1
|
Cohen DJ, Dennis CD, Deng J, Boyan BD, Schwartz Z. Estradiol induces bone osteolysis in triple-negative breast cancer via its membrane-associated receptor ERα36. JBMR Plus 2024; 8:ziae041. [PMID: 38644978 PMCID: PMC11032217 DOI: 10.1093/jbmrpl/ziae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/31/2024] [Accepted: 02/27/2024] [Indexed: 04/23/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is thought to be an estradiol-independent, hormone therapy-resistant cancer because of lack of estrogen receptor alpha 66 (ERα66). We identified a membrane-bound splice variant, ERα36, in TNBC cells that responds to estrogen (E2) and may contribute to bone osteolysis. We demonstrated that the MDA-MB-231 TNBC cell line, which expresses ERα36 similarly to MCF7 cells, is responsive to E2, forming osteolytic tumors in vivo. MDA-MB-231 cells activate osteoclasts in a paracrine manner. Conditioned media (CM) from MDA-MB-231 cells treated with bovine serum albumin-bound E2 (E2-BSA) increased activation of human osteoclast precursor cells; this was blocked by addition of anti-ERα36 antibody to the MDA-MB-231 cultures. Osteoclast activation and bone resorption genes were elevated in RAW 264.7 murine macrophages following treatment with E2-BSA-stimulated MDA-MB-231 CM. E2 and E2-BSA increased phospholipase C (PLC) and protein kinase C (PKC) activity in MDA-MB-231 cells. To examine the role of ERα36 signaling in bone osteolysis in TNBC, we used our bone-cancer interface mouse model in female athymic homozygous Foxn1nu mice. Mice with MDA-MB-231 tumors and treated with tamoxifen (TAM), E2, or TAM/E2 exhibited increased osteolysis, cortical bone breakdown, pathologic fracture, and tumor volume; the combined E2/TAM group also had reduced bone volume. These results suggest that E2 increased osteolytic lesions in TNBC through a membrane-mediated PLC/PKC pathway involving ERα36, which was enhanced by TAM, demonstrating the role of ERα36 and its membrane-associated signaling pathway in bone tumors. This work suggests that ERα36 may be a potential therapeutic target in patients with TNBC.
Collapse
Affiliation(s)
- D Joshua Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Cydney D Dennis
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Jingyao Deng
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Barbara D Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States
- Department of Periodontics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229United States
| |
Collapse
|
2
|
Sharif Swallah M, Bondzie-Quaye P, Wang H, Shao CS, Hua P, Alrasheed Bashir M, Benjamin Holman J, Sossah FL, Huang Q. Potentialities of Ganoderma lucidum extracts as functional ingredients in food formulation. Food Res Int 2023; 172:113161. [PMID: 37689913 DOI: 10.1016/j.foodres.2023.113161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 09/11/2023]
Abstract
Owing to the recognized therapeutic characteristics of G. lucidum, it is one of the most extensively researched mushrooms as a chemopreventive agent and as a functional food. It is a known wood-degrading basidiomycete possessing numerous pharmacological functions and is termed a natural pharmacy store due to its rich number of active compounds which have proved to portray numerous therapeutic properties. This current review highlights studies on the potentialities of G. lucidum extracts as functional ingredients on organoleptic and nutritional properties of food products (e.g., dairy, wine, beverage, bakery, meat, and other products). In addition, the study delved into various aspects of encapsulated G. lucidum extracts, their morphological and rheological characteristics, prebiotic and immunomodulatory importance, the effects on apoptosis, autophagy, cancer therapy, inflammatory responses, oxidative stress, antioxidant activities, and safety concerns. These findings have significant implications for the development of new products in the food and pharmaceutical industries. On the other hand, the various active compounds extracted from G. lucidum exhibited no toxic or adverse effects, and the appeal for it as a dietary food, natural remedy, and health-fortifying food is drastically increasing as well as attracting the interest of both the industrial and scientific communities. Furthermore, the formation of functional foods based on G. lucidum appears to have actual promise and exciting prospects in nutrition, food, and pharmaceutical sciences.
Collapse
Affiliation(s)
- Mohammed Sharif Swallah
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Agriculture, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China
| | - Precious Bondzie-Quaye
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Agriculture, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China
| | - Han Wang
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Agriculture, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China
| | - Chang-Sheng Shao
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Pei Hua
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Mona Alrasheed Bashir
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Agriculture, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China
| | - Joseph Benjamin Holman
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Frederick Leo Sossah
- Council for Scientific and Industrial Research (CSIR), Oil Palm Research Institute, Coconut Research Programme, P.O. Box 245, Sekondi, Ghana
| | - Qing Huang
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Agriculture, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
3
|
Wang S, Wang Z, Wang X, Qu J, Li F, Ji C, Wu H. Histopathological and transcriptomic analyses reveal the reproductive endocrine-disrupting effects of decabromodiphenyl ethane (DBDPE) in mussel Mytilus galloprovincialis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 862:160724. [PMID: 36493811 DOI: 10.1016/j.scitotenv.2022.160724] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/21/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
The novel brominated flame retardant DBDPE has become a widespread environmental contaminant and could affect reproductive endocrine system in vertebrates. However, information about reproductive endocrine-disrupting effects of DBDPE on invertebrates is totally unknown. In this study, mussels Mytilus galloprovincialis were exposed to 1, 10, 50, 200 and 500 μg/L DBDPE for 30 days. Histopathological and transcriptomic analyses were performed to assess the reproductive endocrine-disrupting effects of DBDPE in mussels and the potential mechanisms. DBDPE promoted the gametogenesis in mussels of both sexes according to histological observation, gender-specific gene expression (VERL and VCL) and histological morphometric parameter analysis. Transcriptomic analysis demonstrated that DBDPE suppressed the genes related to cholesterol homeostasis and transport in both sexes via different LRPs- and ABCs-mediated pathways. DBDPE also disturbed nongenomic signaling pathway including signaling cascades (GPR157-IP3-Ca2+) in males and secondary messengers (cGMP) in females, and subsequently altered the expression levels of reproductive genes (VMO1, ZAN, Banf1 and Hook1). Additionally, dysregulation of energy metabolism in male mussels induced by DBDPE might interfere with the reproductive endocrine system. Overall, this is the first report that DBDPE evoked reproductive endocrine-disruptions in marine mussels. These findings will provide important references for ecological risk assessment of DBDPE pollution in marine environment.
Collapse
Affiliation(s)
- Shuang Wang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; College of Life Sciences, Yantai University, Yantai 264005, PR China
| | - Zhiyu Wang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China
| | - Xumin Wang
- College of Life Sciences, Yantai University, Yantai 264005, PR China
| | - Jiangyong Qu
- College of Life Sciences, Yantai University, Yantai 264005, PR China
| | - Fei Li
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China
| | - Chenglong Ji
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China.
| | - Huifeng Wu
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China
| |
Collapse
|
4
|
Xu XL, Huang ZY, Yu K, Li J, Fu XW, Deng SL. Estrogen Biosynthesis and Signal Transduction in Ovarian Disease. Front Endocrinol (Lausanne) 2022; 13:827032. [PMID: 35299973 PMCID: PMC8921451 DOI: 10.3389/fendo.2022.827032] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/03/2022] [Indexed: 12/01/2022] Open
Abstract
Estrogen mainly binds to estrogen receptors (ERs) to regulate menstrual cycles and reproduction. The expression of ERalpha (ERα), ERbeta (ERβ), and G-protein-coupled estrogen receptor (GPER) mRNA could be detected in ovary, suggesting that they play an important role in estrogen signal transduction in ovary. And many studies have revealed that abnormal expression of estrogen and its receptors is closely related to ovarian disease or malignant tumors. With the continuous development and research of animal models, tissue-specific roles of both ERα and ERβ have been demonstrated in animals, which enable people to have a deeper understanding of the potential role of ER in regulating female reproductive diseases. Nevertheless, our current understanding of ERs expression and function in ovarian disease is, however, incomplete. To elucidate the biological mechanism behind ERs in the ovary, this review will focus on the role of ERα and ERβ in polycystic ovary syndrome (PCOS), ovarian cancer and premature ovarian failure (POF) and discuss the major challenges of existing therapies to provide a reference for the treatment of estrogen target tissue ovarian diseases.
Collapse
Affiliation(s)
- Xue-Ling Xu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zheng-Yuan Huang
- Department of Metabolism, Digestion and Reproduction, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Kun Yu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jun Li
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Wei Fu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shou-Long Deng
- National Health Commission of China (NHC) Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Oliveira ML, Mello BP, Gonella-Diaza AM, Scolari SC, Pugliesi G, Martins T, Feltrin IR, Sartori R, Canavessi AMO, Binelli M, Membrive CMB. Unravelling the role of 17β-estradiol on advancing uterine luteolytic cascade in cattle. Domest Anim Endocrinol 2022; 78:106653. [PMID: 34455235 DOI: 10.1016/j.domaniend.2021.106653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 01/10/2023]
Abstract
In cattle, 17β-estradiol (E2) stimulates prostaglandin F2α (PGF2α) synthesis, which causes luteolysis. Except for the well-established upregulation of oxytocin receptor gene (OXTR), molecular mechanisms of E2-induced PGF2α release in vivo remain unknown. We hypothesized that E2-induced PGF2α release requires de novo transcription of components of the PGF2α synthesis machinery. Beef cows (n = 52) were assigned to remain untreated (Control; n = 10), to receive 50% ethanol infusion intravenously (Placebo; n = 21), or 3 mg E2 in 50% ethanol infusion intravenously (Estradiol; n = 21) on day 15 (D15) after estrus. We collected a single endometrial biopsy per animal at the time of the treatment (0h; Control B0h group), 4 hours (4h; Placebo B4h group and Estradiol B4h group), or 7 hours (7h; Placebo B7h group and Estradiol B7h group) post-treatment. Compared to the Placebo group, the Estradiol group presented significantly greater 13,14-dihydro-15-keto-PGF2α concentrations between 4h and 7h and underwent earlier luteolysis. At 4h, the qPCR analysis showed a lower abundance of ESR1, ESR2 and aldo-keto reductase family 1 member B1 (AKR1B1) genes in the Estradiol B4h group, and a greater abundance of OXTR compared to the Placebo B4h group. Similarly, the E2 treatment significantly reduced the abundance of AKR1B1, and AKR1C4 in the Estradiol B7h group, compared to the placebo group. Overall, E2-induced PGF2α release and luteolysis involved an unexpected and transient downregulation of components of the PGF2α-synthesis cascade, except for OXTR, which was upregulated. Collectively, our data suggest that E2 connects newly-synthesized OXTR to pre-existing cellular machinery to synthesize PGF2α and cause luteal regression.
Collapse
Affiliation(s)
- M L Oliveira
- Agrarian Sciences Center, State University of Maranhão Tocantine Region, 1300 Godofredo Viana St, Center, Imperatriz - MA, 65900-000, Brazil..
| | - B P Mello
- Department of Animal Reproduction, University of São Paulo, 225 Duque de Caxias Norte Ave, Pirassununga - SP, 13635900, Brazil
| | - A M Gonella-Diaza
- North Florida Research and Education Center, Institute of Food and Agricultural Sciences, University of Florida, 3925 FL-71, Greenwood, FL 32443, USA
| | - S C Scolari
- Department of Animal Reproduction, University of São Paulo, 225 Duque de Caxias Norte Ave, Pirassununga - SP, 13635900, Brazil
| | - G Pugliesi
- Department of Animal Reproduction, University of São Paulo, 225 Duque de Caxias Norte Ave, Pirassununga - SP, 13635900, Brazil
| | - T Martins
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32611, USA
| | - I R Feltrin
- Department of Pharmacology and Biotechnology, São Paulo State University, Rubião Júnior District no number, Botucatu-SP, 18618-970, Brazil
| | - R Sartori
- Department of Animal Science, Luiz de Queiroz College of Agriculture, University of São Paulo, 11, Pádua Dias Ave, Piracicaba-SP, 13418900, Brazil
| | - A M O Canavessi
- Department of Animal Science, Luiz de Queiroz College of Agriculture, University of São Paulo, 11, Pádua Dias Ave, Piracicaba-SP, 13418900, Brazil
| | - M Binelli
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32611, USA
| | - C M B Membrive
- Department of Animal Sciences, São Paulo State University, 294-SP Commander João Ribeiro de Barros, 651 Road, Dracena-SP, 17900000, Brazil
| |
Collapse
|
6
|
Balthazart J. Membrane-initiated actions of sex steroids and reproductive behavior: A historical account. Mol Cell Endocrinol 2021; 538:111463. [PMID: 34582978 DOI: 10.1016/j.mce.2021.111463] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/25/2023]
Abstract
It was assumed for a long time that sex steroids are activating reproductive behaviors by the same mechanisms that produce their morphological and physiological effects in the periphery. However during the last few decades an increasing number of examples were identified where behavioral effects of steroids were just too fast to be mediated via changes in DNA transcription. This progressively forced behavioral neuroendocrinologists to recognize that part of the effects of steroids on behavior are mediated by membrane-initiated events. In this review we present a selection of these early data that changed the conceptual landscape and we provide a summary the different types of membrane-associated receptors (estrogens, androgens and progestagens receptors) that are playing the most important role in the control of reproductive behaviors. Then we finally describe in more detail three separate behavioral systems in which membrane-initiated events have clearly been established to contribute to behavior control.
Collapse
|
7
|
Cataldi M, Citro V, Resnati C, Manco F, Tarantino G. New Avenues for Treatment and Prevention of Drug-Induced Steatosis and Steatohepatitis: Much More Than Antioxidants. Adv Ther 2021; 38:2094-2113. [PMID: 33761100 PMCID: PMC8107075 DOI: 10.1007/s12325-021-01669-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022]
Abstract
Drug-induced lipid accumulation in the liver may induce two clinically relevant conditions, drug-induced steatosis (DIS) and drug-induced steatohepatitis (DISH). The list of drugs that may cause DIS or DISH is long and heterogeneous and includes therapeutically relevant molecules that cannot be easily replaced by less hepatotoxic medicines, therefore making specific strategies necessary for DIS/DISH prevention or treatment. For years, the only available tools to achieve these goals have been antioxidant drugs and free radical scavengers, which counteract drug-induced mitochondrial dysfunction but, unfortunately, have only limited efficacy. In the present review we illustrate how in vitro preclinical research unraveled new key players in the pathogenesis of specific forms of DISH, and how, in a few cases, proof of concept of the beneficial effects of their pharmacological modulation has been obtained in vivo in animal models of this condition. The key issue emerging from these studies is that, in selected cases, liver toxicity depends on mechanisms unrelated to those responsible for the desired, primary pharmacological effects of the toxic drug and, therefore, specific strategies can be designed to overcome steatogenicity without making the drug ineffective. In particular, the hepatotoxic drug could be given in combination with a second molecule intended to selectively antagonize its liver toxicity whilst, ideally, potentiating its desired pharmacological activity. Although most of the evidence that we discuss is from in vitro or animal models and will need to be further explored and validated in humans, it highlights new avenues to be pursued in order to improve the safety of steatogenic drugs.
Collapse
|
8
|
Li HJ, Goff A, Rudzinskas SA, Jung Y, Dubey N, Hoffman J, Hipolito D, Mazzu M, Rubinow DR, Schmidt PJ, Goldman D. Altered estradiol-dependent cellular Ca 2+ homeostasis and endoplasmic reticulum stress response in Premenstrual Dysphoric Disorder. Mol Psychiatry 2021; 26:6963-6974. [PMID: 34035477 PMCID: PMC8613306 DOI: 10.1038/s41380-021-01144-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/11/2021] [Accepted: 04/21/2021] [Indexed: 02/04/2023]
Abstract
Premenstrual Dysphoric Disorder (PMDD) is characterized by debilitating mood symptoms in the luteal phase of the menstrual cycle. Prior studies of affected women have implicated a differential response to ovarian steroids. However, the molecular basis of these patients' differential response to hormone remains poorly understood. We performed transcriptomic analyses of lymphoblastoid cell lines (LCLs) derived from women with PMDD and asymptomatic controls cultured under untreated (steroid-free), estradiol-treated (E2), and progesterone-treated (P4) conditions. Weighted gene correlation network analysis (WGCNA) of transcriptomes identified four gene modules with significant diagnosis x hormone interactions, including one enriched for neuronal functions. Next, in a gene-level analysis comparing transcriptional response to hormone across diagnoses, a generalized linear model identified 1522 genes differentially responsive to E2 (E2-DRGs). Among the top 10 E2-DRGs was a physically interacting network (NUCB1, DST, GCC2, GOLGB1) involved in endoplasmic reticulum (ER)-Golgi function. qRT-PCR validation reproduced a diagnosis x E2 interaction (F(1,24)=7.01, p = 0.014) for NUCB1, a regulator of cellular Ca2+ and ER stress. Finally, we used a thapsigargin (Tg) challenge assay to test whether E2 induces differences in Ca2+ homeostasis and ER stress response in PMDD. PMDD LCLs had a 1.36-fold decrease in Tg-induced XBP1 splicing response compared to controls, and a 1.62-fold decreased response (p = 0.005), with a diagnosis x treatment interaction (F(3,33)=3.51, p = 0.026) in the E2-exposed condition. Altered hormone-dependent in cellular Ca2+ dynamics and ER stress may contribute to the pathophysiology of PMDD.
Collapse
Affiliation(s)
- Howard J. Li
- grid.47100.320000000419368710Dept. of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT USA ,grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Allison Goff
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| | - Sarah A. Rudzinskas
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Yonwoo Jung
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| | - Neelima Dubey
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Jessica Hoffman
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Dion Hipolito
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| | - Maria Mazzu
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - David R. Rubinow
- grid.410711.20000 0001 1034 1720Dept. of Psychiatry, University of North Carolina, Chapel Hill, NC USA
| | - Peter J. Schmidt
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - David Goldman
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| |
Collapse
|
9
|
Hao E, Chang LY, Wang DH, Chen YF, Huang RI, Chen H. Dietary Supplementation with Ferula Improves Productive Performance, Serum Levels of Reproductive Hormones, and Reproductive Gene Expression in Aged Laying Hens. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2021. [DOI: 10.1590/1806-9061-2020-1319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- E Hao
- Hebei Agricultural University, China
| | - LY Chang
- Hebei Agricultural University, China
| | - DH Wang
- Hebei Agricultural University, China
| | - YF Chen
- Hebei Agricultural University, China
| | - RI Huang
- Hebei Agricultural University, China
| | - H Chen
- Hebei Agricultural University, China
| |
Collapse
|
10
|
Sachs S, Bastidas-Ponce A, Tritschler S, Bakhti M, Böttcher A, Sánchez-Garrido MA, Tarquis-Medina M, Kleinert M, Fischer K, Jall S, Harger A, Bader E, Roscioni S, Ussar S, Feuchtinger A, Yesildag B, Neelakandhan A, Jensen CB, Cornu M, Yang B, Finan B, DiMarchi RD, Tschöp MH, Theis FJ, Hofmann SM, Müller TD, Lickert H. Targeted pharmacological therapy restores β-cell function for diabetes remission. Nat Metab 2020; 2:192-209. [PMID: 32694693 DOI: 10.1038/s42255-020-0171-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/15/2020] [Indexed: 12/27/2022]
Abstract
Dedifferentiation of insulin-secreting β cells in the islets of Langerhans has been proposed to be a major mechanism of β-cell dysfunction. Whether dedifferentiated β cells can be targeted by pharmacological intervention for diabetes remission, and ways in which this could be accomplished, are unknown as yet. Here we report the use of streptozotocin-induced diabetes to study β-cell dedifferentiation in mice. Single-cell RNA sequencing (scRNA-seq) of islets identified markers and pathways associated with β-cell dedifferentiation and dysfunction. Single and combinatorial pharmacology further show that insulin treatment triggers insulin receptor pathway activation in β cells and restores maturation and function for diabetes remission. Additional β-cell selective delivery of oestrogen by Glucagon-like peptide-1 (GLP-1-oestrogen conjugate) decreases daily insulin requirements by 60%, triggers oestrogen-specific activation of the endoplasmic-reticulum-associated protein degradation system, and further increases β-cell survival and regeneration. GLP-1-oestrogen also protects human β cells against cytokine-induced dysfunction. This study not only describes mechanisms of β-cell dedifferentiation and regeneration, but also reveals pharmacological entry points to target dedifferentiated β cells for diabetes remission.
Collapse
Affiliation(s)
- Stephan Sachs
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Miguel A Sánchez-Garrido
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Maximilian Kleinert
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Katrin Fischer
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| | - Sigrid Jall
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| | - Alexandra Harger
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Erik Bader
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Sara Roscioni
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Siegfried Ussar
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, Neuherberg, Germany
| | | | | | | | - Marion Cornu
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Bin Yang
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Matthias H Tschöp
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany.
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Department of Mathematics, Technical University of Munich, Munich, Germany.
| | - Susanna M Hofmann
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medical Clinic and Polyclinic IV, Ludwig Maximilian University of Munich, Munich, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany.
- Department of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
11
|
Guo M, Chen Y, Chen Q, Guo X, Yuan Z, Kang L, Jiang Y. Epigenetic changes associated with increased estrogen receptor alpha mRNA transcript abundance during reproductive maturation in chicken ovaries. Anim Reprod Sci 2020; 214:106287. [PMID: 32087914 DOI: 10.1016/j.anireprosci.2020.106287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/14/2019] [Accepted: 01/16/2020] [Indexed: 10/25/2022]
Abstract
Estrogen receptor alpha (ERα) is a ligand-activated transcription factor that regulates cellular responses to estrogens and transcription processes of target genes. In this study, changes in DNA methylation and histone modifications in the promoter region and Exon 1 of the ERα gene were analyzed to ascertain epigenetic changes associated with increased ERα mRNA abundance during reproductive maturation from 90 (egg production not yet initiated) to 160 (after egg production was initiated) d of age (d post-hatching) in chicken ovaries. The results indicate there was no difference in CpG methylation at the promoter and Exon 1 except at the region analyzed with primer pairs F2 and R2, where percentage of methylated CpG of Sites 2 and 8 after reproductive maturation was greater compared with before reproductive maturation. By using the chromatin immunuoprecipitation (ChIP) assay combined with SYBR green quantitative PCR, effects of histone modifications were evaluated, including histone H3K4 di + tri methylation, H3K9 phosphorylation and trimethylation, H3K36 methylation and H3K27 acetylation on chicken ERα mRNA transcript abundance. The results indicated that there was a greater histone H3K27 acetylation and lesser H3K36 trimethylation associated with increased abundance of ERα mRNA transcript in chicken ovaries after reproductive maturation (90 compared with 160 d of age). In consistent with this finding, the relative abundance of transcriptional coactivator p300 mRNA transcript and protein in the ovaries was markedly greater in reproductively mature than immature chickens. Findings provide insights into the epigenetic regulations of the chicken ERα gene expression that is required for chicken ovarian development.
Collapse
Affiliation(s)
- Miao Guo
- Department of Biology Science and Technology, Shandong First Medical University, Tai'an, 271016, PR China
| | - Yuxia Chen
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, PR China
| | - Qiuyue Chen
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, PR China
| | - Xiaoli Guo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, PR China
| | - Zhenjie Yuan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, PR China
| | - Li Kang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, PR China.
| | - Yunliang Jiang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, PR China.
| |
Collapse
|
12
|
You H, Meng K, Wang ZY. The ER-α36/EGFR signaling loop promotes growth of hepatocellular carcinoma cells. Steroids 2018; 134:78-87. [PMID: 29481815 DOI: 10.1016/j.steroids.2018.02.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/04/2018] [Accepted: 02/20/2018] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is the common primary liver cancer and the third leading cause of cancer related mortality worldwide. It is generally thought that the estrogen-signaling pathway is not related to the development and progression of human HCC. However, accumulating evidences indicate the existence of a rapid estrogen signaling in HCC cells that is able to promote cell growth. However, the receptor that mediates the rapid estrogen signaling in HCC cells has not been established. Previously, our laboratory identified a variant of ER-α, ER-α36, and found that ER-α36 mediates the rapid estrogen signaling such as the activation of the MAPK/ERK signaling in breast carcinoma cells. Our current experiments studied the role of the rapid estrogen signaling mediated by ER-α36 in growth of HCC HepG2 and PLC/PRF/5 cells that highly express ER-α36 and found these cells were strongly responsive to the rapid estrogen signaling. Knockdown of ER-α36 expression in these HCC cells using the shRNA method attenuated their responsiveness to estrogen and destabilized EGFR protein. ER-α36 mediated estrogen-induced phosphorylation of Src and the MAPK/ERK as well as cyclin D1 expression. In addition, there existed an ER-α36/EGFR positive regulatory loop in HCC cells that was important for the maintenance and positive regulation of HCC tumorsphere cells. Our results thus indicated that the rapid estrogen receptor is mediated by ER-α36 in HCC cells through the EGFR/Src/ERK signaling pathway and suggested that the ER-α36/EGFR signaling loop is a potential target to develop novel therapeutic approaches for HCC treatment.
Collapse
Affiliation(s)
- Hui You
- Beijing Shenogen Biomedical Co., Ltd, Beijing, PR China
| | - Kun Meng
- Beijing Shenogen Biomedical Co., Ltd, Beijing, PR China
| | - Zhao-Yi Wang
- Beijing Shenogen Biomedical Co., Ltd, Beijing, PR China.
| |
Collapse
|
13
|
Suvannang N, Preeyanon L, Malik AA, Schaduangrat N, Shoombuatong W, Worachartcheewan A, Tantimongcolwat T, Nantasenamat C. Probing the origin of estrogen receptor alpha inhibition via large-scale QSAR study. RSC Adv 2018; 8:11344-11356. [PMID: 35542807 PMCID: PMC9079045 DOI: 10.1039/c7ra10979b] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/07/2018] [Indexed: 11/21/2022] Open
Abstract
Estrogen is an important component for the sustenance of normal physiological functions of the mammary glands, particularly for growth and differentiation. Approximately, two-thirds of breast cancers are positive for estrogen receptor (ERs), which is a predisposing factor for the growth of breast cancer cells. As such, ERα represents a lucrative therapeutic target for breast cancer that has attracted wide interest in the search for inhibitory agents. However, the conventional laboratory processes are cost- and time-consuming. Thus, it is highly desirable to develop alternative methods such as quantitative structure-activity relationship (QSAR) models for predicting ER-mediated endocrine agitation as to simplify their prioritization for future screening. In this study, we compiled and curated a large, non-redundant data set of 1231 compounds with ERα inhibitory activity (pIC50). Using comprehensive validation tests, it was clearly observed that the model utilizing the substructure count as descriptors, performed well considering two objectives: using less descriptors for model development and achieving high predictive performance (R Tr 2 = 0.94, Q CV 2 = 0.73, and Q Ext 2 = 0.73). It is anticipated that our proposed QSAR model may become a useful high-throughput tool for identifying novel inhibitors against ERα.
Collapse
Affiliation(s)
- Naravut Suvannang
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand +66 2 441 4371 ext. 2715 +66 2 441 4380
| | - Likit Preeyanon
- Department of Community Medical Technology, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand
| | - Aijaz Ahmad Malik
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand +66 2 441 4371 ext. 2715 +66 2 441 4380
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand +66 2 441 4371 ext. 2715 +66 2 441 4380
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand +66 2 441 4371 ext. 2715 +66 2 441 4380
| | - Apilak Worachartcheewan
- Department of Community Medical Technology, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand
| | - Tanawut Tantimongcolwat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University Bangkok 10700 Thailand +66 2 441 4371 ext. 2715 +66 2 441 4380
| |
Collapse
|
14
|
Somayaji MR, Przekwas AJ, Gupta RK. Combination Therapy for Multi-Target Manipulation of Secondary Brain Injury Mechanisms. Curr Neuropharmacol 2018; 16:484-504. [PMID: 28847295 PMCID: PMC6018188 DOI: 10.2174/1570159x15666170828165711] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a major healthcare problem that affects millions of people worldwide. Despite advances in understanding and developing preventative and treatment strategies using preclinical animal models, clinical trials to date have failed, and a 'magic bullet' for effectively treating TBI-induced damage does not exist. Thus, novel pharmacological strategies to effectively manipulate the complex and heterogeneous pathophysiology of secondary injury mechanisms are needed. Given that goal, this paper discusses the relevance and advantages of combination therapies (COMTs) for 'multi-target manipulation' of the secondary injury cascade by administering multiple drugs to achieve an optimal therapeutic window of opportunity (e.g., temporally broad window) and compares these regimens to monotherapies that manipulate a single target with a single drug at a given time. Furthermore, we posit that integrated mechanistic multiscale models that combine primary injury biomechanics, secondary injury mechanobiology/neurobiology, physiology, pharmacology and mathematical programming techniques could account for vast differences in the biological space and time scales and help to accelerate drug development, to optimize pharmacological COMT protocols and to improve treatment outcomes.
Collapse
Affiliation(s)
| | | | - Raj K. Gupta
- Department of Defense Blast Injury Research Program Coordinating Office, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| |
Collapse
|
15
|
Biallelic and monoallelic ESR2 variants associated with 46,XY disorders of sex development. Genet Med 2017; 20:717-727. [PMID: 29261182 DOI: 10.1038/gim.2017.163] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 08/04/2017] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Disorders or differences of sex development (DSDs) are rare congenital conditions characterized by atypical sex development. Despite advances in genomic technologies, the molecular cause remains unknown in 50% of cases. METHODS Homozygosity mapping and whole-exome sequencing revealed an ESR2 variant in an individual with syndromic 46,XY DSD. Additional cases with 46,XY DSD underwent whole-exome sequencing and targeted next-generation sequencing of ESR2. Functional characterization of the identified variants included luciferase assays and protein structure analysis. Gonadal ESR2 expression was assessed in human embryonic data sets and immunostaining of estrogen receptor-β (ER-β) was performed in an 8-week-old human male embryo. RESULTS We identified a homozygous ESR2 variant, c.541_543del p.(Asn181del), located in the highly conserved DNA-binding domain of ER-β, in an individual with syndromic 46,XY DSD. Two additional heterozygous missense variants, c.251G>T p.(Gly84Val) and c.1277T>G p.(Leu426Arg), located in the N-terminus and the ligand-binding domain of ER-β, were found in unrelated, nonsyndromic 46,XY DSD cases. Significantly increased transcriptional activation and an impact on protein conformation were shown for the p.(Asn181del) and p.(Leu426Arg) variants. Testicular ESR2 expression was previously documented and ER-β immunostaining was positive in the developing intestine and eyes. CONCLUSION Our study supports a role for ESR2 as a novel candidate gene for 46,XY DSD.
Collapse
|
16
|
Low concentration of formononetin promotes proliferation of estrogen receptor-positive cells through an ERα-miR-375-PTEN-ERK1/2-bcl-2 pathway. Oncotarget 2017; 8:100045-100055. [PMID: 29245959 PMCID: PMC5725001 DOI: 10.18632/oncotarget.21923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/23/2017] [Indexed: 11/25/2022] Open
Abstract
A low dose of formononetin accelerates the proliferation of nasopharyngeal carcinoma cells in vitro; however, the underlying mechanism remains unknown. Here, we investigated the molecular mechanism of formononetin in CNE2 cell proliferation. CNE2 cells were treated with 0 to 1 μM formononetin. To inhibit mitogen activated protein kinase / extracellular regulate kinase (MAPK/ERK) kinase (MEK) and microRNA (miR)-375, cells were pretreated with either PD98059 or a miR-375 inhibitor, respectively, followed by co-treatment with formononetin (0.3 μM) plus an inhibitor. Female rats were ovariectomized (OVX), and some OVX rats received formononetin or estrogen (E2) injections. Sham operated animals were used as controls. Compared to control, 0.3 μM formononetin accelerated proliferation and decreased late apoptosis of CNE2 cells. However, formononetin-induced pro-growth and anti-apoptosis activity was abolished by PD98059 and the miR-375 inhibitor. In addition, 0.1 and 0.3 μM formononetin significantly increased estrogen receptor-α (ERα) and bcl-2, but decreased protein-phosphatase and tensin homologue (PTEN) protein expression, all of which was reversed by the miR-375 inhibitor. Additionally, formononetin treatment resulted in a transient upregulation of phosphorylated (p)-ERK1/2. In vivo studies indicated that formononetin significantly increased endometrium thickness and down-regulated ERα expression in OVX rats. Taken together, our study demonstrates that a low concentration of formononetin can promote growth of CNE2 cells and uterine tissues, possibly through regulating the ERα-miR-375-PTEN-ERK1/2-bcl-2 signaling pathway.
Collapse
|
17
|
Duda M, Wartalski K, Tabarowski Z, Gorczyca G. The Role of Androgens in Ovarian Follicular Development: From Fertility to Ovarian Cancer. Theriogenology 2017. [DOI: 10.5772/intechopen.68881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Yang H, Ma L, Wang Y, Zuo W, Li B, Yang Y, Chen Y, Chen L, Wang L, Zhu L. Activation of ClC-3 chloride channel by 17β-estradiol relies on the estrogen receptor α expression in breast cancer. J Cell Physiol 2017; 233:1071-1081. [PMID: 28419445 DOI: 10.1002/jcp.25963] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/14/2017] [Indexed: 11/11/2022]
Abstract
Although extensively studied, the mechanisms by which estrogen promotes breast cancer growth remain to be fully elucidated. Tamoxifen, an antiestrogen agent to treat ERα+ breast cancer, is also a high-affinity blocker of the chloride channels. In this study, we explored the involvement of the chloride channels in the action of estrogen in breast cancer. We found that 17β-estradiol (17β-E2) concentration-dependently activated the chloride currents in ERα+ breast cancer MCF-7 cells. Extracellular hypertonic challenge and chloride channel blockers, NPPB and DIDS inhibited the 17β-E2-activated chloride currents. Decreased the ClC-3 protein expression caused the depletion of the 17β-E2-activated chloride currents. 17β-E2-activated chloride currents which relied on the ERα expression were demonstrated by the following evidences. Firstly, 17β-E2-activated chloride currents could not be observed in ERα- breast cancer MDA-MB-231 cells. Secondly, ER antagonists, tamoxifen and ICI 182,780, and downregulation of ERα expression inhibited or abolished the 17β-E2-activated chloride currents. Thirdly, ERα expression was induced in MDA-MB-231 cells by ESR1 gene transfection, and then 17β-E2-activated chloride currents could be observed. In MCF-7 cells, ERα and ClC-3 mainly located in nucleus and translocated to cell plasma and membrane with respect to co-localization following treatment of 17β-E2. Downregulation of ERα expression could decrease the expression of ClC-3 protein. Conversely, downregulation of ClC-3 expression did not influence the ERα expression. Taken together, our findings demonstrated that ClC-3 is a potential target of 17β-E2 and is modulated by the ERα in breast cancer cell. Pharmacological modulation of ClC-3 may provide a deep understanding in antiestrogen treatment of breast cancer patients.
Collapse
Affiliation(s)
- Haifeng Yang
- Department of Pathology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Lianshun Ma
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China.,Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yawei Wang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Wanhong Zuo
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Bingxue Li
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Yaping Yang
- Analysis and Test Center, Jinan University, Guangzhou, China
| | - Yehui Chen
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lixin Chen
- Department of Pathology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Liwei Wang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Linyan Zhu
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
19
|
Sechman A, Batoryna M, Antos PA, Hrabia A. Effects of PCB 126 and PCB 153 on secretion of steroid hormones and mRNA expression of steroidogenic genes (STAR, HSD3B, CYP19A1) and estrogen receptors (ERα, ERβ) in prehierarchical chicken ovarian follicles. Toxicol Lett 2016; 264:29-37. [PMID: 27832956 DOI: 10.1016/j.toxlet.2016.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/26/2016] [Accepted: 11/05/2016] [Indexed: 11/25/2022]
Abstract
The objective of this study was to assess the in vitro effects of dioxin-like PCB 126 and non-dioxin-like PCB 153 on basal and ovine LH (oLH)-stimulated testosterone (T) and estradiol (E2) secretion and expression of steroidogenic genes (STAR, HSD3B and CYP19A1) and estrogen receptors α (ERα) and β (ERβ) in white (WF) and yellowish (YF) prehierarchical follicles of the hen ovary. Steroid concentrations in a medium and gene expression in follicles following 6h of exposition were determined by RIA and real-time qPCR, respectively. Both PCBs increased basal and oLH-stimulated T secretion by the WF follicles. PCB 126 reduced basal E2 secretion by the WF follicles. PCB 153 elevated but PCB 126 reduced oLH-stimulated E2 secretion by the prehierarchical follicles. PCB 126 increased basal STAR and HSD3B and reduced CYP19A1 mRNA expression in these follicles. PCB 153 increased basal expression of STAR and HSD3B in YF follicles, but diminished HSD3B mRNA levels in the WF. The studied PCBs had an opposite effect on basal and oLH-stimulated CYP19A1 mRNA expression in prehierarchical follicles. Both PCBs modulated basal and inhibited oLH-stimulated ERα and ERβ gene expression in the prehierarchical follicles. In conclusion, data of the current study demonstrate the congener-specific effects of PCBs on sex steroid secretion by prehierarchical follicles of the chicken ovary, which are at least partly related to STAR, HSD3B and CYP19A1 gene expression. It is suggested that PCBs, by influencing follicular steroidogenesis and expression of estrogen receptors, may impair development and selection of yellowish follicles to the preovulatory hierarchy.
Collapse
Affiliation(s)
- Andrzej Sechman
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland.
| | - Marta Batoryna
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Piotr A Antos
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Anna Hrabia
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| |
Collapse
|
20
|
L-Type Calcium Channels Modulation by Estradiol. Mol Neurobiol 2016; 54:4996-5007. [PMID: 27525676 DOI: 10.1007/s12035-016-0045-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/08/2016] [Indexed: 01/29/2023]
Abstract
Voltage-gated calcium channels are key regulators of brain function, and their dysfunction has been associated with multiple conditions and neurodegenerative diseases because they couple membrane depolarization to the influx of calcium-and other processes such as gene expression-in excitable cells. L-type calcium channels, one of the three major classes and probably the best characterized of the voltage-gated calcium channels, act as an essential calcium binding proteins with a significant biological relevance. It is well known that estradiol can activate rapidly brain signaling pathways and modulatory/regulatory proteins through non-genomic (or non-transcriptional) mechanisms, which lead to an increase of intracellular calcium that activate multiple kinases and signaling cascades, in the same way as L-type calcium channels responses. In this context, estrogens-L-type calcium channels signaling raises intracellular calcium levels and activates the same signaling cascades in the brain probably through estrogen receptor-independent modulatory mechanisms. In this review, we discuss the available literature on this area, which seems to suggest that estradiol exerts dual effects/modulation on these channels in a concentration-dependent manner (as a potentiator of these channels in pM concentrations and as an inhibitor in nM concentrations). Indeed, estradiol may orchestrate multiple neurotrophic responses, which open a new avenue for the development of novel estrogen-based therapies to alleviate different neuropathologies. We also highlight that it is essential to determine through computational and/or experimental approaches the interaction between estradiol and L-type calcium channels to assist these developments, which is an interesting area of research that deserves a closer look in future biomedical research.
Collapse
|
21
|
Yin L, Wang ZY. Roles of the ER-α36-EGFR/HER2 positive regulatory loops in tamoxifen resistance. Steroids 2016; 111:95-99. [PMID: 26884313 DOI: 10.1016/j.steroids.2016.01.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 11/16/2022]
Abstract
Tamoxifen provided a successful treatment for ER-positive breast cancer for the past four decades. However, most breast tumors are eventually resistant to tamoxifen therapy. Extensive researches were conducted to understand the molecular mechanisms involved in tamoxifen resistance, and have revealed that multiple signaling molecules and pathways such as EGFR and HER2 are involved in tamoxifen resistance. Currently, the mechanisms by which tamoxifen sensitive breast cancer cells acquire these signaling pathways and develop tamoxifen resistance have not been elucidated. The identification of ER-α36, a variant of ER-α, that is able to mediate agonist activity of tamoxifen provided great insights into the underlying mechanisms of tamoxifen resistance. In this review, we will discuss the biological function and the possible underlying mechanisms of ER-α36 in tamoxifen resistance and specifically illustrate a novel cross-talk mechanism; positive regulatory loops between the ER-α36 and EGFR/HER2 in tamoxifen resistance. The function and the underlying mechanisms of ER-α36 in tamoxifen resistance of the breast cancer stem/progenitor cells will also be discussed. Finally, we will postulate a novel approach to restore tamoxifen sensitivity in tamoxifen resistant breast cancer cells.
Collapse
Affiliation(s)
- Li Yin
- Department of Medical Microbiology and Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE, USA
| | - Zhao-Yi Wang
- Department of Medical Microbiology and Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE, USA.
| |
Collapse
|
22
|
Mahaldashtian M, Naghdi M, Ghorbanian MT, Makoolati Z, Movahedin M, Mohamadi SM. In vitro effects of date palm (Phoenix dactylifera L.) pollen on colonization of neonate mouse spermatogonial stem cells. JOURNAL OF ETHNOPHARMACOLOGY 2016; 186:362-368. [PMID: 27084457 DOI: 10.1016/j.jep.2016.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 04/11/2016] [Accepted: 04/11/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Date palm (Phoenix dactylifera L.) pollen (DPP) is widely used as a folk remedy for male infertility treatment, and has well known medicinal effects. AIM OF THE STUDY This study aimed to determine the in vitro effects of DPP on the efficiency of neonate mouse spermatogonial stem cells (SSCs) proliferation. MATERIAL AND METHODS Sertoli and SSCs were isolated from 6 to 10-days-old mouse testes, and their identity was confirmed using immunocytochemistry against cytokeratin for sertoli cells and PLZF, Oct-4 and CDH-1 for SSCs. Isolated testicular cells were cultured in the absence or presence of 0.06, 0.25 and 0.62mg/ml concentrations of DPP aqueous extract for 2 weeks. The number and diameter of SSC colonies were assessed during third, 7th, 9th and 14th day of culture, and the expression of the Mvh, GFRα-1 and Oct-4 was evaluated using quantitative PCR at the end of the culture period. The significance of the data was analyzed using ANOVA and paired samples t-test and Tukey and Bonferroni test as post hoc tests at the level of p≤0.05. RESULTS Pattern assay of colony formation showed that SSCs numbers increased in the present of 0.62mg/ml concentration of DPP extract with higher slop relative to other groups (P <0.05). Colony diameters had no significant difference between groups in 3th, 7th, 9th and 14th days after culture. The Mvh and Oct-4 genes expression had no significant difference between groups, while GFRα1 expression was increased significantly in cells treated with 0.06mg/ml concentration relative to other groups (P<0.05). CONCLUSION It seems that co-culture of SSCs with sertoli sells in the presence of low doses of DPP can increase SSCs proliferation and keep their stemness state, while higher concentrations can differentiate the treated cells.
Collapse
Affiliation(s)
- Maryam Mahaldashtian
- Department of Molecular & Cellular Biology, Faculty of Biology, Damghan University, Semnan, Iran.
| | - Majid Naghdi
- Department of Anatomical Sciences, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Mohamad Taghi Ghorbanian
- Department of Molecular & Cellular Biology, Faculty of Biology, Damghan University, Semnan, Iran.
| | - Zohreh Makoolati
- Department of Anatomical Sciences, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Mansoureh Movahedin
- Department of Anatomical Sciences, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Seyedeh Momeneh Mohamadi
- Department of Anatomical Sciences, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
23
|
Wang ZY, Yin L. Estrogen receptor alpha-36 (ER-α36): A new player in human breast cancer. Mol Cell Endocrinol 2015; 418 Pt 3:193-206. [PMID: 25917453 DOI: 10.1016/j.mce.2015.04.017] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 04/20/2015] [Accepted: 04/20/2015] [Indexed: 01/16/2023]
Abstract
Prevailing wisdom is that estrogen receptor (ER)-α mediated genomic estrogen signaling is responsible for estrogen-stimulated cell proliferation and development of ER-positive breast cancer. However, accumulating evidence indicates that another estrogen signaling pathway, non-genomic or rapid estrogen signaling, also plays an important role in mitogenic estrogen signaling. Previously, our laboratory cloned a 36 kDa variant of ER-α, ER-α36, and found that ER-α36 is mainly expressed in the cytoplasm and at the plasma membrane. ER-α36 mediates rapid estrogen signaling and inhibits genomic estrogen signaling. In this review, we review and update the biological function of ER-α36 in ER-positive and -negative breast cancer, breast cancer stem/progenitor cells and tamoxifen resistance, potential interaction and cross-talk of ER-α36 with other ERs and growth factor receptors, and intracellular pathways of ER-α36-mediated rapid estrogen signaling. The potential function and underlying mechanism of ER-α in development of ER-positive breast cancer will also be discussed.
Collapse
Affiliation(s)
- Zhao-Yi Wang
- Department of Medical Microbiology & Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE 68178, USA.
| | - Li Yin
- Department of Medical Microbiology & Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE 68178, USA
| |
Collapse
|
24
|
Al-Hendy A, Diamond MP, El-Sohemy A, Halder SK. 1,25-dihydroxyvitamin D3 regulates expression of sex steroid receptors in human uterine fibroid cells. J Clin Endocrinol Metab 2015; 100:E572-82. [PMID: 25625804 PMCID: PMC4399292 DOI: 10.1210/jc.2014-4011] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Uterine fibroids (UFs) are the most common benign tumors in premenopausal women. In this study, we evaluated the effects of 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] for the treatment of UFs. OBJECTIVE To determine the role of 1,25(OH)2D3 on the expression of sex steroid receptors in human UF cells. DESIGN Human UFs and their adjacent myometrium were analyzed for expression of estrogen receptor (ER)-α, progesterone receptor (PR)-A, and PR-B, as well as members of the steroid receptor coactivator (SRC) family. Immortalized human uterine fibroid (human uterine leiomyoma [HuLM]) cells were treated with 1,25(OH)2D3 and assayed for the expression and localization of the aforementioned receptors and SRCs using Western blot, immunohistochemistry, immunofluorescence, and immunoprecipitation assays. MAIN OUTCOME MEASURES We discovered a correlation between reduced levels of vitamin D receptor (VDR) and increased levels of ER-α, PR-A, and PR-B in these tissues. We evaluated the effects of 1,25(OH)2D3 on the regulation of the aforementioned sex steroid receptors. RESULTS We observed an inverse correlation between the up-regulated ER-α, PR-A, and PR-B and expression of VDR in UFs. Treatment with 1,25(OH)2D3 significantly decreased levels of ER-α, PR-A, and PR-B, as well as SRCs in HuLM cells (P < .05). In contrast, 1,25(OH)2D3 self-induced its own VDR, which resulted in an induction of VDR-retinoid X receptor-α complex in HuLM cells. Together, these results suggest that 1,25(OH)2D3 functions as an antagonist of sex steroid hormone receptors in HuLM cells. CONCLUSIONS 1,25(OH)2D3 functions as a potent antiestrogenic/antiprogesteronic agent that may have utility as a novel therapeutic option for UF.
Collapse
Affiliation(s)
- Ayman Al-Hendy
- Department of Obstetrics and Gynecology (A.A.-H., M.P.D., S.K.H.), Georgia Regents University, Medical College of Georgia, Augusta, Georgia 30912; and Department of Nutritional Sciences (A.E.-S.), University of Toronto, Toronto, Canada M5S 3E2
| | | | | | | |
Collapse
|
25
|
Mahaldashtian M, Makoolati Z, Ghorbanian MT, Naghdi M, Kouhpayeh SA. In vitro cytotoxicity effects of date palm (Phoenix dactylifera L.) pollen on neonate mouse spermatogonial stem cells. Nat Prod Res 2014; 29:578-81. [PMID: 25189835 DOI: 10.1080/14786419.2014.954115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
There is a fast growing tendency in the use of herbal remedies in developing countries. One of the traditional medicines used for male infertility treatment is date palm (Phoenix dactylifera) pollen (DPP). Isolated spermatogonial stem cells and sertoli cells using enzymatic digestion were grown in Dulbecco's modified Eagle's medium supplemented with 4% foetal bovine serum in the absence or presence of 0.06, 0.25 and 0.62 mg/mL concentrations of aqueous extract of DPP for 2 weeks. The assessment of mean number of the whole cells and the living cells showed that there were no significant differences between the mean viability percentage and proliferation rate between control and experimental groups (P>0.05). As there are no cytotoxicity effects of DPP in our cultural system, this system can be utilised for the enrichment or differentiation of these cells in clinical applications, cell replacement therapy, tissue regeneration and tissue engineering applications.
Collapse
Affiliation(s)
- Maryam Mahaldashtian
- a Department of Molecular & Cellular Biology, Faculty of Biology , Damghan University , Semnan , Iran
| | | | | | | | | |
Collapse
|
26
|
Yang L, Allred CD, Awika JM. Emerging Evidence on the Role of Estrogenic Sorghum Flavonoids in Colon Cancer Prevention. CEREAL FOOD WORLD 2014. [DOI: 10.1094/cfw-59-5-0244] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- L. Yang
- Corresponding author. Department of Soil & Crop Sciences, 2474 TAMU, Texas A&M University, College Station, TX 77843-2474, USA. Current affiliation: Kellogg Company, Global Breakfast R&D.Tel: +1.269.961.6149; Fax: +1.269.961.9107
| | - C. D. Allred
- Texas A&M University, College Station, TX, U.S.A
| | - J. M. Awika
- Texas A&M University, College Station, TX, U.S.A
| |
Collapse
|
27
|
|
28
|
Huang S, Eleniste PP, Wayakanon K, Mandela P, Eipper BA, Mains RE, Allen MR, Bruzzaniti A. The Rho-GEF Kalirin regulates bone mass and the function of osteoblasts and osteoclasts. Bone 2014; 60:235-45. [PMID: 24380811 PMCID: PMC3934571 DOI: 10.1016/j.bone.2013.12.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 12/15/2022]
Abstract
Bone homeostasis is maintained by the balance between bone resorption by osteoclasts and bone formation by osteoblasts. Dysregulation in the activity of the bone cells can lead to osteoporosis, a disease characterized by low bone mass and an increase in bone fragility and risk of fracture. Kalirin is a novel GTP-exchange factor protein that has been shown to play a role in cytoskeletal remodeling and dendritic spine formation in neurons. We examined Kalirin expression in skeletal tissue and found that it was expressed in osteoclasts and osteoblasts. Furthermore, micro-CT analyses of the distal femur of global Kalirin knockout (Kal-KO) mice revealed significantly reduced trabecular and cortical bone parameters in Kal-KO mice, compared to WT mice, with significantly reduced bone mass in 8, 14 and 36week-old female Kal-KO mice. Male mice also exhibited a decrease in bone parameters but not to the level seen in female mice. Histomorphometric analyses also revealed decreased bone formation rate in 14week-old female Kal-KO mice, as well as decreased osteoblast number/bone surface and increased osteoclast surface/bone surface. Consistent with our in vivo findings, the bone resorbing activity and differentiation of Kal-KO osteoclasts was increased in vitro. Although alkaline phosphatase activity by Kal-KO osteoblasts was increased in vitro, Kal-KO osteoblasts showed decreased mineralizing activity, as well as decreased secretion of OPG, which was inversely correlated with ERK activity. Taken together, our findings suggest that deletion of Kalirin directly affects osteoclast and osteoblast activity, leading to decreased OPG secretion by osteoblasts which is likely to alter the RANKL/OPG ratio and promote osteoclastogenesis. Therefore, Kalirin may play a role in paracrine and/or endocrine signaling events that control skeletal bone remodeling and the maintenance of bone mass.
Collapse
Affiliation(s)
- Su Huang
- Department of Oral Biology, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Pierre P Eleniste
- Department of Oral Biology, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Kornchanok Wayakanon
- Department of Oral Biology, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Prashant Mandela
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Angela Bruzzaniti
- Department of Oral Biology, Indiana University School of Dentistry, Indianapolis, IN, USA; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
29
|
Faloppa CC, Baiocchi G, Cunha IW, Fregnani JHTG, Osorio CABT, Fukazawa EM, Kumagai LY, Badiglian-Filho L, Pinto GLS, Soares FA. NF-κB and COX-2 expression in nonmalignant endometrial lesions and cancer. Am J Clin Pathol 2014; 141:196-203. [PMID: 24436266 DOI: 10.1309/ajcpv7u7pghoweqg] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVES To examine the immunohistochemical expression of cyclooxygenase-2 (COX-2) and nuclear factor-κB (NF-κB) in benign endometrial polyps (EPs), endometrial hyperplasia (EH), endometrial intraepithelial neoplasia (EIN), and endometrioid endometrial cancer (EC). METHODS The immunohistochemical expression of COX-2 and NF-κB was performed using an Aperio Scanscope XT automated system in 218 patients with endometrioid EC and 107 patients with nonmalignant endometrial lesions: 53 with benign EPs, 37 with EH, and 17 with EIN. RESULTS COX-2 and NF-κB p50 expression were significantly lower in EC compared with nonmalignant lesions. We observed significant decreased NF-κB p65 expression in EC vs EPs (P < .001) and EH (P = .014) as well as in EIN vs. EPs (P = .01). For patients with EC, COX-2 correlated positively with NF-κB p65 and NF-κB p50 (P < .001). Grade 3 tumors had a higher mean expression of NF-κB p65 (P = .03). NF-κB p50, NF-κB p65, and COX-2 expression had no impact on survival. CONCLUSIONS We conclude that COX-2 and NF-κB expression are lower in EC compared with nonmalignant endometrial lesions. COX-2 and NF-κB expression have no prognostic value in EC.
Collapse
Affiliation(s)
| | - Glauco Baiocchi
- Departments of Gynecologic Oncology, AC Camargo Cancer Hospital, São Paulo, Brazil
| | | | | | | | - Elza Mieko Fukazawa
- Departments of Gynecologic Oncology, AC Camargo Cancer Hospital, São Paulo, Brazil
| | - Lillian Yuri Kumagai
- Departments of Gynecologic Oncology, AC Camargo Cancer Hospital, São Paulo, Brazil
| | | | | | | |
Collapse
|
30
|
Suba Z. Triple-negative breast cancer risk in women is defined by the defect of estrogen signaling: preventive and therapeutic implications. Onco Targets Ther 2014; 7:147-64. [PMID: 24482576 PMCID: PMC3905095 DOI: 10.2147/ott.s52600] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Epidemiologic studies strongly support that triple-negative breast cancers (TNBCs) may be distinct entities as compared with estrogen receptor (ER)+ tumors, suggesting that the etiologic factors, clinical characteristics, and therapeutic possibilities may vary by molecular subtypes. Many investigations propose that reproductive factors and exogenous hormone use differently or even quite inversely affect the risk of TNBCs and ER+ cancers. Controversies concerning the exact role of even the same risk factor in TNBC development justify that the biological mechanisms behind the initiation of both TNBCs and non-TNBCs are completely obscure. To arrive at a comprehensive understanding of the etiology of different breast cancer subtypes, we should also reconsider our traditional concepts and beliefs regarding cancer risk factors. Malignancies are multicausal, but the disturbance of proper estrogen signaling seems to be a crucial risk factor for the development of mammary cancers. The grade of defect in metabolic and hormonal equilibrium is directly associated with TNBC risk for women during their whole life. Inverse impact of menopausal status or parity on the development of ER+ and ER− breast cancers may not be possible; these controversial results derive from the misinterpretation of percentage-based statistical evaluations. Exogenous or parity-associated excessive estrogen supply is suppressive against breast cancer, though the lower the ER expression of tumors, the weaker the anticancer capacity. In women, the most important preventive strategy against breast cancers – included TNBCs – is the strict control and maintenance of hormonal equilibrium from early adolescence through the whole lifetime, particularly during the periods of great hormonal changes.
Collapse
Affiliation(s)
- Zsuzsanna Suba
- National Institute of Oncology, Surgical and Molecular Tumor Pathology Centre, Budapest, Hungary
| |
Collapse
|
31
|
Koutsogiannaki S, Franzellitti S, Fabbri E, Kaloyianni M. Oxidative stress parameters induced by exposure to either cadmium or 17β-estradiol on Mytilus galloprovincialis hemocytes. The role of signaling molecules. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 146:186-195. [PMID: 24316436 DOI: 10.1016/j.aquatox.2013.11.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/24/2013] [Accepted: 11/07/2013] [Indexed: 06/02/2023]
Abstract
The aim of the present study was to determine and compare the possible effects of exposure to an estrogen, 17β-estradiol and to a metal, cadmium on oxidative parameters of Mytilus galloprovincialis hemocytes and to elucidate the signaling pathways that probably mediate the studied effects exerted by these two chemicals. In addition, it was of interest to investigate if the studied parameters could constitute biomarkers for aquatic pollution monitoring. Our results suggest that micromolar concentrations of either cadmium or 17β-estradiol affected the redox status of mussels by modulating oxidative parameters and antioxidant enzymes gene expression in mussel M. galloprovincialis hemocytes. In particular, our results showed that treatment of hemocytes with either 5 μM of cadmium chloride or with 25 nM of 17β-estradiol for 30 min caused significant increased ROS production; this led to oxidative damage exemplified by significant increased DNA damage, protein carbonylation and lipid peroxidation, as well as increased mRNA levels of the antioxidant enzymes catalase (CAT), superoxide dismoutase (SOD) and glutathione S-transferase (GST). Furthermore, our results suggest that either cadmium or 17β-estradiol signal is mediated either through one of the already known pathways initiated by photatidyl-inositol 3-kinase (PI3K) and reaching Na(+)/H(+) exchanger (NHE) probably through protein kinase C (PKC) or a kinase-mediated signaling pathway that involves in most of the cases NHE, PKC, Ca(2+)-dependent PKC isoforms, PI3-K, NADPH oxidase, nitric oxide (NO) synthase, c-Jun N-terminal kinase (JNK) and cyclic adenosine-3'-5'-monophosphate (cAMP). Our results also attribute a protective role to cAMP, since pre-elevated intracellular cAMP levels inhibited the signal induced by each exposure. Finally, since aquatic invertebrates have been the most widely used monitoring organisms for pollution impact evaluation in marine environments and taking under consideration the positive correlation obtained between the studied parameters, we can suggest the simultaneous use of these oxidative stress parameters offering an effective early warning system in biomonitoring of aquatic environments.
Collapse
Affiliation(s)
- Sophia Koutsogiannaki
- Laboratory of Animal Physiology, Zoology Department, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Silvia Franzellitti
- University of Bologna, Interdepartment Centre for Environmental Science Research, via S. Alberto 163, 48123 Ravenna, Italy
| | - Elena Fabbri
- University of Bologna, Interdepartment Centre for Environmental Science Research, via S. Alberto 163, 48123 Ravenna, Italy; University of Bologna, Department of Biological, Geological, and Environmental Sciences, via Selmi 3, 40100 Bologna, Italy
| | - Martha Kaloyianni
- Laboratory of Animal Physiology, Zoology Department, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| |
Collapse
|
32
|
Börjesson AE, Lagerquist MK, Windahl SH, Ohlsson C. The role of estrogen receptor α in the regulation of bone and growth plate cartilage. Cell Mol Life Sci 2013; 70:4023-37. [PMID: 23516016 PMCID: PMC11114058 DOI: 10.1007/s00018-013-1317-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/07/2013] [Accepted: 03/04/2013] [Indexed: 02/02/2023]
Abstract
Estrogens are important endocrine regulators of skeletal growth and maintenance in both females and males. Studies have demonstrated that the estrogen receptor (ER)-α is the main mediator of these estrogenic effects in bone. Therefore, estrogen signaling via ERα is a target both for affecting longitudinal bone growth and bone remodeling. However, treatment with estradiol (E2) leads to an increased risk of side effects such as venous thromboembolism and breast cancer. Thus, an improved understanding of the signaling pathways of ERα will be essential in order to find better bone specific treatments with minimal adverse effects for different estrogen-related bone disorders. This review summarizes the recent data regarding the intracellular signaling mechanisms, in vivo, mediated by the ERα activation functions (AFs), AF-1 and AF-2, and the effect on bone, growth plate and other estrogen responsive tissues. In addition, we review the recent cell-specific ERα-deleted mouse models lacking ERα specifically in neuronal cells or growth plate cartilage. The newly characterized signaling pathways of estrogen, described in this review, provide a better understanding of the ERα signaling pathways, which may facilitate the design of new, bone-specific treatment strategies with minimal adverse effects.
Collapse
Affiliation(s)
- A. E. Börjesson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - M. K. Lagerquist
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S. H. Windahl
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - C. Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
33
|
Zuo W, Zhang W, Chen NH. Sexual dimorphism in cerebral ischemia injury. Eur J Pharmacol 2013; 711:73-9. [PMID: 23652162 DOI: 10.1016/j.ejphar.2013.04.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 04/24/2013] [Accepted: 04/26/2013] [Indexed: 12/19/2022]
Abstract
Stroke is a leading cause of permanent disability and death. A complex series of biochemical and molecular mechanisms (e.g. the release of ROS/NOS, proapoptotic proteins and proinflammatory cytokine; neuronal depolarization, Ca2+ accumulation and so on) impair the neurologic functions of cerebral ischemia and stroke. We have known for some time that the epidemiology of human stroke is sexually dimorphic until late in life, well beyond the years of reproductive senescence and menopause. The principal mammalian estrogen (17β estradiol or E2) is neuroprotective in many types of brain injury and has been the major focus of investigation over the past several decades. However the incidence of stroke in women is lower than in men until decades past menopause, suggesting that factors beyond sex hormone contribute to these epidemiological sex differences. So a new concept is emerging: both sex steroids and biologic sex are important factors in clinical and experimental strokes. In this review, we will address sex steroids and gender differences in influencing the mechanisms and outcomes of brain ischemia stroke. These sex differences need to be identified which could help future translation to human neuroprotection.
Collapse
Affiliation(s)
- Wei Zuo
- Key Laboratory of Bioactive Substances and Resources Utilization, Ministry of Education, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | | | | |
Collapse
|
34
|
Solar P, Velasquez L. Consequences of nongenomic actions of estradiol on pathogenic genital tract response. J Mol Signal 2013; 8:1. [PMID: 23351368 PMCID: PMC3570385 DOI: 10.1186/1750-2187-8-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 01/24/2013] [Indexed: 12/26/2022] Open
Abstract
Estradiol is a steroid hormone that regulates the structure and function of the female reproductive system. In addition to its genomic effects, which are mediated by activated nuclear receptors, estradiol elicits a variety of rapid signaling events independently of transcriptional or genomic regulation. These nongenomic actions influence the milieu of the genital tract, which changes the ability of pathogens to infect the genital tract. This review discusses our current knowledge regarding the mechanisms and relevance of nongenomic estradiol signaling in the genital tract that could change the ability of pathogens to invade epithelial cells. PubMed was searched through January 1980 for papers related to estradiol actions in the ovary, fallopian tube, uterus and cervix. The mechanisms conveying these rapid effects consist of a multitude of signaling molecules and include cross-talk with slower transcriptional actions. The nongenomic actions of estradiol that influence the infectious abilities of pathogens occur either directly on the genital tract cells or indirectly by modulating the local and systemic immune systems. Additional in-depth characterization of the response is required before the normal and pathological reproductive functions of the nongenomic estradiol pathway can be targeted for pharmacological intervention.
Collapse
Affiliation(s)
- Paula Solar
- Center for Integrative Medicine and Innovative Sciences, Facultad de Medicina, Universidad Andrés Bello, Echaurren 183, Santiago, Chile.
| | | |
Collapse
|
35
|
Shi H, Kumar SPDS, Liu X. G protein-coupled estrogen receptor in energy homeostasis and obesity pathogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 114:193-250. [PMID: 23317786 PMCID: PMC3632385 DOI: 10.1016/b978-0-12-386933-3.00006-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity and its related metabolic diseases have reached a pandemic level worldwide. There are sex differences in the prevalence of obesity and its related metabolic diseases, with men being more vulnerable than women; however, the prevalence of these disorders increases dramatically in women after menopause, suggesting that sex steroid hormone estrogens play key protective roles against development of obesity and metabolic diseases. Estrogens are important regulators of several aspects of metabolism, including body weight and body fat, caloric intake and energy expenditure, and glucose and lipid metabolism in both males and females. Estrogens act in complex ways on their nuclear estrogen receptors (ERs) ERα and ERβ and transmembrane ERs such as G protein-coupled estrogen receptor. Genetic tools, such as different lines of knockout mouse models, and pharmacological agents, such as selective agonists and antagonists, are available to study function and signaling mechanisms of ERs. We provide an overview of the evidence for the physiological and cellular actions of ERs in estrogen-dependent processes in the context of energy homeostasis and body fat regulation and discuss its pathology that leads to obesity and related metabolic states.
Collapse
Affiliation(s)
- Haifei Shi
- Department of Biology, Center for Physiology and Neuroscience, Miami University, Oxford, Ohio, USA
| | | | | |
Collapse
|
36
|
Gender and estradiol as major factors in the expression and dimerization of nNOSα in rats with experimental diabetic gastroparesis. Dig Dis Sci 2012; 57:2814-25. [PMID: 22684582 DOI: 10.1007/s10620-012-2230-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 05/01/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND The molecular mechanisms of cellular changes responsible for diabetic gastroparesis, primarily seen in middle-aged women, still remain incompletely defined. We hypothesized that a decrease in the expression, dimerization, and post-translational modification of neuronal nitric oxide synthase alpha (nNOSα) is estrogen mediated and responsible for the gender-specific prevalence of this malady. METHODS We induced diabetes by injecting male and female rats with streptozotocin. Male diabetic rats without gastroparesis were then injected with estrogen for 3 weeks and evaluated for gastroparesis development. Gastric tissues were analyzed for the elucidation of biochemical events associated with diabetes and gastroparetic dysfunction. RESULTS Although male diabetic, gastroparetic (either streptozotocin- or estrogen-induced) rats exhibited similarity in disease pathology to that of females, the molecular mechanisms of development were different. Our results indicate that slow gastric emptying in both male diabetic, gastroparetic rat groups was not associated with the level of expression of nNOSα in gastric tissues. However, nNOSα dimerization, which reflects nNOSα activation, did decline slightly in the antrum of diabetic males with estrogen-induced gastroparesis, suggesting a possible estrogen role. Females with diabetic gastroparesis, in contrast, demonstrated significantly impaired levels and dimerization of nNOSα in the antrum and pylorus. Although the precise mechanism remains unknown, nNOSα dimerization impairment in female antrum is apparently associated with reduced phosphorylation of Ser(1416) in the activation loop of nNOSα. CONCLUSION Taken together, these results demonstrate that gender and estrogens may be leading factors, through molecular changes involved in nitric oxide synthesis down-regulation, within the antrum and pylorus of female diabetic, gastroparetic rats.
Collapse
|
37
|
Yang SH, Liao CC, Chen Y, Syu JP, Jeng CJ, Wang SM. Daidzein induces neuritogenesis in DRG neuronal cultures. J Biomed Sci 2012; 19:80. [PMID: 22931352 PMCID: PMC3500655 DOI: 10.1186/1423-0127-19-80] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 08/21/2012] [Indexed: 11/10/2022] Open
Abstract
Absract
Collapse
Affiliation(s)
- Shih-Hung Yang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
38
|
Fürst RW, Kliem H, Meyer HHD, Ulbrich SE. A differentially methylated single CpG-site is correlated with estrogen receptor alpha transcription. J Steroid Biochem Mol Biol 2012; 130:96-104. [PMID: 22342840 DOI: 10.1016/j.jsbmb.2012.01.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 01/17/2012] [Accepted: 01/21/2012] [Indexed: 12/16/2022]
Abstract
DNA methylation of the promoter region of estrogen receptor alpha (ESR1) is recognized as an epigenetic mechanism that regulates its mRNA abundance. We questioned whether tissues in male growing piglets were influenced in terms of DNA methylation by the developmentally occurring distinct plasma estradiol-17β (E2) concentrations. Additionally, we aimed at broadening the currently limited understanding of the epigenetic regulation of ESR1 in physiological settings. Three distinct genetic regions of ESR1 were analyzed using a combination of methylation-sensitive high resolution melting (MS-HRM) and pyrosequencing. Unexpectedly, major E2 concentration differences were only marginally associated with minor variations in DNA methylation and mRNA abundance. However, by analyzing two tissues showing the greatest differences in transcript abundance, we were able to find one single CpG site in the +1kb intragenic region of ESR1 strikingly differently methylated between heart vs. epididymis. Interestingly, this single CpG-site was identified as a putative binding site for the transcriptional repressor TG-interacting factor 1 (TGIF) which can recruit histone deacetylase 1 (HDAC1) leading to chromatin condensation. Indeed, chromatin immunoprecipitation confirmed a reduced histone H3 presence at the specific ESR1 location in case of higher DNA methylation. We therefore hypothesize that ESR1 expression may be manifested by a single-CpG-site based methylation difference impairing transcription factor binding.
Collapse
Affiliation(s)
- Rainer W Fürst
- Physiology Weihenstephan, Technische Universität München, 85354 Freising-Weihenstephan, Germany
| | | | | | | |
Collapse
|
39
|
Abstract
According to current knowledge, it must be assumed that temporary idiopathic hearing loss and its spontaneous remission are based on mechanical and/or pathological alterations in the inner ear. The causal mechanisms might be based on inter-individual variations. Induced by dose-dependent activators, temporary as well as permanent damage might occur. Sudden hearing loss may be initiated by an increase in the local nitric oxide (NO) concentration. Spontaneous remission, i.e. functional restoration, can be explained by a local decrease in the NO concentration. In this context, regulatory systems such as the gap-junction system, blood vessels or synapses might be affected. In addition, alterations in the hormone level of estrogen and mineralocorticoids, as well as cellular glutathione and vitamin levels, might lead to temporary alterations in the inner ear. Recent experimental findings indicate a role for the shuttle protein Survivin in the spontaneous remission of sudden hearing loss.
Collapse
|
40
|
Zhang XT, Ding L, Kang LG, Wang ZY. Involvement of ER-α36, Src, EGFR and STAT5 in the biphasic estrogen signaling of ER-negative breast cancer cells. Oncol Rep 2012; 27:2057-65. [PMID: 22426783 DOI: 10.3892/or.2012.1722] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 02/10/2012] [Indexed: 01/27/2023] Open
Abstract
It is well established that estrogen is a potent mitogen in cells expressing estrogen receptors (ER). However, a large body of evidence has demonstrated that the effects of mitogenic estrogen signaling exhibit a non-monotonic or biphasic, dose-response curve; estrogen at low concentrations, elicits a mitogenic signaling pathway to stimulate cell proliferation, while at high concentrations, estrogen inhibits cell growth. The molecular mechanism underlying this paradoxical effect of estrogen on cell proliferation remains largely unknown. Recently, we reported that ER-α36, a variant of ER-α, mediates mitogenic estrogen signaling in ER-negative breast cancer cells. Here, we investigated the molecular mechanisms underlying the biphasic estrogen signaling in MDA-MB-231 and MDA-MB-436 ER-negative breast cancer cells. We found that 17β-estradiol (E2β) at l nM induced the phosphorylation of Src-Y416, an event that activates Src, while at 5 µM failed to induce Src-Y416 phosphorylation but induced Src-Y527 phosphorylation an event that inactivates Src. E2β at 1 nM, but not at 5 µM, also induced phosphorylation of MAPK/ERK and activated Cyclin D1 promoter activity through the Src/EGFR/STAT5 pathway. Knockdown of ER‑α36 abrogated the biphasic estrogen signaling in these cells. Our results thus indicate that in ER-negative breast cancer cells Src functions as a switch in ER‑α36-mediated biphasic estrogen signaling through the EGFR/STAT5 pathway.
Collapse
Affiliation(s)
- Xin-Tian Zhang
- Department of Medical Microbiology and Immunology, Creighton University Medical School, Omaha, NE 68178, USA
| | | | | | | |
Collapse
|
41
|
Yang L, Allred KF, Geera B, Allred CD, Awika JM. Sorghum phenolics demonstrate estrogenic action and induce apoptosis in nonmalignant colonocytes. Nutr Cancer 2012; 64:419-27. [PMID: 22369068 DOI: 10.1080/01635581.2012.657333] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Evidence indicates sorghum may be protective against colon cancer; however, the mechanisms are unknown. Estrogen is believed to protect against colon cancer development by inducing apoptosis in damaged nonmalignant colonocytes. Three sorghum extracts (white, red, and black) were screened for estrogenic activity using cell models expressing estrogen receptor α (ER-α; MCF-7 breast cancer cells) and β [ER-β; nonmalignant young adult mouse colonocytes (YAMC)]. Black and white sorghum extracts had significant estrogenic activity mediated through both estrogen receptors at 1-5 and 5-10 μg/mL, respectively; but red sorghum did not. Activation of ER-β in YAMC reduced cell growth via induction of apoptosis. Only the black and red sorghums contained 3-deoxyanthocyanins; however, these compounds were non-estrogenic. Flavones with estrogenic properties, luteolin (0.41-2.12 mg/g) and apigenin (1.1-1.4 mg/g), and their O-methyl derivatives (0.70-0.95 mg/g) were detected in white and black sorghums, but not in the red sorghum. On the other hand, naringenin, a flavanone known to interfere with transcriptional activities of estrogen, was only detected in the red sorghum extract (as its 7-O-glycoside) at relatively high concentration (11.8 mg/g). Sorghum flavonoid composition has important implications on possible modes of chemoprotection by sorghum against colon carcinogenesis.
Collapse
Affiliation(s)
- Liyi Yang
- Cereal Quality Laboratory, Soil & Crop Science Department, Texas A&M University, College Station, Texas 77843-2474, USA
| | | | | | | | | |
Collapse
|
42
|
Suba Z, Kásler M. [Interactions of insulin and estrogen in the regulation of cell proliferation and carcinogenesis]. Orv Hetil 2012; 153:125-36. [PMID: 22257509 DOI: 10.1556/oh.2012.29287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Equilibrium of sexual steroids and metabolic processes has close correlations. Insulin is a potent regulator of human sexual steroid hormone production and modulates their signals at receptor level. Insulin resistance and excessive insulin production provoke hyperandrogenism and estrogen deficiency in women resulting not only in anovulatory dysfunction but also a high risk for cardiovascular diseases and cancer. Physiologic functions of all female organs have higher estrogen demand as compared with men. In healthy women estrogen predominance against androgens is a favor in their reproductive period, which means a strong defense against insulin resistance and its complications. However, in postmenopausal cases the increasing prevalence of insulin resistance and type-2 diabetes associated with estrogen deficiency and androgen excess, result in a gender specific higher risk for precancerous lesions and cancer as compared with men. Estrogen has beneficial effect on the energy metabolism, glucose homeostasis and on the lipid metabolism of liver and of peripheral tissues as well. A moderate or severe decrease in serum estrogen level enhances the prevalence of insulin resistant states. In premenopausal women long or irregular menstrual cycles are predictors for the risk of insulin resistance and type-2 diabetes. Moreover, in postmenopausal estrogen deficient cases elevated fasting glucose, increased body weight and abdominal fat deposition are often observed progressively with age in correlation with an impaired glucose tolerance. In the rare cases of estrogen deficient men severe type-2 diabetes seems to be a characteristic complication. Upon becoming familiar with the cancer risk of insulin resistance and estrogen deficiency, there would be plenty of possibilities for primary cancer prevention. In patients with cancer the treatment of hormonal and metabolic disturbances may become effective adjuvant therapy.
Collapse
Affiliation(s)
- Zsuzsanna Suba
- Országos Onkológiai Intézet Sebészeti és Molekuláris Tumorpatológiai Osztály Budapest Ráth György u.
| | | |
Collapse
|
43
|
Estrogen receptor-alpha 36 mediates mitogenic antiestrogen signaling in ER-negative breast cancer cells. PLoS One 2012; 7:e30174. [PMID: 22276155 PMCID: PMC3261853 DOI: 10.1371/journal.pone.0030174] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 12/14/2011] [Indexed: 11/23/2022] Open
Abstract
It is prevailingly thought that the antiestrogens tamoxifen and ICI 182, 780 are competitive antagonists of the estrogen-binding site of the estrogen receptor-alpha (ER-α). However, a plethora of evidence demonstrated both antiestrogens exhibit agonist activities in different systems such as activation of the membrane-initiated signaling pathways. The mechanisms by which antiestrogens mediate estrogen-like activities have not been fully established. Previously, a variant of ER-α, EP–α36, has been cloned and showed to mediate membrane-initiated estrogen and antiestrogen signaling in cells only expressing ER-α36. Here, we investigated the molecular mechanisms underlying the antiestrogen signaling in ER-negative breast cancer MDA-MB-231 and MDA-MB-436 cells that express high levels of endogenous ER-α36. We found that the effects of both 4-hydoxytamoxifen (4-OHT) and ICI 182, 780 (ICI) exhibited a non-monotonic, or biphasic dose response curve; antiestrogens at low concentrations, elicited a mitogenic signaling pathway to stimulate cell proliferation while at high concentrations, antiestrogens inhibited cell growth. Antiestrogens at l nM induced the phosphorylation of the Src-Y416 residue, an event to activate Src, while at 5 µM induced Src-Y527 phosphorylation that inactivates Src. Antiestrogens at 1 nM also induced phosphorylation of the MAPK/ERK and activated the Cyclin D1 promoter activity through the Src/EGFR/STAT5 pathways but not at 5 µM. Knock-down of ER-α36 abrogated the biphasic antiestrogen signaling in these cells. Our results thus indicated that ER-α36 mediates biphasic antiestrogen signaling in the ER-negative breast cancer cells and Src functions as a switch of antiestrogen signaling dependent on concentrations of antiestrogens through the EGFR/STAT5 pathway.
Collapse
|
44
|
Suba Z. Interplay between insulin resistance and estrogen deficiency as co- activators in carcinogenesis. Pathol Oncol Res 2011; 18:123-33. [PMID: 21984197 DOI: 10.1007/s12253-011-9466-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 09/29/2011] [Indexed: 02/06/2023]
Abstract
Both insulin resistance and estrogen deficiency result in complex metabolic disorder based mainly on defective cellular glucose uptake and on an atherogenic serum lipid profile. These alterations may be regarded as high risks for several life-threatening human diseases, such as type-2 diabetes, cardiovascular lesions and malignancies. Insulin resistance and estrogen deficiency are concomitant disorders with mutual interrelationship. Insulin resistance and the compensatory hyperinsulinemia provoke increased androgen synthesis at the expense of decreased estrogen production. Similarly, a moderate or severe decrease in serum estrogen levels enhances the prevalence of insulin resistant states both in men and women. Healthy premenopausal women enjoy the defensive effect of estrogens against metabolic and hormonal disorders. However, even a slight decrease in their circulatory estrogen levels associated with insulin resistance may increase the risk for cancers, particularly in the organs having high estrogen demand (breast, endometrium and ovary). On the other hand, postmenopausal state with profound estrogen deficiency confers high risk for cancers in different organs with either high or moderate estrogen demand. After menopause, hormone replacement therapy improves insulin sensitivity and decreases the enhanced inclination to malignancies in postmenopausal women. Recognition of the thorough interplay between insulin resistance and estrogen deficiency may illuminate many apparently controversial experimental and clinical findings concerning cancer development and therapeutic possibilities. Moreover, their interactions in the initiation and progression of human malignancies may supply new strategies in primary cancer prevention and cancer cure.
Collapse
Affiliation(s)
- Zsuzsanna Suba
- National Institute of Oncology, 1122 Ráth György u. 7-9, Budapest, Hungary.
| |
Collapse
|
45
|
Chen J, Liu L, Hou R, Shao Z, Wu Y, Chen X, Zhou L. Calycosin promotes proliferation of estrogen receptor-positive cells via estrogen receptors and ERK1/2 activation in vitro and in vivo. Cancer Lett 2011; 308:144-51. [DOI: 10.1016/j.canlet.2011.04.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 04/21/2011] [Accepted: 04/28/2011] [Indexed: 12/23/2022]
|
46
|
Abstract
One segment of the population that is particularly inclined to liver fat accumulation is postmenopausal women. Although nonalcoholic hepatic steatosis is more common in men than in women, after menopause there is a reversal in gender distribution. At the present time, weight loss and exercise are regarded as first line treatments for NAFLD in postmenopausal women, as it is the case for the management of metabolic syndrome. In recent years, there has been substantial evidence coming mostly from the use of the animal model, that indeed estrogens withdrawal is associated with modifications of molecular markers favouring the activity of metabolic pathways ultimately leading to liver fat accumulation. In addition, the use of the animal model has provided physiological and molecular evidence that exercise training provides estrogens-like protective effects on liver fat accumulation and its consequences. The purpose of the present paper is to present information relative to the development of a state of NAFLD resulting from the absence of estrogens and the role of exercise training, emphasizing on the contribution of the animal model on these issues.
Collapse
|
47
|
NAFLD, Estrogens, and Physical Exercise: The Animal Model. J Nutr Metab 2011; 2012:914938. [PMID: 21845221 PMCID: PMC3154523 DOI: 10.1155/2012/914938] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 06/05/2011] [Indexed: 12/17/2022] Open
Abstract
One segment of the population that is particularly inclined to liver fat accumulation is postmenopausal women. Although nonalcoholic hepatic steatosis is more common in men than in women, after menopause there is a reversal in gender distribution. At the present time, weight loss and exercise are regarded as first line treatments for NAFLD in postmenopausal women, as it is the case for the management of metabolic syndrome. In recent years, there has been substantial evidence coming mostly from the use of the animal model, that indeed estrogens withdrawal is associated with modifications of molecular markers favouring the activity of metabolic pathways ultimately leading to liver fat accumulation. In addition, the use of the animal model has provided physiological and molecular evidence that exercise training provides estrogens-like protective effects on liver fat accumulation and its consequences. The purpose of the present paper is to present information relative to the development of a state of NAFLD resulting from the absence of estrogens and the role of exercise training, emphasizing on the contribution of the animal model on these issues.
Collapse
|
48
|
Zhao Y, Deng C, Lu W, Xiao J, Ma D, Guo M, Recker RR, Gatalica Z, Wang Z, Xiao GG. let-7 microRNAs induce tamoxifen sensitivity by downregulation of estrogen receptor α signaling in breast cancer. Mol Med 2011; 17:1233-41. [PMID: 21826373 DOI: 10.2119/molmed.2010.00225] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Accepted: 07/27/2011] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) play an important regulatory role in breast tumorigenesis. Previously, we found that let-7 miRNAs were downregulated significantly in formalin-fixed paraffin-embedded (FFPE) breast cancer tissues. In this study, we further found that endogenous levels of let-7b and let-7i miRNAs are inversely correlated with levels of estrogen receptor (ER)-a36, a new variant of ER-α66, in the FFPE tissue set. Bioinformatic analysis suggested that ER-α36 may be another target of let-7 miRNAs. To test this hypothesis, cotransfection of let-7 mimics or inhibitors together with full-length or a fragment of ER-α36 3'UTR luciferase construct was performed, and we found that let-7b and let-7i mimics suppressed the activity of reporter gene significantly, which was enhanced remarkably by let-7b and let-7i inhibitors. Both mRNA and protein expression of ER-α36 were inhibited by let-7 mimics and enhanced by let-7 inhibitors. Furthermore, ER-α36 mediated nongenomic MAPK and Akt pathways were weakened by let-7b and let-7i mimics in triple negative breast cancer cell line MDA-MB-231. The reverse correlation between let-7 miRNAs and ER-α36 also exists in Tamoxifen (Tam)-resistant MCF7 cell line. Transfection of let-7 mimics to Tam-resistant MCF7 cells downregulated ER-α36 expression and enhanced the sensitivity of MCF7 cells to Tam in estrogen-free medium, which could be restored by overexpression of ER-α36 constructs without 3'UTR. Our results suggested a novel regulatory mechanism of let-7 miRNAs on ER-α36 mediated nongenomic estrogen signal pathways and Tam resistance.
Collapse
Affiliation(s)
- Yingchun Zhao
- Genomics and Functional Proteomics Laboratories, Osteoporosis Research Center, Omaha, Nebraska, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Danesh SM, Kundu P, Lu R, Stefani E, Toro L. Distinct transcriptional regulation of human large conductance voltage- and calcium-activated K+ channel gene (hSlo1) by activated estrogen receptor alpha and c-Src tyrosine kinase. J Biol Chem 2011; 286:31064-71. [PMID: 21757754 DOI: 10.1074/jbc.m111.235457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Estrogen receptor α (ERα) regulates gene transcription via "genomic" (binding directly or indirectly, typically via Sp1 or AP-1 sites, to target genes) and/or "nongenomic" (signaling) mechanisms. ERα activation by estrogen up-regulates the murine Ca(2+)-activated K(+) channel α subunit gene (mSlo1) via genomic mechanisms. Here, we investigated whether ERα also drives transcription of the human (hSlo1) gene. Consistent with this view, estrogen increased hSlo1 transcript levels in primary human smooth muscle cells. Promoter studies revealed that estrogen/hERα-mediated hSlo1 transcription was nearly 6-fold more efficient than for mSlo1 (EC(50), 0.07 versus 0.4 nM). Unlike the genomic transcriptional mechanism employed by mSlo1, hSlo1 exhibits a nongenomic hERα-mediated regulatory mechanism. This is supported by the following: 1) efficient hSlo1 transcription after disruption of the DNA-binding domain of hERα or knockdown of Sp1, and 2) lack of AP-1 sites in the hSlo1 promoter. Three nongenomic signaling pathways were explored: Src, Rho, and PI3K. Inhibition of Src with 10 μM PP2, and reported downstream ERK with 25 μM PD98059 did not prevent estrogen action but caused an increase in hSlo1 basal transcription; conversely, constitutively active c-Src (Y527F) decreased hSlo1 basal transcription even preventing its estrogen/hERα-mediated transcriptional activation. Rho inhibition by coexpressed Clostridium botulinum C3 transferase did not alter estrogen action. In contrast, inhibition of PI3K activity with 10 μM LY294002 decreased estrogen-stimulated hSlo1 transcription by ∼40%. These results indicate that the nongenomic PI3K signaling pathway plays a role in estrogen/hERα-stimulated hSlo1 gene expression; whereas c-Src activity leads to hSlo1 gene tonic repression independently of estrogen, likely through ERK activation.
Collapse
Affiliation(s)
- Shahab M Danesh
- Division of Molecular Medicine, Department of Anesthesiology, UCLA, Los Angeles, California 90095-1778, USA
| | | | | | | | | |
Collapse
|
50
|
Bayne S, Li H, Jones MEE, Pinto AR, van Sinderen M, Drummond A, Simpson ER, Liu JP. Estrogen deficiency reversibly induces telomere shortening in mouse granulosa cells and ovarian aging in vivo. Protein Cell 2011; 2:333-46. [PMID: 21574023 DOI: 10.1007/s13238-011-1033-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 03/21/2011] [Indexed: 12/31/2022] Open
Abstract
Estrogen is implicated as playing an important role in aging and tumorigenesis of estrogen responsive tissues; however the mechanisms underlying the mitogenic actions of estrogen are not fully understood. Here we report that estrogen deficiency in mice caused by targeted disruption of the aromatase gene results in a significant inhibition of telomerase maintenance of telomeres in mouse ovaries in a tissue-specific manner. The inhibition entails a significant shortening of telomeres and compromised proliferation in the follicular granulosa cell compartment of ovary. Gene expression analysis showed decreased levels of proto-oncogene c-Myc and the telomerase catalytic subunit, telomerase reverse transcriptase (TERT), in response to estrogen deficiency. Estrogen replacement therapy led to increases in TERT gene expression, telomerase activity, telomere length and ovarian tissue growth, thereby reinstating ovary development to normal in four weeks. Our data demonstrate for the first time that telomere maintenance is the primary mechanism mediating the mitogenic effect of estrogen on ovarian granulosa cell proliferation by upregulating the genes of c-Myc and TERT in vivo. Estrogen deficiency or over-activity may cause ovarian tissue aging or tumorigenesis, respectively, through estrogen regulation of telomere remodeling.
Collapse
Affiliation(s)
- Sharyn Bayne
- Department of Immunology, Central Eastern Clinical School, Monash University, Victoria 3800, Australia
| | | | | | | | | | | | | | | |
Collapse
|