Please use this identifier to cite or link to this item: https://hdl.handle.net/2440/134487
Citations
Scopus Web of Science® Altmetric
?
?
Type: Journal article
Title: Non-cell autonomous mechanisms control mitochondrial gene dysregulation in polycystic ovary syndrome
Author: Moreno-Asso, A.
Altıntaş, A.
McIlvenna, L.C.
Patten, R.K.
Botella, J.
McAinch, A.J.
Rodgers, R.J.
Barrès, R.
Stepto, N.K.
Citation: Journal of Molecular Endocrinology, 2022; 68(1):63-76
Publisher: BioScientifica
Issue Date: 2022
ISSN: 0952-5041
1479-6813
Statement of
Responsibility: 
Alba Moreno-Asso, Ali Altıntaş, Luke C McIlvenna, Rhiannon K Patten, Javier Botella, Andrew J McAinch, Raymond J Rodgers, Romain Barrès and Nigel K Stepto
Abstract: Polycystic ovary syndrome (PCOS) is a common endocrine disorder associated with insulin resistance and impaired energy metabolism in skeletal muscle, the aetiology of which is currently unclear. Here, we mapped the gene expression profile of skeletal muscle from women with PCOS and determined if cultured primary myotubes retain the gene expression signature of PCOS in vivo. Transcriptomic analysis of vastus lateralis biopsies collected from PCOS women showed lower expression of genes associated with mitochondrial function while the expression of genes associated with the extracellular matrix was higher compared to controls. Altered skeletal muscle mRNA expression of mitochondrial-associated genes in PCOS was associated with lower protein expression of mitochondrial complex II to V, but not complex I, with no difference in mitochondrial DNA content. Transcriptomic analysis of primary myotube cultures established from biopsies did not display any differentially expressed genes between controls and PCOS. Comparison of gene expression profiles in skeletal muscle biopsies and primary myotube cultures showed lower expression of mitochondrial and energy metabolism-related genes in vitro, irrespective of the group. Together, our results show that the altered mitochondrial-associated gene expression in skeletal muscle in PCOS is not preserved in cultured myotubes, indicating that the in vivo extracellular milieu, rather than genetic or epigenetic factors, may drive this alteration. Dysregulation of mitochondrial-associated genes in skeletal muscle by extracellular factors may contribute to the impaired energy metabolism associated with PCOS.
Keywords: Polycystic ovary syndrome; skeletal muscle; myotubes; transcriptomics; mitochondria
Rights: © 2021 The authors. Published by Bioscientifica Ltd. This work is licensed under a Creative Commons Attribution 4.0 International License.
DOI: 10.1530/JME-21-0212
Grant ID: NHMRC
Published version: http://dx.doi.org/10.1530/jme-21-0212
Appears in Collections:Obstetrics and Gynaecology publications

Files in This Item:
File Description SizeFormat 
hdl_134487.pdfPublished version3.19 MBAdobe PDFView/Open


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.