Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2020

Open Access 01-12-2020 | Insulins | Research

Decreased brain and muscle ARNT-like protein 1 expression mediated the contribution of hyperandrogenism to insulin resistance in polycystic ovary syndrome

Authors: Junyu Zhai, Shang Li, Min Hu, Fangfang Di, Jiansheng Liu, Yanzhi Du

Published in: Reproductive Biology and Endocrinology | Issue 1/2020

Login to get access

Abstract

Background

The interface between environmental risk factors and genetic factors could contribute to the pathogenesis of hyperandrogenism and insulin resistance in polycystic ovary syndrome (PCOS); however, the underlying complex mechanism remains to be elucidated.

Methods

We used dehydroepiandrosterone (DHEA)-induced PCOS-like rat model to measure circadian clock genes and insulin resistance-related genes. Additionally, we performed in vitro experiments in mature adipocytes to verify the molecular mechanisms.

Results

DHEA-induced PCOS-like rats exhibited insulin resistance and arrhythmic expression of circadian clock genes in the liver and adipose tissues, particularly showing decreased brain and muscle ARNT-like protein 1 (BMAL1) expression. In addition, hyperandrogenism gave rise to negative regulation of BMAL1 expression to nicotinamide phosphoribosyltransferase and sirtuin 1, which further inhibited downstream glucose transporter type 4, leading to insulin resistance in mature adipocytes, which was consistent with our previous results in HepG2 cells.

Conclusions

Decreased BMAL1 expression in the liver and adipose played a potentially novel role in the contribution of hyperandrogenism to insulin resistance, which might be a possible mechanism accounting for the pathogenesis of PCOS.
Appendix
Available only for authorised users
Literature
1.
go back to reference Elia E, Sander V, Luchetti CG, Solano ME, Di Girolamo G, Gonzalez C, et al. The mechanisms involved in the action of metformin in regulating ovarian function in hyperandrogenized mice. Mol Hum Reprod. 2006;12:475–81.CrossRef Elia E, Sander V, Luchetti CG, Solano ME, Di Girolamo G, Gonzalez C, et al. The mechanisms involved in the action of metformin in regulating ovarian function in hyperandrogenized mice. Mol Hum Reprod. 2006;12:475–81.CrossRef
2.
go back to reference Stepto NK, Cassar S, Joham AE, Hutchison SK, Harrison CL, Goldstein RF, et al. Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycaemic-hyperinsulaemic clamp. Hum Reprod. 2013;28:777–84.CrossRef Stepto NK, Cassar S, Joham AE, Hutchison SK, Harrison CL, Goldstein RF, et al. Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycaemic-hyperinsulaemic clamp. Hum Reprod. 2013;28:777–84.CrossRef
3.
go back to reference Dunaif A, Wu X, Lee A, Diamanti-Kandarakis E. Defects in insulin receptor signaling in vivo in the polycystic ovary syndrome (PCOS). Am J Physiol Endocrinol Metab. 2001;281:E392–9.CrossRef Dunaif A, Wu X, Lee A, Diamanti-Kandarakis E. Defects in insulin receptor signaling in vivo in the polycystic ovary syndrome (PCOS). Am J Physiol Endocrinol Metab. 2001;281:E392–9.CrossRef
4.
go back to reference Merkin SS, Phy JL, Sites CK, Yang D. Environmental determinants of polycystic ovary syndrome. Fertil Steril. 2016;106:16–24.CrossRef Merkin SS, Phy JL, Sites CK, Yang D. Environmental determinants of polycystic ovary syndrome. Fertil Steril. 2016;106:16–24.CrossRef
5.
go back to reference Ueda HR, Hayashi S, Chen W, Sano M, Machida M, Shigeyoshi Y, et al. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat Genet. 2005;37:187–92.CrossRef Ueda HR, Hayashi S, Chen W, Sano M, Machida M, Shigeyoshi Y, et al. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat Genet. 2005;37:187–92.CrossRef
6.
go back to reference Pan X, Taylor MJ, Cohen E, Hanna N, Mota S. Circadian clock, time-restricted feeding and reproduction. Int J Mol Sci. 2020;21(3). Pan X, Taylor MJ, Cohen E, Hanna N, Mota S. Circadian clock, time-restricted feeding and reproduction. Int J Mol Sci. 2020;21(3).
7.
go back to reference Mayeuf-Louchart A, Zecchin M, Staels B, Duez H. Circadian control of metabolism and pathological consequences of clock perturbations. Biochimie. 2017;143:42–50.CrossRef Mayeuf-Louchart A, Zecchin M, Staels B, Duez H. Circadian control of metabolism and pathological consequences of clock perturbations. Biochimie. 2017;143:42–50.CrossRef
8.
go back to reference Vinogradova IA, Anisimov VN, Bukalev AV, Semenchenko AV, Zabezhinski MA. Circadian disruption induced by light-at-night accelerates aging and promotes tumorigenesis in rats. Aging (Albany NY). 2009;1:855–65.CrossRef Vinogradova IA, Anisimov VN, Bukalev AV, Semenchenko AV, Zabezhinski MA. Circadian disruption induced by light-at-night accelerates aging and promotes tumorigenesis in rats. Aging (Albany NY). 2009;1:855–65.CrossRef
9.
go back to reference Green CB, Takahashi JS, Bass J. The meter of metabolism. Cell. 2008;134:728–42.CrossRef Green CB, Takahashi JS, Bass J. The meter of metabolism. Cell. 2008;134:728–42.CrossRef
10.
go back to reference Li S, Zhai J, Chu W, Geng X, Chen ZJ, Du Y. Altered circadian clock as a novel therapeutic target for constant darkness-induced insulin resistance and hyperandrogenism of polycystic ovary syndrome. Transl Res. 2020;219:13–29.CrossRef Li S, Zhai J, Chu W, Geng X, Chen ZJ, Du Y. Altered circadian clock as a novel therapeutic target for constant darkness-induced insulin resistance and hyperandrogenism of polycystic ovary syndrome. Transl Res. 2020;219:13–29.CrossRef
11.
go back to reference Spinedi E, Cardinali DP. The polycystic ovary syndrome and the metabolic syndrome: a possible chronobiotic-cytoprotective adjuvant therapy. Int J Endocrinol. 2018;2018:1349868.CrossRef Spinedi E, Cardinali DP. The polycystic ovary syndrome and the metabolic syndrome: a possible chronobiotic-cytoprotective adjuvant therapy. Int J Endocrinol. 2018;2018:1349868.CrossRef
12.
go back to reference Sellix MT, Murphy ZC, Menaker M. Excess androgen during puberty disrupts circadian organization in female rats. Endocrinology. 2013;154:1636–47.CrossRef Sellix MT, Murphy ZC, Menaker M. Excess androgen during puberty disrupts circadian organization in female rats. Endocrinology. 2013;154:1636–47.CrossRef
13.
go back to reference Mereness AL, Murphy ZC, Sellix MT. Developmental programming by androgen affects the circadian timing system in female mice. Biol Reprod. 2015;92:88.CrossRef Mereness AL, Murphy ZC, Sellix MT. Developmental programming by androgen affects the circadian timing system in female mice. Biol Reprod. 2015;92:88.CrossRef
14.
go back to reference Simon SL, McWhirter L, Diniz BC, Bubar KM, Kaar JL, Pyle L, et al. Morning circadian misalignment is associated with insulin resistance in girls with obesity and polycystic ovarian syndrome. J Clin Endocrinol Metab. 2019;104:3525–34.CrossRef Simon SL, McWhirter L, Diniz BC, Bubar KM, Kaar JL, Pyle L, et al. Morning circadian misalignment is associated with insulin resistance in girls with obesity and polycystic ovarian syndrome. J Clin Endocrinol Metab. 2019;104:3525–34.CrossRef
15.
go back to reference Pan Y, Shu JL, Gu HF, Zhou DC, Liu XL, Qiao QY, et al. Erythropoietin improves insulin resistance via the regulation of its receptor-mediated signaling pathways in 3T3L1 adipocytes. Mol Cell Endocrinol. 2013;367:116–23.CrossRef Pan Y, Shu JL, Gu HF, Zhou DC, Liu XL, Qiao QY, et al. Erythropoietin improves insulin resistance via the regulation of its receptor-mediated signaling pathways in 3T3L1 adipocytes. Mol Cell Endocrinol. 2013;367:116–23.CrossRef
16.
go back to reference Caldwell AS, Middleton LJ, Jimenez M, Desai R, McMahon AC, Allan CM, et al. Characterization of reproductive, metabolic, and endocrine features of polycystic ovary syndrome in female hyperandrogenic mouse models. Endocrinology. 2014;155:3146–59.CrossRef Caldwell AS, Middleton LJ, Jimenez M, Desai R, McMahon AC, Allan CM, et al. Characterization of reproductive, metabolic, and endocrine features of polycystic ovary syndrome in female hyperandrogenic mouse models. Endocrinology. 2014;155:3146–59.CrossRef
17.
go back to reference Stromsdorfer KL, Yamaguchi S, Yoon MJ, Moseley AC, Franczyk MP, Kelly SC, et al. NAMPT-mediated NAD(+) biosynthesis in adipocytes regulates adipose tissue function and multi-organ insulin sensitivity in mice. Cell Rep. 2016;16:1851–60.CrossRef Stromsdorfer KL, Yamaguchi S, Yoon MJ, Moseley AC, Franczyk MP, Kelly SC, et al. NAMPT-mediated NAD(+) biosynthesis in adipocytes regulates adipose tissue function and multi-organ insulin sensitivity in mice. Cell Rep. 2016;16:1851–60.CrossRef
18.
go back to reference Zhou B, Zhang Y, Zhang F, Xia Y, Liu J, Huang R, et al. CLOCK/BMAL1 regulates circadian change of mouse hepatic insulin sensitivity by SIRT1. Hepatology. 2014;59:2196–206.CrossRef Zhou B, Zhang Y, Zhang F, Xia Y, Liu J, Huang R, et al. CLOCK/BMAL1 regulates circadian change of mouse hepatic insulin sensitivity by SIRT1. Hepatology. 2014;59:2196–206.CrossRef
19.
go back to reference Revollo JR, Grimm AA, Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J Biol Chem. 2004;279:50754–63.CrossRef Revollo JR, Grimm AA, Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J Biol Chem. 2004;279:50754–63.CrossRef
20.
go back to reference Bednarska S, Siejka A. The pathogenesis and treatment of polycystic ovary syndrome: What's new? Adv Clin Exp Med. 2017;26:359–67.CrossRef Bednarska S, Siejka A. The pathogenesis and treatment of polycystic ovary syndrome: What's new? Adv Clin Exp Med. 2017;26:359–67.CrossRef
21.
go back to reference Zhang D, Tong X, Arthurs B, Guha A, Rui L, Kamath A, et al. Liver clock protein BMAL1 promotes de novo lipogenesis through insulin-mTORC2-AKT signaling. J Biol Chem. 2014;289:25925–35.CrossRef Zhang D, Tong X, Arthurs B, Guha A, Rui L, Kamath A, et al. Liver clock protein BMAL1 promotes de novo lipogenesis through insulin-mTORC2-AKT signaling. J Biol Chem. 2014;289:25925–35.CrossRef
22.
go back to reference Kennaway DJ, Varcoe TJ, Voultsios A, Boden MJ. Global loss of bmal1 expression alters adipose tissue hormones, gene expression and glucose metabolism. PLoS One. 2013;8:e65255.CrossRef Kennaway DJ, Varcoe TJ, Voultsios A, Boden MJ. Global loss of bmal1 expression alters adipose tissue hormones, gene expression and glucose metabolism. PLoS One. 2013;8:e65255.CrossRef
23.
go back to reference Zhang J, Liu J, Zhu K, Hong Y, Sun Y, Zhao X, et al. Effects of BMAL1-SIRT1-positive cycle on estrogen synthesis in human ovarian granulosa cells: an implicative role of BMAL1 in PCOS. Endocrine. 2016;53:574–84.CrossRef Zhang J, Liu J, Zhu K, Hong Y, Sun Y, Zhao X, et al. Effects of BMAL1-SIRT1-positive cycle on estrogen synthesis in human ovarian granulosa cells: an implicative role of BMAL1 in PCOS. Endocrine. 2016;53:574–84.CrossRef
24.
go back to reference Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B, et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009;324:651–4.CrossRef Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B, et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009;324:651–4.CrossRef
25.
go back to reference Cloix L, Reverchon M, Cornuau M, Froment P, Rame C, Costa C, et al. Expression and regulation of INTELECTIN1 in human granulosa-lutein cells: role in IGF-1-induced steroidogenesis through NAMPT. Biol Reprod. 2014;91:50.CrossRef Cloix L, Reverchon M, Cornuau M, Froment P, Rame C, Costa C, et al. Expression and regulation of INTELECTIN1 in human granulosa-lutein cells: role in IGF-1-induced steroidogenesis through NAMPT. Biol Reprod. 2014;91:50.CrossRef
26.
go back to reference Imai S, Yoshino J. The importance of NAMPT/NAD/SIRT1 in the systemic regulation of metabolism and ageing. Diabetes Obes Metab. 2013;15(Suppl 3):26–33.CrossRef Imai S, Yoshino J. The importance of NAMPT/NAD/SIRT1 in the systemic regulation of metabolism and ageing. Diabetes Obes Metab. 2013;15(Suppl 3):26–33.CrossRef
27.
go back to reference Karatsoreos IN, Wang A, Sasanian J, Silver R. A role for androgens in regulating circadian behavior and the suprachiasmatic nucleus. Endocrinology. 2007;148:5487–95.CrossRef Karatsoreos IN, Wang A, Sasanian J, Silver R. A role for androgens in regulating circadian behavior and the suprachiasmatic nucleus. Endocrinology. 2007;148:5487–95.CrossRef
28.
go back to reference Fujioka K, Kajita K, Wu Z, Hanamoto T, Ikeda T, Mori I, et al. Dehydroepiandrosterone reduces preadipocyte proliferation via androgen receptor. Am J Physiol Endocrinol Metab. 2012;302:E694–704.CrossRef Fujioka K, Kajita K, Wu Z, Hanamoto T, Ikeda T, Mori I, et al. Dehydroepiandrosterone reduces preadipocyte proliferation via androgen receptor. Am J Physiol Endocrinol Metab. 2012;302:E694–704.CrossRef
29.
go back to reference De Gendt K, Verhoeven G. Tissue- and cell-specific functions of the androgen receptor revealed through conditional knockout models in mice. Mol Cell Endocrinol. 2012;352:13–25.CrossRef De Gendt K, Verhoeven G. Tissue- and cell-specific functions of the androgen receptor revealed through conditional knockout models in mice. Mol Cell Endocrinol. 2012;352:13–25.CrossRef
Metadata
Title
Decreased brain and muscle ARNT-like protein 1 expression mediated the contribution of hyperandrogenism to insulin resistance in polycystic ovary syndrome
Authors
Junyu Zhai
Shang Li
Min Hu
Fangfang Di
Jiansheng Liu
Yanzhi Du
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Reproductive Biology and Endocrinology / Issue 1/2020
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-020-00592-1

Other articles of this Issue 1/2020

Reproductive Biology and Endocrinology 1/2020 Go to the issue