1
|
Tang Y, Lu S, Wei J, Xu R, Zhang H, Wei Q, Han B, Gao Y, Zhao X, Peng S, Pan M, Ma B. Growth differentiation factor 9 regulates the expression of estrogen receptors via Smad2/3 signaling in goat cumulus cells. Theriogenology 2024; 219:65-74. [PMID: 38402699 DOI: 10.1016/j.theriogenology.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Both oocyte secretory factors (OSFs) and estrogen are essential for the development and function of mammalian ovarian follicles, playing synergistic role in regulating oocyte growth. OSFs can significantly affect the biological processes regulated by estrogen in cumulus cells (CCs). It is a scientific question worth investigating whether oocyte secretory factors can influence the expression of estrogen receptors in CCs. In our study, we observed a significant increase in the mRNA and protein expressions of estrogen receptor β (Esr2/ERβ) and G-protein-coupled estrogen receptor (GPER) in cumulus cells of goat cumulus-oocyte complexes (COCs) cultured in vitro for 6 h. Furthermore, the addition of 10 ng/mL growth-differentiation factor 9 (GDF9) and 5 ng/mL bone morphogenetic protein 15 (BMP15) to the culture medium of goat COCs resulted in a significant increase in the expressions of ERβ and GPER in cumulus cells. To explore the mechanism further, we performed micromanipulation to remove oocyte contents and co-cultured the oocytectomized complexes (OOXs) with denuded oocytes (DOs) or GDF9/BMP15. The expressions of ERβ and GPER in the co-culture groups were significantly higher than those in the OOXs group, but there was no difference compared to the COCs group. Mechanistically, we found that SB431542 (inhibitor of GDF9 bioactivity), but not LDN193189 (inhibitor of BMP15 bioactivity), abolished the upregulation of ERβ and GPER in cumulus cells and the activation of Smad2/3 signaling. In conclusion, our results demonstrate that the oocyte secretory factor GDF9 promotes the activation of Smad2/3 signaling in cumulus cells during goat COCs culture in vitro, and the phosphorylation of Smad2/3 induces the expression of estrogen receptors ERβ and GPER in cumulus cells.
Collapse
Affiliation(s)
- Yaju Tang
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Sihai Lu
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Juncai Wei
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Rui Xu
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Hui Zhang
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Qiang Wei
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Bin Han
- Yulin Animal Husbandry and Veterinary Service Center, Yulin, 719000, Shaanxi, PR China
| | - Yan Gao
- Yulin Animal Husbandry and Veterinary Service Center, Yulin, 719000, Shaanxi, PR China
| | - Xiaoe Zhao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Sha Peng
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Menghao Pan
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Baohua Ma
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, PR China.
| |
Collapse
|
2
|
Li LH, Ling DD, Lin H, Wang ZC, Sun ZR, Zhang YQ, Yang L, Zhang J, Cao H. Ovariectomy induces hyperalgesia accompanied by upregulated estrogen receptor α and protein kinase B in the rat spinal cord. Physiol Behav 2023; 271:114342. [PMID: 37673233 DOI: 10.1016/j.physbeh.2023.114342] [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: 06/27/2023] [Revised: 08/20/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
Hormone supplementation is one of the common therapies for menopause-related disorders. Among different tools, the ovariectomy (OVX) rodents are widely accepted as an appropriate menopausal pain model. Our previous study has showed that OVX produces a significant pain facilitation in both acute pain and tonic pain, however, the underlying mechanisms remain unclear. In this study, we examined the effects of OVX treatment and estradiol (E2) supplementation on formalin-induced nociceptive responses, and explored the associated spinal mechanisms. Female Sprague-Dawley rats underwent bilateral OVX, and E2 supplementation was given subcutaneously from the 5th week after surgery (30 μg/day for 7 days). Our results showed that formalin-induced nociceptive behaviors did not differ between diestrus and proestrus stages of the estrous in intact rats. However, OVX exacerbated formalin-evoked inflammatory pain, especially in the late phase at 4-5 weeks but not 2 weeks post-surgery. E2 supplementation significantly reversed the OVX-triggered hyperalgesia. Double immunofluorescence staining revealed that both ERα and ERβ in the spinal dorsal horn were co-labeled with the neuronal markers, but not with markers of astrocytes or microglia. The spinal ERα (but not ERβ) expression significantly increased in the OVX group, which was reversed by E2 supplementation. Moreover, the OVX individuals showed an increased protein kinase B (AKT) level in lumbar spinal cord, and E2 supplementation diminished the AKT expression in OVX rats. Finally, intrathecal injection Wortmannin, an inhibitor for AKT signaling, effectively reduced the nociceptive behaviors in the late phase and the number of c-fos positive cells. Together, our findings indicate that E2 supplementation alleviates the OVX-induced hyperalgesia, which might be involved in spinal ERα and AKT mechanisms.
Collapse
Affiliation(s)
- Li-Hong Li
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Dan-Dan Ling
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Hong Lin
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhe-Chen Wang
- Department of Psychology, School of Social Development and Public Policy, Fudan University, Shanghai 200032, China
| | - Zhi-Rong Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Liu Yang
- Shanghai Dunlu Biomedical Technology Co. Ltd. Shanghai 201611, China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China.
| | - Hong Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Wang M, Yang Y, Xu Y. Brain nuclear receptors and cardiovascular function. Cell Biosci 2023; 13:14. [PMID: 36670468 PMCID: PMC9854230 DOI: 10.1186/s13578-023-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Brain-heart interaction has raised up increasing attentions. Nuclear receptors (NRs) are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of cardiovascular diseases (CVDs), including hypertension, heart failure, atherosclerosis, etc. In this review, we will elaborate recent findings that have established the physiological relevance of brain NRs in the context of cardiovascular function. In addition, we will discuss the currently available evidence regarding the distinct neuronal populations that respond to brain NRs in the cardiovascular control. These findings suggest connections between cardiac control and brain dynamics through NR signaling, which may lead to novel tools for the treatment of pathological changes in the CVDs.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yongjie Yang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
4
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
5
|
Molecular dissection of cellular response of pancreatic islet cells to Bisphenol-A (BPA): a comprehensive review. Biochem Pharmacol 2022; 201:115068. [DOI: 10.1016/j.bcp.2022.115068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
|
6
|
Tsugami Y, Wakasa H, Nishimura T, Kobayashi K. Genistein Directly Represses the Phosphorylation of STAT5 in Lactating Mammary Epithelial Cells. ACS OMEGA 2021; 6:22765-22772. [PMID: 34514247 PMCID: PMC8427774 DOI: 10.1021/acsomega.1c03107] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/16/2021] [Indexed: 06/13/2023]
Abstract
Genistein is a soy isoflavone and shows various physiological activities, such as affinities for estrogen receptors (ERs) and inhibitory effects on the epidermal growth factor receptor (EGFR) pathway. A previous study reported that genistein downregulates milk production ability in mammary epithelial cells (MECs) while decreasing the phosphorylation of STAT5. The ER and EGFR pathways indirectly regulate STAT5. In this study, the repressing mechanism of genistein against the phosphorylation of STAT5 was investigated using a culture model of mouse MECs with milk production ability. The results revealed that genistein did not influence the behavior of ERα and ERβ, whereas genistein immediately repressed the phosphorylation of ERK1/2. However, the decrease in phosphorylated STAT5 occurred independent of the phosphorylation of EGFR. Genistein repressed new phosphorylation of STAT5 by prolactin without influencing the phosphorylation of JAK2. In conclusion, this study indicates that genistein directly inhibits the phosphorylation of STAT5 in lactating MECs.
Collapse
Affiliation(s)
- Yusaku Tsugami
- Laboratory
of Animal Histophysiology, Graduate School of Integrated Sciences
for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima 739-8528 Hiroshima, Japan
| | - Haruka Wakasa
- Laboratory
of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9 060-8589 Sapporo, Japan
| | - Takanori Nishimura
- Laboratory
of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9 060-8589 Sapporo, Japan
| | - Ken Kobayashi
- Laboratory
of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9 060-8589 Sapporo, Japan
| |
Collapse
|
7
|
Hevener AL, Ribas V, Moore TM, Zhou Z. The Impact of Skeletal Muscle ERα on Mitochondrial Function and Metabolic Health. Endocrinology 2020; 161:5735479. [PMID: 32053721 PMCID: PMC7017798 DOI: 10.1210/endocr/bqz017] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 02/11/2020] [Indexed: 12/16/2022]
Abstract
The incidence of chronic disease is elevated in women after menopause. Increased expression of ESR1 (the gene that encodes the estrogen receptor alpha, ERα) in muscle is highly associated with metabolic health and insulin sensitivity. Moreover, reduced muscle expression levels of ESR1 are observed in women, men, and animals presenting clinical features of the metabolic syndrome (MetSyn). Considering that metabolic dysfunction elevates chronic disease risk, including type 2 diabetes, heart disease, and certain cancers, treatment strategies to combat metabolic dysfunction and associated pathologies are desperately needed. This review will provide published work supporting a critical and protective role for skeletal muscle ERα in the regulation of mitochondrial function, metabolic homeostasis, and insulin action. We will provide evidence that muscle-selective targeting of ERα may be effective for the preservation of mitochondrial and metabolic health. Collectively published findings support a compelling role for ERα in the control of muscle metabolism via its regulation of mitochondrial function and quality control. Studies identifying ERα-regulated pathways essential for disease prevention will lay the important foundation for the design of novel therapeutics to improve metabolic health of women while limiting secondary complications that have historically plagued traditional hormone replacement interventions.
Collapse
Affiliation(s)
- Andrea L Hevener
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, University of California, Los Angeles, California
- Iris Cantor-UCLA Women’s Health Research Center, University of California, Los Angeles, California
- Correspondence: Andrea L. Hevener, PhD, University of California, Los Angeles, David Geffen School of Medicine, Division of Endocrinology, Diabetes, and Hypertension, 650 Charles E. Young Drive, CHS Suite 34-115B, Los Angeles, California 90095–7073. E-mail:
| | - Vicent Ribas
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, University of California, Los Angeles, California
- Current Affiliation: Vicent Ribas, Department of cell death and proliferation Instituto de Investigaciones Biomédicas de Barcelona, (IIBB-CSIC) Spanish National Research Council C/Rosselló 179, 6th floor 08036, Barcelona Spain
| | - Timothy M Moore
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, University of California, Los Angeles, California
| | - Zhenqi Zhou
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, University of California, Los Angeles, California
| |
Collapse
|
8
|
Hevener AL, Zhou Z, Moore TM, Drew BG, Ribas V. The impact of ERα action on muscle metabolism and insulin sensitivity - Strong enough for a man, made for a woman. Mol Metab 2018; 15:20-34. [PMID: 30005878 PMCID: PMC6066787 DOI: 10.1016/j.molmet.2018.06.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/16/2018] [Accepted: 06/18/2018] [Indexed: 12/25/2022] Open
Abstract
Background The incidence of chronic disease is elevated in women after menopause. Natural variation in muscle expression of the estrogen receptor (ER)α is inversely associated with plasma insulin and adiposity. Moreover, reduced muscle ERα expression levels are observed in women and animals presenting clinical features of the metabolic syndrome (MetSyn). Considering that metabolic dysfunction impacts nearly a quarter of the U.S. adult population and elevates chronic disease risk including type 2 diabetes, heart disease, and certain cancers, treatment strategies to combat metabolic dysfunction and associated pathologies are desperately needed. Scope of the review This review will provide evidence supporting a critical and protective role for skeletal muscle ERα in the regulation of metabolic homeostasis and insulin sensitivity, and propose novel ERα targets involved in the maintenance of metabolic health. Major conclusions Studies identifying ERα-regulated pathways essential for disease prevention will lay the important foundation for the rational design of novel therapeutics to improve the metabolic health of women while limiting secondary complications that have plagued traditional hormone replacement interventions.
Collapse
Affiliation(s)
- Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Zhenqi Zhou
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Timothy M Moore
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Brian G Drew
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Vicent Ribas
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
9
|
Hevener AL, Zhou Z, Drew BG, Ribas V. The Role of Skeletal Muscle Estrogen Receptors in Metabolic Homeostasis and Insulin Sensitivity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:257-284. [PMID: 29224099 DOI: 10.1007/978-3-319-70178-3_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Women in the modern era are challenged with facing menopausal symptoms as well as heightened disease risk associated with increasing adiposity and metabolic dysfunction for up to three decades of life. Treatment strategies to combat metabolic dysfunction and associated pathologies have been hampered by our lack of understanding regarding the biological causes of these clinical conditions and our incomplete understanding regarding the effects of estrogens and the tissue-specific functions and molecular actions of its receptors. In this chapter we provide evidence supporting a critical and protective role for skeletal muscle estrogen receptor α in the maintenance of metabolic homeostasis and insulin sensitivity. Studies identifying the critical ER-regulated pathways essential for disease prevention will lay the important foundation for the rational design of novel therapeutic strategies to improve the health of women while limiting secondary complications that have plagued traditional hormone replacement interventions.
Collapse
Affiliation(s)
- Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Zhenqi Zhou
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Brian G Drew
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Vicent Ribas
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Estrogen Receptor Signaling and the PI3K/Akt Pathway Are Involved in Betulinic Acid-Induced eNOS Activation. Molecules 2016; 21:molecules21080973. [PMID: 27463705 PMCID: PMC6273205 DOI: 10.3390/molecules21080973] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 11/16/2022] Open
Abstract
Betulinic acid (BA) is a naturally occurring pentacyclic triterpenoid with anti-inflammatory, antiviral and anti-cancer properties. Beneficial cardiovascular effects such as increased nitric oxide (NO) production through enhancement of endothelial NO synthase (eNOS) activity and upregulation of eNOS expression have been demonstrated for this compound. In the present study, immortalized human EA.hy 926 endothelial cells were incubated for up to 1 h with 1–100 µM BA and with the phosphatidylinositol-3-kinase (PI3K) inhibitors LY294002 and wortmannin, or the estrogen receptor (ER) antagonist ICI 182,780. Phosphorylation status of eNOS and total eNOS protein were analyzed by Western blotting using a serine 1177 phosphosite-specific antibody. Bioactive NO production was assessed by determination of cGMP content in rat lung fibroblasts (RFL-6) reporter cells. Short-term incubation of EA.hy 926 cells with BA resulted in eNOS phosphorylation at the serine 1177 residue in a concentration- and time-dependent manner with a half-maximal effective concentration of 0.57 µM. This was associated with an enhanced production of NO. BA-induced eNOS phosphorylation and NO production was completely blocked by pretreatment with ICI 182,780, and was attenuated by pretreatment with the PI3K inhibitors wortmannin and LY294002. These results indicate that fast non-genomic effects of ER with downstream signaling through the PI3K/Akt pathway and consecutive eNOS phosphorylation at serine 1177 are involved in BA-induced eNOS activation.
Collapse
|
11
|
Hevener AL, Clegg DJ, Mauvais-Jarvis F. Impaired estrogen receptor action in the pathogenesis of the metabolic syndrome. Mol Cell Endocrinol 2015; 418 Pt 3:306-21. [PMID: 26033249 PMCID: PMC5965692 DOI: 10.1016/j.mce.2015.05.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 12/13/2022]
Abstract
Considering the current trends in life expectancy, women in the modern era are challenged with facing menopausal symptoms as well as heightened disease risk associated with increasing adiposity and metabolic dysfunction for up to three decades of life. Treatment strategies to combat metabolic dysfunction and associated pathologies have been hampered by our lack of understanding regarding the biological underpinnings of these clinical conditions and our incomplete understanding of the effects of estrogens and the tissue-specific functions and molecular actions of its receptors. In this review we provide evidence supporting a critical and protective role for the estrogen receptor α specific form in the maintenance of metabolic homeostasis and insulin sensitivity. Studies identifying the ER-regulated pathways required for disease prevention will lay the important foundation for the rational design of targeted therapeutics to improve women's health while limiting complications that have plagued traditional hormone replacement interventions.
Collapse
Affiliation(s)
- Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, Iris Cantor-UCLA Women's Health Center, University of California, Los Angeles, CA 90095, USA.
| | - Deborah J Clegg
- Department of Biomedical Sciences, Diabetes and Obesity Research Institute Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Franck Mauvais-Jarvis
- Section of Endocrinology, Department of Medicine Tulane University, Health Science Center New Orleans, New Orleans, LA 70112, USA
| |
Collapse
|
12
|
Efstathiadou ZA, Sakka C, Polyzos SA, Goutou M, Stakias N, Bargiota A, Koukoulis GN. Associations of estrogen receptor alpha and Beta gene polymorphisms with lipid levels and insulin resistance in men. Metabolism 2015; 64:611-7. [PMID: 25665486 DOI: 10.1016/j.metabol.2015.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/29/2014] [Accepted: 01/16/2015] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The association of four single nucleotide polymorphisms in estrogen receptor alpha (ESR1) and beta (ESR2) genes with lipid levels and insulin resistance in men. DESIGN AND METHODS Lipids, glucose, insulin and HOMA-IR were determined, in a population-based, cross-sectional, cohort of 170 apparently healthy middle-aged Greek men, along with body mass index (BMI), waist circumference (WC) and percentage of body fat content (%fat). Genotyping of ESR1 for PvuII and XbaI and ESR2 for RsaI and AluI polymorphisms was performed. RESULTS Associations of AluI with LDL-Chol (mean ± SD, aa 4.3 ± 1.1 vs. Aa 3.7 ± 1.0 and ΑΑ 4.2 ± 1.1, p = 0.023) and RsaI with HOMA-IR [median (IQR), RR 1.55 (0.88-2.49) vs. Rr/rr 1.69 (0.72-2.29), p = 0.032] were found. Synergistic effects of RsaI and AluI of ESR2 gene on LDL-Chol levels, %fat and WC, as well as a synergistic effect of both ESR1 and ESR2 genes on levels of TChol (p = 0.01) and LDL-Chol (p = 0.027) were also shown. These findings remained significant after adjustment for potential confounders. Significant independent associations of PvuII with %fat (mean ± SD, pp 24.6 ± 5.3 vs Pp 22.4 ± 5.2 and PP 21.2 ± 6.7, p = 0.044), and RsaI with %fat (RR 22.6 ± 5.5 vs. Rr/rr 25.2 ± 6.3, p = 0.015) and WC (mean ± SD, RR 97.4 ± 10.4 vs. Rr/rr 102.6 ± 12.6, p = 0.013) were found. Synergistic effects on %fat, between the ESR1 polymorphisms (p = 0.004), between the ESR2 polymorphisms and among all four ESR polymorphisms studied were also present. CONCLUSIONS ESR2 is associated with LDL-Chol levels and HOMA-IR in men independently of confounders. Body fat is affected by both genes. Furthermore, a synergistic effect of ESR1 and ESR2 on TChol, LDL-Chol and %fat, was shown.
Collapse
Affiliation(s)
- Zoe A Efstathiadou
- Endocrine Clinic, "Hippokration" General Hospital of Thessaloniki, Thessaloniki, Greece.
| | - Christina Sakka
- Department of Endocrinology, University of Thessaly School of Medicine, Larissa, Greece
| | - Stergios A Polyzos
- Endocrine Clinic, "Hippokration" General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Maria Goutou
- Department of Endocrinology, University of Thessaly School of Medicine, Larissa, Greece
| | - Nikolaos Stakias
- Department of Endocrinology, University of Thessaly School of Medicine, Larissa, Greece
| | - Alexandra Bargiota
- Department of Endocrinology, University of Thessaly School of Medicine, Larissa, Greece
| | - George N Koukoulis
- Department of Endocrinology, University of Thessaly School of Medicine, Larissa, Greece
| |
Collapse
|
13
|
Wakui S, Shirai M, Motohashi M, Mutou T, Oyama N, Wempe MF, Takahashi H, Inomata T, Ikegami M, Endou H, Asari M. Effects of in utero exposure to di(n-butyl) phthalate for estrogen receptors α, β, and androgen receptor of Leydig cell on rats. Toxicol Pathol 2014; 42:877-87. [PMID: 24067674 DOI: 10.1177/0192623313502879] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Estrogens and androgens affect male and female reproductive systems. Recently, we reported that prenatal di(n-butyl) phthalate (DBP) exposure induced atypical Leydig cells (LCs) hyperplasia during adulthood. The present study investigated the expression of estrogen receptor α (ERα), estrogen receptor β (ERβ), and androgen receptor (AR) in LCs of 5-, 7-, 9-, 14-, and 17-week-old Sprague-Dawley (srl) rats whose dams had been administered DBP intragastrically at 100 mg/kg/day or the vehicle (corn oil) from days 12 to 21 postconception. Immunohistochemical, Western blotting, and reverse transcription polymerase chain reaction analyses revealed that the expressions of ERα, ERβ, and AR proteins and mRNAs in the DBP group were similar to those of the vehicle group at 5 and 7 weeks, but significantly higher ERα and lower ERβ and AR levels were observed in the DBP group at 9 to 17 weeks. The rats prenatally exposed to DBP had seminiferous tubule degeneration and atypical hyperplasia of LCs during adulthood, which was associated with an increase in expression of ERα and a decrease of ERβ and AR in the testis.
Collapse
Affiliation(s)
- Shin Wakui
- Department of Toxicology, Laboratory Animal Science and Veterinary Anatomy, Azabu University School of Veterinary Medicine, Kanagawa, Japan
| | - Masaru Shirai
- Department of Toxicology, Laboratory Animal Science and Veterinary Anatomy, Azabu University School of Veterinary Medicine, Kanagawa, Japan
| | - Masaya Motohashi
- Department of Toxicology, Laboratory Animal Science and Veterinary Anatomy, Azabu University School of Veterinary Medicine, Kanagawa, Japan
| | | | | | - Michael F Wempe
- School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomoo Inomata
- Department of Toxicology, Laboratory Animal Science and Veterinary Anatomy, Azabu University School of Veterinary Medicine, Kanagawa, Japan
| | - Masahiro Ikegami
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Masao Asari
- Department of Toxicology, Laboratory Animal Science and Veterinary Anatomy, Azabu University School of Veterinary Medicine, Kanagawa, Japan
| |
Collapse
|
14
|
Obradovic M, Stewart AJ, Pitt SJ, Labudovic-Borovic M, Sudar E, Petrovic V, Zafirovic S, Maravic-Stojkovic V, Vasic V, Isenovic ER. In vivo effects of 17β-estradiol on cardiac Na(+)/K(+)-ATPase expression and activity in rat heart. Mol Cell Endocrinol 2014; 388:58-68. [PMID: 24662727 DOI: 10.1016/j.mce.2014.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 03/15/2014] [Accepted: 03/15/2014] [Indexed: 12/24/2022]
Abstract
In this study the in vivo effects of estradiol in regulating Na(+)/K(+)-ATPase function in rat heart was studied. Adult male Wistar rats were treated with estradiol (40μg/kg, i.p.) and after 24h the animals were sacrificed and the heart excised. Following estradiol administration, cardiac Na(+)/K(+)-ATPase activity, expression of the α1 subunit, and phosphorylation of the α1 subunit were significantly increased. These animals also had significantly decreased levels of digoxin-like immunoreactive factor(s). Na(+) levels were also significantly reduced but to a level that was still within the normal physiological range, highlighting the ability of the Na(+)/K(+)-ATPase to balance the ionic composition following treatment with estradiol. Estradiol treated rats also showed increased phosphorylation of protein kinase B (Akt), and extracellular-signal-regulated kinase 1/2 (ERK1/2). We therefore suggest a role for Akt and/or ERK1/2 in estradiol-mediated regulation of cardiac Na(+)/K(+)-ATPase expression and activity in rat heart.
Collapse
Affiliation(s)
- Milan Obradovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Alan J Stewart
- School of Medicine, University of St Andrews, North Haugh, St Andrews KY16 9TF, United Kingdom.
| | - Samantha J Pitt
- School of Medicine, University of St Andrews, North Haugh, St Andrews KY16 9TF, United Kingdom.
| | - Milica Labudovic-Borovic
- Institute of Histology and Embryology "Aleksandar Đ. Kostić", Faculty of Medicine, University of Belgrade, Visegradska 26, 11000 Belgrade, Serbia.
| | - Emina Sudar
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Voin Petrovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Physical Chemistry, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Sonja Zafirovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Vera Maravic-Stojkovic
- Dedinje Cardiovascular Institute, Belgrade University, School of Medicine, Heroja Milana Tepica 1, 11000 Belgrade, Serbia.
| | - Vesna Vasic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Physical Chemistry, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Esma R Isenovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| |
Collapse
|
15
|
Kypreos KE, Zafirovic S, Petropoulou PI, Bjelogrlic P, Resanovic I, Traish A, Isenovic ER. Regulation of endothelial nitric oxide synthase and high-density lipoprotein quality by estradiol in cardiovascular pathology. J Cardiovasc Pharmacol Ther 2014; 19:256-68. [PMID: 24414281 DOI: 10.1177/1074248413513499] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Estrogens have been recognized, in the last 3 decades, as important hormones in direct and indirect modulation of vascular health. In addition to their direct benefit on cardiovascular health, the presence of esterified estrogen in the lipid core of high-density lipoprotein (HDL) particles indirectly contributes to atheroprotection by significantly improving HDL quality and functionality. Estrogens modulate their physiological activity via genomic and nongenomic mechanisms. Genomic mechanisms are thought to be mediated directly by interaction of the hormone receptor complex with the hormone response elements that regulate gene expression. Nongenomic mechanisms are thought to occur via interaction of the estrogen with membrane-bound receptors, which rapidly activate intracellular signaling without binding of the hormone receptor complex to its hormone response elements. Estradiol in particular mediates early and late endothelial nitric oxide synthase (eNOS) activation via interaction with estrogen receptors through both nongenomic and genomic mechanisms. In the vascular system, the primary endogenous source of nitric oxide (NO) generation is eNOS. Nitric oxide primarily influences blood vessel relaxation, the heart rate, and myocyte contractility. The abnormalities in expression and/or functions of eNOS lead to the development of cardiovascular diseases, both in animals and in humans. Although considerable research efforts have been dedicated to understanding the mechanisms of action of estradiol in regulating cardiac eNOS, more research is needed to fully understand the details of such mechanisms. This review focuses on recent findings from animal and human studies on the regulation of eNOS and HDL quality by estradiol in cardiovascular pathology.
Collapse
Affiliation(s)
- Kyriakos E Kypreos
- 1Department of Medicine, University of Patras Medical School, Pharmacology Laboratory, Panepistimioupolis, Rio, Greece
| | | | | | | | | | | | | |
Collapse
|
16
|
Reichardt HM, Gold R, Lühder F. Glucocorticoids in multiple sclerosis and experimental autoimmune encephalomyelitis. Expert Rev Neurother 2014; 6:1657-70. [PMID: 17144780 DOI: 10.1586/14737175.6.11.1657] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glucocorticoids exert a variety of immunomodulatory activities. Since changes in glucocorticoid homeostasis impact on susceptibility to autoimmune diseases, and synthetic glucocorticoids are widely used in the treatment of multiple sclerosis, a detailed understanding of their mechanism of action is desirable. Experimental autoimmune encephalomyelitis is a common animal model that mirrors many hallmarks of multiple sclerosis, a chronic inflammatory disease of the CNS with presumed autoimmune origin. Experimental autoimmune encephalomyelitis has been instrumental for many years in studying multiple sclerosis, revealing the blood-brain barrier, the microglia and T-cell apoptosis as major targets of glucocorticoids in this disease. Despite the great advances in the field, the answers to many questions concerning the mechanism of glucocorticoids; for example, the contribution of nongenomic effects or the cell-type specificity of their action, remain elusive. This review will critically discuss what we have learned so far from the analysis of animal models of the molecular mode of therapeutic and endogenous glucocorticoid action in multiple sclerosis. With this knowledge in mind, we should be able to further improve the management of multiple sclerosis using this class of drugs.
Collapse
Affiliation(s)
- Holger M Reichardt
- University of Würzburg, Molecular Immunology, Institute for Virology and Immunobiology, Versbacher Strasse 7, 97078 Würzburg, Germany.
| | | | | |
Collapse
|
17
|
Mauvais-Jarvis F, Clegg DJ, Hevener AL. The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev 2013; 34:309-38. [PMID: 23460719 PMCID: PMC3660717 DOI: 10.1210/er.2012-1055] [Citation(s) in RCA: 823] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Estrogens play a fundamental role in the physiology of the reproductive, cardiovascular, skeletal, and central nervous systems. In this report, we review the literature in both rodents and humans on the role of estrogens and their receptors in the control of energy homeostasis and glucose metabolism in health and metabolic diseases. Estrogen actions in hypothalamic nuclei differentially control food intake, energy expenditure, and white adipose tissue distribution. Estrogen actions in skeletal muscle, liver, adipose tissue, and immune cells are involved in insulin sensitivity as well as prevention of lipid accumulation and inflammation. Estrogen actions in pancreatic islet β-cells also regulate insulin secretion, nutrient homeostasis, and survival. Estrogen deficiency promotes metabolic dysfunction predisposing to obesity, the metabolic syndrome, and type 2 diabetes. We also discuss the effect of selective estrogen receptor modulators on metabolic disorders.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA.
| | | | | |
Collapse
|
18
|
McCarthy PL, Paterno GD, Gillespie LL. Protein expression pattern of human MIER1 alpha, a novel estrogen receptor binding protein. J Mol Histol 2013; 44:469-79. [PMID: 23277184 DOI: 10.1007/s10735-012-9478-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 12/14/2012] [Indexed: 10/27/2022]
Abstract
MIER1 is a transcriptional regulator that exists as several isoforms. Of particular interest is the MIER1α isoform, which contains in its unique C-terminus an LXXLL motif for interaction with nuclear hormone receptors. Indeed, MIER1α has been shown to interact with ERα and inhibit estrogen-stimulated growth of breast carcinoma cells. Moreover, the subcellular localization of MIER1α changes dramatically, from nuclear to cytoplasmic, during progression to invasive breast carcinoma. While human MIER1 RNA and protein expression pattern data have been posted on several websites, none of these studies use probes or antibodies that distinguish between the α and β isoforms. We report here the first immunohistochemical study of the MIER1α protein expression pattern in human tissues. Our analysis revealed intense staining of specific cell types within virtually every endocrine and reproductive tissue except for the thyroid gland. In particular, we detected intense staining of ovarian follicles and germinal epithelium, ductal epithelial cells of the breast, pancreatic islet cells, all areas of the anterior pituitary and all zones of the adrenal cortex; moderate staining of germ cells and Leydig cells within the testis, patches of chromaffin cells in the adrenal medulla and weak staining of the fibromuscular stroma within the prostate. Immunoreactivity was limited to the cytoplasm in all positive cells except for oocytes and germinal epithelial cells in which the nucleus was also stained and in ductal epithelial cells of the breast in which staining was exclusively nuclear. In general, non-endocrine tissues were negative, however a few exceptions were noted. These included hepatocytes, myocardial fibers and neurons in all regions of the brain examined, with the exception of the thalamus. Neuronal staining was restricted to the cell bodies and dendrites, as most axons were negative. These data suggest that human MIER1α functions specifically in endocrine tissues and in a limited number of non-endocrine organs.
Collapse
Affiliation(s)
- Patti L McCarthy
- Terry Fox Cancer Research Labs, Division of BioMedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, A1B 3V6, Canada
| | | | | |
Collapse
|
19
|
Kampa M, Pelekanou V, Notas G, Stathopoulos EN, Castanas E. The estrogen receptor: two or more molecules, multiple variants, diverse localizations, signaling and functions. Are we undergoing a paradigm-shift as regards their significance in breast cancer? Hormones (Athens) 2013; 12:69-85. [PMID: 23624133 DOI: 10.1007/bf03401288] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Marilena Kampa
- Department of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Crete, Greece
| | | | | | | | | |
Collapse
|
20
|
Estradiol receptors agonists induced effects in rat intestinal microcirculation during sepsis. Microvasc Res 2012; 85:118-27. [PMID: 23063870 DOI: 10.1016/j.mvr.2012.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/02/2012] [Accepted: 10/03/2012] [Indexed: 01/26/2023]
Abstract
The steroid hormone estradiol is suggested to play a protective role in intestinal injury during systemic inflammation (sepsis). Our aim was to determine the effects of specific estradiol receptor (ER-α and ER-ß) agonists on the intestinal microcirculation during experimental sepsis. Male and sham ovariectomized female rats were subjected to sham colon ascendens stent peritonitis (CASP), and they were compared to male and ovariectomized female rats underwent CASP and either estradiol receptor α (ER-α) agonist propyl pyrazole triol (PPT), estradiol receptor ß (ER-ß) agonist diarylpropiolnitrile (DPN), or vehicle treatment. Intravital microscopy was performed, which is sufficiently sensitive to measure changes in the functional capillary density (FCD) as well as the major steps in leukocyte recruitment (rolling and adhesion). The leukocyte extravasations were also quantified by using histological paraffin sections of formalin fixed intestine. We found that either DPN (ER-β) or PPT (ER-α) significantly reduced (P<0.05) sepsis-induced leukocyte-endothelial interaction (rolling, adherent leukocytes and neutrophil extravasations) and improved the intestinal muscular FCD. [PPT: Female; Leukocyte rolling (n/min): V(3) 3.7±0.7 vs 0.8±0.2, Leukocyte adhesion(n/mm(2)): V(3) 131.3±22.6 vs 57.2±13.5, Neutrophil extravasations (n/10000 μm(2)): 3.1±0.7 vs 6 ±1. Male; Leukocyte adhesion (n/mm(2)): V(1) 154.8±19.2 vs 81.3±11.2, V(3) 115.5±23.1 vs 37.8±12]. [DPN: Female; neutrophil extravasations (n/10000 μm(2)) 3.8±0.6 vs 6 ±1. Male; Leukocyte adhesion (n/mm(2)) V(1) 154.8±19.2 vs 70±10.5, V(3) 115.5±23.1 vs 52.8±9.6].Those results suggest that the observed effects of estradiol receptors on different phases of leukocytes recruitment with the improvement of the functional capillary density could partially explain the previous demonstrated salutary effects of estradiol on the intestinal microcirculation during sepsis. The observed activity of this class of compounds could open up a new avenue of research into the potential treatment of sepsis.
Collapse
|
21
|
Wu KLH, Chen CH, Shih CD. Nontranscriptional activation of PI3K/Akt signaling mediates hypotensive effect following activation of estrogen receptor β in the rostral ventrolateral medulla of rats. J Biomed Sci 2012; 19:76. [PMID: 22897791 PMCID: PMC3438069 DOI: 10.1186/1423-0127-19-76] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 08/01/2012] [Indexed: 12/22/2022] Open
Abstract
Background Estrogen acts on the rostral ventrolateral medulla (RVLM), where sympathetic premotor neurons are located, to elicit vasodepressor effects via an estrogen receptor (ER)β-dependent mechanism. We investigated in the present study nontranscriptional mechanism on cardiovascular effects following activation of ERβ in the RVLM, and delineated the involvement of phosphatidylinositol 3-kinase (PI3K)/serine/threonine kinase (Akt) signaling pathway in the effects. Methods In male Sprague–Dawley rats maintained under propofol anesthesia, changes in arterial pressure, heart rate and sympathetic neurogenic vasomotor tone were examined after microinjection bilaterally into RVLM of 17β-estradiol (E2β) or a selective ERα or ERβ agonist. Involvement of ER subtypes and PI3K/Akt signaling pathway in the induced cardiovascular effects were studied using pharmacological tools of antagonists or inhibitors, gene manipulation with antisense oligonucleotide (ASON) or adenovirus-mediated gene transfection. Results Similar to E2β (1 pmol), microinjection of ERβ agonist, diarylpropionitrile (DPN, 1, 2 or 5 pmol), into bilateral RVLM evoked dose-dependent hypotension and reduction in sympathetic neurogenic vasomotor tone. These vasodepressive effects of DPN (2 pmol) were inhibited by ERβ antagonist, R,R-tetrahydrochrysene (50 pmol), ASON against ERβ mRNA (250 pmol), PI3K inhibitor LY294002 (5 pmol), or Akt inhibitor (250 pmol), but not by ERα inhibitor, methyl-piperidino-pyrazole (1 nmol), or transcription inhibitor, actinomycin D (5 or 10 nmol). Gene transfer by microinjection into bilateral RVLM of adenovirus encoding phosphatase and tensin homologues deleted on chromosome 10 (5 × 108 pfu) reversed the vasodepressive effects of DPN. Conclusions Our results indicate that vasodepressive effects following activation of ERβ in RVLM are mediated by nongenomic activation of PI3K/Akt signaling pathway. This study provides new insight in the intracellular signaling cascades involved in central vasodepressive functions of estrogen.
Collapse
Affiliation(s)
- Kay L H Wu
- Department of Pharmacy, Graduate Institute of Pharmaceutical Technology, Tajen University, 20 Weishin Road, Yanpu Township, Pingtung County 90741, Taiwan
| | | | | |
Collapse
|
22
|
Otto C, Kantner I, Nubbemeyer R, Schkoldow J, Fuchs I, Krahl E, Vonk R, Schüler C, Fritzemeier KH, Erben RG. Estradiol release kinetics determine tissue response in ovariectomized rats. Endocrinology 2012; 153:1725-33. [PMID: 22334713 DOI: 10.1210/en.2011-1801] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogen replacement is an effective therapy of postmenopausal symptoms such as hot flushes, bone loss, and vaginal dryness. Undesired estrogen effects are the stimulation of uterine and mammary gland epithelial cell proliferation as well as hepatic estrogenicity. In this study, we examined the influence of different estradiol release kinetics on tissue responsivity in ovariectomized (OVX) rats. Pulsed release kinetics was achieved by ip or sc administration of estradiol dissolved in physiological saline containing 10% ethanol (EtOH/NaCl) whereas continuous release kinetics was achieved by sc injection of estradiol dissolved in benzylbenzoate/ricinus oil (1+4, vol/vol). Initial 3-d experiments in OVX rats showed that pulsed ip estradiol administration had profoundly reduced stimulatory effects on the uterus and the liver compared with continuous release kinetics. On the other hand, both administration forms prevented severe vaginal atrophy. Based on these results, we compared the effects of pulsed (sc in EtOH/NaCl) vs. continuous (sc in benzylbenzoate/ricinus oil) estradiol release kinetics on bone, uterus, mammary gland, and liver in a 4-month study in OVX rats. Ovariectomy-induced bone loss was prevented by both administration regimes. However, pulsed estradiol resulted in lower uterine weight, reduced induction of hepatic gene expression, and reduced mammary epithelial hyperplasia relative to continuous estradiol exposure. We conclude that organ responsivity is influenced by different hormone release kinetics, a fact that might be exploited to reduce undesired estradiol effects in postmenopausal women.
Collapse
Affiliation(s)
- Christiane Otto
- TRG Oncology & Gynecological Therapy, Bayer Pharma AG, 13342 Berlin.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Tanaka R, Tsutsui H, Kobuchi S, Sugiura T, Yamagata M, Ohkita M, Takaoka M, Yukimura T, Matsumura Y. Protective effect of 17β-estradiol on ischemic acute kidney injury through the renal sympathetic nervous system. Eur J Pharmacol 2012; 683:270-5. [PMID: 22426161 DOI: 10.1016/j.ejphar.2012.02.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/13/2012] [Accepted: 02/26/2012] [Indexed: 10/28/2022]
Abstract
Enhanced renal sympathetic nerve activity during an ischemic period and renal venous norepinephrine overflow after reperfusion play important roles in the development of ischemic acute kidney injury. In this study, we examined the effect of 17β-estradiol on the renal sympathetic nervous system and kidney function in ischemia/reperfusion-induced acute kidney injury in anesthetized rats. Ischemic acute kidney injury was induced by clamping the left renal artery and vein for 45 min followed by reperfusion, 2 weeks after a contralateral nephrectomy. Intravenous injection of 17β-estradiol (100 μg/kg) 15 min before reperfusion suppressed enhanced renal sympathetic nerve activity during renal ischemia, also suppressed renal venous norepinephrine overflow after reperfusion, and attenuated ischemia/reperfusion-induced renal dysfunction with histological damage. The above renoprotective effects of 17β-estradiol were reversed by pretreatment with tamoxifen (5 mg/kg), an estrogen receptor antagonist, or N(G)-nitro-L-arginine methyl ester (0.3 mg/kg), a non-selective nitric oxide synthase inhibitor. These results indicate that 17β-estradiol can suppress enhanced renal sympathetic nerve activity during renal ischemia, and its consequent effect on norepinephrine overflow from nerve endings, by nitric oxide production via estrogen receptors. These effects appear to contribute to renoprotection against ischemia/reperfusion-induced renal injury.
Collapse
Affiliation(s)
- Ryosuke Tanaka
- Laboratory of Pathological and Molecular Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wallacides A, Chesnel A, Ajj H, Chillet M, Flament S, Dumond H. Estrogens promote proliferation of the seminoma-like TCam-2 cell line through a GPER-dependent ERα36 induction. Mol Cell Endocrinol 2012; 350:61-71. [PMID: 22138413 DOI: 10.1016/j.mce.2011.11.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 11/17/2011] [Accepted: 11/17/2011] [Indexed: 01/17/2023]
Abstract
Seminoma, originated from carcinoma in situ cells (CIS), is one of the main causes of cancer in young men. Postpubertal development of these testicular germ cell tumors suggests a hormone-sensitive way of CIS cell proliferation induction. Using the unique seminoma TCam-2 cell line, we demonstrate that both estradiol and testosterone can stimulate TCam-2 cell proliferation in the absence of the estradiol receptor ERα. We establish that estradiol can activate GPER-cAMP/PKA signalling pathway. TCam-2 cells express ERα36, a truncated isoform of the canonical ERα receptor, the expression of which is rapidly induced after estrogen treatment in a GPER-dependent manner. ERα36 knockdown indicates that ERα36 is (i) a downstream target of E(2)-activated GPER/PKA/CREB pathway, (ii) required for estradiol-dependent EGFR expression, (iii) necessary for cell proliferation. Colocalization of ERα36 with cytoskeleton microfilaments suggests a role of estrogens in cell motility. Our results highlight the functional role of ERα36 in context of seminoma cell proliferation and the importance of testing ERα36 in vivo as a possible future prognostic marker.
Collapse
Affiliation(s)
- Angelina Wallacides
- EA4421, Signalisation, Génomique et Recherche Translationnelle en Oncologie, Nancy-Université, Nancy, France
| | | | | | | | | | | |
Collapse
|
25
|
Differential vasoactive effects of oestrogen, oestrogen receptor agonists and selective oestrogen receptor modulators in rat middle cerebral artery. Neurosci Res 2011; 71:78-84. [PMID: 21624404 DOI: 10.1016/j.neures.2011.05.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 05/13/2011] [Accepted: 05/16/2011] [Indexed: 12/29/2022]
Abstract
Cerebrovascular disorders are less common in pre-menopausal than post-menopausal women and in females than males. This protection may be due, in part at least, to direct effects of oestrogens on blood vessels. Oestrogen's vasodilatory mechanisms have been reported to be via the endothelium, vascular smooth muscle and extracellular matrix, depending on the vascular bed studied. Herein we investigated the vasoactive effects of oestrogen, oestrogen receptor (ER) and GPR30 agonists and selective ER modulators (SERMs) in the rat middle cerebral artery(MCA), an artery affected in focal ischaemia. MCAs isolated from male Sprague Dawley rats were mounted on a wire myograph. Concentration response curves were constructed to 17β-oestradiol, ERα agonist-PPT, ERβ agonist-DPN, GPR30 agonist-G1 and novel SERMs (LY362321 and LY2120310) in pre-constricted vessels, in the presence and absence of endothelium, blocking agents for nitric oxide synthase (L-NAME), classic ER antagonist (ICI182,780) or plasma membrane specific ERα (ERα-36) antibody. 17β-oestradiol induced rapid vasorelaxation of the MCA which was not affected by endothelium removal, L-NAME or ICI182,780. Vasorelaxation was mimicked by PPT, DPN and G1 but not by the SERMs. Using ERα-36 antibody, effects of oestrogen were partially blocked. PPT had a greater vasorelaxation, while DPN and G1 had a lesser effect than 17β-oestradiol. These findings indicate that activation of plasma membrane bound ERα, β and GPR30 elicits rapid, endothelial-nitric oxide-independent relaxation of the rat MCA.
Collapse
|
26
|
Millas I, Liquidato BM, Buck HDS, Barros MD, Paes RAP, Dolci JEL. Evaluation of estrogenic receptors in the nasal mucosa of women taking oral contraceptives. Contraception 2010; 83:571-7. [PMID: 21570556 DOI: 10.1016/j.contraception.2010.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 08/11/2010] [Accepted: 09/15/2010] [Indexed: 10/18/2022]
Abstract
BACKGROUND There is evidence that the nasal mucosa is affected by estrogen influence. Some authors have already detected estrogen receptors (ER) in the nasal mucosa. However, there doesn't seem to be a consensus about the concentration and distribution of the ER or the possible influence of hormonal contraceptives in the nasal mucosa. OBJECTIVE The study was conducted to evaluate the influence of oral contraceptives on the distribution and concentration of estrogenic receptors in nasal mucosa. STUDY DESIGN Two groups of 32 women with regular menstrual cycles were selected. One group of women was taking oral contraceptives and the other was not. Samples of mucosa of inferior nasal turbinate were analyzed by immunohistochemical staining for alpha and beta ER. RESULTS The use of oral contraceptives induced a decrease of beta-receptors only in lamina propria cells. In both groups, there was a predominance of beta-receptors. CONCLUSION Women who took oral contraceptives showed a decrease of beta-receptors in some cells of the lamina propria. These findings show us the possibility of effects of contraceptive pills on the cells such as fibroblasts, mast cells, plasmocytes, and other inflammatory cells.
Collapse
Affiliation(s)
- Ieda Millas
- Faculty of Medical Sciences, Otorhinolaryngology Department of Santa Casa de São Paulo, São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
27
|
Pae HO, Son Y, Kim NH, Jeong HJ, Chang KC, Chung HT. Role of heme oxygenase in preserving vascular bioactive NO. Nitric Oxide 2010; 23:251-7. [PMID: 20713168 DOI: 10.1016/j.niox.2010.08.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 07/22/2010] [Accepted: 08/11/2010] [Indexed: 01/12/2023]
Abstract
Beyond its vasodilator role, vascular nitric oxide (NO), which is synthesized by endothelial NO synthase (eNOS) via its activation, has been shown to play a number of other beneficial roles in the vascular system; it inhibits proliferation of vascular smooth muscle cells, prevents platelet aggregation, and regulates endothelial apoptosis. Such beneficial roles have been shown to be implicated in the regulation of endothelial functions. A loss of NO bioavailability that may result either from decreased eNOS expression and activity or from increased NO degradation is associated with endothelial dysfunction, a key factor in the development of vascular diseases. Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. In the vascular system, HO-1 and heme degradation products perform important physiological functions, which are ultimately linked to the protection of vascular cells. Studies have shown that HO-1 and heme degradation products exert vasodilatory, antioxidant, anti-inflammatory, antiproliferative and anti-apoptotic effects on vascular cells. Interestingly, these effects of HO-1 and its by-products are similar, at least in part, to those of eNOS-derived NO; this similarity may prompt investigators to study a possible relationship between eNOS-derived NO and HO-1 pathways. Many studies have been reported, and accumulating evidence suggests that HO-1 and heme degradation products can improve vascular function, at least in part, by compensating for the loss of NO bioavailability. This paper will provide the possible pathway explaining how HO-1 and heme degradation products can preserve vascular NO.
Collapse
Affiliation(s)
- Hyun-Ock Pae
- Department of Microbiology and Immunology, Wonkwang University School of Medicine, Iksan 570-749, Republic of Korea
| | | | | | | | | | | |
Collapse
|
28
|
Ádori M, Kiss E, Barad Z, Barabás K, Kiszely E, Schneider A, Sziksz E, Ábrahám IM, Matkó J, Sármay G. Estrogen augments the T cell-dependent but not the T-independent immune response. Cell Mol Life Sci 2010; 67:1661-74. [PMID: 20140748 PMCID: PMC11115714 DOI: 10.1007/s00018-010-0270-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Revised: 01/06/2010] [Accepted: 01/13/2010] [Indexed: 11/25/2022]
Abstract
Estrogen plays a critical regulatory role in the development and maintenance of immunity. Its role in the regulation of antibody synthesis in vivo is still not completely clear. Here, we have compared the effect of estrogen on T cell-dependent (TD) and T cell-independent type 2 (TI-2) antibody responses. The results provide the first evidence that estrogen enhances the TD but not the TI-2 response. Ovariectomy significantly decreased, while estrogen re-administration increased the number of hapten-specific IgM- and IgG-producing cells in response to TD antigen. In vitro experiments also show that estrogen may have a direct impact on B and T cells by inducing rapid signaling events, such as Erk and AKT phosphorylation, cell-specific Ca(2+) signal, and NFkappaB activation. These non-transcriptional effects are mediated by classical estrogen receptors and partly by an as yet unidentified plasma membrane estrogen receptor. Such receptor- mediated rapid signals may modulate the in vivo T cell-dependent immune response.
Collapse
Affiliation(s)
- Mónika Ádori
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Endre Kiss
- Immunology Research Group of the Hungarian Academy of Sciences, Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsanna Barad
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
- Department of Physiology, Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Klaudia Barabás
- Proteomics Laboratory, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Edda Kiszely
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Andrea Schneider
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Erna Sziksz
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - István M. Ábrahám
- Department of Physiology, Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - János Matkó
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
- Immunology Research Group of the Hungarian Academy of Sciences, Eötvös Loránd University, Budapest, Hungary
| | - Gabriella Sármay
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
- Immunology Research Group of the Hungarian Academy of Sciences, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
29
|
Abstract
As women enter menopause, the concentration of estrogen and other female hormones declines. This hormonal decrease has been associated with a number of negative outcomes, including a greater incidence of injury as well as a delay in recovery from these injuries. Over the past two decades, our understanding of the protective effects of estrogen against various types of injury and disease states has grown immensely. In skeletal muscle, studies with animals have demonstrated that sex and estrogen may potentially influence muscle contractile properties and attenuate indices of post-exercise muscle damage, including the release of creatine kinase into the bloodstream and activity of the intramuscular lysosomal acid hydrolase, beta-glucuronidase. Furthermore, numerous studies have revealed an estrogen-mediated attenuation of infiltration of inflammatory cells such as neutrophils and macrophages into the skeletal muscles of rats following exercise or injury. Estrogen has also been shown to play a significant role in stimulating muscle repair and regenerative processes, including the activation and proliferation of satellite cells. Although the mechanisms by which estrogen exerts its influence upon indices of skeletal muscle damage, inflammation and repair have not been fully elucidated, it is thought that estrogen may potentially exert its protective effects by: (i) acting as an antioxidant, thus limiting oxidative damage; (ii) acting as a membrane stabilizer by intercalating within membrane phospholipids; and (iii) binding to estrogen receptors, thus governing the regulation of a number of downstream genes and molecular targets. In contrast to animal studies, studies with humans have not as clearly delineated an effect of estrogen on muscle contractile function or on indices of post-exercise muscle damage and inflammation. These inconsistencies have been attributed to a number of factors, including age and fitness level of subjects, the type and intensity of exercise protocols, and a focus on sex differences that typically involve factors and hormones in addition to estrogen. In recent years, hormone replacement therapy (HRT) or estrogen combined with exercise have been proposed as potentially therapeutic agents for postmenopausal women, as these agents may potentially limit muscle damage and inflammation and stimulate repair in this population. While the benefits and potential health risks of long-term HRT use have been widely debated, controlled studies using short-term HRT or other estrogen agonists may provide future new and valuable insights into understanding the effects of estrogen on skeletal muscle, and greatly benefit the aging female population. Recent studies with older females have begun to demonstrate their benefits.
Collapse
Affiliation(s)
- Deborah L Enns
- Department of Kinesiology and Physical Education, Faculty of Science, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | | |
Collapse
|
30
|
Shih CD. Activation of estrogen receptor beta-dependent nitric oxide signaling mediates the hypotensive effects of estrogen in the rostral ventrolateral medulla of anesthetized rats. J Biomed Sci 2009; 16:60. [PMID: 19583861 PMCID: PMC2717931 DOI: 10.1186/1423-0127-16-60] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Accepted: 07/07/2009] [Indexed: 12/02/2022] Open
Abstract
Background Apart from their well-known peripheral cardiovascular effects, emerging evidence indicates that estrogen acts as a modulator in the brain to regulate cardiovascular functions. The underlying mechanisms of estrogen in central cardiovascular regulation, however, are poorly understood. The present study investigated the cardiovascular effects of 17β-estradiol (E2β) in the rostral ventrolateral medulla (RVLM), where sympathetic premotor neurons are located, and delineated the engagement of nitric oxide (NO) in E2β-induced cardiovascular responses. Methods In male Sprague-Dawley rats maintained under propofol anesthesia, the changes of blood pressure, heart rate and sympathetic vasomotor tone after microinjection bilaterally into the RVLM of a synthetic estrogen, E2β were examined for at least 120 min. The involvement of ERα and/or ERβ subtypes was determined by microinjection of selective ERα or ERβ agonist into bilateral RVLM. Different NO synthase (NOS) inhibitors were used to evaluate the involvement of differential of NOS isoforms in the cardiovascular effects of E2β. Results Bilateral microinjection of E2β (0.5, 1, or 5 pmol) into the RVLM dose-dependently decreased systemic arterial pressure (SAP) and the power density of the vasomotor components of SAP signals, our experimental index for sympathetic neurogenic vasomotor tone. These cardiovascular depressive effects of E2β (1 pmol) were abolished by co-injection of ER antagonist ICI 182780 (0.25 or 0.5 pmol), but not a transcription inhibitor actinomycin D (10 nmol). Like E2β, microinjection bilaterally into the RVLM of a selective ERβ agonist 2,3-bis(4-hydroxyphenyl) propionitrile (DPN, 1, 2, or 5 pmol) induced significant decreases in these hemodynamic parameters in a dose-dependent manner. In contrast, the selective ERα agonist 1,3,5-tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole (5 pmol) did not influence the same cardiovascular parameters. Co-administration bilaterally into the RVLM of NOS inhibitor NG-nitro-L-arginine methyl ester (5 nmol) or selective inducible NOS (iNOS) inhibitor S-methylisothiourea (25 pmol), but not selective neuronal NOS inhibitor 7-nitroindazole (0.5 pmol) or endothelial NOS inhibitor N5-(1-Iminoethyl)-L-ornithine (2.5 pmol), significantly attenuated the cardiovascular depressive effects elicited by DPN (2 pmol). Conclusion Our results indicate that E2β in the RVLM elicited short-term cardiovascular depressive effects via an ERβ-dependent nontranscriptional mechanism. These vasodepressor effects of E2β are likely to be mediated by the iNOS-derived NO in the RVLM.
Collapse
Affiliation(s)
- Cheng-Dean Shih
- Department of Pharmacy & Graduate Institute of Pharmaceutical Technology, Tajen University, Pingtung 90741, Taiwan, Republic of China.
| |
Collapse
|
31
|
Bednarek-Tupikowska G, Tworowska-Bardzinska U, Tupikowski K. Effects of estrogen and estrogen-progesteron on serum nitric oxide metabolite concentrations in post-menopausal women. J Endocrinol Invest 2008; 31:877-81. [PMID: 19092292 DOI: 10.1007/bf03346435] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
UNLABELLED Estrogens have some anti-atherosclerotic properties and they influence nitric oxide (NO) production. The aim of this study was to determine NOx levels in post-menopausal women and the effect of estrogen/estrogen-progesteron therapy (ET/EPT) on plasma NO levels. Eighty postmenopausal women (M1) comprising 26 with surgically induced menopause (ET1), mean age 50.9+/-2.9 yr, and 54 with physiological menopause (EPT1), mean age 50.5+/-3.0 yr, were studied. Forty healthy pre-menopausal women, mean age 48.3+/-2.3 yr were the controls (C). The post-menopausal women were treated for 4 months: group ET1 with ET and group EPT1 with EPT. Serum estradiol (E2), FSH, NOx and lipid profile before and after therapy were measured. NOx levels were lower in group M1 than in group C (8.75+/-1.57 vs 10.27+/-2.62, p<0.01) and increased after hormonal therapy (10.65+/-2.38). NOx concentration showed significant positive correlation with E2 (r=0.25, p<0.05). Total cholesterol (240.9+/-43.2), LDL-cholesterol (155.2+/-33.6), triglycerides (124.8+/-54.1), and apolipoprotein B (1.52+/-0.33) were higher in group M1 than in group C (223.1+/-44.3, 133.0+/-38.2, 108.3+/-52.9, and 1.12+/-0.36, respectively), and after ET/EPT they decreased to the values observed in group C. There were no correlations between NO and lipids or apolipoproteins. CONCLUSIONS ET and EPT improve NOx synthesis and endothelial relaxation. Medroxyprogesterone acetate added to E2 does not significantly influence NOx levels.
Collapse
Affiliation(s)
- G Bednarek-Tupikowska
- Department of Endocrinology, Diabetology and Isotope Treatment, Wroclaw Medical University, Wroclaw, Poland.
| | | | | |
Collapse
|
32
|
Enns DL, Iqbal S, Tiidus PM. Oestrogen receptors mediate oestrogen-induced increases in post-exercise rat skeletal muscle satellite cells. Acta Physiol (Oxf) 2008; 194:81-93. [PMID: 18397384 DOI: 10.1111/j.1748-1716.2008.01861.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM Our laboratory recently demonstrated that increases in post-exercise muscle satellite cell numbers are augmented by oestrogen. We investigated whether muscle oestrogen receptors (ORs) mediate this effect through administration of an OR antagonist, ICI 182,780. METHODS Ovariectomized female rats were divided into three groups: sham, oestrogen (0.25 mg pellet) and oestrogen plus OR blocker (ICI 182,780). Each group was divided into control and exercised groups. ICI 182,780 (5 mg kg(-1) sc) was administered 1 day prior to and 6 days following oestrogen pellet implantation. After 8 days of oestrogen exposure, animals ran downhill for 90 min (17 m min(-1), -13.5 degrees grade) on a treadmill. Soleus and white vastus muscles were removed 24 and 72 h post-exercise and immunostained for total (Pax7), activated (MyoD) and proliferating (BrdU) satellite cells. Muscle damage was indirectly assessed by measuring beta-glucuronidase activity. Two markers (His48 and ED1) of leucocyte infiltration were also examined. RESULTS beta-Glucuronidase activities and His48+ and ED1+ leucocytes increased post-exercise, and these increases were attenuated with oestrogen. ICI 182,780 did not influence the attenuating effect of oestrogen on leucocyte infiltration or beta-glucuronidase activities in muscle. Total (Pax7+), activated (MyoD+) and proliferating (BrdU+) satellite cells increased post-exercise, and these increases were augmented with oestrogen. Interestingly, ICI 182,780 abolished both exercise- and oestrogen-mediated increases in these satellite cell markers. CONCLUSION Oestrogen may augment increases in muscle satellite cells following exercise through OR-mediated mechanisms; furthermore, the attenuation of post-exercise muscle damage and leucocyte infiltration by oestrogen appears to be a non-OR-mediated process.
Collapse
Affiliation(s)
- D L Enns
- Department of Kinesiology & Physical Education, Wilfrid Laurier University, Waterloo, ON, Canada
| | | | | |
Collapse
|
33
|
Góñez LJ, Naselli G, Banakh I, Niwa H, Harrison LC. Pancreatic expression and mitochondrial localization of the progestin-adipoQ receptor PAQR10. Mol Med 2008; 14:697-704. [PMID: 18769639 DOI: 10.2119/2008-00072.gonez] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 08/18/2008] [Indexed: 11/06/2022] Open
Abstract
Steroid hormones induce changes in gene expression by binding to intracellular receptors that then translocate to the nucleus. Steroids have also been shown to rapidly modify cell function by binding to surface membrane receptors. We identified a candidate steroid membrane receptor, the progestin and adipoQ receptor (PAQR) 10, a member of the PAQR family, in a screen for genes differentially expressed in mouse pancreatic beta-cells. PAQR10 gene expression was tissue restricted compared with other PAQRs. In the mouse embryonic pancreas, PAQR10 expression mirrored development of the endocrine lineage, with PAQR10 protein expression confined to endocrine islet-duct structures in the late embryo and neonate. In the adult mouse pancreas, PAQR10 was expressed exclusively in islet cells except for its reappearance in ducts of maternal islets during pregnancy. PAQR10 has a predicted molecular mass of 29 kDa, comprises seven transmembrane domains, and, like other PAQRs, is predicted to have an intracellular N-terminus and an extracellular C-terminus. In silico analysis indicated that three members of the PAQR family, PAQRs 9, 10, and 11, have a candidate mitochondrial localization signal (MLS) at the N-terminus. We showed that PAQR10 has a functional N-terminal MLS and that the native protein localizes to mitochondria. PAQR10 is structurally related to some bacterial hemolysins, pore-forming virulence factors that target mitochondria and regulate apoptosis. We propose that PAQR10 may act at the level of the mitochondrion to regulate pancreatic endocrine cell development/survival.
Collapse
Affiliation(s)
- L Jorge Góñez
- Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
34
|
Ishunina TA, Swaab DF. Age-dependent ERα MB1 splice variant expression in discrete areas of the human brain. Neurobiol Aging 2008; 29:1177-89. [PMID: 17368651 DOI: 10.1016/j.neurobiolaging.2007.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 01/08/2007] [Accepted: 02/10/2007] [Indexed: 11/29/2022]
Abstract
A role of estrogens in brain aging and Alzheimer's disease (AD) is a hot topic of research. We show in material of 71 patients that the estrogen receptor alpha (ERalpha) splice variant MB1 is expressed at the protein and mRNA level in the human brain. MB1 is mainly confined to astrocytes, membranes and cytoplasm of projecting neurons and endothelial cells. It was consistently observed in the thalamus, colliculus inferior, pontine nuclei, dorsal motor nucleus of vagus, some motor neurons in the anterior and lateral horns of the spinal cord and rarely in pyramidal neurons of the cerebral cortex. The highest level of MB1 immunoreactivity (MB1-ir) was noted in the caudal hypothalamus, in particular in the tuberomamillary nucleus (TMN). MB1-ir in the TMN increased during aging in women. MB1-ir was higher in young (<50 years of age) men than in premenopausal women. No significant changes of this variant were observed in the TMN of AD cases. In conclusion, MB1 may function as a dominant negative isoform in the human brain.
Collapse
Affiliation(s)
- Tatjana A Ishunina
- Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | | |
Collapse
|
35
|
Otto C, Fuchs I, Altmann H, Klewer M, Schwarz G, Bohlmann R, Nguyen D, Zorn L, Vonk R, Prelle K, Osterman T, Malmström C, Fritzemeier KH. In vivo characterization of estrogen receptor modulators with reduced genomic versus nongenomic activity in vitro. J Steroid Biochem Mol Biol 2008; 111:95-100. [PMID: 18606537 DOI: 10.1016/j.jsbmb.2008.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Revised: 04/08/2008] [Accepted: 05/26/2008] [Indexed: 11/21/2022]
Abstract
Estrogen receptor (ER) ligands that are able to prevent postmenopausal bone loss, but have reduced activity in the uterus and the mammary gland might be of great value for hormone therapy. It is well established that the classical ER can activate genomic as well as nongenomic signal transduction pathways. In this study, we analyse the in vivo behaviour of ER ligands that stimulate nongenomic ER effects to the same extent as estradiol, but show clearly reduced activation of genomic ER effects in vitro. Using different readout parameters such as morphological changes, cellular proliferation, and target gene induction, we are able to demonstrate that ER ligands with reduced genomic activity in vitro show a better dissociation of bone versus uterine and mammary gland effects than estradiol that stimulates genomic and nongenomic effects to the same extent. We conclude that pathway-selective ER ligands may represent an interesting option for hormone therapy.
Collapse
Affiliation(s)
- Christiane Otto
- Research Laboratories, Bayer Schering Pharma AG, D-13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bourdeau V, Deschênes J, Laperrière D, Aid M, White JH, Mader S. Mechanisms of primary and secondary estrogen target gene regulation in breast cancer cells. Nucleic Acids Res 2007; 36:76-93. [PMID: 17986456 PMCID: PMC2248750 DOI: 10.1093/nar/gkm945] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Estrogen receptors (ERs), which mediate the proliferative action of estrogens in breast cancer cells, are ligand-dependent transcription factors that regulate expression of their primary target genes through several mechanisms. In addition to direct binding to cognate DNA sequences, ERs can be recruited to DNA through other transcription factors (tethering), or affect gene transcription through modulation of signaling cascades by non-genomic mechanisms of action. To better characterize the mechanisms of gene regulation by estrogens, we have identified more than 700 putative primary and about 1300 putative secondary target genes of estradiol in MCF-7 cells through microarray analysis performed in the presence or absence of the translation inhibitor cycloheximide. Although siRNA-mediated inhibition of ERalpha expression antagonized the effects of estradiol on up- and down-regulated primary target genes, estrogen response elements (EREs) were enriched only in the vicinity of up-regulated genes. Binding sites for several other transcription factors, including proteins known to tether ERalpha, were enriched in up- and/or down-regulated primary targets. Secondary estrogen targets were particularly enriched in sites for E2F family members, several of which were transcriptionally regulated by estradiol, consistent with a major role of these factors in mediating the effects of estrogens on gene expression and cellular growth.
Collapse
Affiliation(s)
- Véronique Bourdeau
- Institute for Research in Immunology and Cancer and Biochemistry Department, Université de Montréal, C.P. 6128 Succursale Centre Ville, Montréal, QC H3C 3J7, Canada
| | | | | | | | | | | |
Collapse
|
37
|
Zhang H, Zhang L. Role of protein kinase C isozymes in the regulation of alpha1-adrenergic receptor-mediated contractions in ovine uterine arteries. Biol Reprod 2007; 78:35-42. [PMID: 17901075 PMCID: PMC2391137 DOI: 10.1095/biolreprod.107.063479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Previously, we demonstrated that activation of protein kinase C (PRKC) enhanced alpha(1)-adrenergic receptor-induced contractions in nonpregnant ovine uterine arteries but inhibited the contractions in pregnant ovine uterine arteries. The present study tested the hypothesis that differential regulation of PRKC isozyme activities contributes to the different effects of phorbol 12, 13-dibutyrate (PDBu) on alpha(1)-adrenergic receptor-mediated contractions between the pregnant and nonpregnant ovine uterine arteries. Phenylephrine-induced contractions of ovine nonpregnant and pregnant uterine arteries were determined in the absence or presence of the PRKC activator PDBu and/or in combination with conventional and novel PRKC isozyme inhibitor GF109203X, PRKC isozyme-selective inhibitory peptides for conventional PRKC, PRKCB1, PRKCB2, and PRKCE. GF109203X produced a concentration-dependent inhibition of phenylephrine-induced contractions in both nonpregnant and pregnant uterine arteries, and it reversed the PDBu-mediated potentiation and inhibition of phenylephrine-induced contractions in nonpregnant and pregnant uterine artieries, respectively. In addition, PRKCB1, PRKCB2, and PRKCE inhibitory peptides blocked the PDBu-mediated responses in both nonpregnant and pregnant uterine arteries. Western blot analysis showed that PDBu induced a membrane translocation of PRKCA, PRKCB1, PRKCB2, and PRKCE in pregnant uterine arteries, and PRKCB1, PRKCB2, and PRKCE in nonpregnant uterine arteries. The results disprove the hypothesis that the dichotomy of PRKC mechanisms in the regulation of alpha(1)-adrenergic receptor-induced contractions in nonpregnant and pregnant uterine arteries is caused by the activation of different PRKC isozymes, and suggest downstream mechanisms of differential subcellular distributions for the distinct functional effects of PRKC isozymes in the adaptation of uterine arteries to pregnancy.
Collapse
Affiliation(s)
| | - Lubo Zhang
- Correspondence: Lubo Zhang, Center for Perinatal Biology, Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350. FAX: 909 558 4029; e-mail:
| |
Collapse
|
38
|
Chen N, Napoli JL. All-trans-retinoic acid stimulates translation and induces spine formation in hippocampal neurons through a membrane-associated RARalpha. FASEB J 2007; 22:236-45. [PMID: 17712061 DOI: 10.1096/fj.07-8739com] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Differentiation and patterning in the developing nervous system require the vitamin A metabolite all-trans-retinoic acid (atRA). Recent data suggest that higher cognitive functions, such as creation of hippocampal memory, also require atRA and its receptors, RAR, through affecting synaptic plasticity. Here we show that within 30 min atRA increased dendritic growth approximately 2-fold, and PSD-95 and synaptophysin puncta intensity approximately 3-fold, in cultured mouse hippocampal neurons, suggesting increased synapse formation. atRA (10 nM) increased ERK1/2 phosphorylation within 10 min. In synaptoneurosomes, atRA rapidly increased phosphorylation of ERK1/2, its target 4E-BP, and p70S6K, and its substrate, ribosome protein S6, indicating activation of MAPK and mammalian target of rapamycin (mTOR). Immunofluorescence revealed intense dendritic expression of RARalpha in the mouse hippocampus and localization of RARalpha on the surfaces of primary cultures of hippocampal neurons, with bright puncta along soma and neurites. Surface biotinylation confirmed the locus of RARalpha expression. Knockdown of RARalpha by shRNA impaired atRA-induced spine formation and abolished dendritic growth. Prolonged atRA stimulation reduced surface/total RARalpha by 43%, suggesting internalization, whereas brain-derived nerve growth factor or bicuculline increased the ratio by approximately 1.8-fold. atRA increased translation in the somatodendritic compartment, similar to brain-derived nerve growth factor. atRA specifically increased dendritic translation and surface expression of the alpha-amino-3-hydroxyl-5-methyl-4-isoxazole propionate receptor (AMPAR) subunit 1 (GluR1), without affecting GluR2. These data provide mechanistic insight into atRA function in the hippocampus and identify a unique membrane-associated RARalpha that mediates rapid induction of neuronal translation by atRA.
Collapse
Affiliation(s)
- Na Chen
- Nutritional Science and Toxicology, University of California, Berkeley, California 94720, USA
| | | |
Collapse
|
39
|
Watson CS, Alyea RA, Jeng YJ, Kochukov MY. Nongenomic actions of low concentration estrogens and xenoestrogens on multiple tissues. Mol Cell Endocrinol 2007; 274:1-7. [PMID: 17601655 PMCID: PMC1986712 DOI: 10.1016/j.mce.2007.05.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 05/17/2007] [Indexed: 10/23/2022]
Abstract
Nongenomic estrogenic mechanisms offer an opportunity to explain the conundrum of environmental estrogen and plant estrogen effects on cells and animals at the very low concentrations which are prevalent in our environments and diets. Heretofore the actions of these compounds have not been adequately accounted for by laboratory tests utilizing assays for actions only via the genomic pathway of steroid action and the nuclear forms of estrogen receptor alpha and beta. Membrane versions of these receptors, and the newly described GPR30 (7TMER) receptor protein provide explanations for the more potent actions of xenoestrogens. The effects of estrogens on many tissues demand a comprehensive assessment of the receptors, receptor levels, and mechanisms that might be involved, to determine which of these estrogen mimetic compounds are harmful and which might even be used therapeutically, depending upon the life stage at which we are exposed to them.
Collapse
Affiliation(s)
- C S Watson
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-0645, USA.
| | | | | | | |
Collapse
|
40
|
Otto C, Wessler S, Fritzemeier KH. Exploiting Nongenomic Estrogen Receptor-Mediated Signaling for the Development of Pathway-Selective Estrogen Receptor Ligands. ACTA ACUST UNITED AC 2007:163-81. [PMID: 17824177 DOI: 10.1007/2789_2006_022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
Different molecular mechanisms mediate the diverse biological effects of estrogens. The classical genomic mechanism is based on the function of the ER as a ligand-dependent transcription factor that binds to estrogen-response elements (EREs) in promoters of target genes to initiate gene expression. These direct genomic effects play a prominent role in the regulation of reproductive function. In contrast, nongenomic effects mediated by the classical ER have been demonstrated to activate PI3K, leading to the activation of endothelial NOS (eNOS) and hence vasorelaxation. Pathway-selective ER ligands might represent a novel option for hormone replacement therapy. Here we describe the identification and in vitro characterization of tool compounds that bind the ER reasonably well but exhibit low transcriptional activity on ERE-driven promoters. However, these compounds behave as potent stimulators of PI3K/Akt activation in vitro and lead to aortic vessel relaxation, a mechanism that is thought to be driven by nongenomic ER action. In a second set of experiments, we analyze how the in vitro pathway selectivity translates into the in vivo situation. We examine our tool compounds in comparison to estradiol and estren in the following paradigms: bone protection, uterine growth assays, and mammary gland assays.
Collapse
Affiliation(s)
- C Otto
- TRG Gynecology and Andrology, Bayer Schering Pharma AG, Müllerstr. 178, 13342 Berlin, Germany.
| | | | | |
Collapse
|
41
|
Lengyel F, Vértes Z, Kovács KA, Környei JL, Sümegi B, Vértes M. Effect of estrogen and inhibition of phosphatidylinositol-3 kinase on Akt and FOXO1 in rat uterus. Steroids 2007; 72:422-8. [PMID: 17433823 DOI: 10.1016/j.steroids.2007.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 02/06/2007] [Accepted: 03/02/2007] [Indexed: 11/26/2022]
Abstract
The importance of FOXO transcription factors in regulating different aspects of cellular homeostasis and apoptosis has become apparent. Akt/protein kinase B has been shown to phosphorylate and inactivate members of FOXO family of transcription factors. Akt and its upstream regulator, phosphatidylinositol-3 kinase (PI3K) are involved in rapid action of estrogen (E2) in different cells and tissues. The aim of the present study was to analyze the E2/PI3K/Akt/FOXO pathway in rat uterus. In response to E2, phosphorylation of Akt/PKB on Ser473 and FOXO1 on Ser256 and Thr24 residues increased but with distinct kinetics, regulating the activation and inactivation of Akt and FOXO1 proteins, respectively. The antiestrogen ICI 182,780 prevented E2 induced Akt activation suggesting that estrogen receptors mediate this effect of E2. Intrauterine injection of Wortmannin caused a decrease in the phosphorylation of Ser473 of Akt, and attenuated phosphorylation of its downstream target FOXO1 at Ser256 and at Thr24. However, the effect of E2 on phosphorylation of Thr24 showed a kinetic pattern distinct from that of Ser256. Our results suggest that the E2/PI3K/Akt/FOXO1 pathway in rat uterus is functioning even at the lack of ovarian hormones and responses to E2 treatment. Estradiol increases Akt phosphorylation through a Wortmannin sensitive way, presumably involving PI3K. The present work shows that PI3K plays a crucial role in the phosphorylation and inactivation of FOXO1 in vivo, indicating that the regulation of this transcription factor is a more complex event in uterine cells requiring further investigations.
Collapse
Affiliation(s)
- Ferenc Lengyel
- Institute of Physiology, Pécs University Medical School, Szigeti út 12, Pécs H7624, Hungary.
| | | | | | | | | | | |
Collapse
|
42
|
Sun K, Montana V, Chellappa K, Brelivet Y, Moras D, Maeda Y, Parpura V, Paschal BM, Sladek FM. Phosphorylation of a conserved serine in the deoxyribonucleic acid binding domain of nuclear receptors alters intracellular localization. Mol Endocrinol 2007; 21:1297-311. [PMID: 17389749 DOI: 10.1210/me.2006-0300] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nuclear receptors (NRs) are a superfamily of transcription factors whose genomic functions are known to be activated by lipophilic ligands, but little is known about how to deactivate them or how to turn on their nongenomic functions. One obvious mechanism is to alter the nuclear localization of the receptors. Here, we show that protein kinase C (PKC) phosphorylates a highly conserved serine (Ser) between the two zinc fingers of the DNA binding domain of orphan receptor hepatocyte nuclear factor 4alpha (HNF4alpha). This Ser (S78) is adjacent to several positively charged residues (Arg or Lys), which we show here are involved in nuclear localization of HNF4alpha and are conserved in nearly all other NRs, along with the Ser/threonine (Thr). A phosphomimetic mutant of HNF4alpha (S78D) reduced DNA binding, transactivation ability, and protein stability. It also impaired nuclear localization, an effect that was greatly enhanced in the MODY1 mutant Q268X. Treatment of the hepatocellular carcinoma cell line HepG2 with PKC activator phorbol 12-myristate 13-acetate also resulted in increased cytoplasmic localization of HNF4alpha as well as decreased endogenous HNF4alpha protein levels in a proteasome-dependent fashion. We also show that PKC phosphorylates the DNA binding domain of other NRs (retinoic acid receptor alpha, retinoid X receptor alpha, and thyroid hormone receptor beta) and that phosphomimetic mutants of the same Ser/Thr result in cytoplasmic localization of retinoid X receptor alpha and peroxisome proliferator-activated receptor alpha. Thus, phosphorylation of this conserved Ser between the two zinc fingers may be a common mechanism for regulating the function of NRs.
Collapse
Affiliation(s)
- Kai Sun
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhu JTT, Choi RCY, Chu GKY, Cheung AWH, Gao QT, Li J, Jiang ZY, Dong TTX, Tsim KWK. Flavonoids possess neuroprotective effects on cultured pheochromocytoma PC12 cells: a comparison of different flavonoids in activating estrogenic effect and in preventing beta-amyloid-induced cell death. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2007; 55:2438-45. [PMID: 17323972 DOI: 10.1021/jf063299z] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Despite the classical hormonal effect, estrogen possesses a neuroprotective effect in the brain, which has led many to search for novel treatments for neurodegenerative diseases. Flavonoids, a group of compounds mainly derived from vegetables, share a resemblance, chemically, to estrogen, and indeed, some have been used as estrogen substitutes. To search for potential therapeutic agents against neurodegenerative diseases, different subclasses of flavonoids were analyzed and compared with estrogen. First, the estrogenic activities of these flavonoids were determined by activating the estrogen-responsive elements in cultured MCF-7 breast cancer cells. Second, the neuroprotective effects of flavonoids were revealed by measuring its inhibition effects on the formation of reactive oxygen species, the aggregation of beta-amyloid, and the induction of cell death by beta-amyloid in cultured neuronal PC12 cells. Among these flavonoids, baicalein, scutellarin, hibifolin, and quercetin-3'-glucoside possessed the strongest effect in neuroprotection; however, the neuroprotective activity did not directly correlate with the estrogenic activity of the flavonoids. Identification of these flavonoids could be very useful in finding potential drugs, or food supplements, for treating Alzheimer's disease.
Collapse
Affiliation(s)
- Judy T T Zhu
- Department of Biology and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Victor NA, Wanderi EW, Gamboa J, Zhao X, Aronowski J, Deininger K, Lust WD, Landreth GE, Sundararajan S. Altered PPARgamma expression and activation after transient focal ischemia in rats. Eur J Neurosci 2007; 24:1653-63. [PMID: 17004929 DOI: 10.1111/j.1460-9568.2006.05037.x] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stroke is a devastating disease with limited treatment options. Recently, we found that the peroxisome proliferator-activated receptor-gamma (PPARgamma) agonists troglitazone and pioglitazone reduce injury and inflammation in a rat model of transient cerebral ischemia. The mechanism of this protection is unclear, as these agents can act through PPAR-gamma activation or through PPAR-gamma-independent mechanisms. Therefore, we examined PPAR-gamma expression, DNA binding and transcriptional activity following stroke. In addition, we used a PPAR-gamma antagonist, T0070907, to determine the role of PPAR-gamma during ischemia. Using immunohistochemical techniques and real-time PCR, we found low levels of PPAR-gamma mRNA and PPAR-gamma immunoreactivity in nonischemic brain; however, PPAR-gamma expression dramatically increased in ischemic neurons, peaking 24 h following middle cerebral artery occlusion. Interestingly, we found that in both vehicle- and agonist-treated brains, DNA binding was reduced in the ischemic hemisphere relative to the contralateral hemisphere. Expression of a PPAR-gamma target gene, lipoprotein lipase, was also reduced in ischemic relative to nonischemic brain. Both DNA binding and lipoprotein lipase expression were increased by the addition of the PPAR-gamma agonist rosiglitazone. Finally, we found that rosiglitazone-mediated protection after stroke was reversed by the PPAR-gamma antagonist T0070907. Interestingly, infarction size was also increased by T0070907 in the absence of PPAR-gamma agonist, suggesting that endogenous PPAR-gamma ligands may mitigate the effects of cerebral ischemia.
Collapse
Affiliation(s)
- N A Victor
- Department of Neurology, Case Western Reserve University, 11100 Euclid Ave., Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang R, Zhang QG, Han D, Xu J, Lü Q, Zhang GY. Inhibition of MLK3-MKK4/7-JNK1/2 pathway by Akt1 in exogenous estrogen-induced neuroprotection against transient global cerebral ischemia by a non-genomic mechanism in male rats. J Neurochem 2006; 99:1543-54. [PMID: 17064355 DOI: 10.1111/j.1471-4159.2006.04201.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Numerous studies have demonstrated the neuroprotective effects of estrogen in experimental cerebral ischemia. To investigate molecular mechanisms of estrogen neuroprotection in global ischemia, immunoblotting, immunohistochemistry and Nissel-staining analysis were used. Our results showed that chronic pretreatment with beta-estradiol 3-benzoate (E2) enhanced Akt1 activation and reduced the activation of mixed-lineage kinase 3 (MLK3), mitogen-activated protein kinase kinase 4/7 (MKK4/7), and c-Jun N-terminal kinase 1/2 (JNK1/2) in the hippocampal CA1 subfield during reperfusion after 15 min of global ischemia. In addition, E2 reduced downstream JNK nuclear and non-nuclear components, c-Jun and Bcl-2 phosphorylation and Fas ligand protein expression induced by ischemia/reperfusion. Administration of phosphoinositide 3-kinase (PI3K) inhibitor LY 294,002 prevented both activation of Akt1 and inhibition of MLK3, MKK4/7 and JNK1/2. The interaction between ERalpha and the p85 subunit of PI3K was also examined. E2 and antiestrogen ICI 182,780 promoted and prevented this interaction, respectively. Furthermore, ICI 182,780 blocked both the activation of Akt1 and the inhibition of MLK3, MKK4/7 and JNK1/2. Photomicrographs of cresyl violet-stained brain sections showed that E2 reduced CA1 neuron loss after 5 days of reperfusion, which was abolished by ICI 182,780 and LY 294,002. Our data indicate that in response to estrogen, ERalpha interacts with PI3K to activate Akt1, which may inhibit the MLK3-MKK4/7-JNK1/2 pathway to protect hippocampal CA1 neurons against global cerebral ischemia in male rats.
Collapse
Affiliation(s)
- Ran Wang
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory for Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, China
| | | | | | | | | | | |
Collapse
|
46
|
Skøtt O, Uhrenholt TR, Schjerning J, Hansen PBL, Rasmussen LE, Jensen BL. Rapid actions of aldosterone in vascular health and disease—friend or foe? Pharmacol Ther 2006; 111:495-507. [PMID: 16413609 DOI: 10.1016/j.pharmthera.2005.10.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2005] [Accepted: 10/28/2005] [Indexed: 02/07/2023]
Abstract
The mineralocorticoid receptor (MR) and the enzyme 11betahydroxysteroid dehydrogenase type 2, which confers aldosterone specificity to the MR, are present in endothelium and vascular smooth muscle. In several pathological conditions aldosterone promotes vascular damage by formation of reactive oxygen species. The effect of aldosterone on vascular function, however, is far from clear. By rapid non-genomic mechanisms aldosterone may cause calcium mobilization and vasoconstriction, or may stimulate nitric oxide formation through the PI-3 kinase/Akt pathway and thereby counteract vasoconstriction. Vasoconstrictor, vasodilator or no effects of aldosterone have been reported from studies on human forearm blood flow. Inhibition of MR with spironolactone improves endothelial function in patients with heart failure but worsens endothelial function in type 2 diabetic patients. The aim of the present review is to reconcile some of the apparently conflicting data. A key observation is that reactive oxygen and nitrogen species serve as physiological signaling molecules at low concentrations, while they initiate pathological processes at higher concentrations. The net effect of aldosterone, which stimulates ROS production, therefore depends on the ambient level of oxidative stress. Thus, in situations with low levels of oxidative stress aldosterone may promote vasodilatation, while at higher oxidative stress (high NaCl intake, pre-existing vascular pathological conditions, high oxygen tension in vitro) aldosterone is likely to be associated with vasoconstriction and oxidative damage, and in this setting inhibition of the MR is likely to be beneficial.
Collapse
Affiliation(s)
- Ole Skøtt
- Department of Physiology and Pharmacology, University of Southern Denmark, DK-5000 Odense, Denmark.
| | | | | | | | | | | |
Collapse
|
47
|
Banka CL, Lund CV, Nguyen MTN, Pakchoian AJ, Mueller BM, Eliceiri BP. Estrogen induces lung metastasis through a host compartment-specific response. Cancer Res 2006; 66:3667-72. [PMID: 16585192 DOI: 10.1158/0008-5472.can-05-4416] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Direct proliferative effects of estrogen (E(2)) on estrogen receptor-positive tumors are well documented; however, the potential for E(2) to mediate effects selective for the host (i.e., angiogenesis, vascular permeability, or stromal effects), which influence tumor growth and/or metastasis, has received less attention. In this study, we examine the capacity for E(2) to promote tumor growth and/or metastasis independent of direct effects on tumor cells. In these studies, we distinguish host versus tumor compartment components of E(2) action in tumor growth and metastasis by analysis of E(2)-nonresponsive tumor cells implanted in ovariectomized (OVX) mice that contain s.c. implants of placebo (OVX) or E(2)-containing slow-release pellets (OVX + E(2)). We show that the D121 lung carcinoma cell line is E(2)-nonresponsive, and following s.c. implantation in OVX versus OVX + E(2) mice, E(2) action on the host compartment leads to an increase in spontaneous metastasis but not primary tumor growth or neovascularization. Similarly, experimental lung metastasis of E(2)-nonresponsive 4T1 mammary carcinoma cells also leads to increased tumor burden in the lungs of OVX + E(2) mice. These results suggest that the E(2) status of the host compartment influences late steps in tumor cell metastasis that can provide important insights into the role of E(2) in the tumor versus host compartments.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/blood supply
- Breast Neoplasms/pathology
- Cell Growth Processes/drug effects
- Cell Line, Tumor
- Estradiol/toxicity
- Estrogen Receptor alpha/biosynthesis
- Female
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/chemically induced
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neoplasms, Hormone-Dependent/blood supply
- Neoplasms, Hormone-Dependent/chemically induced
- Neoplasms, Hormone-Dependent/pathology
- Neoplasms, Hormone-Dependent/secondary
- Neovascularization, Pathologic/pathology
Collapse
Affiliation(s)
- Carole L Banka
- Division of Cancer and Vascular Biology, La Jolla Institute for Molecular Medicine, 4570 Executive Drive, Ste. 100, San Diego, CA 92121, USA
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Estrogens have a profound influence on skin. The relative hypoestrogenism that accompanies menopause exacerbates the deleterious effects of both intrinsic and environmental aging. Estrogens clearly have a key role in skin aging homeostasis as evidenced by the accelerated decline in skin appearance seen in the perimenopausal years. Estrogens improve skin in many ways. Among these, they increase collagen content and skin thickness and improve skin moisture. However, despite the knowledge that estrogens have such important effects on skin, the cellular and subcellular sites and mechanisms of estrogen action are still poorly understood. Estrogen receptors (ERs) have been detected in skin, and recent studies suggest that estrogens exert their effect in skin through the same molecular pathways used in other non-reproductive tissues. Although systemic hormone replacement therapy (HRT) has been used for many years, recent trials have reported a significant increased risk of breast cancer and other pathologies with this treatment. This has led to reconsider the risks and benefits of HRT. For this reason, systemic HRT cannot be recommended today to treat skin aging. Currently, intensive research is conducted to develop new drugs called selective ER modulators (SERMs). These drugs exert mixed estrogenic and antiestrogenic effects depending on the tissue and cell type. One might expect in the future such a drug targeting specifically the skin without systemic side effects.
Collapse
|
49
|
Muñoz-Castañeda JR, Muntané J, Herencia C, Muñoz MC, Bujalance I, Montilla P, Túnez I. Ovariectomy exacerbates oxidative stress and cardiopathy induced by adriamycin. Gynecol Endocrinol 2006; 22:74-9. [PMID: 16603431 DOI: 10.1080/09513590500490249] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Ovarian hormone depletion in ovariectomized experimental animals is a useful model with which to study the physiopathological consequences of menopause in women. It has been suggested that menopause is a risk factor for the induction of several cardiovascular disorders. In the present study we analyzed the effects of ovarian hormone depletion by ovariectomy (OVX) in a model of oxidative stress and cardiopathy induced by adriamycin (AD). To evaluate these effects, we measured parameters related to cardiac damage (creatinine kinase, lactate dehydrogenase, aspartate aminotransferase and alanine aminotransferase) and oxidative stress (malondialdehyde, catalase, superoxide dismutase, glutathione peroxidase, reduced glutathione, nitric oxide and carbonyl proteins) in cardiac tissue and erythrocytes. OVX was found to alter all markers of oxidative stress and cell damage in cardiac tissue. Similarly, the OVX-derived loss of ovarian hormones enhanced cardiac damage and oxidative stress induced by AD. Our results suggest that antioxidant status in cardiac tissue and erythrocytes is seriously compromised by OVX during the cardiomyopathy induced by AD in experimental animals. In conclusion, the absence of hormones caused by OVX or menopause may induce or accelerate pre-existing cardiovascular dysfunctions.
Collapse
Affiliation(s)
- Juan Rafael Muñoz-Castañeda
- Department of Biochemistry and Molecular Biology, School of Medicine, Reina Sofia University Hospital, Cordoba, Spain.
| | | | | | | | | | | | | |
Collapse
|
50
|
Wessler S, Otto C, Wilck N, Stangl V, Fritzemeier KH. Identification of estrogen receptor ligands leading to activation of non-genomic signaling pathways while exhibiting only weak transcriptional activity. J Steroid Biochem Mol Biol 2006; 98:25-35. [PMID: 16203130 DOI: 10.1016/j.jsbmb.2005.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Accepted: 08/11/2005] [Indexed: 12/27/2022]
Abstract
Estrogen receptors (ERs) stimulate genomic effects by acting as nuclear transcription factors as well as non-genomic effects by activating distinct cytoplasmic protein kinase cascades. Non-genomic effects have been implicated in numerous cellular processes, such as proliferation, differentiation, apoptosis and vasorelaxation. To exploit non-genomic effects mediated by ERalpha for novel hormone replacement regimens, we screened a focused library of steroid receptor ligands to identify compounds exhibiting properties different from estradiol, i.e. substances that selectively stimulate non-genomic signal transduction pathways while exhibiting low genomic activities. Treatment of breast cancer cells and osteosarcoma cells with estradiol, estren, substance A and substance B led to non-genomic activation of Akt (protein kinase B) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling cascades mediated by Src (Rous Sarcoma Virus, non-receptor tyrosine kinase) and phosphatidylinositol-3-kinase (PI3K) stimulation. Such compounds leading to prominent Akt/ERK activation but exhibiting only weak genomic properties were applied in vasorelaxation assays, modeling physiological non-genomic ER responses. As expected from PI3K and Src activation data, substances were as effective as estradiol in mediating vasorelaxation. We assume that these pathway-selective estrogen receptor ligands may serve as potent lead structures for novel hormone replacement strategies exhibiting lesser side effects than the existing treatment paradigms.
Collapse
Affiliation(s)
- Silja Wessler
- Paul-Ehrlich-Institute, Paul-Ehrlich Strasse 51-59, D-63225 Langen, Germany.
| | | | | | | | | |
Collapse
|