1
|
Kubota S, Yamamoto Y, Kimura K. A Chinese Medicine, Tokishakuyakusan, Increases Bovine Oviductal Tonus <i>via</i> G Protein-Coupled Estrogen Receptor 1. Biol Pharm Bull 2022; 45:1133-1141. [DOI: 10.1248/bpb.b22-00201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Sayaka Kubota
- Department of Animal Production Science, Graduate School of Environmental and Life Science, Okayama University
| | - Yuki Yamamoto
- Department of Animal Production Science, Graduate School of Environmental and Life Science, Okayama University
| | - Koji Kimura
- Department of Animal Production Science, Graduate School of Environmental and Life Science, Okayama University
| |
Collapse
|
2
|
Hazano K, Haneda S, Kayano M, Matsui M. Local sex steroid hormone milieu in the bovine oviduct ipsilateral and contralateral to preovulatory follicle or corpus luteum during the periovulatory phase. Domest Anim Endocrinol 2021; 74:106515. [PMID: 32711284 DOI: 10.1016/j.domaniend.2020.106515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/27/2020] [Accepted: 06/21/2020] [Indexed: 11/28/2022]
Abstract
Estradiol-17β (E2) and progesterone (P4) regulate oviductal functions, providing a suitable environment for the transport and maturation of gametes, fertilization, and embryonic development. In addition to the E2 and P4 nuclear receptors, estrogen receptor (ESR) α and β, nuclear progesterone receptor (PGR), nongenomic mechanisms through G protein-coupled estrogen receptor (GPER1), and progesterone receptor membrane component (PGRMC) 1 and 2 mediate E2 and P4 actions. This study aimed to characterize the local endocrine environment of the oviduct by examining the oviductal E2 and P4 concentrations and their receptors' mRNA expression during the periovulatory phase. The bovine oviducts were collected in a slaughterhouse and the days postovulation were estimated according to state of the ovaries and the uterus. Samples of the ampulla and isthmus ipsilateral and contralateral to the preovulatory follicle or corpus luteum were collected on Days 19 to 21, Days 0 to 1, Days 2 to 4, and Days 5 to 7 of the estrous cycle. The effects of the estrous cycle phase and oviductal region (ampulla and isthmus) and side (ipsilateral and contralateral) were analyzed by 3-way ANOVA. Moreover, to clarify the regulatory mechanisms of the mRNA expression of hormone receptors, the effects of E2 and P4 on mRNA expression in the oviduct were examined by multiple linear regression. The oviductal endocrine milieu on Days 19 to 21 was characterized by an E2-dominant environment with high E2 and low P4, high ESR1 and PGR mRNA expression, and low ESR2, GPER1, and PGRMC2 mRNA expression, whereas the corresponding on Days 0 to 1 was characterized by the endocrine milieu without hormone dominance. The environment on Days 2 to 4 and Day 5 to 7 was characterized by opposite tendency of oviductal hormone concentrations and their receptors' mRNA expression to Days 19 to 21. Additionally, the ipsilateral oviduct had the more P4-dominant endocrine milieu, with lower E2 and higher P4 concentrations, and different expression of ESR1/2, GPER1, PGR, and PGRMC2 mRNA when compared with the contralateral oviduct on Days 2 to 4 and Days 5 to 7, except for PGRMC1. Although oviductal E2 and P4 influenced the mRNA expression of ESR1/2, GPER1, PGR, and PGRMC1/2, their effects were different between regions and sides. In summary, the oviductal endocrine milieu varies according to the estrous cycle phase and the oviductal region and side, which may be involved in the estrous cycle phase-specific and oviductal region-specific and side-specific functions.
Collapse
Affiliation(s)
- K Hazano
- The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - S Haneda
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - M Kayano
- Research Center for Global Agromedicine, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - M Matsui
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan.
| |
Collapse
|
3
|
Huang W, Ai W, Lin W, Fang F, Wang X, Huang H, Dahlgren RA, Wang H. Identification of receptors for eight endocrine disrupting chemicals and their underlying mechanisms using zebrafish as a model organism. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 204:111068. [PMID: 32745784 DOI: 10.1016/j.ecoenv.2020.111068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 06/11/2023]
Abstract
Herein, eight common endocrine disrupting chemicals (EDCs) were exposed to zebrafish (Danio rerio) to investigate the relationship between different EDCs and their activated estrogen receptors. Under acute exposure, we identified five major malformation types whose incidence and deformity modes differed among EDCs. Luciferase analysis divided the EDC receptors into four categories: (i) triclosan (TCS), 17ß-estradiol (E2) and estriol (E3) mainly activated GPER expression; (ii) bisphenol A (BPA), p-(tert-octyl) phenol (POP), 17α-ethynylestradiol (EE2), E2 and E3 activated ERβ expression; (iii) E2 and E3 acted on both GPER and ERβ; and (iv) estrone (E1) and 9,9-bis(4-hydroxyphenyl)fluorene (BHPF) had little effect on the two receptors. In vivo immunofluorescence experiments on 96-hpf larvae provided evidence that TCS and POP acted on GPER and ERβ, respectively, while E2 acted on the two receptors simultaneously. Luciferase activities in the promoter regions of gper (-986 to -488) and erβ (-1998 to -1496) were higher than those in other regions, identifying these key regions as targets for transcription activity. TCS promoted GPER expression by acting on the JUND transcription factor, while POP promoted ERβ expression by activating the Foxl1 transcription factor. In contrast, E2 mainly regulated transcription of GPER and ERβ by Arid3a. These findings provide compelling evidence that different EDCs possess varying estrogen receptors, leading to differential regulatory pathways and abnormality symptoms. These results offer an experimental strategy and fundamental information to assess the molecular mechanisms of EDC-induced estrogen effects.
Collapse
Affiliation(s)
- Wenhao Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Weiming Ai
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Weiwei Lin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Fang Fang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xuedong Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Randy A Dahlgren
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Huili Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
4
|
Sun S, Shin J, Jang J, Hwang S, Kim J, Kong J, Yang H. 17Beta-Estradiol Regulates NUCB2/ Nesfatin-1 Expression in Mouse Oviduct. Dev Reprod 2020; 24:43-52. [PMID: 32411917 PMCID: PMC7201062 DOI: 10.12717/dr.2020.24.1.43] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/26/2022]
Abstract
NUCB2/nesfatin-1 known to regulate appetite and energy homeostasis is expressed not only in the hypothalamus, but also in various organs and tissues. Our previous reports also demonstrated that NUCB2/nesfatin-1 was expressed in the reproductive organs, including the ovaries, uterus, and testes of mice. However, it is yet known whether NUCB2/nesfatin-1 is expressed in the oviduct and how its expression is regulated. Therefore, we investigated the expression of NUCB2/nesfatin-1 in the oviduct and its expression is regulated by gonadotropin. Immunohistochemical staining results showed that nesfatin-1 protein was localized in epithelial cells of the oviduct. As a result of quantitative real-time PCR (qRT-PCR) and Western blot, NUCB2/nesfatin-1 was detected strongly in the oviducts. During the estrus cycle, NUCB2/nesfatin-1 expression in the oviducts was markedly higher in the proestrus stage than in other estrus stages. In order to elucidate whether the expression of NUCB2 mRNA is controlled by the gonadotropins, we injected PMSG and hCG and measured NUCB2 mRNA level in the oviduct after injection. Its level was increased in the oviduct after PMSG injection, but no significant change after hCG injection. In addition, NUCB2 mRNA levels were markedly reduced after ovariectomy, while recovered after 17β-estradiol (E2) injection, but not by progesterone (P4). This study demonstrated that NUCB2/nesfatin-1 is highly expressed in the oviduct of mouse and its expression is regulated by E2 secreted by the ovaries. These results suggest that NUCB2/nesfatin-1 expressed by the oviduct may affect the function of the oviduct regulated by the ovaries.
Collapse
Affiliation(s)
- Sojung Sun
- Dept. of Bioenvironmental Technology, Seoul Women's University, Seoul 01797, Korea
| | - Jungwoo Shin
- Dept. of Bioenvironmental Technology, Seoul Women's University, Seoul 01797, Korea
| | - Jiwon Jang
- Dept. of Bioenvironmental Technology, Seoul Women's University, Seoul 01797, Korea
| | - Seungyeon Hwang
- Dept. of Bioenvironmental Technology, Seoul Women's University, Seoul 01797, Korea
| | - Jeongwoo Kim
- Dept. of Bioenvironmental Technology, Seoul Women's University, Seoul 01797, Korea
| | - Jinseong Kong
- Dept. of Bioenvironmental Technology, Seoul Women's University, Seoul 01797, Korea
| | - Hyunwon Yang
- Dept. of Bioenvironmental Technology, Seoul Women's University, Seoul 01797, Korea
| |
Collapse
|
5
|
Wu T, Tang C, Chen Y, Yong X, Liu Z, Jiang L, Zeng Q, Tao R. Regulatory effect of 17β-estradiol on the expression of β-defensin-2 and proinflammatory cytokines in human oral epithelial cells. J Oral Pathol Med 2020; 49:365-372. [PMID: 32176389 DOI: 10.1111/jop.13016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/24/2020] [Accepted: 03/10/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Although estrogen deficiency has been proposed as a risk factor for oral mucosal inflammatory diseases in post-menopausal women, the mechanisms involved remain unclear. This study aimed to investigate the effect of 17β-estradiol (E2) on the inflammatory response stimulated by interleukin-1 beta (IL-1β) in human oral mucosal epithelial cells (hOMECs) and its possible mechanism. METHODS Primary hOMECs were obtained from female infants and cultured in keratinocyte growth medium. The hOMECs at second passage were collected and stimulated by 10-7 mol/L ICI182,780 or 10-7 mol/L G1 for 1 hour, E2 (10-7 mol/L, 10-8 mol/L, 10-9 mol/L) for 36 hour, 100 ng/mL IL-1β for 12 hours, respectively. Human beta-2 defensin (hBD-2), tumor necrosis factor-alpha (TNF)-α, IL-6, IL-8, estrogen receptor-alpha (ERα), estrogen receptor-beta (ERβ), and G protein-coupled receptor 30 (GPR30) mRNA levels and protein levels were measured by real-time quantitative polymerase chain reaction (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), and Western Blot (WB), respectively. RESULTS Expression of hBD-2 and inflammatory cytokines increased after IL-1β stimulation, which was down-regulated by E2 pre-treatment. With ICI182,780, the suppression of E2 on hBD-2 mRNA was attenuated. With G1, the mRNA expression and protein expression of hBD-2 were reduced. CONCLUSION Pre-treatment of hOMECs with E2 at physiological concentrations inhibited the IL-1β-induced expression of hBD-2 and inflammatory cytokines. The protective effects of E2 suggest its potential use treating oral inflammatory diseases in clinical practice.
Collapse
Affiliation(s)
- Tiantian Wu
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Caijin Tang
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Yi Chen
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Xiangzhi Yong
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Zhenmin Liu
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
| | - Lanlan Jiang
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Qixin Zeng
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Renchuan Tao
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| |
Collapse
|
6
|
Imam Aliagan A, Madungwe NB, Tombo N, Feng Y, Bopassa JC. Chronic GPER1 Activation Protects Against Oxidative Stress-Induced Cardiomyoblast Death via Preservation of Mitochondrial Integrity and Deactivation of Mammalian Sterile-20-Like Kinase/Yes-Associated Protein Pathway. Front Endocrinol (Lausanne) 2020; 11:579161. [PMID: 33193095 PMCID: PMC7604496 DOI: 10.3389/fendo.2020.579161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction: Estrogen (17β-estradiol, E2) is well-known to induce cardioprotective effects against ischemia/reperfusion (I/R) injury. We recently reported that acute application of E2 at the onset of reperfusion in vivo induces cardioprotective effects against I/R injury via activation of its non-steroidal receptor, G protein-coupled estrogen receptor 1 (GPER1). Here, we investigated the impact and mechanism underlying chronic GPER1 activation in cultured H9c2 rat cardiomyoblasts. Methods: H9c2 rat cardiomyoblasts were cultured and pretreated with the cytotoxic agent H2O2 for 24 h and incubated in the presence of vehicle (control), GPER1 agonists E2 and G1, or GPER1 agonists supplemented with G15 (GPER1 antagonist) for 48 or 96 h. After treatment, cells were collected to measure the rate of cell death and viability using flow cytometry and Calcein AM assay or MTT assay, respectively. The resistance to opening of the mitochondrial permeability transition pore (mPTP), the mitochondrial membrane potential, and ATP production was assessed using fluorescence microscopy, and the mitochondrial structural integrity was observed with electron microscopy. The levels of the phosphorylation of mammalian sterile-20-like kinase (MST1) and yes-associated protein (YAP) were assessed by Western blot analysis in whole-cell lysate, while the expression levels of mitochondrial biogenesis genes, YAP target genes, and proapoptotic genes were measured by qRT-PCR. Results: We found that after H2O2 treatment, chronic E2/G1 treatment decreased cell death effect was associated with the prevention of the S phase of the cell cycle arrest compared to control. In the mitochondria, chronic E2/G1 activation treatment preserved the cristae morphology, and increased resistance to opening of mPTP, but with little change to mitochondrial fusion/fission. Additionally, chronic E2/G1 treatment predominantly reduced phosphorylation of MST1 and YAP, as well as increased MST1 and YAP protein levels. E2 treatment also upregulated the expression levels of TGF-β and PGC-1α mRNAs and downregulated PUMA and Bim mRNAs. Except for ATP production, all the E2 or G1 effects were prevented by the cotreatment with the GPER1 antagonist, G15. Conclusion: Together, these results indicate that chronic GPER1 activation with its agonists E2 or G1 treatment protects H9c2 cardiomyoblasts against oxidative stress-induced cell death and increases cell viability by preserving mitochondrial structure and function as well as delaying the opening of mPTP. These chronic GPER1 effects are associated with the deactivation of the non-canonical MST1/YAP mechanism that leads to genetic upregulation of cell growth genes (CTGF, CYR61, PGC-1α, and ANKRD1), and downregulation of proapoptotic genes (PUMA and Bim).
Collapse
Affiliation(s)
- Abdulhafiz Imam Aliagan
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ngonidzashe B. Madungwe
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| | - Nathalie Tombo
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Yansheng Feng
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Jean C. Bopassa
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- *Correspondence: Jean C. Bopassa
| |
Collapse
|