1
|
Hegner P, Lebek S, Maier LS, Arzt M, Wagner S. The Effect of Gender and Sex Hormones on Cardiovascular Disease, Heart Failure, Diabetes, and Atrial Fibrillation in Sleep Apnea. Front Physiol 2021; 12:741896. [PMID: 34744785 PMCID: PMC8564381 DOI: 10.3389/fphys.2021.741896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Sleep apnea is a highly prevalent disorder with increasing impact on healthcare systems worldwide. Previous studies have been conducted primarily with male subjects, and prevalence and severity of sleep apnea in women are underestimated. Recent clinical and basic science evidence increasingly points to different mechanisms in men and women with sleep-disordered breathing (SDB). SDB is associated with a variety of comorbidities, including cardiovascular disease, heart failure, diabetes, and atrial fibrillation. In this review, we discuss sex-dependent mechanisms of SDB in select associated conditions to sharpen our clinical understanding of these sex-dependent inherent differences.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
2
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Hong S, Chang J, Jeong K, Lee W. Raloxifene as a treatment option for viral infections. J Microbiol 2021; 59:124-131. [PMID: 33527314 PMCID: PMC7849956 DOI: 10.1007/s12275-021-0617-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 01/31/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused corona virus disease 2019 (COVID-19) pandemic and led to mass casualty. Even though much effort has been put into development of vaccine and treatment methods to combat COVID-19, no safe and efficient cure has been discovered. Drug repurposing or drug repositioning which is a process of investigating pre-existing drug candidates for novel applications outside their original medical indication can speed up the drug development process. Raloxifene is a selective estrogen receptor modulator (SERM) that has been approved by FDA in 1997 for treatment and prevention of postmenopausal osteoporosis and cancer. Recently, raloxifene demonstrates efficacy in treating viral infections by Ebola, influenza A, and hepatitis C viruses and shows potential for drug repurposing for the treatment of SARS-CoV-2 infection. This review will provide an overview of raloxifene's mechanism of action as a SERM and present proposed mechanisms of action in treatment of viral infections.
Collapse
Affiliation(s)
- Subin Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - JuOae Chang
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Kwiwan Jeong
- Bio-center, Gyeonggido Business & Science Accelerator, Suwon, 16229 Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| |
Collapse
|
4
|
Uehara IA, Soldi LR, Silva MJB. Current perspectives of osteoclastogenesis through estrogen modulated immune cell cytokines. Life Sci 2020; 256:117921. [DOI: 10.1016/j.lfs.2020.117921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
|
5
|
Feng C, Ji P, Luo P, Xu J. Estrogen-Mediated MicroRNA-101-3p Expression Represses Hyaluronan Synthase 2 in Synovial Fibroblasts From Idiopathic Condylar Resorption Patients. J Oral Maxillofac Surg 2019; 77:1582-1593. [PMID: 30904552 DOI: 10.1016/j.joms.2019.02.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Idiopathic condylar resorption (ICR) is an aggressive form of osteoarthritis that is frequently observed in adolescent female patients. We hypothesized that an estrogen-mediated pathway may contribute to ICR development. MATERIALS AND METHODS An enzyme-linked immunosorbent assay was used to detect the levels of estradiol (E2) and hyaluronan in synovial fluid. Immunohistochemistry, real-time polymerase chain reaction, and Western blotting were used to detect the expression of microRNAs (miRNAs) and related genes after transfection of miRNA-101-3p mimics, inhibitor, or short interfering RNA into synovial fibroblasts. Dual-luciferase activity was determined to identify the direct effect of miRNA-101-3p on hyaluronan synthase 2 (HAS2). Linear regression analysis, the nonparametric Mann-Whitney U test, the Student t test, and 1-way analysis of variance were carried out to analyze the results of each group. RESULTS The relationship between hyaluronan and E2 was negatively correlated in synovial fluid (Pearson r = -0.3179, P = .0230). Among the screened miRNAs, miRNA-101-3p was the most overexpressed in ICR. E2 mostly upregulated the expression of miRNA-101-3p at a dose of 10 nmol/L 12 hours after transfection in synovial fibroblasts of patients with ICR. However, E2 induction of miRNA-101-3p expression was significantly repressed by estrogen receptor α interference (P = 0.0286). The dual-luciferase assay showed that miRNA-101-3p regulated the expression of HAS2 by directly targeting its 3' untranslated region. CONCLUSIONS We speculate that E2 regulates HAS2 expression by targeting miRNA-101-3p in synovial fibroblasts of patients with ICR. Thus, the E2-miRNA-101-3p-HAS2 pathway might play an important role in the pathogenesis of ICR.
Collapse
Affiliation(s)
- Chi Feng
- Resident, Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ping Ji
- Professor, Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ping Luo
- Resident, Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Xu
- Resident, Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
6
|
Liu M, Ke H, Sun C, Wang G, Wang Y, Zhao G. A simple and highly selective electrochemical label-free aptasensor of 17β-estradiol based on signal amplification of bi-functional graphene. Talanta 2018; 194:266-272. [PMID: 30609530 DOI: 10.1016/j.talanta.2018.10.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 01/13/2023]
Abstract
In the present work, a convenient signal-on electrochemical label-free aptasensor for 17β-estradiol (E2), a typical steroidal hormones endocrine disrupting chemicals, was proposed. 6-mercapto-1-hexanol (MCH) self-assembled monolayer (SAM) modified Au (MCH/Au) electrode was used as the substrate electrode. Graphene is used with bi-functions, not only to adsorb E2 binding aptamer, serving as the recognition element to E2, but also to be assembled onto MCH/Au electrode when sensing E2, to controllably turn on the electron transfer (eT), and further indicate the signal to E2 concentration. With the synergistic effect of DNase I enzyme, highly sensitive detection of E2 was achieved at this aptasensing system, with a linear range from 0.07 to 10 pM and a detection limit of 50 fM. An outstanding selectivity towards E2 was proven for the sensing system by simultaneously detecting 100-fold potential co-existing interferences. The stability and reproducibility were also evaluated to be satisfactory. Spiked real water analysis further indicated its reliability and potential in practical environmental monitoring.
Collapse
Affiliation(s)
- Meichuan Liu
- School of Chemical Science and Engineering, Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Hongyang Ke
- School of Chemical Science and Engineering, Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Caiqin Sun
- School of Chemical Science and Engineering, Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Guoqiang Wang
- School of Chemical Science and Engineering, Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Wang
- School of Chemical Science and Engineering, Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Guohua Zhao
- School of Chemical Science and Engineering, Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
7
|
Song C, Li W, Leng J, Wang L, Li W, Shi F, Liu G, Zhou J, Yang X. Passive smoking and postpartum depression among Chinese women: A prospective cohort study in Tianjin, China. Women Health 2018; 59:281-293. [PMID: 29924720 DOI: 10.1080/03630242.2018.1478365] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Smoking has been associated with depression in the general population. Whether passive smoking is also associated with postpartum depression (PPD) is uncertain. From 2010 to 2012, we recruited 8,842 pregnant women in Tianjin, China. The mainland Chinese version of the Edinburgh Postnatal Depression Scale was used to evaluate postpartum depressive symptoms after birth, with a score of >9 defining PPD. Data were collected using specially designed questionnaires or data from the electronic database of Tianjin Maternal and Child Health Information System. Odds ratios (OR) and 95 percent confidence intervals (CI) were obtained for the association of smoking status with PPD using binary logistic regression. Passive smoke exposure rates before and during pregnancy were 40.9 percent and 52.1 percent, respectively. A total of 8.5% (n = 747) of participants had PPD. Compared with those not exposed, women passively exposed to smoke before and during pregnancy had higher odds of PPD (before pregnancy: OR: 1.24, 95 percent CI: 1.03-1.50; during pregnancy: OR: 1.43, 95 percent CI: 1.16-1.77) after adjustment for confounding factors. Passive smoking before and during pregnancy were associated with PPD in Chinese women. Reducing passive smoke exposure may reduce PPD in Chinese women; further longitudinal studies are warranted to replicate these findings.
Collapse
Affiliation(s)
- Chunhua Song
- a Department of Epidemiology and Biostatistics, School of Public Health , Tianjin Medical University , Tianjin , China.,b Center for Disease Control and Prevention of Tianjin Economic-Technological Development Area (TEDA) , Tianjin , China
| | - Wei Li
- c Tianjin Women and Children's Health Center , Tianjin , China
| | - Junhong Leng
- c Tianjin Women and Children's Health Center , Tianjin , China
| | - Leishen Wang
- c Tianjin Women and Children's Health Center , Tianjin , China
| | - Weiqin Li
- c Tianjin Women and Children's Health Center , Tianjin , China
| | - Fenglan Shi
- d Tianjin He Ping Women and Children's Health Centre , Tianjin , China
| | - Guifeng Liu
- e Tianjin Nan Kai Women and Children's Health Centre , Tianjin , China
| | - Juan Zhou
- f Tianjin Hong Qiao Disease Control and Prevention Center , Tianjin , China
| | - Xilin Yang
- a Department of Epidemiology and Biostatistics, School of Public Health , Tianjin Medical University , Tianjin , China
| |
Collapse
|
8
|
Xu X, Jia X, Mo L, Liu C, Zheng L, Yuan Q, Zhou X. Intestinal microbiota: a potential target for the treatment of postmenopausal osteoporosis. Bone Res 2017; 5:17046. [PMID: 28983411 PMCID: PMC5627629 DOI: 10.1038/boneres.2017.46] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Postmenopausal osteoporosis (PMO) is a prevalent metabolic bone disease characterized by bone loss and structural destruction, which increases the risk of fracture in postmenopausal women. Owing to the high morbidity and serious complications of PMO, many efforts have been devoted to its prophylaxis and treatment. The intestinal microbiota is the complex community of microorganisms colonizing the gastrointestinal tract. Probiotics, which are dietary or medical supplements consisting of beneficial intestinal bacteria, work in concert with endogenous intestinal microorganisms to maintain host health. Recent studies have revealed that bone loss in PMO is closely related to host immunity, which is influenced by the intestinal microbiota. The curative effects of probiotics on metabolic bone diseases have also been demonstrated. The effects of the intestinal microbiota on bone metabolism suggest a promising target for PMO management. This review seeks to summarize the critical effects of the intestinal microbiota and probiotics on PMO, with a focus on the molecular mechanisms underlying the pathogenic relationship between bacteria and host, and to define the possible treatment options.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyue Jia
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Longyi Mo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Dental Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Abstract
Experimental ovarian carcinogenesis has been investigated in inbred and hybrid strains of mice and induced by a diversity of mechanisms including X-irradiation, oocytotoxic xenobiotic chemicals, ovarian grafting to ectopic or orthotopic sites, neonatal thymectomy, mutant genes reducing germ cell populations, and aging. Disruptions in the function of graafian follicles by a variety of mechanisms results in a spectrum of ovarian proliferative lesions, including tumors. The findings in mutant and genetically engineered mice support the concept of a secondary (hormonally mediated) mechanism of ovarian tumorigenesis in mice associated with sterility. Multiple pathogenic factors that either destroy or diminish the numbers of graafian follicles in the ovary result in decreased sex hormone secretion, especially estradiol-17 β, leading to a compensatory overproduction of pituitary gonadotrophins, particularly LH, which places the mouse ovary at an increased risk for developing tumors in chronic studies. The intense proliferation of ovarian surface epithelium and stromal (interstitial) cells with the development of unique tubular adenomas in response to sterility does not appear to have a counterpart in the ovaries of adult human females.
Collapse
Affiliation(s)
- Charles C Capen
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio 43210, USA.
| |
Collapse
|
10
|
Simsa P, Mihalyi A, Kyama CM, Mwenda JM, Fülöp V, D'Hooghe TM. Selective Estrogen-Receptor Modulators and Aromatase Inhibitors: Promising New Medical Therapies for Endometriosis? WOMENS HEALTH 2016; 3:617-28. [DOI: 10.2217/17455057.3.5.617] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Endometriosis is an estrogen-dependent disease and estrogen-related pathways are imbalanced in women with endometriosis. One of the key enzymes in estrogen synthesis is aromatase. Inhibiting this pathway at several points is a promising idea for the treatment of endometriosis. The third generation of aromatase inhibitors is becoming more potent in efficacy, with fewer side effects than previous generations, but cotreatment with other hormones is needed to inhibit ovarian stimulation. Other components that promote estrogen synthesis such as COX-2 can also be potentially targeted. Selective estrogen-receptor modulators could also be interesting in view of their tissue-specific effect. However, all these new drugs are still in an early phase of development. At present, it is too early to conclude that aromatase inhibitors, COX-2 inhibitors or selective estrogen-receptor modulators really present any added value compared with the existing drugs that can be used to achieve hormonal suppression in the medical treatment of endometriosis.
Collapse
Affiliation(s)
- Peter Simsa
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- National Institute of Health, Budapest, Hungary
| | - Attila Mihalyi
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
| | - Cleophas M Kyama
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- Institute of Primate Research, Division of Reproduction, PO Box 24481, Karen, Nairobi, Kenya
| | | | | | - Thomas M D'Hooghe
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- Institute of Primate Research, Division of Reproduction, PO Box 24481, Karen, Nairobi, Kenya
| |
Collapse
|
11
|
Abstract
Gonadal steroids seem to regulate affective state in some people (but not all), despite the absence of abnormal steroid hormone levels or dysfunction of the reproductive endocrine axis. In this article, we attempt to explain this paradox 1) by describing the molecular mechanisms by which gonadal steroids can regulate neuronal function; 2) by describing the specific regulatory impact of gonadal steroids on two systems im plicated in the pathophysiology of mood disorders; and 3) by defining the role of gonadal steroids in several mood disorders linked to periods of reproductive change. We suggest that the context in which the neuro- regulatory actions of gonadal steroids occur determines the impact of steroid signaling on the regulation of affective state. NEUROSCIENTIST 5:227-237, 1999
Collapse
Affiliation(s)
- Catherine A. Roca
- Behavioral Endocnnology Branch National Institute of
Mental Health Bethesda, Maryland
| | - Peter J. Schmidt
- Behavioral Endocnnology Branch National Institute of
Mental Health Bethesda, Maryland
| | - David R. Rubinow
- Behavioral Endocnnology Branch National Institute of
Mental Health Bethesda, Maryland
| |
Collapse
|
12
|
Liang Y, Duan L, Xiong J, Zhu W, Liu Q, Wang D, Liu W, Li Z, Wang D. E2 regulates MMP-13 via targeting miR-140 in IL-1β-induced extracellular matrix degradation in human chondrocytes. Arthritis Res Ther 2016; 18:105. [PMID: 27165343 PMCID: PMC4863330 DOI: 10.1186/s13075-016-0997-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Estrogen deficiency is closely related to the development of menopausal arthritis. Estrogen replacement therapy (ERT) shows a protective effect against the osteoarthritis. However, the underlying mechanism of this protective effect is unknown. This study aimed to determine the role of miR-140 in the estrogen-dependent regulation of MMP-13 in human chondrocytes. METHODS Primary human articular chondrocytes were obtained from female OA patients undergoing knee replacement surgery. Normal articular chondrocytes were isolated from the knee joints of female donors after trauma and treated with interleukin-1 beta (IL-1β). Gene expression levels of miR-140, MMP-13, and ADAMTS-5 were detected by quantitative real-time PCR (qRT-PCR). miR-140 levels were upregulated or downregulated by transfecting cells with a miRNA mimic and inhibitor, respectively, prior to treatment with IL-1β. MMP-13 expression was then evaluated by Western blotting and immunofluorescence. Luciferase reporter assays were performed to verify the interaction between miR-140 and ER. RESULTS 17-β-estradiol (E2) suppressed MMP-13 expression in human articular chondrocytes. miR-140 expression was upregulated after estrogen treatment. Knockdown of miR-140 expression abolished the inhibitory effect of estrogen on MMP-13. In addition, the estrogen/ER/miR-140 pathway showed an inhibitory effect on IL-1β-induced cartilage matrix degradation. CONCLUSIONS This study suggests that estrogen acts via ER and miR-140 to inhibit the catabolic activity of proteases within the chondrocyte extracellular matrix. These findings provide new insight into the mechanism of menopausal arthritis and indicate that the ER/miR-140 signaling pathway may be a potential target for therapeutic interventions for menopausal arthritis.
Collapse
Affiliation(s)
- Yujie Liang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, Guangdong Province, China
- Departments of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
| | - Jianyi Xiong
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
| | - Weiming Zhu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
| | - Qisong Liu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
| | - Daming Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
| | - Wei Liu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China
| | - Zigang Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, Guangdong Province, China.
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China.
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, 518035, Guangdong Province, China.
| |
Collapse
|
13
|
Weng YS, Wang HF, Pai PY, Jong GP, Lai CH, Chung LC, Hsieh DJY, HsuanDay C, Kuo WW, Huang CY. Tanshinone IIA Prevents Leu27IGF-II-Induced Cardiomyocyte Hypertrophy Mediated by Estrogen Receptor and Subsequent Akt Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 43:1567-91. [PMID: 26621443 DOI: 10.1142/s0192415x15500895] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
IGF-IIR plays important roles as a key regulator in myocardial pathological hypertrophy and apoptosis, which subsequently lead to heart failure. Salvia miltiorrhiza Bunge (Danshen) is a traditional Chinese medicinal herb used to treat cardiovascular diseases. Tanshinone IIA is an active compound in Danshen and is structurally similar to 17[Formula: see text]-estradiol (E[Formula: see text]. However, whether tanshinone IIA improves cardiomyocyte survival in pathological hypertrophy through estrogen receptor (ER) regulation remains unclear. This study investigates the role of ER signaling in mediating the protective effects of tanshinone IIA on IGF-IIR-induced myocardial hypertrophy. Leu27IGF-II (IGF-II analog) was shown in this study to specifically activate IGF-IIR expression and ICI 182,780 (ICI), an ER antagonist used to investigate tanshinone IIA estrogenic activity. We demonstrated that tanshinone IIA significantly enhanced Akt phosphorylation through ER activation to inhibit Leu27IGF-II-induced calcineurin expression and subsequent NFATc3 nuclear translocation to suppress myocardial hypertrophy. Tanshinone IIA reduced the cell size and suppressed ANP and BNP, inhibiting antihypertrophic effects induced by Leu27IGF-II. The cardioprotective properties of tanshinone IIA that inhibit Leu27IGF-II-induced cell hypertrophy and promote cell survival were reversed by ICI. Furthermore, ICI significantly reduced phospho-Akt, Ly294002 (PI3K inhibitor), and PI3K siRNA significantly reduced the tanshinone IIA-induced protective effect. The above results suggest that tanshinone IIA inhibited cardiomyocyte hypertrophy, which was mediated through ER, by activating the PI3K/Akt pathway and inhibiting Leu27IGF-II-induced calcineurin and NFATC3. Tanshinone IIA exerted strong estrogenic activity and therefore represented a novel selective ER modulator that inhibits IGF-IIR signaling to block cardiac hypertrophy.
Collapse
Affiliation(s)
- Yueh-Shan Weng
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hsueh-Fang Wang
- Institute of Biomedical Nutrition, Hungkuang University, Taichung, Taiwan
| | - Pei-Ying Pai
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Gwo-Ping Jong
- Division of Cardiology, Armed Force Taichung General Hospital, Taichung, Taiwan
| | - Chao-Hung Lai
- Graduate Institute of Aging Medicine, China Medical University, Taichung, Taiwan
- Division of Cardiology, Armed Force Taichung General Hospital, Taichung, Taiwan
| | - Li-Chin Chung
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy & Science, Tainan County, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Cecilia HsuanDay
- Department of Nursing, Mei Ho University, Pingguang Road, Pingtung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
14
|
Alvarez P, Bogen O, Levine JD. Role of nociceptor estrogen receptor GPR30 in a rat model of endometriosis pain. Pain 2014; 155:2680-2686. [PMID: 25280432 DOI: 10.1016/j.pain.2014.09.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 12/16/2022]
Abstract
Endometriosis, the most common cause of chronic pelvic pain, is an estrogen-dependent disease in which classic estrogen receptors (ERα, ERβ) play an important role. Although recent evidence suggests that the novel G protein-coupled estrogen receptor (GPR30) also plays a key role in the progression of endometriosis, whether it is also involved in endometriosis pain is still unknown. Here we tested the hypothesis that GPR30 expressed by nociceptors contributes to endometriosis pain. Intramuscular injection of the GPR30 agonists raloxifene or 17β-estradiol produced a fast-onset, persistent, mechanical hyperalgesia at the site of the injection. Intrathecal antisense (AS) oligodeoxynucleotides (ODN), but not mismatch (MM) ODN, targeting mRNA for GPR30 markedly inhibited its protein expression in nociceptors and attenuated the mechanical hyperalgesia induced by local raloxifene or 17β-estradiol. Pretreatment with the GPR30 antagonist G-36 also inhibited the hyperalgesia induced by raloxifene or 17β-estradiol in naive control rats. Surgical implant of autologous uterine tissue onto the gastrocnemius muscle, which induces endometriosis-like lesions, produced local mechanical hyperalgesia. Intrathecal AS, but not MM, ODN targeting GPR30 mRNA reversibly inhibited the mechanical hyperalgesia at the site of endometriotic lesions. Finally, intralesional injection of the GPR30 antagonist G-36 also inhibited the mechanical hyperalgesia at the site of ectopic uterine tissue. We conclude that local GPR30 agonists produce persistent mechanical hyperalgesia in naive female rats, whereas local GPR30 antagonists inhibit mechanical hyperalgesia in a model of endometriosis pain. Thus, GPR30 expressed by nociceptors innervating ectopic uterine lesions might play a major role in endometriosis pain.
Collapse
Affiliation(s)
- Pedro Alvarez
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA Division of Neuroscience, University of California San Francisco, San Francisco, CA, USA Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
15
|
Choi HJ, Park HK, Jo W, Ryu JE, Jang S, Gu HK, Kim KS, Chung BJ, Son WC. Carcinogenicity of metamifop, a novel herbicide, in Wistar rats following oral administration for 104 weeks. Regul Toxicol Pharmacol 2014; 70:535-44. [PMID: 25194188 DOI: 10.1016/j.yrtph.2014.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/15/2014] [Accepted: 08/18/2014] [Indexed: 10/24/2022]
Abstract
Metamifop is a novel herbicide with as yet undetermined properties. To assess its carcinogenicity, metamifop was mixed into standard rodent chow and fed to male and female Wistar rats at doses of 10, 100 and 750ppm for 104weeks. The viability/mortality of these rats was not affected by treatment with metamifop. Treatment had no significant effects on clinical parameters, and food consumption. Males and females fed 750ppm of metamifop for 104weeks showed decreases in body weight and body weight gain. Histopathological examination revealed that treatment with metamifop reduced non-neoplastic findings (chronic progressive nephropathy, tubular basophilia, tubular casts, glomerulosclerosis, basophilic and clear cell foci, senile atrophy, and mesothelial hyperplasia) and reduced neoplastic findings (thymoma, pituitary adenoma, and mammary fibroadenoma and adenocarcinoma in females, and mesenteric lymph node hemangioma in males) compared with control groups. Benign granulosa cell tumors were increased in a dose-dependent manner. As metamifop did not show any genotoxic potential, and there was no correlation between ovarian cancer and increased gonadal hormone levels in humans, the granulosa cell tumors observed in female rats fed a high dose of metamifop were considered not relevant to humans.
Collapse
Affiliation(s)
- Hyun-Ji Choi
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Republic of Korea
| | - Hyun-Kyu Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Republic of Korea
| | - Woori Jo
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Republic of Korea
| | - Jae-Eun Ryu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Republic of Korea
| | - Sungwoong Jang
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Republic of Korea
| | - Hee-Kyung Gu
- Dongbu Farm Hannong, Daejeon 305-708, Republic of Korea
| | | | | | - Woo-Chan Son
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Republic of Korea.
| |
Collapse
|
16
|
Birzniece V, Magnusson NE, Ho KKY, Frystyk J. Effects of raloxifene and estrogen on bioactive IGF1 in GH-deficient women. Eur J Endocrinol 2014; 170:375-83. [PMID: 24347426 DOI: 10.1530/eje-13-0835] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CONTEXT GH action is attenuated by estrogens and selective estrogen receptor modulators (SERMs) administered orally. During GH therapy in hypopituitary women, co-treatment with raloxifene, a SERM, induced a smaller gain in lean body mass (LBM) compared with estrogen, despite an equal reduction in IGF1. As a higher IGF-binding protein-3 (IGFBP3) level was observed with raloxifene co-treatment, we hypothesize that an increase in IGFBP3 reduced IGF1 bioactivity causing the attenuated anabolic effect. OBJECTIVE To assess the effects of 17β-estradiol (E₂) and raloxifene on bioactive IGF1. DESIGN In study 1, 12 GH-deficient (GHD) women were randomized to raloxifene 120 mg/day or E₂ 4 mg/day for 1 month. In study 2, 16 GHD women were randomized to 1 month GH treatment alone (0.5 mg/day) and in combination with raloxifene (60 mg/day) or E₂ (2 mg/day). We measured bioactive IGF1, immunoreactive IGF1 and IGF2, and IGFBP3 immunoreactivity and fragmentation. RESULTS Raloxifene and estrogen suppressed (P<0.05) total IGF1 equally in GHD and GH-replaced hypopituitary women. In GHD patients, neither raloxifene nor estrogen affected bioactive IGF1. GH significantly increased IGF1 bioactivity, an effect attenuated by co-treatment with raloxifene (Δ -23 ± 7%, P<0.01) and estrogen (Δ -26 ± 3%, P=0.06). Total IGF1 correlated (r(2)=0.54, P<0.001) with bioactive IGF1, which represented 3.1 ± 0.2% of the total IGF1, irrespective of the treatments. Total IGF2 was unchanged by raloxifene and estrogen treatment. IGFBP3 was significantly higher during raloxifene administration, whereas no differences in IGFBP3 fragmentation were observed. CONCLUSION Raloxifene effect on bioactive IGF1 is similar to that of estrogen despite higher IGFBP3 levels during raloxifene administration. We conclude that the observed different effects on LBM between raloxifene and estrogen treatments cannot be explained by differences in IGF1 bioactivity.
Collapse
Affiliation(s)
- Vita Birzniece
- Department of Endocrinology, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
17
|
Changes in expression levels of oxidative stress-related genes in mouse epididymides by neonatal exposure to low-dose decabromodiphenyl ether. Reprod Med Biol 2013; 13:127-134. [PMID: 29699156 DOI: 10.1007/s12522-013-0173-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/07/2013] [Indexed: 10/25/2022] Open
Abstract
Decabromodiphenyl ether (decaBDE), one of the polybrominated diphenyl ethers (PBDEs), is the most well-known flame retardant and is used worldwide. In a previous study, we identified adverse effects of neonatal decaBDE exposure on mouse epididymides, such as decreased epididymal weight. On the other hand, neonatal exposure to diethylstilbestrol (DES), an artificial estrogenic compound, also causes several adverse effects on epididymides. DES exposure results in decreased epididymal weight, morphological abnormalities, and permanent alterations in the expression levels of several genes. The molecular mechanisms underlying the harmful effects of decaBDE exposure remain unclear. Many studies have reported that PBDEs have estrogenic activity, which may contribute to the induction of the adverse effects of decaBDE exposure. We aimed to examine the effects of neonatal decaBDE exposure on epididymides. Our data showed that (1) no histological change was observed on epididymal tissues from neonatal decaBDE exposure, unlike the effect of DES, (2) decaBDE exposure did not induce the alterations in gene expression observed with DES exposure; instead alterations in gene expression of certain oxidative stress-related genes were observed, and (3) the expression of ubiquitin C increased in decaBDE-exposed mouse epididymides. Our present data suggest the possibility that increased oxidative stress plays a role in the harmful effects observed in mouse epididymides after decaBDE-exposure.
Collapse
|
18
|
Fernandez JW, Grizzell JA, Wecker L. The role of estrogen receptor β and nicotinic cholinergic receptors in postpartum depression. Prog Neuropsychopharmacol Biol Psychiatry 2013; 40:199-206. [PMID: 23063492 DOI: 10.1016/j.pnpbp.2012.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 10/02/2012] [Accepted: 10/02/2012] [Indexed: 01/19/2023]
Abstract
Postpartum depression (PPD) is a devastating disease occurring in approximately 20% of women. Women who suffer from PPD appear to be more sensitive to postpartum hormonal changes than women who do not experience this form of depression. Furthermore, women who quit smoking prior to or during pregnancy, and who develop PPD, are at an increased risk of smoking relapse. Unfortunately, the mechanistic relationship between the pathophysiology of PPD and smoking relapse is unknown. Here we review the roles of both estrogen receptor beta (ERβ) and cholinergic nicotinic receptors (nAChRs) in the pathogenesis of depression and propose a mechanistic rationale to explain the high rate of smoking relapse exhibited by women who develop PPD.
Collapse
Affiliation(s)
- Jamie Winderbaum Fernandez
- Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, 3515 E. Fletcher Avenue, Tampa, FL, 33611, USA.
| | | | | |
Collapse
|
19
|
Desaulniers D, Leingartner K, Zacharewski T, Foster WG. Optimization of an MCF7-E3 Cell Proliferation Assay and Effects of Environmental Pollutants and Industrial Chemicals. Toxicol In Vitro 2012; 12:409-22. [PMID: 20654424 DOI: 10.1016/s0887-2333(98)00004-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/1997] [Indexed: 10/18/2022]
Abstract
Environmental contaminants might adversely affect human health by acting as endocrine disruptors and thus need to be identified. Our objective was to optimize the MCF7 cell proliferation assay to screen industrial chemicals for potential oestrogenic effects. Growth conditions, performance of the clone E3 and WT-MCF7 cells and five methods to derive proliferation indices were compared. The E3 cells were further characterized by testing the effects of transforming growth factorbeta (TGFbeta), epidermal growth factor (EGF), insulin, testosterone, the anti-oestrogen ICI 164,384 (ICI) and environmental contaminants with known oestrogenic potential. Industrial chemicals with unknown oestrogenic effects were then tested. As expected, induction of proliferation by estradiol-17beta (E2) was greater and less variable using the clone E3. To generate proliferation indices, the alamarBlue assay had a sensitivity comparable to that of [(3)H]thymidine incorporation ((3)H-TI). The E3 cells were not responsive to EGF (0-100 ng/ml) or insulin (0-313 ng/ml) but their proliferation was decreased (P<0.05) by TGFbeta (45 ng/ml) and testosterone (10(-8)m), which might be typical of highly oestrogen-responsive MCF7 cells. ICI (5x10(-7)m) inhibited the proliferative effects of 10(-10)m E2 and that of 10(-6)m 4-tert-octylphenol (Op) but not the proliferative effect of 10(-5)m Op, suggesting displacement of ICI by Op or induction of oestrogen-receptor independent proliferation. N-oxydiethylene-2-benzothiazole sulfenamide (OBTS) altered (3)H-TI in the MCF7 cells, although not in a dose related manner. OBTS did not induce uterotrophic effects in immature female rats, or any response in a human oestrogen chimeric receptor/reporter gene assay, suggesting that its effects were not mediated through the binding of the oestrogen-receptor. Seven other industrial chemicals were tested and had no effects. In conclusion, the MCF7 cell proliferation assay is one screening tool that permits identification of chemicals with oestrogenic potential which thus require further testing.
Collapse
Affiliation(s)
- D Desaulniers
- Reproductive Toxicology Section, Environmental and Occupational Toxicology Division, Bureau of Chemical Hazards, Environmental Health Directorate, Health Protection Branch, Department of Health, Ottawa, Ontario, Canada, K1A 0L2
| | | | | | | |
Collapse
|
20
|
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
21
|
Abstract
The immune system and its orchestrated response are affected by a multitude of endogenous and exogenous factors, modulators and challenges. One of the most frequent differences described in the immune response is its vigor and activity in females compared to males, leading to the consequent increase in autoimmune conditions seen in the female population as well as differences in the immune response to pathogens and viruses. The following review summarizes our present knowledge on sex differences in the immune response, detailing the hormonal and genetic effects that have been proposed as explanatory mechanisms. Sexual hormones, mostly estrogen but also progesterone and testosterone, affect immune cells quantitatively and qualitatively. Relevant research has focused on the impact of hormones on cytokine production by the different effector cells, as well as impact on immunoglobulin production by B lymphocytes and activity of granulocytes and NK cells. The biological aspects are complemented by research data on the possible modulatory role of the X chromosome. In addition to biological differences, the frequently neglected role of gender as an immunomodulator is introduced and explored. Gender affects all areas of human life and consequently affects the different steps of an immune response. Exposure to various types of antigens, access to health promotion programs and health care, as well as prioritization of health needs and household resource allocation all affect the different response of females and males to immunologic challenges.
Collapse
|
22
|
Jochems C, Islander U, Erlandsson M, Engdahl C, Lagerquist M, Ohlsson C, Nandakumar KS, Holmdahl R, Carlsten H. Effects of oestradiol and raloxifene on the induction and effector phases of experimental postmenopausal arthritis and secondary osteoporosis. Clin Exp Immunol 2011; 165:121-9. [PMID: 21501150 DOI: 10.1111/j.1365-2249.2011.04397.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Oestradiol and the selective oestrogen receptor modulator (SERM) raloxifene have been shown to ameliorate collagen-induced arthritis (CIA) in rats and in mice. One aim was to investigate if raloxifene exerts its anti-arthritic and anti-osteoporotic effects during the induction or effector phase of arthritis. A second aim was to analyse if raloxifene activates the oestrogen response element (ERE) to produce its immune-modulator effects. CIA or collagen-antibody-induced arthritis (CAIA) was induced in ovariectomized DBA/1-mice. CIA was used for evaluation of treatment during the induction, and CAIA for the effector phase of arthritis and osteoporosis development. Raloxifene, oestradiol or vehicle was administered 5 days/week. The clinical disease was evaluated continuously. Bone marrow density (BMD) was analysed with peripheral quantitative computer tomography, paws were collected for histological examination, and sera were analysed for markers of bone and cartilage turnover and proinflammatory cytokines. Transgenic luciferase (Luc)-ERE mice were immunized with collagen (CII), and after 10 days injected once with raloxifene, oestradiol or vehicle before termination. Spleens were analysed for luciferase activity to measure ERE activation. Treatment with oestradiol or raloxifene during the induction phase of CIA failed to affect arthritis. Raloxifene did not hamper disease activity in CAIA, whereas oestradiol delayed the onset and ameliorated the severity. Both raloxifene and oestradiol preserved BMD in CAIA. CII-immunization increased the oestradiol-induced ERE activation in spleen, and raloxifene activated the ERE at about 25% the intensity of oestradiol. Further experiments are needed to elucidate the exact mechanisms behind this finding.
Collapse
Affiliation(s)
- C Jochems
- Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy at Göteborg University, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
D’Amelio P, Fornelli G, Roato I, Isaia GC. Interactions between the immune system and bone. World J Orthop 2011; 2:25-30. [PMID: 22474632 PMCID: PMC3302038 DOI: 10.5312/wjo.v2.i3.25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 01/12/2011] [Accepted: 01/19/2011] [Indexed: 02/06/2023] Open
Abstract
The relationship between the immune system, estrogen deficiency and bone loss is an intriguing and, as yet, unexplained challenge of the past two decades. Here we summarize the evidence that links immune cells, inflammation, cytokine production and osteoclast formation and activity with particular regard to humans.
Collapse
|
24
|
Rojo Venegas K, Aguilera Gómez M, Eisman JA, García Sánchez A, Faus Dader MJ, Calleja Hernández MA. Pharmacogenetics of osteoporosis-related bone fractures: moving towards the harmonization and validation of polymorphism diagnostic tools. Pharmacogenomics 2011; 11:1287-303. [PMID: 20860468 DOI: 10.2217/pgs.10.116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Osteoporosis is one of the most common skeletal chronic conditions in developed countries, hip fracture being one of its major healthcare outcomes. There is considerable variation in the implementation of current pharmacological treatment and prevention, despite consistent recommendations and guidelines. Many studies have reported conflicting findings of genetic associations with bone density and turnover that might predict fracture risk. Moreover, it is not clear whether genetic differences exist in relation to the morbidity and efficiency of the pharmacotherapy treatments. Clinical response, including beneficial and adverse events associated with osteoporosis treatments, is highly variable among individuals. In this context, the present article intends to summarize putative candidate genes and genome-wide association studies that have been related with BMD and fracture risk, and to draw the attention to the need for pharmacogenetic methodology that could be applicable in clinical translational research after an adequate validation process. This article mainly compiles analysis of important polymorphisms in osteoporosis documented previously, and it describes the simple molecular biology tools for routine genotype acquisition. Validation of methods for the easy, fast and accessible identification of SNPs is necessary for evolving pharmacogenetic diagnostic tools in order to contribute to the discovery of clinically relevant genetic variation with an impact on osteoporosis and its personalized treatment.
Collapse
Affiliation(s)
- Karen Rojo Venegas
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avenida de las Fuerzas Armadas 2, CP:18014, Granada, Spain.
| | | | | | | | | | | |
Collapse
|
25
|
Giroux V, Bernatchez G, Carrier JC. Chemopreventive effect of ERβ-Selective agonist on intestinal tumorigenesis in Apc(Min/+) mice. Mol Carcinog 2010; 50:359-69. [PMID: 21480389 DOI: 10.1002/mc.20719] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 11/02/2010] [Accepted: 11/03/2010] [Indexed: 12/17/2022]
Abstract
Epidemiological and experimental evidence suggests that estrogen replacement therapy reduces the risk of colon cancer in postmenopausal women. Estrogen receptor beta (ERβ) is thought to be the principal mediator of the estrogen effect in the colon. Recent studies by our team suggested positive regulation of the transforming growth factor (TGF)β pathway by estrogen in mice colonocytes. We therefore wanted to investigate the effects of ERβ agonist treatment on intestinal tumorigenesis in Apc(Min/+) mice. Weaned Apc(Min/+) mice were injected subcutaneously three times a week for 12 wk with vehicle or ERβ-selective agonist, diarylpropionitrile (DPN, 5 mg/kg). DPN administration resulted in a significant reduction in small intestinal polyp multiplicity in both Apc(Min/+) male and female mice. Furthermore, the mean diameter of small intestinal polyps was lower in DPN-treated than vehicle-treated males, along with lower BrdU incorporation indices in jejunal and colon epithelial cells of both sexes. DPN treatment also increased apoptosis in colon epithelium as measured by TUNEL assay and cleaved caspase 3 quantification. The effect of DPN on various components of the TGFβ pathway was also studied in colonocytes. DPN treatment increased expression of TGFβ1 and TGFβ3 transcripts, levels of nuclear and phosphorylated Smad2 as well as p27 cell-cycle inhibitor, a TGFβ pathway target gene. Our results demonstrate that DPN treatment reduces intestinal tumorigenesis in Apc(Min/+) mice. Furthermore, we suggest that positive regulation of the TGFβ pathway by ERβ activation could contribute to the protective role of estrogen in intestinal tumor development.
Collapse
Affiliation(s)
- Véronique Giroux
- Faculty of Medicine and Health Sciences, Department of Medicine and Anatomy and Cellular Biology, CIHR Team on Digestive Epithelium, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | | |
Collapse
|
26
|
Shibata MA, Morimoto J, Shibata E, Kurose H, Akamatsu K, Li ZL, Kusakabe M, Ohmichi M, Otsuki Y. Raloxifene inhibits tumor growth and lymph node metastasis in a xenograft model of metastatic mammary cancer. BMC Cancer 2010; 10:566. [PMID: 20958960 PMCID: PMC2978204 DOI: 10.1186/1471-2407-10-566] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 10/19/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The effects of raloxifene, a novel selective estrogen receptor modulator, were studied in a mouse metastatic mammary cancer model expressing cytoplasmic ERα. METHODS Mammary tumors, induced by inoculation of syngeneic BALB/c mice with BJMC3879luc2 cells, were subsequently treated with raloxifene at 0, 18 and 27 mg/kg/day using mini-osmotic pumps. RESULTS In vitro study demonstrated that the ERα in BJMC3879luc2 cells was smaller (between 50 and 64 kDa) than the normal-sized ERα (66 kDa) and showed cytoplasmic localization. A statistically significant but weak estradiol response was observed in this cell line. When BJMC3879luc2 tumors were implanted into mice, the ERα mRNA levels were significantly higher in females than in males. In vitro studies showed that raloxifene induced mitochondria-mediated apoptosis and cell-cycle arrest in the G1-phase and a decrease in the cell population in the S-phase. In animal experiments, tumor volumes were significantly suppressed in the raloxifene-treated groups. The multiplicity of lymph node metastasis was significantly decreased in the 27 mg/kg group. Levels of apoptosis were significantly increased in the raloxifene-treated groups, whereas the levels of DNA synthesis were significantly decreased in these groups. No differences in microvessel density in tumors were observed between the control and raloxifene-treated groups. The numbers of dilated lymphatic vessels containing intraluminal tumor cells were significantly reduced in mammary tumors in the raloxifene-treated groups. The levels of ERα mRNA in mammary tumors tended to be decreased in the raloxifene-treated groups. CONCLUSION These results suggest that the antimetastatic activity of raloxifene in mammary cancer expressing cytoplasmic ERα may be a crucial finding with clinical applications and that raloxifene may be useful as an adjuvant therapy and for the chemoprevention of breast cancer development.
Collapse
Affiliation(s)
- Masa-Aki Shibata
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Junji Morimoto
- Laboratory Animal Center, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Eiko Shibata
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
- Department of Bioscience, National Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Hitomi Kurose
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Kanako Akamatsu
- Department of Systems Bioscience for Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Zhong-Lian Li
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Moriaki Kusakabe
- Research Center for Food Safety, University of Tokyo Graduate School of Agricultural and Life Sciences, Tokyo, Japan
| | - Masahide Ohmichi
- Department of Gynecology, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Yoshinori Otsuki
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
27
|
Mechanisms involved in the inhibition of osteoclast generation by the benzothiophene SERM LY117018. Wien Klin Wochenschr 2010; 122:626-32. [DOI: 10.1007/s00508-010-1469-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 09/09/2010] [Indexed: 11/25/2022]
|
28
|
Pacifici R. T cells: critical bone regulators in health and disease. Bone 2010; 47:461-71. [PMID: 20452473 PMCID: PMC2926258 DOI: 10.1016/j.bone.2010.04.611] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 04/27/2010] [Accepted: 04/30/2010] [Indexed: 01/16/2023]
Abstract
Postmenopausal osteoporosis and hyperparathyroidism are to two common forms of bone loss caused primarily by an expansion of the osteoclastic pool only partially compensated by a stimulation of bone formation. The intimate mechanisms by which estrogen deficiency and excessive production of PTH cause bone loss remain to be determined in part because in vitro studies do not provide the means to adequately reproduce the effects of ovx and PTH overproduction observed in vivo. This article examines the connection between T cells and bone in health and disease and reviews the evidence in favor of the hypothesis that T cells play an unexpected critical role in the mechanism of action of estrogen and PTH in bone.
Collapse
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA.
| |
Collapse
|
29
|
Alfonzo R, Reyna E, Guerra M, Mejia J, Reyna N, Torres D, Santos J, Schloeter J. Concentraciones de lípidos y lipoproteínas en menopáusicas tratadas con raloxifeno. CLINICA E INVESTIGACION EN GINECOLOGIA Y OBSTETRICIA 2010. [DOI: 10.1016/j.gine.2009.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
30
|
Pacifici R. The immune system and bone. Arch Biochem Biophys 2010; 503:41-53. [PMID: 20599675 DOI: 10.1016/j.abb.2010.05.027] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 05/26/2010] [Accepted: 05/27/2010] [Indexed: 02/06/2023]
Abstract
T cells and B cells produce large amounts of cytokines which regulate bone resorption and bone formation. These factors play a critical role in the regulation of bone turnover in health and disease. In addition, immune cells of the bone marrow regulate bone homeostasis by cross-talking with bone marrow stromal cells and osteoblastic cells via cell surface molecules. These regulatory mechanisms are particularly relevant for postmenopausal osteoporosis and hyperparathyroidism, two common forms of bone loss caused primarily by an expansion of the osteoclastic pool only partially compensated by a stimulation of bone formation. This article describes the cytokines and immune factors that regulate bone cells, the immune cells relevant to bone, examines the connection between T cells and bone in health and disease, and reviews the evidence in favor of a link between T cells and the mechanism of action of estrogen and PTH in bone.
Collapse
|
31
|
Genome-wide analysis of estrogen receptor alpha DNA binding and tethering mechanisms identifies Runx1 as a novel tethering factor in receptor-mediated transcriptional activation. Mol Cell Biol 2010; 30:3943-55. [PMID: 20547749 DOI: 10.1128/mcb.00118-10] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Nuclear receptor estrogen receptor alpha (ER alpha) controls the expression of hundreds of genes responsible for target cell phenotypic properties, but the relative importance of direct versus tethering mechanisms of DNA binding has not been established. In this first report, we examine the genome-wide chromatin localization of an altered-specificity mutant ER with a DNA binding domain deficient in binding to estrogen response element (ERE)-containing DNA (DBDmut ER) versus wild-type ER alpha. Using high-throughput sequencing of ER chromatin immunoprecipitations (ChIP-Seq) and mRNA transcriptional profiling, we show that direct ERE binding is required for most of (75%) estrogen-dependent gene regulation and 90% of hormone-dependent recruitment of ER to genomic binding sites. De novo motif analysis of the chromatin binding regions in MDA-MB-231 human breast cancer cells defined unique transcription factor profiles responsible for genes regulated through tethering versus direct ERE binding, with Runx motifs enriched in ER-tethered sites. We confirmed a role for Runx1 in mediating ER alpha genomic recruitment and regulation of tethering genes. Our findings delineate the contributions of direct receptor ERE binding versus binding through response elements for other transcription factors in chromatin localization and ER-dependent gene regulation, paradigms likely to underlie the gene regulatory actions of other nuclear receptors as well.
Collapse
|
32
|
Abstract
BACKGROUND Fractures are a significant problem in geriatric patients, and understanding the evidence for benefit and possible harm of osteoporosis treatments is critical to appropriate management of this patient population. OBJECTIVE The purpose of this article was to review the evidence and treatment considerations related to use of the approved osteoporosis treatments in the United States across the continuum of ages in the geriatric population. METHODS MEDLINE and the Web of Science were searched to find English-language articles published from 2000 through July 2009. Search terms included: practice guideline, osteoporosis, calcium, vitamin D, pharmacoeconomics, ethnicity, and treatment. The generic names of each of the osteoporosis treatments approved in the United States were searched to find relevant clinical trials and randomized controlled trials (RCTs). Pivotal trials that included fracture data or focused specifically on elderly patients (> or = 60 years of age) were selected. Bibliographies in the identified articles were searched for additional articles, and the prescribing information for each of the approved treatments was reviewed. RESULTS Many osteoporosis studies have a mean patient age >60 years, but data for older patients are limited. Subanalyses of older patient groups have found risedronate to be beneficial for vertebral fractures in patients aged 70 to 79 years (absolute risk reduction [ARR], 8.4%; P < 0.001) and teriparatide to be beneficial for both vertebral (ARR, 6.4%; P < 0.05) and new nonvertebral fragility fractures (ARR, 9.9%; P < 0.05) in women aged > or = 75 years. However, no RCTs of geriatric patients who were either nonambulatory or had multiple comorbidities were identified in the literature. CONCLUSIONS Evidence indicates that the osteoporosis treatments currently available in the United States are beneficial for treating osteoporosis in geriatric patients. However, data are limited for the oldest patients (> or = 80 years) and those with significant comorbidities. Because of the limited availability of data for geriatric patients with significant comorbidities, the properties of the various agents, including efficacy, tolerability, and potential contraindications, should be considered carefully for each geriatric patient.
Collapse
|
33
|
Vico L, Vanacker JM. Sex hormones and their receptors in bone homeostasis: insights from genetically modified mouse models. Osteoporos Int 2010; 21:365-72. [PMID: 19495826 DOI: 10.1007/s00198-009-0963-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 04/29/2009] [Indexed: 10/20/2022]
Abstract
In this review, we summarize available data regarding bone phenotypes in estrogen receptors alpha and beta, androgen receptor, and aromatase enzyme-deficient mice. We examine sex differences in the trabecular and cortical bone compartments and we discuss these findings in relation to known estrogen effects in humans. We also report how estrogen influences the responsiveness of the skeleton to exercise. Although uncertainties remain, it is clear that both estrogen and androgen are important for both male and female skeleton. Estrogen receptor alpha mainly through its classical signaling pathway is particularly important for the male mice skeleton while both estrogen receptors alpha and beta are required for female mice skeleton. These deletions also induce major hormonal alterations themselves impacting on bone metabolism. More investigations are needed to fully understand the respective role of all these receptors in periosteal expansion in both sexes and the way they affect the mechanical sensitivity of the periosteum.
Collapse
Affiliation(s)
- L Vico
- Université de Lyon, INSERM, U890, Université Jean Monnet, St-Etienne, 42023, France.
| | | |
Collapse
|
34
|
Yan MZ, Xu Y, Gong YX, Liu JM, Lu SY, Huang L, Wang ZG, Zhao YJ, Pang XF. Raloxifene inhibits bone loss and improves bone strength through an Opg-independent mechanism. Endocrine 2010; 37:55-61. [PMID: 20963556 DOI: 10.1007/s12020-009-9267-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Accepted: 09/11/2009] [Indexed: 10/20/2022]
Abstract
The osteoblast-derived paracrine factor osteoprotegerin (OPG) is considered to play a key role in inhibition of osteoclast formation and activity. Recently, raloxifene, a nonsteroidal benzothiophene, was found to exert anti-resorptive effects via modulating OPG expression in osteoblasts. To explore whether raloxifene regulates bone metabolism via an OPG-dependant pathway in vivo, we investigated the effects of raloxifene on bone loss in Opg-deficient mice. The results show that bone mineral density and bone strength are increased in mice deficient for Opg after treatment with raloxifene for 30 days. Histomorphometric analysis shows that raloxifene can increase bone trabecular area and decrease the number of osteoclasts in Opg (-/-) mice. Moreover, raloxifene reduces Rankl transcription and serum level of Rankl, which is dramatically increased in Opg knockout mice. These results suggest that raloxifene-induced inhibition of bone resorption may be independent of Opg pathway in mice.
Collapse
Affiliation(s)
- Mei-zhu Yan
- Department of Geriatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Omwandho CO, Konrad L, Halis G, Oehmke F, Tinneberg HR. Role of TGF- s in normal human endometrium and endometriosis. Hum Reprod 2009; 25:101-9. [DOI: 10.1093/humrep/dep382] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
36
|
|
37
|
Tsuruoka S, Hasegawa G, Kaneda T, Maeda A, Fujimura A. Dosing Time‐Dependent Effect of Raloxifene on Plasma Fibrinogen Concentration in Ovariectomized Rats. Chronobiol Int 2009; 25:808-18. [DOI: 10.1080/07420520802387682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
38
|
Piperi C, Zisaki K, Skenderi K, Kalofoutis C, Kalofoutis A. Evidence of improved serum fatty acid profile of postmenopausal women receiving atorvastatin and raloxifene. J OBSTET GYNAECOL 2009; 25:476-81. [PMID: 16183584 DOI: 10.1080/01443610500171060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Raloxifene and atorvastatin have been shown to reduce the risk of cardiovascular disease associated with postmenopausal status and it has been postulated that their effects may be partly mediated by favourable changes in serum lipids and fatty acid composition. In the present study, individual administration of either raloxifene (Group A) or atorvastatin (Group B) or both (Group C) was compared for a period of 3 months and their effects on total lipids and fatty acids composition was evaluated. Postmenopausal women receiving both raloxifene and atorvastatin showed significant changes in the majority of serum lipids with important reductions in total cholesterol (p < 0.001), triglycerides (p < 0.001), LDL-C (p < 0.001) and Apo B levels (p < 0.001). Phospholipids concentrations (p < 0.01) as well as Apo A-I were also significantly raised (p < 0.001). Furthermore, oleic acid (18:1) and linoleic acid (18:2) levels were significantly increased (p < 0.01 and p < 0.001 respectively) followed by a marked reduction in palmitic acid (16:0) and arachidonic acid (20:4) concentrations (p < 0.01 and p < 0.001 respectively). The results of the study indicate that the serum lipid and fatty acid composition in postmenopausal women is influenced by the combined treatment of raloxifene and atorvastatin and a further attempt to evaluate the significance of these results is discussed.
Collapse
Affiliation(s)
- C Piperi
- Department of Biological Chemistry, University of Athens, School of Medicine, Athens, Greece
| | | | | | | | | |
Collapse
|
39
|
Rey JRC, Cervino EV, Rentero ML, Crespo EC, Alvaro AO, Casillas M. Raloxifene: mechanism of action, effects on bone tissue, and applicability in clinical traumatology practice. Open Orthop J 2009; 3:14-21. [PMID: 19516920 PMCID: PMC2687107 DOI: 10.2174/1874325000903010014] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 01/05/2009] [Accepted: 01/14/2009] [Indexed: 11/22/2022] Open
Abstract
Raloxifene, a member of the class of selective estrogen receptor modulators (SERM), reproduces the beneficial effects of estrogens on the skeletal systems, without the negative effects estrogens on breast and endometrium. This is a review article summarizing its mechanism, effects on bone and its applicability in traumatology clinical practice. In postmenopausal osteoporosis, this drug has been proven to decrease accelerated bone turnover, increase bone mineral density (BMD), and to structurally recover bone, decreasing the risk of vertebral fractures and the risk of non-vertebral fractures in patients with previous, severe vertebral fractures. Moreover, raloxifene appears to lower the risk of invasive breast cancer. Raloxifene would be efficacious in the prevention and treatment of postmenopausal osteoporosis.We can therefore conclude that raloxifene would be efficacious in the prevention and treatment of postmenopausal osteoporosis, while reducing the risk of breast cancer when used at the indicated dose of 60 mg/day and with a low incidence of side effects.
Collapse
Affiliation(s)
- Jose R Caeiro Rey
- Servicio de C.O.T. Complexo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
A large body of evidence suggests hormone replacement therapy (HRT) reduces cardiovascular risk in postmenopausal women. It is, however, associated with serious side effects, such as increased risk of breast and endometrial cancer. This has likely caused uneasiness among both women and health care providers. A new class of compounds, called selective estrogen receptor modulators (SERMs), have emerged. Through their interactions at the estrogen receptor level they have become a class of compounds distinct from estrogen. While they share similar effects with estrogen on such factors as lipid profile and bone density, they affect other tissues differently. Specifically, they do not induce endometrial hyperplasia and are therefore not associated with endometrial cancer. In vitro studies have also shown that they inhibit lipoprotein oxidation and vascular smooth muscle cell proliferation. The cumulative effects of these compounds may prove quite beneficial in reducing cardiovascular risk in postmenopausal women while avoiding serious side effects. This may, in turn, ease much of the anxiety surrounding the issue of HRT. Clinical trials are presently being conducted to evaluate the effectiveness of raloxifene, a SERM, on cardiovascular risk reduction in postmenopausal women.
Collapse
Affiliation(s)
- J A Guzzo
- Department of Medicine, University of North Carolina, Chapel Hill 27599, USA
| |
Collapse
|
41
|
Maru BS, Tobias JH, Rivers C, Caunt CJ, Norman MR, McArdle CA. Potential use of an estrogen-glucocorticoid receptor chimera as a drug screen for tissue selective estrogenic activity. Bone 2009; 44:102-12. [PMID: 18976723 DOI: 10.1016/j.bone.2008.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 09/24/2008] [Accepted: 09/29/2008] [Indexed: 11/20/2022]
Abstract
SERMs act as ER agonists in bone despite their antagonistic properties in other tissues. As well as inhibiting bone remodelling, this effect may involve stimulation of osteoblast activity, in light of evidence from recent in vivo studies. However, progress in exploring this action has been hampered by a lack of accurate in vitro models. For example, ER antagonists are reported to stimulate reporter assays based on estrogen target genes in osteoblasts, contrary to their inhibitory effects in vivo. We examined whether evaluating global aspects of ER function provides a more accurate reflection of ER activation in osteoblasts, based on the use of morphological and/or transcriptional read-outs with green fluorescent protein (GFP)-receptor chimeras. Osteoblast-like (ROS and U2OS) and breast cancer (MCF7) cells were transfected with a human ERalpha-GFP fusion protein, and treated with ER agonists (17beta-estradiol, and dienestrol), antagonists (ICI 182,780 and ZK 164015) and SERMs (tamoxifen, raloxifene, 4-hydroxytamoxifen (4-HT) and hexestrol). We investigated cellular compartmentalisation of these constructs by fluorescence microscopy, nuclear mobility by fluorescence recovery after photobleaching (FRAP), and global activation of estrogenic transcription using a ERE-luc reporter. SERMs caused a modest increase in ERE-luc activity in osteoblast-like cells (but not in breast cells), and a reduction in nuclear mobility in breast (but not osteoblast-like) cells. These studies were then repeated using a GFP chimera where the human GR ligand binding domain (LBD) was replaced by the human ERalpha LBD (ERGR-GFP), combined with a GRE-luc reporter. Interestingly, SERMs increased both cytoplasmic to nuclear translocation of ERGR-GFP, and GRE-luc reporter activity, in osteoblast-like (but not breast) cells. Indeed, transcriptional responses to SERMs in osteoblast-like cells were considerably greater with the ERGR/GRE-luc than the ERalpha/ERE-luc system, 4-HT inducing 300 and 25% increases in reporter activity respectively. ER antagonists were entirely without effect. We conclude that evaluation of global estrogenic activity, as opposed to activation of a specific target gene, provides a more accurate read-out for osteoblast stimulation. In particular, ERGR-mediated GRE-luc activity provides a high signal response to estrogen agonists and SERMs, in a cell context dependent manner closely resembling that observed in vivo. Further studies utilising this system are justified to explore the mechanistic basis for estrogenic stimulation of osteoblast activity, and to identify newer SERMs capable of targeting this activity.
Collapse
Affiliation(s)
- Benit S Maru
- Laboratory for Integrated Neurosciences and Endocrinology, University of Bristol, UK
| | | | | | | | | | | |
Collapse
|
42
|
Scafonas A, Reszka AA, Kimmel DB, Hou XS, Su Q, Birzin ET, Kim S, Chen HY, Tan Q, Roher SP, Dininno F, Hammond ML, Rodan GA, Towler DA, Schmidt A. Agonist-like SERM effects on ERalpha-mediated repression of MMP1 promoter activity predict in vivo effects on bone and uterus. J Steroid Biochem Mol Biol 2008; 110:197-206. [PMID: 18508261 DOI: 10.1016/j.jsbmb.2007.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 10/25/2007] [Indexed: 10/22/2022]
Abstract
Estradiol receptors (ER), ERalpha and ERbeta, are ligand-dependent transcription factors that regulate gene expression. Human and murine genetics suggest that ERalpha is the key target for estradiol action on bone, uterus and breast. To date, the molecular mode of action of estradiol and selective estradiol receptor modulators (SERMs) on bone is not fully understood. This is exemplified by a lack of in vitro assays that reliably predict SERM agonist activities in vivo. We hypothesized that ligand-dependent ERalpha transrepression, via protein-protein interactions at AP1, may predict estrogenic effects on bone. We modeled this using the MMP1 promoter, which encodes an AP1 binding site. We show that ICI-182780, raloxifene, 4-hydroxytamoxifen and estradiol all exhibit differential agonistic activities on the MMP1 promoter by suppressing activity by 20-80%. Transrepression efficacy and potency correlated with both uterotrophic (R(2)=0.98) and osteoprotective (R(2)=0.80) potential in the ovariectomized rat. This identifies MMP1 promoter transrepression as an agonist activity commonly shared by AF2 agonists and "antagonists" alike. Mutation analysis showed that the repression by estradiol and SERMs required correct amino acid sequences in the AF-2 domain. For instance, L540Q AF2 mutation did not alter responses to raloxifene, although it greatly increased responses to ICI-182780 (threefold) and reduced estradiol's effect by 20%. Furthermore, all tested ligands repressed the MMP1 promoter through the L540Q mutant with identical efficacy. Together, these data suggest that estradiol and SERMs share common agonist transcriptional activity via protein-protein interactions at AP1.
Collapse
Affiliation(s)
- Angela Scafonas
- Molecular Endocrinology, Merck Research Laboratories, West Point, 770 Sumneytown Pike, PA 19486, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Bone is a dynamic organ constantly remodeled to support calcium homeostasis and structural needs. The osteoclast is the cell responsible for removing both the organic and inorganic components of bone. It is derived from hematopoietic progenitors in the macrophage lineage and differentiates in response to the tumor necrosis factor family cytokine receptor activator of NF kappa B ligand. alpha v beta 3 integrin mediates cell adhesion necessary for polarization and formation of an isolated, acidified resorptive microenvironment. Defects in osteoclast function, whether genetic or iatrogenic, may increase bone mass but lead to poor bone quality and a high fracture risk. Pathological stimulation of osteoclast formation and resorption occurs in postmenopausal osteoporosis, inflammatory arthritis, and metastasis of tumors to bone. In these diseases, osteoclast activity causes bone loss that leads to pain, deformity, and fracture. Thus, osteoclasts are critical for normal bone function, but their activity must be controlled.
Collapse
Affiliation(s)
- Deborah V Novack
- Department of Pathology and Immunology, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
44
|
Nalbandian G, Kovats S. Understanding sex biases in immunity: effects of estrogen on the differentiation and function of antigen-presenting cells. Immunol Res 2008; 31:91-106. [PMID: 15778508 DOI: 10.1385/ir:31:2:091] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The initiation and perpetuation of innate and adaptive immunity is dependent on the ability of professional antigen-presenting cells (APCs) to sense inflammatory stimuli; produce cytokines; and internalize, degrade, and present antigens via surface major histocompatibility complex (MHC) molecules. Dendritic cells (DCs), macrophages, and B lymphocytes express estrogen receptors, indicating that the steroid sex hormone estrogen might directly modulate the function of these cells during immune responses. Sex-specific parameters of immune function have been identified during autoimmunity and the pathogenesis of infectious disease, which show sex biases in their incidence and manifestation; female immunity also varies as estrogen levels change. In this article, we summarize studies that demonstrate effects of estrogen on the differentiation or function of APCs in model in vitro systems, or under circumstances of natural or imposed variation in estrogen levels in vivo.
Collapse
Affiliation(s)
- Greg Nalbandian
- Division of Immunology, Beckman Research Institute, City of Hope National Medical Center, CA 91010, USA
| | | |
Collapse
|
45
|
Pacifici R. Estrogen deficiency, T cells and bone loss. Cell Immunol 2008; 252:68-80. [PMID: 17888417 DOI: 10.1016/j.cellimm.2007.06.008] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 06/01/2007] [Accepted: 06/05/2007] [Indexed: 12/11/2022]
Abstract
Estrogen plays a fundamental role in the maintenance of skeletal homeostasis. Although estrogen is established to have direct effects on bone cells, animal studies have identified additional regulatory effects of estrogen centered at the level of the adaptive immune response. Furthermore, a potential role for reactive oxygen species has now been identified in both humans and animals. One of the major challenges has been to integrate a multitude of redundant pathways and cytokines, that all appear capable of playing a relevant role, into a global model of postmenopausal osteoporosis. This review presents our current understanding of the process of estrogen deficiency mediated bone destruction and explores some of the most recent findings and hypotheses to explain estrogen action in bone.
Collapse
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and Molecular Pathogenesis Program, Emory University School of Medicine, 101 Woodruff Circle, Room 1307, Atlanta, GA 30322, USA.
| |
Collapse
|
46
|
Leung LK, Yuen YM, Leung HY, Wang Y. Dietary soya isoflavones and breast carcinogenesis: a perspective from a cell-culture model. Nutr Res Rev 2007; 18:202-11. [DOI: 10.1079/nrr2005111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Southeast Asian women have a lower incidence of breast cancer than their counterparts in the West. Epidemiological studies have indicated that soya consumption may be a contributing factor. Carcinogenesis is a process involving multiple stages. The present review attempts to fit the cellular mechanisms attributed to soya isoflavones into these different stages. Many cell-culture studies have reported the growth-inhibitory effect of soya isoflavones; however, with the non-physiological concentrations employed in these studies it would be difficult to explain the protection mechanisms observed in epidemiological studies. Our laboratory has previously found that genistein inhibits cytochrome P450 (CYP)1A1 and CYP1B1. The inhibition implies that soya consumption may have the potential to prevent chemical carcinogenesis. The preferential inhibition of CYP1B1 may also block the oestrogen-initiated carcinogenesis. The antagonism of oestrogen receptor (ER) binding can affect the cell-proliferative phase, which is likely to be important in the promotion stage of breast cancer. Since our laboratory and others have indicated that genistein at physiological concentrations has no effect on the downstream activities of ER binding, the antagonism of ER is not likely to be a contributing factor in the disease prevention. Moreover, soya isoflavones cannot inhibit aromatase (CYP19), which is the enzyme responsible for oestrogen synthesis. In the present review various cellular activities altered by soya isoflavones are discussed
Collapse
|
47
|
Abstract
Estrogen deficiency is one of the most frequent causes of osteoporosis in women and a possible cause of bone loss in men. But the mechanism involved remains largely unknown. Estrogen deficiency leads to an increase in the immune function, which culminates in an increased production of tumor necrosis factor (TNF) by activated T cells. TNF increases osteoclast formation and bone resorption both directly and by augmenting the sensitivity of maturing osteoclasts to the essential osteoclastogenic factor RANKL (the RANK ligand). Increased T cell production of TNF is induced by estrogen deficiency via a complex mechanism mediated by antigen presenting cells and the cytokines IFNγ, IL-7 and transforming growth factor-β. The experimental evidence that suggests that estrogen prevents bone loss by regulating T cell function and the interactions between immune cells and bone is reviewed here.
Collapse
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
48
|
Abstract
There is still an unresolved paradox with respect to the immunomodulating role of estrogens. On one side, we recognize inhibition of bone resorption and suppression of inflammation in several animal models of chronic inflammatory diseases. On the other hand, we realize the immunosupportive role of estrogens in trauma/sepsis and the proinflammatory effects in some chronic autoimmune diseases in humans. This review examines possible causes for this paradox. This review delineates how the effects of estrogens are dependent on criteria such as: 1) the immune stimulus (foreign antigens or autoantigens) and subsequent antigen-specific immune responses (e.g., T cell inhibited by estrogens vs. activation of B cell); 2) the cell types involved during different phases of the disease; 3) the target organ with its specific microenvironment; 4) timing of 17beta-estradiol administration in relation to the disease course (and the reproductive status of a woman); 5) the concentration of estrogens; 6) the variability in expression of estrogen receptor alpha and beta depending on the microenvironment and the cell type; and 7) intracellular metabolism of estrogens leading to important biologically active metabolites with quite different anti- and proinflammatory function. Also mentioned are systemic supersystems such as the hypothalamic-pituitary-adrenal axis, the sensory nervous system, and the sympathetic nervous system and how they are influenced by estrogens. This review reinforces the concept that estrogens have antiinflammatory but also proinflammatory roles depending on above-mentioned criteria. It also explains that a uniform concept as to the action of estrogens cannot be found for all inflammatory diseases due to the enormous variable responses of immune and repair systems.
Collapse
Affiliation(s)
- Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrino-Immunology, Division of Rheumatology, Department of Internal Medicine I, University Hospital, 93042 Regensburg, Germany.
| |
Collapse
|
49
|
Michael H, Härkönen PL, Kangas L, Väänänen HK, Hentunen TA. Differential effects of selective oestrogen receptor modulators (SERMs) tamoxifen, ospemifene and raloxifene on human osteoclasts in vitro. Br J Pharmacol 2007; 151:384-95. [PMID: 17420779 PMCID: PMC2013975 DOI: 10.1038/sj.bjp.0707232] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Several selective oestrogen receptor modulators (SERMs) with oestrogen agonist effects in bone cells and without increased risk of breast and endometrial cancer have been developed. Here, we have investigated the effects of different types of SERMs on osteoclast differentiation, bone resorption and apoptosis in vitro. EXPERIMENTAL APPROACH Human peripheral blood-derived CD14+ monocytes were cultured on bovine bone slices in the presence of RANKL, M-CSF, TNF-alpha and dexamethasone for seven days. Also, CD14+ monocytes were co-cultured either with human SaOS-2 or MG-63 osteosarcoma cells, in the presence of parathyroid hormone. Osteoclast cultures were treated with different SERMs. TRACP+ multinucleated cells and C-terminal telopeptide of type I collagen were used as markers for osteoclast formation and bone resorption, respectively. KEY RESULTS In CD14+ monocyte cultures, tamoxifen directly inhibited human osteoclast formation and bone resorption, while raloxifene and ospemifene had no inhibitory effect. In the co-cultures either with SaOS-2 or MG-63 cells, ospemifene and raloxifene as well as tamoxifen inhibited osteoclast formation in a concentration-dependent manner. The inhibitory effect was associated with an increased production of osteoprotegerin. The anti-oestrogen ICI 182 780 completely reversed the effects of these SERMs. CONCLUSION AND IMPLICATIONS Tamoxifen had an oestrogen receptor dependent, direct, inhibitory effect on human osteoclast differentiation and bone resorption, whereas ospemifene and raloxifene required osteoblastic cells to achieve a similar inhibition. The effects of ospemifene and raloxifene were mediated by oestrogen receptors by a mechanism involving paracrine induction of osteoprotegerin in cultures with osteoblast derived osteosarcoma cells.
Collapse
Affiliation(s)
- H Michael
- Department of Anatomy, Institute of Biomedicine, University of Turku Turku, Finland
| | - P L Härkönen
- Department of Anatomy, Institute of Biomedicine, University of Turku Turku, Finland
- Department of Laboratory Medicine, Tumor Biology, MAS University Hospital, Lund University Malmö, Sweden
| | - L Kangas
- Hormos Medical Corp., Itäinen Pitkäkatu 4B Turku, Finland
| | - H K Väänänen
- Department of Anatomy, Institute of Biomedicine, University of Turku Turku, Finland
| | - T A Hentunen
- Department of Anatomy, Institute of Biomedicine, University of Turku Turku, Finland
- Author for correspondence:
| |
Collapse
|
50
|
Wang TH, Xiang QL, Chen JW, Pan H, Cui YH. Raloxifene plus 17beta-estradiol inhibits proliferation of primary cultured vascular smooth muscle cells and human mammary endothelial cells via the janus kinase/signal transducer and activator of transcription3 cascade. Eur J Pharmacol 2007; 561:7-13. [PMID: 17336289 DOI: 10.1016/j.ejphar.2007.01.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2006] [Revised: 12/25/2006] [Accepted: 01/08/2007] [Indexed: 11/16/2022]
Abstract
Long-term use of estrogen replacement therapy increases the risk of breast cancer. Presently, we investigated the effects and mechanisms of Raloxifene, a second generation selective estrogen receptor modulator, plus 17beta-estradiol on the proliferation of primary cultured vascular smooth muscle cells (VSMC) and human mammary endothelial cells (HMEC). Raloxifene plus 17beta-estradiol inhibited angiotensin II-induced VSMC proliferation and rapid phosphorylation of STAT(3); these effects were blocked by AG490, the janus kinase/signal transducer and activator of transcription3 (JAK/STAT(3)) inhibitor. STAT(3) production was not affected. In primary cultured HMEC, immunofluorescence identified the ERbeta subtype, but not the ERalpha subtype, in the nucleus. Raloxifene plus 17beta-estradiol inhibited 17beta-estradiol-induced proliferation of HMEC. Western blot analysis established that Raloxifene attenuated the 17beta-estradiol-induced phosphorylation of STAT(3), and that this effect was blocked by AG490. We conclude that Raloxifene plus 17beta-estradiol inhibits the proliferation of VSMC and HMEC through the JAK/STAT(3) cascade, which in primary cultured HMEC may be implemented through ERbeta.
Collapse
Affiliation(s)
- Ting Huai Wang
- Department of Physiology, Sun Yat-Sen Medical College, Sun Yat-Sen University, Guang Zhou 510080, People's Republic of China.
| | | | | | | | | |
Collapse
|