Cargando…

Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway

Granulosa cells (GCs) are essential for follicular growth, development, and atresia. The orexin-A (OXA) neuropeptide is widely involved in the regulation of various biological functions. OXA selectively binds to orexin receptor type 1 (OX1R) and mediates all its biological actions via OX1R. This stu...

Descripción completa

Detalles Bibliográficos
Autores principales: Safdar, Muhammad, Liang, Aixin, Rajput, Shahid Ali, Abbas, Nasir, Zubair, Muhammad, Shaukat, Aftab, Rehman, Aziz ur, Jamil, Huma, Guo, Yan, Ullah, Farman, Yang, Liguo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8467508/
https://www.ncbi.nlm.nih.gov/pubmed/34577105
http://dx.doi.org/10.3390/molecules26185635
_version_ 1784573415850508288
author Safdar, Muhammad
Liang, Aixin
Rajput, Shahid Ali
Abbas, Nasir
Zubair, Muhammad
Shaukat, Aftab
Rehman, Aziz ur
Jamil, Huma
Guo, Yan
Ullah, Farman
Yang, Liguo
author_facet Safdar, Muhammad
Liang, Aixin
Rajput, Shahid Ali
Abbas, Nasir
Zubair, Muhammad
Shaukat, Aftab
Rehman, Aziz ur
Jamil, Huma
Guo, Yan
Ullah, Farman
Yang, Liguo
author_sort Safdar, Muhammad
collection PubMed
description Granulosa cells (GCs) are essential for follicular growth, development, and atresia. The orexin-A (OXA) neuropeptide is widely involved in the regulation of various biological functions. OXA selectively binds to orexin receptor type 1 (OX1R) and mediates all its biological actions via OX1R. This study aimed to explore the expression of OXA and OX1R and their regulatory role in GCs proliferation, cell cycle progression, apoptosis, oocyte maturation, and underlying molecular mechanisms of these processes and elucidate its novel signaling pathway. Western blotting and RT-qPCR showed that OXA and OX1R were expressed during different developmental stages of GCs, and siRNA transfection successfully inhibited the expression of OX1R at the translational and transcriptional levels. Flow cytometry revealed that OX1R knockdown upregulated GCs apoptosis and triggered S-phase arrest in cell cycle progression. RT-qPCR and Western blotting showed significantly reduced expression of Bcl-2 and elevated expression of Bax, caspase-3, TNF-α, and P21 in OX1R-silenced GCs. Furthermore, the CCK-8 assay showed that knockdown of OX1R suppressed GCs proliferation by downregulating the expression of PCNA, a proliferation marker gene, at the translational and transcriptional levels. Western blotting revealed that knockdown of OX1R resulted in a considerable decrease of the phosphorylation level of the AKT and ERK1/2 proteins, indicating that the AKT/ERK1/2 pathway is involved in regulating GCs proliferation and apoptosis. In addition, OX1R silencing enhanced the mRNA expression of GDF9 and suppressed the mRNA expression of BMP15 in mouse GCs. Collectively, these results reveal a novel regulatory role of OXA in the development of GCs and folliculogenesis by regulating proliferation, apoptosis, and cell cycle progression. Therefore, OXA can be a promising therapeutic agent for female infertility.
format Online
Article
Text
id pubmed-8467508
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-84675082021-09-27 Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway Safdar, Muhammad Liang, Aixin Rajput, Shahid Ali Abbas, Nasir Zubair, Muhammad Shaukat, Aftab Rehman, Aziz ur Jamil, Huma Guo, Yan Ullah, Farman Yang, Liguo Molecules Article Granulosa cells (GCs) are essential for follicular growth, development, and atresia. The orexin-A (OXA) neuropeptide is widely involved in the regulation of various biological functions. OXA selectively binds to orexin receptor type 1 (OX1R) and mediates all its biological actions via OX1R. This study aimed to explore the expression of OXA and OX1R and their regulatory role in GCs proliferation, cell cycle progression, apoptosis, oocyte maturation, and underlying molecular mechanisms of these processes and elucidate its novel signaling pathway. Western blotting and RT-qPCR showed that OXA and OX1R were expressed during different developmental stages of GCs, and siRNA transfection successfully inhibited the expression of OX1R at the translational and transcriptional levels. Flow cytometry revealed that OX1R knockdown upregulated GCs apoptosis and triggered S-phase arrest in cell cycle progression. RT-qPCR and Western blotting showed significantly reduced expression of Bcl-2 and elevated expression of Bax, caspase-3, TNF-α, and P21 in OX1R-silenced GCs. Furthermore, the CCK-8 assay showed that knockdown of OX1R suppressed GCs proliferation by downregulating the expression of PCNA, a proliferation marker gene, at the translational and transcriptional levels. Western blotting revealed that knockdown of OX1R resulted in a considerable decrease of the phosphorylation level of the AKT and ERK1/2 proteins, indicating that the AKT/ERK1/2 pathway is involved in regulating GCs proliferation and apoptosis. In addition, OX1R silencing enhanced the mRNA expression of GDF9 and suppressed the mRNA expression of BMP15 in mouse GCs. Collectively, these results reveal a novel regulatory role of OXA in the development of GCs and folliculogenesis by regulating proliferation, apoptosis, and cell cycle progression. Therefore, OXA can be a promising therapeutic agent for female infertility. MDPI 2021-09-16 /pmc/articles/PMC8467508/ /pubmed/34577105 http://dx.doi.org/10.3390/molecules26185635 Text en © 2021 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Safdar, Muhammad
Liang, Aixin
Rajput, Shahid Ali
Abbas, Nasir
Zubair, Muhammad
Shaukat, Aftab
Rehman, Aziz ur
Jamil, Huma
Guo, Yan
Ullah, Farman
Yang, Liguo
Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway
title Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway
title_full Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway
title_fullStr Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway
title_full_unstemmed Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway
title_short Orexin-A Regulates Follicular Growth, Proliferation, Cell Cycle and Apoptosis in Mouse Primary Granulosa Cells via the AKT/ERK Signaling Pathway
title_sort orexin-a regulates follicular growth, proliferation, cell cycle and apoptosis in mouse primary granulosa cells via the akt/erk signaling pathway
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8467508/
https://www.ncbi.nlm.nih.gov/pubmed/34577105
http://dx.doi.org/10.3390/molecules26185635
work_keys_str_mv AT safdarmuhammad orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT liangaixin orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT rajputshahidali orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT abbasnasir orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT zubairmuhammad orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT shaukataftab orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT rehmanazizur orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT jamilhuma orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT guoyan orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT ullahfarman orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway
AT yangliguo orexinaregulatesfolliculargrowthproliferationcellcycleandapoptosisinmouseprimarygranulosacellsviatheakterksignalingpathway