Cargando…

Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells

Melatonin receptors MT1 and MT2 (genes officially named MTNR1A and MTNR1B, respectively) play crucial roles in melatonin-mediated regulation of circadian rhythms, the immune system, and control of reproduction in seasonally breeding animals. In this study, immunolocalization assay showed that MT1 an...

Descripción completa

Detalles Bibliográficos
Autores principales: Gao, Yuan, Wu, Xiaochun, Zhao, Shuqin, Zhang, Yujun, Ma, Hailong, Yang, Zhen, Yang, Wanghao, Zhao, Chen, Wang, Li, Zhang, Quanwei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6416941/
https://www.ncbi.nlm.nih.gov/pubmed/30915128
http://dx.doi.org/10.1186/s11658-019-0147-z
_version_ 1783403460773806080
author Gao, Yuan
Wu, Xiaochun
Zhao, Shuqin
Zhang, Yujun
Ma, Hailong
Yang, Zhen
Yang, Wanghao
Zhao, Chen
Wang, Li
Zhang, Quanwei
author_facet Gao, Yuan
Wu, Xiaochun
Zhao, Shuqin
Zhang, Yujun
Ma, Hailong
Yang, Zhen
Yang, Wanghao
Zhao, Chen
Wang, Li
Zhang, Quanwei
author_sort Gao, Yuan
collection PubMed
description Melatonin receptors MT1 and MT2 (genes officially named MTNR1A and MTNR1B, respectively) play crucial roles in melatonin-mediated regulation of circadian rhythms, the immune system, and control of reproduction in seasonally breeding animals. In this study, immunolocalization assay showed that MT1 and MT2 are highly expressed in Leydig cell membrane. To understand the biological function of melatonin receptors in hCG-induced testosterone synthesis, we generated melatonin receptor knockdown cells using specific siRNA and performed testosterone detection after hCG treatment. We found that knockdown of melatonin receptors, especially MTNR1A, led to an obvious decrease (> 60%) of testosterone level. Our further study revealed that knockdown of melatonin receptors repressed expression, at both the mRNA level and the protein level, of key steroidogenic genes, such as p450scc, p450c17 and StAR, which are essential for testosterone synthesis. hCG triggered endoplasmic reticulum (ER) stress to regulate steroidogenic genes’ expression and apoptosis. To further investigate the potential roles of melatonin receptors in hCG-induced regulation of ER stress and apoptosis, we examined expression of some crucial ER stress markers, including Grp78, Chop, ATF4, Xbp1, and IRE1. We found that inhibition of melatonin receptors increased hCG-induced expression of Grp78, Chop and ATF4, but not Xbp1 and IRE1, suggesting that hCG may modulate IRE1 signaling pathways in a melatonin receptor-dependent manner. In addition, our further data showed that knockdown of MTNR1A and MTNR1B promoted hCG-induced expression of apoptosis markers, including p53, caspase-3 and Bcl-2. These results suggested that the melatonin receptors MTNR1A and MTNR1B are essential to repress hCG-induced ER stress and cell apoptosis. Our studies demonstrated that the mammalian melatonin receptors MT1 and MT2 are involved in testosterone synthesis via mediating multiple cell pathways.
format Online
Article
Text
id pubmed-6416941
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-64169412019-03-26 Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells Gao, Yuan Wu, Xiaochun Zhao, Shuqin Zhang, Yujun Ma, Hailong Yang, Zhen Yang, Wanghao Zhao, Chen Wang, Li Zhang, Quanwei Cell Mol Biol Lett Research Melatonin receptors MT1 and MT2 (genes officially named MTNR1A and MTNR1B, respectively) play crucial roles in melatonin-mediated regulation of circadian rhythms, the immune system, and control of reproduction in seasonally breeding animals. In this study, immunolocalization assay showed that MT1 and MT2 are highly expressed in Leydig cell membrane. To understand the biological function of melatonin receptors in hCG-induced testosterone synthesis, we generated melatonin receptor knockdown cells using specific siRNA and performed testosterone detection after hCG treatment. We found that knockdown of melatonin receptors, especially MTNR1A, led to an obvious decrease (> 60%) of testosterone level. Our further study revealed that knockdown of melatonin receptors repressed expression, at both the mRNA level and the protein level, of key steroidogenic genes, such as p450scc, p450c17 and StAR, which are essential for testosterone synthesis. hCG triggered endoplasmic reticulum (ER) stress to regulate steroidogenic genes’ expression and apoptosis. To further investigate the potential roles of melatonin receptors in hCG-induced regulation of ER stress and apoptosis, we examined expression of some crucial ER stress markers, including Grp78, Chop, ATF4, Xbp1, and IRE1. We found that inhibition of melatonin receptors increased hCG-induced expression of Grp78, Chop and ATF4, but not Xbp1 and IRE1, suggesting that hCG may modulate IRE1 signaling pathways in a melatonin receptor-dependent manner. In addition, our further data showed that knockdown of MTNR1A and MTNR1B promoted hCG-induced expression of apoptosis markers, including p53, caspase-3 and Bcl-2. These results suggested that the melatonin receptors MTNR1A and MTNR1B are essential to repress hCG-induced ER stress and cell apoptosis. Our studies demonstrated that the mammalian melatonin receptors MT1 and MT2 are involved in testosterone synthesis via mediating multiple cell pathways. BioMed Central 2019-03-12 /pmc/articles/PMC6416941/ /pubmed/30915128 http://dx.doi.org/10.1186/s11658-019-0147-z Text en © The Author(s) 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Gao, Yuan
Wu, Xiaochun
Zhao, Shuqin
Zhang, Yujun
Ma, Hailong
Yang, Zhen
Yang, Wanghao
Zhao, Chen
Wang, Li
Zhang, Quanwei
Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells
title Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells
title_full Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells
title_fullStr Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells
title_full_unstemmed Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells
title_short Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells
title_sort melatonin receptor depletion suppressed hcg-induced testosterone expression in mouse leydig cells
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6416941/
https://www.ncbi.nlm.nih.gov/pubmed/30915128
http://dx.doi.org/10.1186/s11658-019-0147-z
work_keys_str_mv AT gaoyuan melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT wuxiaochun melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT zhaoshuqin melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT zhangyujun melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT mahailong melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT yangzhen melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT yangwanghao melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT zhaochen melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT wangli melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells
AT zhangquanwei melatoninreceptordepletionsuppressedhcginducedtestosteroneexpressioninmouseleydigcells