesis and lipid metabolite accumulation. The same genes regulate lipid metabolism abnormalities in the bone marrow. In this review, abnormalities in lipid metabolism caused by E2 deficiency were investigated, with a focus on genes able to simultaneously regulate not only skeletal muscle and adipose tissue but also bone metabolism (e.g., genes encoding PGC-1α and ERRα). In addition, the mechanisms through which mesenchymal stem cells lead to adipocyte differentiation in the bone marrow as well as metabolic processes related to bone marrow adiposity, bone loss, and osteoporosis were evaluated, focusing on the loss of E2 and lipid metabolic alterations. The work reviewed here suggests that genes underlying lipid metabolism and bone marrow adiposity are candidate therapeutic targets for bone loss and osteoporosis in postmenopausal women.
Collapse
Affiliation(s)
- Dae-Yong Kim
- CEO, N- BIOTEK, Inc., 402-803, Technopark, 655, Pyeongcheon-ro, Bucheon-si 14502, Gyeonggi-do, Republic of Korea
| | - Seong-Hee Ko
- Regenerative Medicine Research Team, N- BIOTEK, Inc., 104-706, Technopark Ssangyong 3Cha, 397, Seokcheon-ro, Bucheon-si 14449, Gyeonggi-do, Republic of Korea
| |
Collapse
|
5
|
Abikar A, Saimon C, Ranganathan P. To block or not to block-hormonal signaling in the treatment of cancers. Front Endocrinol (Lausanne) 2023; 14:1129332. [PMID: 36891053 PMCID: PMC9986485 DOI: 10.3389/fendo.2023.1129332] [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: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
The breast and prostate glands are the two major organs that are highly dependent on the gonadal steroid hormones for their development and homeostasis. The cancers of these organs also show a large dependence on steroid hormones and have formed the basis of endocrine therapy. Estrogen deprivation by oophorectomy has been in active practice since the 1970s, and androgen deprivation therapy for prostate cancer was a major breakthrough in medicine in 1941. Since then, several improvisations have happened in these modes of therapy. However, the development of resistance to this deprivation and the emergence of hormone independence are major problems in both cancers. The lessons learned from rodent models have made it clear that the male hormone has a role in females and vice versa. Also, the metabolic products of these hormones may have unintentional effects including proliferative conditions in both sexes. Hence, administering estrogen as a method of chemical castration in males and administering DHT in females may not be the ideal scenario. It would be important to consider the status of the opposite sex hormone signaling and its effects and come up with a combinatorial regime to strike a balance between androgen and estrogen signaling. This review summarizes the current understanding and developments in this field in the context of prostate cancer.
Collapse
|
6
|
White AA, Lin A, Bickendorf X, Cavve BS, Moore JK, Siafarikas A, Strickland DH, Leffler J. Potential immunological effects of gender-affirming hormone therapy in transgender people - an unexplored area of research. Ther Adv Endocrinol Metab 2022; 13:20420188221139612. [PMID: 36533187 PMCID: PMC9747891 DOI: 10.1177/20420188221139612] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/31/2022] [Indexed: 12/14/2022] Open
Abstract
There are well-described sex-based differences in how the immune system operates. In particular, cisgender (cis) females have a more easily activated immune system; associated with an increased prevalence of autoimmune diseases and adverse events following vaccinations. Conversely, cis males have a higher threshold for immune activation, and are more prone to certain infectious diseases, such as coronavirus disease (COVID-19). Oestrogen and testosterone have immune-modulatory properties, and it is likely that these contribute to the sexual dimorphism of the immune system. There are also important immune-related genes located on the X chromosome, such as toll-like receptor (TLR) 7/8; and the mosaic bi-allelic expression of such genes may contribute to the state of immune hyperactivation in cis females. The scientific literature strongly suggests that sex-based differences in the functioning of the immune system are related to both X-linked genes and immune modulation by sex hormones. However, it is currently not clear how this impacts transgender (trans) people receiving gender-affirming hormonal therapy. Moreover, it is estimated that in Australia, at least 2.3% of adolescents identify as trans and/or gender diverse, and referrals to specialist gender-affirming care are increasing each year. Despite the improving social awareness of trans people, they remain chronically underrepresented in the scientific literature. In addition, a small number of case studies describe new onset autoimmune disorders in adult trans females following oestrogen use. However, there is currently minimal long-term research with an immunological focus on trans people. Therefore, to ensure the positive health outcomes of trans people, it is crucial that the role of sex hormones in immune modulation is investigated further.
Collapse
Affiliation(s)
- Alice A. White
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Ashleigh Lin
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Xander Bickendorf
- Telethon Kids Institute, University of Western Australia, WA, Australia
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
| | - Blake S. Cavve
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
| | - Julia K. Moore
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Nedlands, WA, Australia
| | - Aris Siafarikas
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- Gender Diversity Service, Child and Adolescent Health Service, Nedlands, WA, Australia
- Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia
| | | | - Jonatan Leffler
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, 15 Hospital Ave., Nedlands, WA 6009, Australia
| |
Collapse
|
7
|
Estrogen as a key regulator of energy homeostasis and metabolic health. Biomed Pharmacother 2022; 156:113808. [DOI: 10.1016/j.biopha.2022.113808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
|
8
|
Fisher JL, Jones EF, Flanary VL, Williams AS, Ramsey EJ, Lasseigne BN. Considerations and challenges for sex-aware drug repurposing. Biol Sex Differ 2022; 13:13. [PMID: 35337371 PMCID: PMC8949654 DOI: 10.1186/s13293-022-00420-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/06/2022] [Indexed: 01/09/2023] Open
Abstract
Sex differences are essential factors in disease etiology and manifestation in many diseases such as cardiovascular disease, cancer, and neurodegeneration [33]. The biological influence of sex differences (including genomic, epigenetic, hormonal, immunological, and metabolic differences between males and females) and the lack of biomedical studies considering sex differences in their study design has led to several policies. For example, the National Institute of Health's (NIH) sex as a biological variable (SABV) and Sex and Gender Equity in Research (SAGER) policies to motivate researchers to consider sex differences [204]. However, drug repurposing, a promising alternative to traditional drug discovery by identifying novel uses for FDA-approved drugs, lacks sex-aware methods that can improve the identification of drugs that have sex-specific responses [7, 11, 14, 33]. Sex-aware drug repurposing methods either select drug candidates that are more efficacious in one sex or deprioritize drug candidates based on if they are predicted to cause a sex-bias adverse event (SBAE), unintended therapeutic effects that are more likely to occur in one sex. Computational drug repurposing methods are encouraging approaches to develop for sex-aware drug repurposing because they can prioritize sex-specific drug candidates or SBAEs at lower cost and time than traditional drug discovery. Sex-aware methods currently exist for clinical, genomic, and transcriptomic information [1, 7, 155]. They have not expanded to other data types, such as DNA variation, which has been beneficial in other drug repurposing methods that do not consider sex [114]. Additionally, some sex-aware methods suffer from poorer performance because a disproportionate number of male and female samples are available to train computational methods [7]. However, there is development potential for several different categories (i.e., data mining, ligand binding predictions, molecular associations, and networks). Low-dimensional representations of molecular association and network approaches are also especially promising candidates for future sex-aware drug repurposing methodologies because they reduce the multiple hypothesis testing burden and capture sex-specific variation better than the other methods [151, 159]. Here we review how sex influences drug response, the current state of drug repurposing including with respect to sex-bias drug response, and how model organism study design choices influence drug repurposing validation.
Collapse
Affiliation(s)
- Jennifer L. Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Emma F. Jones
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Victoria L. Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Avery S. Williams
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Elizabeth J. Ramsey
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| |
Collapse
|
9
|
Bedir Ö, Gram A, Dorsam ST, Grazul-Bilska AT, Kowalewski MP. Plane of nutrition and FSH-induced superovulation affect the expression of steroid hormone receptors and growth factors in caruncular tissue of non-pregnant sheep. Domest Anim Endocrinol 2022; 78:106683. [PMID: 34688215 DOI: 10.1016/j.domaniend.2021.106683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 11/03/2022]
Abstract
Implantation is a critical step in the establishment of pregnancy and an important part of embryo-maternal contact. Uterine receptivity can be affected by changes in body condition and the maternal endocrine milieu, including those caused by the use of exogenous gonadotropins in controlled ovarian hyperstimulation to induce the development of multiple follicles. This study demonstrates the effects of FSH-mediated ovarian hyperstimulation on the caruncles of ewes under various feeding regimes. Sheep were classified into 3 categories: control fed (CF), overfed (OF), or underfed (UF). In each group, animals were superovulated with FSH or injected with a saline solution (non-treated control). Uterine caruncles were collected at the early (d 5) and mid-luteal phase (d 10) of the estrous cycle. The transcript levels of steroid hormone receptors (ESR1, ESR2, PGR) and growth factors (IGF1, IGF2, VEGFA) were investigated and their expression localized by immunohistochemical staining. As for the main findings, day of the estrous cycle affected expression of ESR1, IGF1 and IGF2, but not of ESR2, PGR and VEGFA; both feeding and superovulation had modulatory effects, with feeding (UF/OF) stimulating expression of all genes studied, and superovulation altering expression of some genes, eg IGF1, PGR and ESR1 and ESR2, in CF animals. Similarly, feeding (UF/OF) altered responsiveness to superovulation for PGR on d 5 and ESR1/ESR2 on d 5 and/or 10. Our data emphasize possible effects of dietary and/or hormonal stimuli on uterine physiology, which may affect pregnancy outcomes by disrupting uterine functionality.
Collapse
Affiliation(s)
- Özlem Bedir
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| | - Aykut Gram
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland; Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, 38280 Kayseri, Turkey.
| | - Sheri T Dorsam
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA.
| | - Anna T Grazul-Bilska
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA.
| | - Mariusz P Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
10
|
Duan L, Xu X, Xu L, Wen C, Ouyang K, Li Z, Liang Y. ERα-Targeting PROTAC as a Chemical Knockdown Tool to Investigate the Estrogen Receptor Function in Rat Menopausal Arthritis. Front Pharmacol 2021; 12:764154. [PMID: 34916941 PMCID: PMC8669996 DOI: 10.3389/fphar.2021.764154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/09/2021] [Indexed: 12/02/2022] Open
Abstract
Proteolytic targeting chimeras (PROTACs) is a rapid and reversible chemical knockout method. Compared with traditional gene-editing tools, it can avoid potential genetic compensation, misunderstandings caused by spontaneous mutations, or gene knockouts that lead to embryonic death. To study the role of estrogen receptor alpha (ERα) in the occurrence and progression of menopausal arthritis, we report a chemical knockout strategy in which stable peptide-based (PROTACs) against ERα to inhibit their function. This chemical knockdown strategy can effectively and quickly inhibit ERα protein in vivo and in vitro. In the rat menopausal arthritis model, this study showed that inhibiting estrogen function by degrading ERα can significantly interfere with cartilage matrix metabolism and cause menopausal arthritis by up-regulating matrix metalloproteinase (MMP-13). The results of this study indicate that ERα is a crucial estrogen receptor for maintaining cartilage metabolism. Inhibition of ERα function by PROTACs can promote the progression of osteoarthritis.
Collapse
Affiliation(s)
- Li Duan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiao Xu
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Limei Xu
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Caining Wen
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Kan Ouyang
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zigang Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
| |
Collapse
|
11
|
Sexually Dimorphic Neurosteroid Synthesis Regulates Neuronal Activity in the Murine Brain. J Neurosci 2021; 41:9177-9191. [PMID: 34561233 DOI: 10.1523/jneurosci.0885-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/12/2021] [Accepted: 09/10/2021] [Indexed: 11/21/2022] Open
Abstract
Sex steroid hormones act on hypothalamic kisspeptin neurons to regulate reproductive neural circuits in the brain. Kisspeptin neurons start to express estrogen receptors in utero, suggesting steroid hormone action on these cells early during development. Whether neurosteroids are locally produced in the embryonic brain and impinge onto kisspeptin/reproductive neural circuitry is not known. To address this question, we analyzed aromatase expression, a key enzyme in estrogen synthesis, in male and female mouse embryos. We identified an aromatase neuronal network comprising ∼6000 neurons in the hypothalamus and amygdala. By birth, this network has become sexually dimorphic in a cluster of aromatase neurons in the arcuate nucleus adjacent to kisspeptin neurons. We demonstrate that male arcuate aromatase neurons convert testosterone to estrogen to regulate kisspeptin neuron activity. We provide spatiotemporal information on aromatase neuronal network development and highlight a novel mechanism whereby aromatase neurons regulate the activity of distinct neuronal populations expressing estrogen receptors.SIGNIFICANCE STATEMENT Sex steroid hormones, such as estradiol, are important regulators of neural circuits controlling reproductive physiology in the brain. Embryonic kisspeptin neurons in the hypothalamus express steroid hormone receptors, suggesting hormone action on these cells in utero Whether neurosteroids are locally produced in the brain and impinge onto reproductive neural circuitry is insufficiently understood. To address this question, we analyzed aromatase expression, a key enzyme in estradiol synthesis, in mouse embryos and identified a network comprising ∼6000 neurons in the brain. By birth, this network has become sexually dimorphic in a cluster of aromatase neurons in the arcuate nucleus adjacent to kisspeptin neurons. We demonstrate that male aromatase neurons convert testosterone to estradiol to regulate kisspeptin neuron activity.
Collapse
|
12
|
Zhang FL, Kong L, Zhao AH, Ge W, Yan ZH, Li L, De Felici M, Shen W. Inflammatory cytokines as key players of apoptosis induced by environmental estrogens in the ovary. ENVIRONMENTAL RESEARCH 2021; 198:111225. [PMID: 33971129 DOI: 10.1016/j.envres.2021.111225] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/02/2021] [Accepted: 04/22/2021] [Indexed: 06/12/2023]
Abstract
Natural and synthetic environmental estrogens (EEs), interfering with the physiological functions of the body's estrogens, are widespread and are rising much concern for their possible deleterious effects on human and animal health, in particular on reproduction. In fact, increasing evidence indicate that EEs can be responsible for a variety of disfunctions of the reproductive system especially in females such as premature ovarian insufficiency (POI). Because of their great structural diversity, the modes of action of EEs are controversial. One important way through which EEs exert their effects on reproduction is the induction of apoptosis in the ovary. In general, EEs can exert pro-and anti-apoptotic effects by agonizing or antagonizing numerous estrogen-dependent signaling pathways. In the present work, results concerning apoptotic pathways and diseases induced by representative EEs (such as zearalenone, bisphenol A and di-2-ethylhexyl phthalate), in ovaries throughout development are presented into an integrated network. By reviewing and elaborating these studies, we propose inflammatory factors, centered on the production of tumor necrosis factor (TNF), as a major cause of the induction of apoptosis by EEs in the mammalian ovary. As a consequence, potential strategies to prevent such EE effect are suggested.
Collapse
Affiliation(s)
- Fa-Li Zhang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Li Kong
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ai-Hong Zhao
- Qingdao Academy of Agricultural Sciences, Qingdao, 266100, China
| | - Wei Ge
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zi-Hui Yan
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Lan Li
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy.
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
13
|
Lee MY, Kim EJ, Shin A, Kim YS. [How to Study the Sex and Gender Effect in Biomedical Research?]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2021; 77:104-114. [PMID: 33758108 DOI: 10.4166/kjg.2021.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/19/2021] [Accepted: 02/19/2021] [Indexed: 12/16/2022]
Abstract
Men and women are different, but this difference has not been well reflected in clinical trials and preclinical studies of biomedical science. Gender medicine, which systematically analyzes research results according to sex and gender, has been emphasized to overcome this problem. On the other hand, researchers still have difficulty in applying gender medicine to their research. To perform rigorous gender medicine, using correct terms, a thorough literature review during research planning, appropriate statistical analysis and reporting, and cautious interpretation of the results are necessary. Applying gender medicine will increase the reproducibility of studies, promote discoveries, expand the study relevance, and ultimately improve patient care in both men and women. Here, this study reviewed the practical issues on applying gender medicine to both preclinical and clinical studies in the field of biomedical science.
Collapse
Affiliation(s)
- Moon Young Lee
- Department of Physiology, Wonkwang Digestive Disease Research Institute, Wonkwang University School of Medicine, Iksan, Korea
| | - Eui Joong Kim
- Department of Physiology, Wonkwang Digestive Disease Research Institute, Wonkwang University School of Medicine, Iksan, Korea
| | - Aesun Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea.,Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea
| | - Yong Sung Kim
- Department of Physiology, Wonkwang Digestive Disease Research Institute, Wonkwang University School of Medicine, Iksan, Korea.,Sanbon Good Breath Clinic, Gunpo, Korea.,Diversity Committee of the Korean Society of Gastroenterology, Seoul, Korea
| |
Collapse
|
14
|
Shin EM, Huynh VT, Neja SA, Liu CY, Raju A, Tan K, Tan NS, Gunaratne J, Bi X, Iyer LM, Aravind L, Tergaonkar V. GREB1: An evolutionarily conserved protein with a glycosyltransferase domain links ERα glycosylation and stability to cancer. SCIENCE ADVANCES 2021; 7:7/12/eabe2470. [PMID: 33731348 PMCID: PMC7968844 DOI: 10.1126/sciadv.abe2470] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 01/29/2021] [Indexed: 05/03/2023]
Abstract
What covalent modifications control the temporal ubiquitination of ERα and hence the duration of its transcriptional activity remain poorly understood. We show that GREB1, an ERα-inducible enzyme, catalyzes O-GlcNAcylation of ERα at residues T553/S554, which stabilizes ERα protein by inhibiting association with the ubiquitin ligase ZNF598. Loss of GREB1-mediated glycosylation of ERα results in reduced cellular ERα levels and insensitivity to estrogen. Higher GREB1 expression in ERα+ve breast cancer is associated with greater survival in response to tamoxifen, an ERα agonist. Mice lacking Greb1 exhibit growth and fertility defects reminiscent of phenotypes in ERα-null mice. In summary, this study identifies GREB1, a protein with an evolutionarily conserved domain related to DNA-modifying glycosyltransferases of bacteriophages and kinetoplastids, as the first inducible and the only other (apart from OGT) O-GlcNAc glycosyltransferase in mammalian cytoplasm and ERα as its first substrate.
Collapse
Affiliation(s)
- Eun Myoung Shin
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Vinh Thang Huynh
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Sultan Abda Neja
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Chia Yi Liu
- Bioprocessing Technology Institute (BTI), A*STAR, Singapore, Singapore
| | - Anandhkumar Raju
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Kelly Tan
- Bioprocessing Technology Institute (BTI), A*STAR, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive,, Singapore 637551, Singapore
| | - Jayantha Gunaratne
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117594, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute (BTI), A*STAR, Singapore, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Lakshminarayan M Iyer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Vinay Tergaonkar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore.
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| |
Collapse
|
15
|
Khatpe AS, Adebayo AK, Herodotou CA, Kumar B, Nakshatri H. Nexus between PI3K/AKT and Estrogen Receptor Signaling in Breast Cancer. Cancers (Basel) 2021; 13:369. [PMID: 33498407 PMCID: PMC7864210 DOI: 10.3390/cancers13030369] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
Signaling from estrogen receptor alpha (ERα) and its ligand estradiol (E2) is critical for growth of ≈70% of breast cancers. Therefore, several drugs that inhibit ERα functions have been in clinical use for decades and new classes of anti-estrogens are continuously being developed. Although a significant number of ERα+ breast cancers respond to anti-estrogen therapy, ≈30% of these breast cancers recur, sometimes even after 20 years of initial diagnosis. Mechanism of resistance to anti-estrogens is one of the intensely studied disciplines in breast cancer. Several mechanisms have been proposed including mutations in ESR1, crosstalk between growth factor and ERα signaling, and interplay between cell cycle machinery and ERα signaling. ESR1 mutations as well as crosstalk with other signaling networks lead to ligand independent activation of ERα thus rendering anti-estrogens ineffective, particularly when treatment involved anti-estrogens that do not degrade ERα. As a result of these studies, several therapies that combine anti-estrogens that degrade ERα with PI3K/AKT/mTOR inhibitors targeting growth factor signaling or CDK4/6 inhibitors targeting cell cycle machinery are used clinically to treat recurrent ERα+ breast cancers. In this review, we discuss the nexus between ERα-PI3K/AKT/mTOR pathways and how understanding of this nexus has helped to develop combination therapies.
Collapse
Affiliation(s)
- Aditi S. Khatpe
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.K.); (A.K.A.); (C.A.H.); (B.K.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Adedeji K. Adebayo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.K.); (A.K.A.); (C.A.H.); (B.K.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Christopher A. Herodotou
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.K.); (A.K.A.); (C.A.H.); (B.K.)
| | - Brijesh Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.K.); (A.K.A.); (C.A.H.); (B.K.)
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.K.); (A.K.A.); (C.A.H.); (B.K.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- VA Roudebush Medical Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Lacouture A, Jobin C, Weidmann C, Berthiaume L, Bastien D, Laverdière I, Pelletier M, Audet-Walsh É. A FACS-Free Purification Method to Study Estrogen Signaling, Organoid Formation, and Metabolic Reprogramming in Mammary Epithelial Cells. Front Endocrinol (Lausanne) 2021; 12:672466. [PMID: 34456857 PMCID: PMC8397380 DOI: 10.3389/fendo.2021.672466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Few in vitro models are used to study mammary epithelial cells (MECs), and most of these do not express the estrogen receptor α (ERα). Primary MECs can be used to overcome this issue, but methods to purify these cells generally require flow cytometry and fluorescence-activated cell sorting (FACS), which require specialized instruments and expertise. Herein, we present in detail a FACS-free protocol for purification and primary culture of mouse MECs. These MECs remain differentiated for up to six days with >85% luminal epithelial cells in two-dimensional culture. When seeded in Matrigel, they form organoids that recapitulate the mammary gland's morphology in vivo by developing lumens, contractile cells, and lobular structures. MECs express a functional ERα signaling pathway in both two- and three-dimensional cell culture, as shown at the mRNA and protein levels and by the phenotypic characterization. Extracellular metabolic flux analysis showed that estrogens induce a metabolic switch favoring aerobic glycolysis over mitochondrial respiration in MECs grown in two-dimensions, a phenomenon known as the Warburg effect. We also performed mass spectrometry (MS)-based metabolomics in organoids. Estrogens altered the levels of metabolites from various pathways, including aerobic glycolysis, citric acid cycle, urea cycle, and amino acid metabolism, demonstrating that ERα reprograms cell metabolism in mammary organoids. Overall, we have optimized mouse MEC isolation and purification for two- and three-dimensional cultures. This model represents a valuable tool to study how estrogens modulate mammary gland biology, and particularly how these hormones reprogram metabolism during lactation and breast carcinogenesis.
Collapse
Affiliation(s)
- Aurélie Lacouture
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec City, QC, Canada
| | - Cynthia Jobin
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec City, QC, Canada
| | - Cindy Weidmann
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec City, QC, Canada
| | - Line Berthiaume
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec City, QC, Canada
| | - Dominic Bastien
- Centre de recherche sur le cancer de l’Université Laval, Québec City, QC, Canada
- Faculty of Pharmacy, University Laval, Quebec City, QC, Canada
| | - Isabelle Laverdière
- Centre de recherche sur le cancer de l’Université Laval, Québec City, QC, Canada
- Faculty of Pharmacy, University Laval, Quebec City, QC, Canada
- Oncology Axis, Centre de recherche du CHU de Québec - Université Laval, Quebec City, QC, Canada
- Department of Pharmacy, CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Martin Pelletier
- Infectious and Immune Disease Axis, CHU de Québec-Université Laval Research Center, Québec, QC, Canada
- ARThrite Research Center, Laval University, Québec, QC, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec City, QC, Canada
- *Correspondence: Étienne Audet-Walsh,
| |
Collapse
|
17
|
Gustafsson KL, Farman HH, Nilsson KH, Henning P, Movérare-Skrtic S, Lionikaite V, Lawenius L, Engdahl C, Ohlsson C, Lagerquist MK. Arginine site 264 in murine estrogen receptor-α is dispensable for the regulation of the skeleton. Am J Physiol Endocrinol Metab 2021; 320:E160-E168. [PMID: 33225718 DOI: 10.1152/ajpendo.00349.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mutation of arginine 264 in ERα has been shown to abrogate rapid membrane ERα-mediated endothelial effects. Our novel finding that mutation of R264 is dispensable for ERα-mediated skeletal effects supports the concept that R264 determines tissue specificity of ERα. Estrogen protects against bone loss but is not a suitable treatment due to adverse effects in other tissues. Therefore, increased knowledge regarding estrogen signaling in estrogen-responsive tissues is warranted to aid the development of bone-specific estrogen treatments. Estrogen receptor-α (ERα), the main mediator of estrogenic effects in bone, is widely subjected to posttranslational modifications (PTMs). In vitro studies have shown that methylation at site R260 in the human ERα affects receptor localization and intracellular signaling. The corresponding amino acid R264 in murine ERα has been shown to have a functional role in endothelium in vivo, although the methylation of R264 in the murine gene is yet to be empirically demonstrated. The aim of this study was to investigate whether R264 in ERα is involved in the regulation of the skeleton in vivo. Dual-energy X-ray absorptiometry (DEXA) analysis at 3, 6, 9, and 12 mo of age showed no differences in total body areal bone mineral density (BMD) between R264A and wild type (WT) in either female or male mice. Furthermore, analyses using computed tomography (CT) demonstrated that trabecular bone mass in tibia and vertebra and cortical thickness in tibia were similar between R264A and WT mice. In addition, R264A females displayed a normal estrogen treatment response in trabecular bone mass as well as in cortical thickness. Furthermore, uterus, thymus, and adipose tissue responded similarly in R264A and WT female mice after estrogen treatment. In conclusion, our novel finding that mutation of R264 in ERα does not affect the regulation of the skeleton, together with the known role of R264 for ERα-mediated endothelial effects, supports the concept that R264 determines tissue specificity of ERα.NEW & NOTEWORTHY Mutation of arginine 264 in ERα has been shown to abrogate rapid membrane ERα-mediated endothelial effects. Our novel finding that mutation of R264 is dispensable for ERα-mediated skeletal effects supports the concept that R264 determines tissue specificity of ERα.
Collapse
Affiliation(s)
- Karin L Gustafsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Helen H Farman
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin H Nilsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vikte Lionikaite
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lina Lawenius
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Engdahl
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Pharmacology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marie K Lagerquist
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
18
|
Brandi ML, Giustina A. Sexual Dimorphism of Coronavirus 19 Morbidity and Lethality. Trends Endocrinol Metab 2020; 31:918-927. [PMID: 33082024 PMCID: PMC7513816 DOI: 10.1016/j.tem.2020.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/30/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
The recent coronavirus disease 2019 (COVID-19) pandemic showed a different severity in the disease between males and females. Men have been becoming severely ill at a higher rate than women. These data along with an age-dependent disease susceptibility and mortality in the elderly suggest that sex hormones are the main factors in determining the clinical course of the infection. The differences in aging males versus females and the role of sex hormones in key phenotypes of COVID-19 infection are described in this review. Recommendations based on a dimorphic approach for males and females suggest a sex-specific management the disease.
Collapse
Affiliation(s)
- Maria Luisa Brandi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy.
| | - Andrea Giustina
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and Division of Endocrinology IRCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
19
|
Rouhimoghadam M, Lu AS, Salem AK, Filardo EJ. Therapeutic Perspectives on the Modulation of G-Protein Coupled Estrogen Receptor, GPER, Function. Front Endocrinol (Lausanne) 2020; 11:591217. [PMID: 33329395 PMCID: PMC7719807 DOI: 10.3389/fendo.2020.591217] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Estrogens exert their physiological and pathophysiological effects via cellular receptors, named ERα, ERβ, and G-protein coupled estrogen receptor (GPER). Estrogen-regulated physiology is tightly controlled by factors that regulate estrogen bioavailability and receptor sensitivity, while disruption of these control mechanisms can result in loss of reproductive function, cancer, cardiovascular and neurodegenerative disease, obesity, insulin resistance, endometriosis, and systemic lupus erythematosus. Restoration of estrogen physiology by modulating estrogen bioavailability or receptor activity is an effective approach for treating these pathological conditions. Therapeutic interventions that block estrogen action are employed effectively for the treatment of breast and prostate cancer as well as for precocious puberty and anovulatory infertility. Theoretically, treatments that block estrogen biosynthesis should prevent estrogen action at ERs and GPER, although drug resistance and ligand-independent receptor activation may still occur. In addition, blockade of estrogen biosynthesis does not prevent activation of estrogen receptors by naturally occurring or man-made exogenous estrogens. A more complicated scenario is provided by anti-estrogen drugs that antagonize ERs since these drugs function as GPER agonists. Based upon its association with metabolic dysregulation and advanced cancer, GPER represents a therapeutic target with promise for the treatment of several critical health concerns facing Western society. Selective ligands that specifically target GPER have been developed and may soon serve as pharmacological agents for treating human disease. Here, we review current forms of estrogen therapy and the implications that GPER holds for these therapies. We also discuss existing GPER targeted drugs, additional approaches towards developing GPER-targeted therapies and how these therapies may complement existing modalities of estrogen-targeted therapy.
Collapse
Affiliation(s)
- Milad Rouhimoghadam
- Department of Surgery, University of Iowa, Carver College of Medicine, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Anh S. Lu
- College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Aliasger K. Salem
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
- College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Edward J. Filardo
- Department of Surgery, University of Iowa, Carver College of Medicine, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
20
|
Ohlsson C, Gustafsson KL, Farman HH, Henning P, Lionikaite V, Movérare-Skrtic S, Sjögren K, Törnqvist AE, Andersson A, Islander U, Bernardi AI, Poutanen M, Chambon P, Lagerquist MK. Phosphorylation site S122 in estrogen receptor α has a tissue-dependent role in female mice. FASEB J 2020; 34:15991-16002. [PMID: 33067917 DOI: 10.1096/fj.201901376rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Estrogen treatment increases bone mass and reduces fat mass but is associated with adverse effects in postmenopausal women. Knowledge regarding tissue-specific estrogen signaling is important to aid the development of new tissue-specific treatments. We hypothesized that the posttranslational modification phosphorylation in estrogen receptor alpha (ERα) may modulate ERα activity in a tissue-dependent manner. Phosphorylation of site S122 in ERα has been shown in vitro to affect ERα activity, but the tissue-specific role in vivo is unknown. We herein developed and phenotyped a novel mouse model with a point mutation at the phosphorylation site 122 in ERα (S122A). Female S122A mice had increased fat mass and serum insulin levels but unchanged serum sex steroid levels, uterus weight, bone mass, thymus weight, and lymphocyte maturation compared to WT mice. In conclusion, phosphorylation site S122 in ERα has a tissue-dependent role with an impact specifically on fat mass in female mice. This study is the first to demonstrate in vivo that a phosphorylation site in a transactivation domain in a nuclear steroid receptor modulates the receptor activity in a tissue-dependent manner.
Collapse
Affiliation(s)
- Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin L Gustafsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Helen H Farman
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vikte Lionikaite
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Klara Sjögren
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna E Törnqvist
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Annica Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulrika Islander
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Angelina I Bernardi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Matti Poutanen
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Centre National de la Recherche Scientifique, National de la Sante et de la Recherche Medicale, ULP, Collège de France, Illkirch-Strasbourg, France
| | - Marie K Lagerquist
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
21
|
Pajkos M, Zeke A, Dosztányi Z. Ancient Evolutionary Origin of Intrinsically Disordered Cancer Risk Regions. Biomolecules 2020; 10:biom10081115. [PMID: 32731489 PMCID: PMC7465906 DOI: 10.3390/biom10081115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is a heterogeneous genetic disease that alters the proper functioning of proteins involved in key regulatory processes such as cell cycle, DNA repair, survival, or apoptosis. Mutations often accumulate in hot-spots regions, highlighting critical functional modules within these proteins that need to be altered, amplified, or abolished for tumor formation. Recent evidence suggests that these mutational hotspots can correspond not only to globular domains, but also to intrinsically disordered regions (IDRs), which play a significant role in a subset of cancer types. IDRs have distinct functional properties that originate from their inherent flexibility. Generally, they correspond to more recent evolutionary inventions and show larger sequence variations across species. In this work, we analyzed the evolutionary origin of disordered regions that are specifically targeted in cancer. Surprisingly, the majority of these disordered cancer risk regions showed remarkable conservation with ancient evolutionary origin, stemming from the earliest multicellular animals or even beyond. Nevertheless, we encountered several examples where the mutated region emerged at a later stage compared with the origin of the gene family. We also showed the cancer risk regions become quickly fixated after their emergence, but evolution continues to tinker with their genes with novel regulatory elements introduced even at the level of humans. Our concise analysis provides a much clearer picture of the emergence of key regulatory elements in proteins and highlights the importance of taking into account the modular organisation of proteins for the analyses of evolutionary origin.
Collapse
Affiliation(s)
- Mátyás Pajkos
- Department of Biochemistry, ELTE Eötvös Loránd University, Pázmány Péter stny 1/c, H-1117 Budapest, Hungary;
| | - András Zeke
- Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary;
| | - Zsuzsanna Dosztányi
- Department of Biochemistry, ELTE Eötvös Loránd University, Pázmány Péter stny 1/c, H-1117 Budapest, Hungary;
- Correspondence:
| |
Collapse
|
22
|
Vasquez YM, Nandu TS, Kelleher AM, Ramos EI, Gadad SS, Kraus WL. Genome-wide analysis and functional prediction of the estrogen-regulated transcriptional response in the mouse uterus†. Biol Reprod 2020; 102:327-338. [PMID: 31511857 PMCID: PMC7016285 DOI: 10.1093/biolre/ioz183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/21/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
The ovarian hormones estrogen and progesterone orchestrate the transcriptional programs required to direct functions of the uterus for initiation and maintenance of pregnancy. Estrogen, acting via estrogen receptor alpha, regulates gene expression by activating and repressing distinct genes involved in signaling pathways that regulate cellular and physiological responses including cell division, water influx, and immune cell recruitment. Historically, these transcriptional responses have been postulated to reflect a biphasic physiological response. In this study, we explored the transcriptional responses of the ovariectomized mouse uterus to 17β-estradiol (E2) by RNA-seq to obtain global expression profiles of protein-coding transcripts (mRNAs) and long noncoding RNAs (lncRNAs) following 0.5, 1, 2, and 6 hours of treatment. The E2-regulated mRNA and lncRNA expression profiles in the mouse uterus indicate an association between lncRNAs and mRNAs that regulate E2-driven pathways and reproductive phenotypes in the mouse. The transient E2-regulated transcriptome is reflected in the time-dependent shifting of biological processes regulated in the uterus in response to E2. Moreover, high expression of some conserved lncRNAs that are E2 regulated in the mouse uterus are predictive of low overall survival in endometrial carcinoma patients (e.g., H19, KCNQ1OT1, MIR17HG, and FTX). Collectively, this study (1) describes a genomic approach for identifying E2-regulated lncRNAs that may serve critical function in the uterus and (2) provides new insights into our understanding of the regulation of hormone-regulated transcriptional responses with implications in pregnancy and endometrial pathologies.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Databases, Genetic
- Estradiol/pharmacology
- Female
- Gene Expression Regulation/drug effects
- Gene Ontology
- Genital Neoplasms, Female/genetics
- Genital Neoplasms, Female/metabolism
- Genital Neoplasms, Female/mortality
- Humans
- Mice
- Ovariectomy
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Survival Rate
- Transcriptome/drug effects
- Uterus/drug effects
- Uterus/metabolism
Collapse
Affiliation(s)
- Yasmin M Vasquez
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tulip S Nandu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew M Kelleher
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Enrique I Ramos
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Shrikanth S Gadad
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
23
|
Jones BG, Sealy RE, Penkert RR, Surman SL, Maul RW, Neale G, Xu B, Gearhart PJ, Hurwitz JL. Complex sex-biased antibody responses: estrogen receptors bind estrogen response elements centered within immunoglobulin heavy chain gene enhancers. Int Immunol 2020; 31:141-156. [PMID: 30407507 DOI: 10.1093/intimm/dxy074] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 11/02/2018] [Indexed: 01/10/2023] Open
Abstract
Nuclear hormone receptors including the estrogen receptor (ERα) and the retinoic acid receptor regulate a plethora of biological functions including reproduction, circulation and immunity. To understand how estrogen and other nuclear hormones influence antibody production, we characterized total serum antibody isotypes in female and male mice of C57BL/6J, BALB/cJ and C3H/HeJ mouse strains. Antibody levels were higher in females compared to males in all strains and there was a female preference for IgG2b production. Sex-biased patterns were influenced by vitamin levels, and by antigen specificity toward influenza virus or pneumococcus antigens. To help explain sex biases, we examined the direct effects of estrogen on immunoglobulin heavy chain sterile transcript production among purified, lipopolysaccharide-stimulated B cells. Supplemental estrogen in B-cell cultures significantly increased immunoglobulin heavy chain sterile transcripts. Chromatin immunoprecipitation analyses of activated B cells identified significant ERα binding to estrogen response elements (EREs) centered within enhancer elements of the immunoglobulin heavy chain locus, including the Eµ enhancer and hypersensitive site 1,2 (HS1,2) in the 3' regulatory region. The ERE in HS1,2 was conserved across animal species, and in humans marked a site of polymorphism associated with the estrogen-augmented autoimmune disease, lupus. Taken together, the results highlight: (i) the important targets of ERα in regulatory regions of the immunoglobulin heavy chain locus that influence antibody production, and (ii) the complexity of mechanisms by which estrogen instructs sex-biased antibody production profiles.
Collapse
Affiliation(s)
- Bart G Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Danny Thomas Place, Memphis, USA
| | - Robert E Sealy
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Danny Thomas Place, Memphis, USA
| | - Rhiannon R Penkert
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Danny Thomas Place, Memphis, USA
| | - Sherri L Surman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Danny Thomas Place, Memphis, USA
| | - Robert W Maul
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Geoff Neale
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children's Research Hospital, Memphis, USA
| | - Beisi Xu
- Computational Biology, St. Jude Children's Research Hospital, Memphis, USA
| | - Patricia J Gearhart
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Julia L Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Danny Thomas Place, Memphis, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, USA
| |
Collapse
|
24
|
|
25
|
Guercio G, Saraco N, Costanzo M, Marino R, Ramirez P, Berensztein E, Rivarola MA, Belgorosky A. Estrogens in Human Male Gonadotropin Secretion and Testicular Physiology From Infancy to Late Puberty. Front Endocrinol (Lausanne) 2020; 11:72. [PMID: 32158430 PMCID: PMC7051936 DOI: 10.3389/fendo.2020.00072] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/03/2020] [Indexed: 12/13/2022] Open
Abstract
Several reports in humans as well as transgenic mouse models have shown that estrogens play an important role in male reproduction and fertility. Estrogen receptor alpha (ERα) and beta (ERβ) are expressed in different male tissues including the brain. The estradiol-binding protein GPER1 also mediates estrogen action in target tissues. In human testes a minimal ERα expression during prepuberty along with a marked pubertal up-regulation in germ cells has been reported. ERβ expression was detected mostly in spermatogonia, primary spermatocytes, and immature spermatids. In Sertoli cells ERβ expression increases with age. The aromatase enzyme (cP450arom), which converts androgens to estrogens, is widely expressed in human tissues (including gonads and hypothalamus), even during fetal life, suggesting that estrogens are also involved in human fetal physiology. Moreover, cP450arom is expressed in the early postnatal testicular Leydig cells and spermatogonia. Even though the aromatase complex is required for estrogen synthesis, its biological relevance is also related to the regulation of the balance between androgens and estrogens in different tissues. Knockout mouse models of aromatase (ArKO) and estrogen receptors (ERKOα, ERKOβ, and ERKOαβ) provide an important tool to study the effects of estrogens on the male reproductive physiology including the gonadal axis. High basal serum FSH levels were reported in adult aromatase-deficient men, suggesting that estrogens are involved in the negative regulatory gonadotropin feedback. However, normal serum gonadotropin levels were observed in an aromatase-deficient boy, suggesting a maturational pattern role of estrogen in the regulation of gonadotropin secretion. Nevertheless, the role of estrogens in primate testis development and function is controversial and poorly understood. This review addresses the role of estrogens in gonadotropin secretion and testicular physiology in male humans especially during childhood and puberty.
Collapse
Affiliation(s)
- Gabriela Guercio
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
- Research Institute Garrahan-CONICET, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
| | - Nora Saraco
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
- Research Institute Garrahan-CONICET, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
| | - Mariana Costanzo
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
| | - Roxana Marino
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
| | - Pablo Ramirez
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
| | - Esperanza Berensztein
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
- Facultad de Medicina, Department of Cellular Biology and Histology, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marco A. Rivarola
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
- Research Institute Garrahan-CONICET, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
| | - Alicia Belgorosky
- Endocrinology Department, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
- Research Institute Garrahan-CONICET, Hospital de Pediatría “Prof. Dr. Juan P. Garrahan”, Buenos Aires, Argentina
- *Correspondence: Alicia Belgorosky
| |
Collapse
|
26
|
Kammel LG, Correa SM. Selective sexual differentiation of neurone populations may contribute to sex-specific outputs of the ventromedial nucleus of the hypothalamus. J Neuroendocrinol 2020; 32:e12801. [PMID: 31605642 PMCID: PMC6982598 DOI: 10.1111/jne.12801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/26/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022]
Abstract
Sex differences among neurones in the ventrolateral region of the ventromedial hypothalamic nucleus (VMHvl) allow for the display of a diversity of sex-typical behaviours and physiological responses, ranging from mating behaviour to metabolism. Here, we review recent studies that interrogate the relationship between sex-typical responses and changes in cellular phenotypes. We discuss technologies that increase the resolution of molecular profiling or targeting of cell populations, including single-cell transcriptional profiling and conditional viral genetic approaches to manipulate neurone survival or activity. Overall, emerging studies indicate that sex-typical functions of the VMH may be mediated by phenotypically distinct and sexually differentiated neurone populations within the VMHvl. Future studies in this and other brain regions could exploit cell-type-specific tools to reveal the cell populations and molecular mediators that modulate sex-typical responses. Furthermore, cell-type-specific analyses of the effects of sexually differentiating factors, including sex hormones, can test the hypothesis that distinct cell types within a single brain region vary with respect to sexual differentiation.
Collapse
Affiliation(s)
- Laura G Kammel
- Department of Integrative Biology and Physiology, Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, USA
- Molecular, Cellular, Integrative Physiology Graduate Program, University of California, Los Angeles, CA, USA
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
27
|
Trexler AW, Knudsen GA, Nicklisch SCT, Birnbaum LS, Cannon RE. 2,4,6-Tribromophenol Exposure Decreases P-Glycoprotein Transport at the Blood-Brain Barrier. Toxicol Sci 2019; 171:463-472. [PMID: 31368499 PMCID: PMC6760274 DOI: 10.1093/toxsci/kfz155] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/12/2019] [Accepted: 07/09/2019] [Indexed: 01/24/2023] Open
Abstract
2,4,6-Tribromophenol (TBP, CAS No. 118-79-6) is a brominated chemical used in the production of flame-retardant epoxy resins and as a wood preservative. In marine environments, TBP is incorporated into shellfish and consumed by predatory fish. Food processing and water treatment facilities produce TBP as a byproduct. 2,4,6-Tribromophenol has been detected in human blood and breast milk. Biologically, TBP interferes with estrogen and thyroid hormone signaling, which regulate important transporters of the blood-brain barrier (BBB). The BBB is a selectively permeable barrier characterized by brain microvessels which are composed of endothelial cells mortared by tight-junction proteins. ATP-binding cassette (ABC) efflux transporters on the luminal membrane facilitate the removal of unwanted endobiotics and xenobiotics from the brain. In this study, we examined the in vivo and ex vivo effects of TBP on two important transporters of the BBB: P-glycoprotein (P-gp, ABCB1) and Multidrug Resistance-associated Protein 2 (MRP2, ABCC2), using male and female rats and mice. 2,4,6-Tribromophenol exposure ex vivo resulted in a time- (1-3 h) and dose- (1-100 nM) dependent decrease in P-gp transport activity. MRP2 transport activity was unchanged under identical conditions. Immunofluorescence and western blotting measured decreases in P-gp expression after TBP treatment. ATPase assays indicate that TBP is not a substrate and does not directly interact with P-gp. In vivo dosing with TBP (0.4 µmol/kg) produced decreases in P-gp transport. Co-treatment with selective protein kinase C (PKC) inhibitors prevented the TBP-mediated decreases in P-gp transport activity.
Collapse
Affiliation(s)
- Andrew W Trexler
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| | - Gabriel A Knudsen
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| | - Sascha C T Nicklisch
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, La Jolla 92093, California
- Department of Environmental Toxicology, University of California Davis 95616, Davis, California
| | - Linda S Birnbaum
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| | - Ronald E Cannon
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| |
Collapse
|
28
|
Cripps SM, Mattiske DM, Black JR, Risbridger GP, Govers LC, Phillips TR, Pask AJ. A loss of estrogen signaling in the aromatase deficient mouse penis results in mild hypospadias. Differentiation 2019; 109:42-52. [PMID: 31520742 DOI: 10.1016/j.diff.2019.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 11/18/2022]
Abstract
Hypospadias is the abnormal opening of the urethra on the underside of the penis and occurs in approximately 1/125 live male births worldwide. The incidence rate of hypospadias has dramatically increased over the past few decades. This is now attributed, at least in part, to our exposure to endocrine-disrupting chemicals (EDCs) which alter the hormonal signals required for development of the penis. In humans androgens are the main drivers of fusion of the urethral folds to form the urethra within the shaft of the penis, a process required for termination of the urethra in its normal location at the tip of the penis. However, recent research has suggested that estrogen also plays a role in this process. To better understand how EDCs impact urethral development it is essential that we understand the normal function of hormones during development of the penis. To define the role of estrogen in urethral development we examined development of the penis in the aromatase (Cyp19a1) Knockout (ArKO) mouse strain in which endogenous estrogen production is completely ablated. We found that the ArKO penis had a mild hypospadias phenotype. The developing ArKO postnatal penis displayed an early disruption in preputial development, which likely causes the mild hypospadias observed in adults. Using qPCR, we found altered expression of keratin genes and key urethral patterning genes in response to the disrupted estrogen signaling. The hypospadias phenotype was almost identical to that reported for the estrogen receptor α (ERα) knockout confirming that ERα is the predominant receptor for mediating estrogen action during development of the mouse penis. Our results show that estrogen is required for normal prepucial development and placement of the mature urethral opening at the distal aspect of the penis. We also identified several genes which are potential downstream targets of estrogen during normal urethral closure. With this knowledge, we can now better understand how anti-estrogenic as well as estrogenic EDCs disrupt urethral closure to cause mild hypospadias in both mice and humans.
Collapse
Affiliation(s)
- Samuel M Cripps
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Deidre M Mattiske
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Jay R Black
- School of Earth Sciences, The University of Melbourne, Victoria, Australia
| | - Gail P Risbridger
- Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| | - Luke C Govers
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | | | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
29
|
Balla B, Sárvári M, Kósa JP, Kocsis-Deák B, Tobiás B, Árvai K, Takács I, Podani J, Liposits Z, Lakatos P. Long-term selective estrogen receptor-beta agonist treatment modulates gene expression in bone and bone marrow of ovariectomized rats. J Steroid Biochem Mol Biol 2019; 188:185-194. [PMID: 30685384 DOI: 10.1016/j.jsbmb.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 01/20/2023]
Abstract
Gonadal hormones including 17β-estradiol exert important protective functions in skeletal homeostasis. However, numerous details of ovarian hormone deficiency in the common bone marrow microenvironment have not yet been revealed and little information is available on the tissue-specific acts either, especially those via estrogen receptor beta (ERβ). The aim of the present study was therefore to examine the bone-related gene expression changes after ovariectomy (OVX) and long-term ERβ agonist diarylpropionitrile (DPN) administration. We found that OVX produced strong and widespread changes of gene expression in both femoral bone and bone marrow. In the bone out of 22 genes, 20 genes were up- and 2 were downregulated after OVX. It is noteworthy that DPN restored mRNA expression of 10 OVX-induced changes (Aldh2, Col1a1, Daam1, Fgf12, Igf1, Il6r, Nfkb1, Notch1, Notch2 and Psen1) suggesting a modulatory role of ERβ in bone physiology. In bone marrow, out of 37 categorized genes, transcription of 25 genes were up- and 12 were downregulated indicating that the marrow is highly responsive to gonadal hormones. DPN modestly affected transcription, only expression of two genes (Nfatc1 and Tgfb1) was restored by DPN action. The PI3K/Akt signaling pathway was the most affected gene cluster following the interventions in bone and bone marrow, as demonstrated by canonical variates analysis (CVA). We suggested that our results contribute to a deeper understanding of alterations in gene expression of bone and bone marrow niche elicited by ERβ and selective ERβ analogs.
Collapse
Affiliation(s)
- Bernadett Balla
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| | - Miklós Sárvári
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - János P Kósa
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Barbara Kocsis-Deák
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Bálint Tobiás
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Kristóf Árvai
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - István Takács
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - János Podani
- Biological Institute, Eötvös Loránd University, Budapest, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary; Department of Neuroscience, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Péter Lakatos
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
30
|
Winn NC, Jurrissen TJ, Grunewald ZI, Cunningham RP, Woodford ML, Kanaley JA, Lubahn DB, Manrique-Acevedo C, Rector RS, Vieira-Potter VJ, Padilla J. Estrogen receptor-α signaling maintains immunometabolic function in males and is obligatory for exercise-induced amelioration of nonalcoholic fatty liver. Am J Physiol Endocrinol Metab 2019; 316:E156-E167. [PMID: 30512987 PMCID: PMC6397364 DOI: 10.1152/ajpendo.00259.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The role of estrogen receptor-α (ERα) signaling in immunometabolic function is established in females. However, its necessity in males, while appreciated, requires further study. Accordingly, we first determined whether lower metabolic function in male mice compared with females is related to reduced ERα expression. ERα protein expression in metabolically active tissues was lower in males than in females, and this lower expression was associated with worse glucose tolerance. Second, we determined whether ERα is required for optimal immunometabolic function in male mice consuming a chow diet. Despite lower expression of ERα in males, its genetic ablation (KO) caused an insulin-resistant phenotype characterized by enhanced adiposity, glucose intolerance, hepatic steatosis, and metaflammation in adipose tissue and liver. Last, we determined whether ERα is essential for exercise-induced metabolic adaptations. Twelve-week-old wild-type (WT) and ERα KO mice either remained sedentary (SED) or were given access to running wheels (WR) for 10 wk while fed an obesogenic diet. Body weight and fat mass were lower in WR mice regardless of genotype. Daily exercise obliterated immune cell infiltration and inflammatory gene transcripts in adipose tissue in both genotypes. In the liver, however, wheel running suppressed hepatic steatosis and inflammatory gene transcripts in WT but not in KO mice. In conclusion, the present findings indicate that ERα is required for optimal immunometabolic function in male mice despite their reduced ERα protein expression in metabolically active tissues. Furthermore, for the first time, we show that ERα signaling appears to be obligatory for exercise-induced prevention of hepatic steatosis.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Thomas J Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Zachary I Grunewald
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Rory P Cunningham
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Makenzie L Woodford
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Dennis B Lubahn
- Department of Biochemistry, University of Missouri , Columbia, Missouri
| | - Camila Manrique-Acevedo
- Department of Medicine, Division of Endocrinology, University of Missouri , Columbia, Missouri
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri , Columbia, Missouri
- Research Service, Harry S. Truman Memorial Hospital, University of Missouri , Columbia, Missouri
| | | | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
- Department of Child Health, University of Missouri , Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri
| |
Collapse
|
31
|
Issigonis M, Newmark PA. From worm to germ: Germ cell development and regeneration in planarians. Curr Top Dev Biol 2019; 135:127-153. [DOI: 10.1016/bs.ctdb.2019.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Becerra-Díaz M, Strickland AB, Keselman A, Heller NM. Androgen and Androgen Receptor as Enhancers of M2 Macrophage Polarization in Allergic Lung Inflammation. THE JOURNAL OF IMMUNOLOGY 2018; 201:2923-2933. [PMID: 30305328 DOI: 10.4049/jimmunol.1800352] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 09/10/2018] [Indexed: 02/06/2023]
Abstract
Allergic asthma is a disease initiated by a breach of the lung mucosal barrier and an inappropriate Th2 inflammatory immune response that results in M2 polarization of alveolar macrophages (AM). The number of M2 macrophages in the airway correlates with asthma severity in humans. Sex differences in asthma suggest that sex hormones modify lung inflammation and macrophage polarization. Asthmatic women have more M2 macrophages than asthmatic men and androgens have been used as an experimental asthma treatment. In this study, we demonstrate that although androgen (dihydrotestosterone) reconstitution of castrated mice reduced lung inflammation in a mouse model of allergic lung inflammation, it enhanced M2 polarization of AM. This indicates a cell-specific role for androgens. Dihydrotestosterone also enhanced IL-4-stimulated M2 macrophage polarization in vitro. Using mice lacking androgen receptor (AR) in monocytes/macrophages (ARfloxLysMCre), we found that male but not female mice exhibited less eosinophil recruitment and lung inflammation due to impaired M2 polarization. There was a reduction in eosinophil-recruiting chemokines and IL-5 in AR-deficient AM. These data reveal an unexpected and novel role for androgen/AR in promoting M2 macrophage polarization. Our findings are also important for understanding pathology in diseases promoted by M2 macrophages and androgens, such as asthma, eosinophilic esophagitis, and prostate cancer, and for designing new approaches to treatment.
Collapse
Affiliation(s)
- Mireya Becerra-Díaz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and
| | - Ashley B Strickland
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and
| | - Aleksander Keselman
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and .,Allergy and Clinical Immunology, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
33
|
Konings G, Brentjens L, Delvoux B, Linnanen T, Cornel K, Koskimies P, Bongers M, Kruitwagen R, Xanthoulea S, Romano A. Intracrine Regulation of Estrogen and Other Sex Steroid Levels in Endometrium and Non-gynecological Tissues; Pathology, Physiology, and Drug Discovery. Front Pharmacol 2018; 9:940. [PMID: 30283331 PMCID: PMC6157328 DOI: 10.3389/fphar.2018.00940] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022] Open
Abstract
Our understanding of the intracrine (or local) regulation of estrogen and other steroid synthesis and degradation expanded in the last decades, also thanks to recent technological advances in chromatography mass-spectrometry. Estrogen responsive tissues and organs are not passive receivers of the pool of steroids present in the blood but they can actively modify the intra-tissue steroid concentrations. This allows fine-tuning the exposure of responsive tissues and organs to estrogens and other steroids in order to best respond to the physiological needs of each specific organ. Deviations in such intracrine control can lead to unbalanced steroid hormone exposure and disturbances. Through a systematic bibliographic search on the expression of the intracrine enzymes in various tissues, this review gives an up-to-date view of the intracrine estrogen metabolisms, and to a lesser extent that of progestogens and androgens, in the lower female genital tract, including the physiological control of endometrial functions, receptivity, menopausal status and related pathological conditions. An overview of the intracrine regulation in extra gynecological tissues such as the lungs, gastrointestinal tract, brain, colon and bone is given. Current therapeutic approaches aimed at interfering with these metabolisms and future perspectives are discussed.
Collapse
Affiliation(s)
- Gonda Konings
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Linda Brentjens
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Bert Delvoux
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Karlijn Cornel
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Marlies Bongers
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Roy Kruitwagen
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Sofia Xanthoulea
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Andrea Romano
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
34
|
Della Torre S, Rando G, Meda C, Ciana P, Ottobrini L, Maggi A. Transcriptional activity of oestrogen receptors in the course of embryo development. J Endocrinol 2018; 238:165-176. [PMID: 30012715 PMCID: PMC6084787 DOI: 10.1530/joe-18-0003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 06/11/2018] [Indexed: 12/16/2022]
Abstract
Oestrogens are well-known proliferation and differentiation factors that play an essential role in the correct development of sex-related organs and behaviour in mammals. With the use of the ERE-Luc reporter mouse model, we show herein that throughout mouse development, oestrogen receptors (ERs) are active starting from day 12 post conception. Most interestingly, we show that prenatal luciferase expression in each organ is proportionally different in relation to the germ layer of the origin. The luciferase content is highest in ectoderm-derived organs (such as brain and skin) and is lowest in endoderm-derived organs (such as liver, lung, thymus and intestine). Consistent with the testosterone surge occurring in male mice at the end of pregnancy, in the first 2 days after birth, we observed a significant increase in the luciferase content in several organs, including the liver, bone, gonads and hindbrain. The results of the present study show a widespread transcriptional activity of ERs in developing embryos, pointing to the potential contribution of these receptors in the development of non-reproductive as well as reproductive organs. Consequently, the findings reported here might be relevant in explaining the significant differences in male and female physiopathology reported by a growing number of studies and may underline the necessity for more systematic analyses aimed at the identification of the prenatal effects of drugs interfering with ER signalling, such as aromatase inhibitors or endocrine disrupter chemicals.
Collapse
Affiliation(s)
- Sara Della Torre
- Center of Excellence on Neurodegenerative DiseasesUniversity of Milan, Milan, Italy
- Department of Pharmacological and Biomolecular SciencesUniversity of Milan, Milan, Italy
| | - Gianpaolo Rando
- Center of Excellence on Neurodegenerative DiseasesUniversity of Milan, Milan, Italy
- Department of Pharmacological and Biomolecular SciencesUniversity of Milan, Milan, Italy
| | - Clara Meda
- Center of Excellence on Neurodegenerative DiseasesUniversity of Milan, Milan, Italy
- Department of Pharmacological and Biomolecular SciencesUniversity of Milan, Milan, Italy
| | - Paolo Ciana
- Department of Oncology and Hemato-OncologyUniversity of Milan, Milan, Italy
| | - Luisa Ottobrini
- Department of Pathophysiology and TransplantationUniversity of Milan, Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative DiseasesUniversity of Milan, Milan, Italy
- Department of Pharmacological and Biomolecular SciencesUniversity of Milan, Milan, Italy
| |
Collapse
|
35
|
Gustafsson KL, Nilsson KH, Farman HH, Andersson A, Lionikaite V, Henning P, Wu J, Windahl SH, Islander U, Movérare-Skrtic S, Sjögren K, Carlsten H, Gustafsson JÅ, Ohlsson C, Lagerquist MK. ERα expression in T lymphocytes is dispensable for estrogenic effects in bone. J Endocrinol 2018; 238:129-136. [PMID: 29848607 PMCID: PMC6026922 DOI: 10.1530/joe-18-0183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022]
Abstract
Estrogen treatment has positive effects on the skeleton, and we have shown that estrogen receptor alpha (ERα) expression in cells of hematopoietic origin contributes to a normal estrogen treatment response in bone tissue. T lymphocytes are implicated in the estrogenic regulation of bone mass, but it is not known whether T lymphocytes are direct estrogen target cells. Therefore, the aim of this study was to determine the importance of ERα expression in T lymphocytes for the estrogenic regulation of the skeleton using female mice lacking ERα expression specifically in T lymphocytes (Lck-ERα-/-) and ERαflox/flox littermate (control) mice. Deletion of ERα expression in T lymphocytes did not affect bone mineral density (BMD) in sham-operated Lck-ERα-/- compared to control mice, and ovariectomy (ovx) resulted in a similar decrease in BMD in control and Lck-ERα-/- mice compared to sham-operated mice. Furthermore, estrogen treatment of ovx Lck-ERα-/- led to an increased BMD that was indistinguishable from the increase seen after estrogen treatment of ovx control mice. Detailed analysis of both the appendicular (femur) and axial (vertebrae) skeleton showed that both trabecular and cortical bone parameters responded to a similar extent regardless of the presence of ERα in T lymphocytes. In conclusion, ERα expression in T lymphocytes is dispensable for normal estrogenic regulation of bone mass in female mice.
Collapse
Affiliation(s)
- K L Gustafsson
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - K H Nilsson
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - H H Farman
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - A Andersson
- Center for Bone and Arthritis ResearchDepartment of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - V Lionikaite
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - P Henning
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - J Wu
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S H Windahl
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - U Islander
- Center for Bone and Arthritis ResearchDepartment of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S Movérare-Skrtic
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - K Sjögren
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - H Carlsten
- Center for Bone and Arthritis ResearchDepartment of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - J-Å Gustafsson
- Center for Nuclear Receptors and Cell SignalingDepartment of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - C Ohlsson
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - M K Lagerquist
- Center for Bone and Arthritis ResearchDepartment of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
36
|
Chatkupt TT, Libal NL, Mader SL, Murphy SJ, Saunders KE. Effect of Continuous Trio Breeding Compared with Continuous Pair Breeding in 'Shoebox' Caging on Measures of Reproductive Performance in Estrogen Receptor Knockout Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2018; 57:328-334. [PMID: 30049297 PMCID: PMC6059215 DOI: 10.30802/aalas-jaalas-17-000125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/15/2017] [Accepted: 10/27/2017] [Indexed: 06/08/2023]
Abstract
Some performance standards for continuous trio breeding in 'shoebox' cages for inbred stocks and outbred strains of mice challenge the minimum floor space recommendations in the 8th edition of the Guide for the Care and Use of Laboratory Animals. In our study, we evaluated whether continuous trio breeding could be successfully applied to a breeding colony of genetically engineered mice housed in shoebox cages with a floor area of 67.6 in2. Mice heterozygous for genetically engineered mutations to estrogen receptors and their wildtype counterparts were continuously bred as trios or pairs. Confounding environmental factors were controlled through standardized husbandry practices and husbandry, and all mice were bred simultaneously to control for temporal factors. Several measures of reproductive performance-including number of litters per female, production index, interlitter interval, litter size at birth, litter size at weaning, weaning rate, and body weight of pups at weaning- were evaluated over approximately 6 mo. Regardless of genotype, interlitter interval, litter size at birth, and litter size at weaning were significantly lower for trio-bred mice than for pair-bred mice. In addition, significant interactions emerged between genotype and breeding strategy for these reproductive measures. Furthermore, significant differences between genotypes occurred for interlitter interval and weaning rate, regardless of breeding strategy. Underlying mechanisms to account for effects of genotype on interlitter interval and the interaction of genotype with breeding strategy were unclear but may reflect effects of overcrowding and reproductive suppression.
Collapse
Affiliation(s)
- Tom T Chatkupt
- Departments of Comparative Medicine, Oregon Health and Science University, Portland, Oregon;,
| | - Nicole L Libal
- Departments of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
| | - Sarah L Mader
- Departments of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
| | - Stephanie J Murphy
- Departments of Comparative Medicine, Departments of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, Division of Comparative Medicine; Office of Research Infrastructure Programs; Division of Program Coordination, Planning, and Strategic Initiative; Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Kim E Saunders
- Departments of Comparative Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
37
|
Morselli E, Santos RDS, Gao S, Ávalos Y, Criollo A, Palmer BF, Clegg DJ. Impact of estrogens and estrogen receptor-α in brain lipid metabolism. Am J Physiol Endocrinol Metab 2018; 315:E7-E14. [PMID: 29509437 PMCID: PMC7717113 DOI: 10.1152/ajpendo.00473.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Estrogens and their receptors play key roles in regulating body weight, energy expenditure, and metabolic homeostasis. It is known that lack of estrogens promotes increased food intake and induces the expansion of adipose tissues, for which much is known. An area of estrogenic research that has received less attention is the role of estrogens and their receptors in influencing intermediary lipid metabolism in organs such as the brain. In this review, we highlight the actions of estrogens and their receptors in regulating their impact on modulating fatty acid content, utilization, and oxidation through their direct impact on intracellular signaling cascades within the central nervous system.
Collapse
Affiliation(s)
- Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Roberta de Souza Santos
- Cedars-Sinai Diabetes and Obesity Research Institute, Department of Biomedical Research , Los Angeles, California
| | - Su Gao
- Cedars-Sinai Diabetes and Obesity Research Institute, Department of Biomedical Research , Los Angeles, California
- Department of Medicine, Columbia University Medical Center , New York, New York
| | - Yenniffer Ávalos
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell , Santiago , Chile
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile , Santiago , Chile
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Deborah J Clegg
- Cedars-Sinai Diabetes and Obesity Research Institute, Department of Biomedical Research , Los Angeles, California
| |
Collapse
|
38
|
Wetendorf M, Wu SP, Wang X, Creighton CJ, Wang T, Lanz RB, Blok L, Tsai SY, Tsai MJ, Lydon JP, DeMayo FJ. Decreased epithelial progesterone receptor A at the window of receptivity is required for preparation of the endometrium for embryo attachment. Biol Reprod 2018; 96:313-326. [PMID: 28203817 DOI: 10.1095/biolreprod.116.144410] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 12/23/2016] [Accepted: 01/03/2017] [Indexed: 01/25/2023] Open
Abstract
The precise timing of progesterone signaling through its cognate receptor, the progesterone receptor (PGR), is critical for the establishment and maintenance of pregnancy. Loss of PGR expression in the murine uterine epithelium during the preimplantation period is a marker for uterine receptivity and embryo attachment. We hypothesized that the decrease in progesterone receptor A (PGRA) expression is necessary for successful embryo implantation. To test this hypothesis, a mouse model constitutively expressing PGRA (mPgrALsL/+) was generated. Expression of PGRA in all uterine compartments (Pgrcre) or uterine epithelium (Wnt7acre) resulted in infertility with defects in embryo attachment and stromal decidualization. Expression of critical PGRA target genes, indian hedgehog, and amphiregulin (Areg), was maintained through the window of receptivity while the estrogen receptor target gene, the leukemia inhibitory factor (Lif), a key regulator of embryo receptivity, was decreased. Transcriptomic and cistromic analyses of the mouse uterus at day 4.5 of pregnancy identified an altered group of genes regulating molecular transport in the control of fluid and ion levels within the uterine interstitial space. Additionally, LIF and its cognate receptor, the leukemia inhibitory factor receptor (LIFR), exhibited PGR-binding events in regions upstream of the transcriptional start sites, suggesting PGRA is inhibiting transcription at these loci. Therefore, downregulation of the PGRA isoform at the window of receptivity is necessary for the attenuation of hedgehog signaling, transcriptional activation of LIF signaling, and modulation of solutes and fluid, producing a receptive environment for the attaching embryo.
Collapse
Affiliation(s)
- Margeaux Wetendorf
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.,Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, USA
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, USA
| | - Xiaoqiu Wang
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Tianyuan Wang
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, USA
| | - Rainer B Lanz
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Leen Blok
- Department of Obstetrics and Gynaecology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sophia Y Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, USA
| |
Collapse
|
39
|
Rivera HM, Stincic TL. Estradiol and the control of feeding behavior. Steroids 2018; 133:44-52. [PMID: 29180290 PMCID: PMC5864536 DOI: 10.1016/j.steroids.2017.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022]
Abstract
This review lays out the evidence for the role of E2 in homeostatic and hedonic feeding across several species. While significant effort has been expended on homeostatic feeding research, more studies for hedonic feeding need to be conducted (i.e. are there increases in meal size and enhanced motivation to natural food rewards). By identifying the underlying neural circuitry involved, one can better delineate the mechanisms by which E2 influences feeding behavior. By utilizing more selective neural targeting techniques, such as optogenetics, significant progress can be made toward this goal. Together, behavioral and physiological techniques will help us to better understand neural deficits that can increase the risk for obesity in the absence of E2 (menopause) and aid in developing therapeutic strategies.
Collapse
Affiliation(s)
- H M Rivera
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - T L Stincic
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
40
|
Bartemes K, Chen CC, Iijima K, Drake L, Kita H. IL-33-Responsive Group 2 Innate Lymphoid Cells Are Regulated by Female Sex Hormones in the Uterus. THE JOURNAL OF IMMUNOLOGY 2017; 200:229-236. [PMID: 29133293 DOI: 10.4049/jimmunol.1602085] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 10/14/2017] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) reside in multiple organs in the body, where they play roles in immunity, tissue homeostasis, and metabolic regulation. However, little is known about the regulatory mechanisms of ILC2s in different organs. Here, we identified ILC2s in the mouse uterus and found that they express cell surface molecules, including the IL-33 receptor, ST2, that are roughly comparable to those expressed by lung ILC2s. Both in vivo and in vitro treatment with IL-33 induced type 2 cytokine production in uterine ILC2s, suggesting that they respond to IL-33 in a manner similar to ILC2s in other organs. Importantly, uterine ILC2s were nearly absent in ovariectomized mice and were increased in wild-type mice by estrogen administration, whereas lung ILC2s were unaffected by both ovariectomy and estrogen administration. Likewise, a marked reduction in uterine ILC2s was observed in mice deficient in estrogen receptor α or estrogen receptor β. Furthermore, uterine ILC2s highly expressed estrogen receptor α, and in vitro culture of isolated uterine ILC2s with 17β-estradiol modified expression of a number of genes. Finally, an increased prevalence in neonatal mortality was observed in litters from dams lacking the IL-33 receptor, ST2. Taken together, our findings indicate that unlike lung IL2Cs, uterine ILC2s are regulated by female sex hormones, which may specialize them for specific physiological functions.
Collapse
Affiliation(s)
- Kathleen Bartemes
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and.,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Chien-Chang Chen
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and.,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Koji Iijima
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Li Drake
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and .,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
41
|
Clayton JA. Applying the new SABV (sex as a biological variable) policy to research and clinical care. Physiol Behav 2017; 187:2-5. [PMID: 28823546 DOI: 10.1016/j.physbeh.2017.08.012] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 11/27/2022]
Abstract
Sex as a biological variable (SABV) is a key part of the new National Institutes of Health (NIH) initiative to enhance reproducibility through rigor and transparency. The SABV policy requires researchers to factor sex into the design, analysis, and reporting of vertebrate animal and human studies. The policy was implemented as it has become increasingly clear that male/female differences extend well beyond reproductive and hormonal issues. Implementation of the policy is also meant to address inattention to sex influences in biomedical research. Sex affects: cell physiology, metabolism, and many other biological functions; symptoms and manifestations of disease; and responses to treatment. For example, sex has profound influences in neuroscience, from circuitry to physiology to pain perception. Extending beyond the robust efforts of NIH to ensure that women are included in clinical trials, the SABV policy also includes rigorous preclinical experimental designs that inform clinical research. Additionally, the NIH has engaged journal editors and publishers to facilitate reproducibility by addressing rigor and promoting transparency through scientifically appropriate sex-specific study results reporting. The Sex And Gender Equity in Research (SAGER) guidelines were developed to assist researchers and journal editors in reporting sex and gender information in publications [1].
Collapse
Affiliation(s)
- Janine Austin Clayton
- Office of Research on Women's Health, National Institutes of Health, 6707 Democracy Boulevard, Bethesda, MD 20817, United States.
| |
Collapse
|
42
|
Bado I, Gugala Z, Fuqua SAW, Zhang XHF. Estrogen receptors in breast and bone: from virtue of remodeling to vileness of metastasis. Oncogene 2017; 36:4527-4537. [PMID: 28368409 PMCID: PMC5552443 DOI: 10.1038/onc.2017.94] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 12/11/2022]
Abstract
Bone metastasis is a prominent cause of morbidity and mortality in cancer. High rates of bone colonization in breast cancer, especially in the subtype expressing estrogen receptors (ERs), suggest tissue-specific proclivities for metastatic tumor formation. The mechanisms behind this subtype-specific organ-tropism remains largely elusive. Interestingly, as the major driver of ER+ breast cancer, ERs also have important roles in bone development and homeostasis. Thus, any agents targeting ER will also inevitably affect the microenvironment, which involves the osteoblasts and osteoclasts. Yet, how such microenvironmental effects are integrated with direct therapeutic responses of cancer cells remain poorly understood. Recent findings on ER mutations, especially their enrichment in bone metastasis, raised even more provocative questions on the role of ER in cancer-bone interaction. In this review, we evaluate the importance of ERs in bone metastasis and discuss new avenues of investigation for bone metastasis treatment based on current knowledge.
Collapse
Affiliation(s)
- Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Zbigniew Gugala
- Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555
| | - Suzanne A. W. Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| |
Collapse
|
43
|
Newell-Fugate AE. The role of sex steroids in white adipose tissue adipocyte function. Reproduction 2017; 153:R133-R149. [PMID: 28115579 DOI: 10.1530/rep-16-0417] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/16/2017] [Accepted: 01/20/2017] [Indexed: 12/13/2022]
Abstract
With the increasing knowledge that gender influences normal physiology, much biomedical research has begun to focus on the differential effects of sex on tissue function. Sexual dimorphism in mammals is due to the combined effects of both genetic and hormonal factors. Hormonal factors are mutable particularly in females in whom the estrous cycle dominates the hormonal milieu. Given the severity of the obesity epidemic and the fact that there are differences in the obesity rates in men and women, the role of sex in white adipose tissue function is being recognized as increasingly important. Although sex differences in white adipose tissue distribution are well established, the mechanisms affecting differential function of adipocytes within white adipose tissue in males and females remain largely understudied and poorly understood. One of the largest differences in the endocrine environment in males and females is the concentration of circulating androgens and estrogens. This review examines the effects of androgens and estrogens on lipolysis/lipogenesis, adipocyte differentiation, insulin sensitivity and adipokine production in adipocytes from white adipose tissue with a specific emphasis on the sexual dimorphism of adipocyte function in white adipose tissue during both health and disease.
Collapse
Affiliation(s)
- A E Newell-Fugate
- Department of Veterinary Physiology and PharmacologyTexas A&M University, College Station, Texas, USA
| |
Collapse
|
44
|
Epidemiology of Endometrial Carcinoma: Etiologic Importance of Hormonal and Metabolic Influences. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 943:3-46. [PMID: 27910063 DOI: 10.1007/978-3-319-43139-0_1] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Endometrial carcinoma is the most common gynecologic cancer in developed nations, and the annual incidence is projected to increase, secondary to the high prevalence of obesity, a strong endometrial carcinoma risk factor. Although endometrial carcinomas are etiologically, biologically, and clinically diverse, hormonal and metabolic mechanisms are particularly strongly implicated in the pathogenesis of endometrioid carcinoma, the numerically predominant subtype. The centrality of hormonal and metabolic disturbances in the pathogenesis of endometrial carcinoma, combined with its slow development from well-characterized precursors in most cases, offers a substantial opportunity to reduce endometrial carcinoma mortality through early detection, lifestyle modification, and chemoprevention. In this chapter, we review the epidemiology of endometrial carcinoma, emphasizing theories that link risk factors for these tumors to hormonal and metabolic mechanisms. Future translational research opportunities related to prevention are discussed.
Collapse
|
45
|
Abstract
When orthodontic patients desire shorter treatment times with aesthetic results and long-term stability, it is important for the orthodontist to understand the potential limitations and problems that may arise during standard and/or technology-assisted accelerated treatment. Bone density plays an important role in facilitating orthodontic tooth movement (OTM), such that reductions in bone density can significantly increase movement velocity. Lifestyle, genetic background, environmental factors, and disease status all can influence a patients' overall health and bone density. In some individuals, these factors may create specific conditions that influence systemic-wide bone metabolism. Both genetic variation and the onset of a bone-related disease can influence systemic bone density and local bone density, such as observed in the mandible and maxilla. These types of localized density changes can affect the rate of OTM and may also influence the risk of unwanted outcomes, i.e., the occurrence of dental external apical root resorption (EARR).
Collapse
Affiliation(s)
- Alejandro Iglesias-Linares
- Department of Orthodontics, Complutense University of Madrid, Plaza Ramon y Cajal sn, Phone: +34636705246,
| | - Lorri Ann Morford
- University of Kentucky Center for the Biologic Basis of Oral/Systemic Diseases, 1095 Veterans Administration Drive, HSRB Room 414, Lexington, KY 40536-0305 USA, Phone: 859-323-2595 Fax: 859-257-6566,
| | - James Kennedy Hartsfield
- University of Kentucky Center for the Biologic Basis of Oral/Systemic Diseases, 1095 Veterans Administration Drive, HSRB Room 414, Lexington, KY 40536-0305 USA, Phone: 859-323-0296 Fax: 859-257-6566,
| |
Collapse
|
46
|
|
47
|
Seleit I, Bakry OA, El Repey HS, Ali R. Intrinsic versus Extrinsic Aging: A Histopathological, Morphometric and Immunohistochemical Study of Estrogen Receptor β and Androgen Receptor. Skin Pharmacol Physiol 2016; 29:178-89. [PMID: 27399919 DOI: 10.1159/000446662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/28/2016] [Indexed: 11/19/2022]
Abstract
Skin is a target organ of sex steroids which play important roles in skin health and disease. The aim of this study is to investigate the expression of estrogen receptor β (ERβ) and androgen receptor (AR) in human skin from different age groups for a better understanding of the hormonal regulation of skin aging. Using standard immunohistochemical techniques, biopsies of sun-unprotected and sun-protected skin were taken from 60 normal subjects. Sun-protected skin showed significantly higher immunoreactivity for ERβ and AR compared to sun-unprotected skin of all age groups. Significantly higher ERβ H score and percent of expression were associated with the 20-35 years age group compared to the groups that were 35-50 years and >50 years old (p < 0.02, p = 0.03, respectively) in sun-unprotected and sun-protected skin (p < 0.001, p = 0.01, respectively). AR H score showed a negative correlation with age (p = 0.04) with no significant difference in immunoreactivity in different age groups, either in sun-unprotected or sun-protected skin. There was also a significant correlation between ERβ H score and epidermal thickness in sun-unprotected (p = 0.04) and sun-protected skin (p = 0.04) in studied subjects regardless of age. The same relationships did not reach significance with AR expression. However, a significant positive correlation was detected between H scores and percent of expression of ERβ and AR in sun-unprotected (p = 0.01, p = 0.02, respectively) and sun-protected skin (p = 0.005, p = 0.02, respectively) regardless of age. In conclusion, both ERβ and AR decline gradually with intrinsic and extrinsic aging. This decline is more obvious with extrinsic aging. Further large-scaled studies are recommended to expand, validate and translate current findings to clinically significant, diagnostic and therapeutic applications. Molecular studies to investigate the probable ligand-independent action of both receptors are warranted. In addition, their gene expression patterns and associated signaling and metabolic pathways can also be tackled to provide a basis for further interventions in pathological processes that involve their dysregulation.
Collapse
Affiliation(s)
- Iman Seleit
- Department of Dermatology, Andrology and STDs, Faculty of Medicine, Menoufiya University, Shibin El Koom, Egypt
| | | | | | | |
Collapse
|
48
|
The role of membrane ERα signaling in bone and other major estrogen responsive tissues. Sci Rep 2016; 6:29473. [PMID: 27388455 PMCID: PMC4937452 DOI: 10.1038/srep29473] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/20/2016] [Indexed: 12/03/2022] Open
Abstract
Estrogen receptor α (ERα) signaling leads to cellular responses in several tissues and in addition to nuclear ERα-mediated effects, membrane ERα (mERα) signaling may be of importance. To elucidate the significance, in vivo, of mERα signaling in multiple estrogen-responsive tissues, we have used female mice lacking the ability to localize ERα to the membrane due to a point mutation in the palmitoylation site (C451A), so called Nuclear-Only-ER (NOER) mice. Interestingly, the role of mERα signaling for the estrogen response was highly tissue-dependent, with trabecular bone in the axial skeleton being strongly dependent (>80% reduction in estrogen response in NOER mice), cortical and trabecular bone in long bones, as well as uterus and thymus being partly dependent (40–70% reduction in estrogen response in NOER mice) and effects on liver weight and total body fat mass being essentially independent of mERα (<35% reduction in estrogen response in NOER mice). In conclusion, mERα signaling is important for the estrogenic response in female mice in a tissue-dependent manner. Increased knowledge regarding membrane initiated ERα actions may provide means to develop new selective estrogen receptor modulators with improved profiles.
Collapse
|
49
|
Differential expression of estrogen receptor α and progesterone receptor in the normal and cryptorchid testis of a dog. Lab Anim Res 2016; 32:128-32. [PMID: 27382382 PMCID: PMC4931037 DOI: 10.5625/lar.2016.32.2.128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/27/2016] [Accepted: 05/26/2016] [Indexed: 01/13/2023] Open
Abstract
Descending of the testes is an important process for spermatogenesis and cryptorchidism is one of the most relevant genital defects in dogs. In a previous study, we observed abnormal morphology and proliferation of Sertoli cells in a cryptorchid testis. In the present study, we investigated the expression of estrogen and progesterone receptors in the normal and cryptorchid testis of a dog. Elective orchidectomy was performed on the dog's abdominal right testis (undescended, cryptorchid) and scrotal left testis (descended, normal). In the normal testis, estrogen receptor α immunoreactivity was detected in Leydig cells alone, while estrogen receptor α immunoreactivity in the cryptorchid testis was significantly prominent in the Sertoli cells as well. In addition, progesterone receptor immunoreactivity in the control testis was detected in the spermatids, but was not detected in the cryptorchid testis. This result suggests that unilateral cryptorchidism causes increases of estrogen receptor α expression in Sertoli cells.
Collapse
|
50
|
Macari S, Ajay Sharma L, Wyatt A, Knowles P, Szawka R, Garlet G, Grattan D, Dias G, Silva T. Osteoprotective Effects of Estrogen in the Maxillary Bone Depend on ERα. J Dent Res 2016; 95:689-96. [DOI: 10.1177/0022034516633154] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Estrogen deficiency results in disruption of maxillary alveolar bone microarchitecture. Most of the actions of estrogen in long bones occur via estrogen receptor α (ERα). However, the function of ERα in the maxillary bone has not been defined. We aimed to investigate the role and underlying mechanisms of ERα in the physiological and mechanically induced alveolar bone remodeling in female and male mice. Wild-type (WT) and ERα−/− (ERKOα) mice were subjected to mechanically stimulated bone remodeling by inducing orthodontic tooth movement (OTM). The maxillary bone was analyzed using histomorphometric analysis, micro–computed tomography, quantitative polymerase chain reaction, and energy-dispersive spectroscopy. Bone marrow cells (BMCs) from WT and ERKOα mice were tested for their capacity to differentiate into osteoblasts and osteoclasts. Both male and female ERKOα mice exhibited marked reduction of alveolar bone mass and increased OTM. This response was associated with an increased number of osteoclasts and reduced number of apoptotic cells and osteoblasts in the periodontium and alveolar bone. Consistently, ERKOα mice exhibited lower levels of calcium in bone and increased expression of IL-33 (interleukin-33), TNF-α (tumor necrosis factor α), and IL-1β (interleukin-1β) and decreased expression of dentin matrix acidic phosphoprotein and alkaline phosphatase in periodontal tissues. Moreover, the differentiation of osteoclasts and osteoblasts in vitro was significantly higher in BMCs obtained from ERKOα. ERα is required to maintain the microarchitecture of maxillary alveolar bone. This process is linked to bone cell differentiation and apoptosis, as well as local production of inflammatory molecules such as IL-33, TNF-α, and IL-1β.
Collapse
Affiliation(s)
- S. Macari
- Department of Oral Pathology and Surgery, Faculty of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - L. Ajay Sharma
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - A. Wyatt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - P. Knowles
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - R.E. Szawka
- Department of Physiology and Biophysics, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - G.P. Garlet
- Department of Biological Sciences, School of Dentistry of Bauru, São Paulo University, Bauru, SP, Brazil
| | - D.R. Grattan
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - G.J. Dias
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - T.A. Silva
- Department of Oral Pathology and Surgery, Faculty of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|