Cargando…

Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway

Mechanical stimulation induces bone growth and remodeling by the secondary messenger, cyclic guanosine 3’, 5’-monophosphate (cGMP), in osteoblasts. However, the role of cGMP in the regulation of estrogen biosynthesis, whose deficiency is a major cause of osteoporosis, remains unclear. Here, we found...

Descripción completa

Detalles Bibliográficos
Autores principales: Wisanwattana, Wisanee, Wongkrajang, Kanjana, Cao, Dong-yi, Shi, Xiao-ke, Zhang, Zhong-hui, Zhou, Zong-yuan, Li, Fu, Mei, Qing-gang, Wang, Chun, Suksamrarn, Apichart, Zhang, Guo-lin, Wang, Fei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7995890/
https://www.ncbi.nlm.nih.gov/pubmed/33776932
http://dx.doi.org/10.3389/fendo.2021.636784
_version_ 1783670002279251968
author Wisanwattana, Wisanee
Wongkrajang, Kanjana
Cao, Dong-yi
Shi, Xiao-ke
Zhang, Zhong-hui
Zhou, Zong-yuan
Li, Fu
Mei, Qing-gang
Wang, Chun
Suksamrarn, Apichart
Zhang, Guo-lin
Wang, Fei
author_facet Wisanwattana, Wisanee
Wongkrajang, Kanjana
Cao, Dong-yi
Shi, Xiao-ke
Zhang, Zhong-hui
Zhou, Zong-yuan
Li, Fu
Mei, Qing-gang
Wang, Chun
Suksamrarn, Apichart
Zhang, Guo-lin
Wang, Fei
author_sort Wisanwattana, Wisanee
collection PubMed
description Mechanical stimulation induces bone growth and remodeling by the secondary messenger, cyclic guanosine 3’, 5’-monophosphate (cGMP), in osteoblasts. However, the role of cGMP in the regulation of estrogen biosynthesis, whose deficiency is a major cause of osteoporosis, remains unclear. Here, we found that the prenylated flavonoids, 3-O-methoxymethyl-7-O-benzylicaritin (13), 7-O-benzylicaritin (14), and 4'-O-methyl-8-isopentylkaempferol (15), which were synthesized using icariin analogs, promoted estrogen biosynthesis in osteoblastic UMR106 cells, with calculated EC(50) values of 1.53, 3.45, and 10.57 µM, respectively. 14 and 15 increased the expression level of the bone specific promoter I.4-driven aromatase, the only enzyme that catalyzes estrogen formation by using androgens as substrates, in osteoblastic cells. 14 inhibited phosphodiesterase 5 (PDE5), stimulated intracellular cGMP level and promoted osteoblast cell differentiation. Inhibition of cGMP dependent-protein kinase G (PKG) abolished the stimulatory effect of 14 on estrogen biosynthesis and osteoblast cell differentiation. Further, PKG activation by 14 stimulated the activity of SHP2 (Src homology 2 domain-containing tyrosine phosphatase 2), thereby activating Src and ERK (extracellular signal-regulated kinase) signaling and increasing ERK-dependent aromatase expression in osteoblasts. Our findings reveal a previously unknown role of cGMP in the regulation of estrogen biosynthesis in the bone. These results support the further development of 14 as a PKG-activating drug to mimic the anabolic effects of mechanical stimulation of bone in the treatment of osteoporosis.
format Online
Article
Text
id pubmed-7995890
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-79958902021-03-27 Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway Wisanwattana, Wisanee Wongkrajang, Kanjana Cao, Dong-yi Shi, Xiao-ke Zhang, Zhong-hui Zhou, Zong-yuan Li, Fu Mei, Qing-gang Wang, Chun Suksamrarn, Apichart Zhang, Guo-lin Wang, Fei Front Endocrinol (Lausanne) Endocrinology Mechanical stimulation induces bone growth and remodeling by the secondary messenger, cyclic guanosine 3’, 5’-monophosphate (cGMP), in osteoblasts. However, the role of cGMP in the regulation of estrogen biosynthesis, whose deficiency is a major cause of osteoporosis, remains unclear. Here, we found that the prenylated flavonoids, 3-O-methoxymethyl-7-O-benzylicaritin (13), 7-O-benzylicaritin (14), and 4'-O-methyl-8-isopentylkaempferol (15), which were synthesized using icariin analogs, promoted estrogen biosynthesis in osteoblastic UMR106 cells, with calculated EC(50) values of 1.53, 3.45, and 10.57 µM, respectively. 14 and 15 increased the expression level of the bone specific promoter I.4-driven aromatase, the only enzyme that catalyzes estrogen formation by using androgens as substrates, in osteoblastic cells. 14 inhibited phosphodiesterase 5 (PDE5), stimulated intracellular cGMP level and promoted osteoblast cell differentiation. Inhibition of cGMP dependent-protein kinase G (PKG) abolished the stimulatory effect of 14 on estrogen biosynthesis and osteoblast cell differentiation. Further, PKG activation by 14 stimulated the activity of SHP2 (Src homology 2 domain-containing tyrosine phosphatase 2), thereby activating Src and ERK (extracellular signal-regulated kinase) signaling and increasing ERK-dependent aromatase expression in osteoblasts. Our findings reveal a previously unknown role of cGMP in the regulation of estrogen biosynthesis in the bone. These results support the further development of 14 as a PKG-activating drug to mimic the anabolic effects of mechanical stimulation of bone in the treatment of osteoporosis. Frontiers Media S.A. 2021-03-12 /pmc/articles/PMC7995890/ /pubmed/33776932 http://dx.doi.org/10.3389/fendo.2021.636784 Text en Copyright © 2021 Wisanwattana, Wongkrajang, Cao, Shi, Zhang, Zhou, Li, Mei, Wang, Suksamrarn, Zhang and Wang http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Endocrinology
Wisanwattana, Wisanee
Wongkrajang, Kanjana
Cao, Dong-yi
Shi, Xiao-ke
Zhang, Zhong-hui
Zhou, Zong-yuan
Li, Fu
Mei, Qing-gang
Wang, Chun
Suksamrarn, Apichart
Zhang, Guo-lin
Wang, Fei
Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway
title Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway
title_full Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway
title_fullStr Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway
title_full_unstemmed Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway
title_short Inhibition of Phosphodiesterase 5 Promotes the Aromatase-Mediated Estrogen Biosynthesis in Osteoblastic Cells by Activation of cGMP/PKG/SHP2 Pathway
title_sort inhibition of phosphodiesterase 5 promotes the aromatase-mediated estrogen biosynthesis in osteoblastic cells by activation of cgmp/pkg/shp2 pathway
topic Endocrinology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7995890/
https://www.ncbi.nlm.nih.gov/pubmed/33776932
http://dx.doi.org/10.3389/fendo.2021.636784
work_keys_str_mv AT wisanwattanawisanee inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT wongkrajangkanjana inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT caodongyi inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT shixiaoke inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT zhangzhonghui inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT zhouzongyuan inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT lifu inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT meiqinggang inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT wangchun inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT suksamrarnapichart inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT zhangguolin inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway
AT wangfei inhibitionofphosphodiesterase5promotesthearomatasemediatedestrogenbiosynthesisinosteoblasticcellsbyactivationofcgmppkgshp2pathway