1
|
Tatone C, Di Emidio G, Battaglia R, Di Pietro C. Building a Human Ovarian Antioxidant ceRNA Network "OvAnOx": A Bioinformatic Perspective for Research on Redox-Related Ovarian Functions and Dysfunctions. Antioxidants (Basel) 2024; 13:1101. [PMID: 39334761 PMCID: PMC11428640 DOI: 10.3390/antiox13091101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
The ovary is a major determinant of female reproductive health. Ovarian functions are mainly related to the primordial follicle pool, which is gradually lost with aging. Ovarian aging and reproductive dysfunctions share oxidative stress as a common underlying mechanism. ROS signaling is essential for normal ovarian processes, yet it can contribute to various ovarian disorders when disrupted. Therefore, balance in the redox system is crucial for proper ovarian functions. In the present study, by focusing on mRNAs and ncRNAs described in the ovary and taking into account only validated ncRNA interactions, we built an ovarian antioxidant ceRNA network, named OvAnOx ceRNA, composed of 5 mRNAs (SOD1, SOD2, CAT, PRDX3, GR), 10 miRNAs and 5 lncRNAs (XIST, FGD5-AS1, MALAT1, NEAT1, SNHG1). Our bioinformatic analysis indicated that the components of OvAnOx ceRNA not only contribute to antioxidant defense but are also involved in other ovarian functions. Indeed, antioxidant enzymes encoded by mRNAs of OvAnOx ceRNA operate within a regulatory network that impacts ovarian reserve, follicular dynamics, and oocyte maturation in normal and pathological conditions. The OvAnOx ceRNA network represents a promising tool to unravel the complex dialog between redox potential and ovarian signaling pathways involved in reproductive health, aging, and diseases.
Collapse
Affiliation(s)
- Carla Tatone
- Department of Life, Health and Experimental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (C.T.); (G.D.E.)
| | - Giovanna Di Emidio
- Department of Life, Health and Experimental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (C.T.); (G.D.E.)
| | - Rosalia Battaglia
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics, University of Catania, 95123 Catania, Italy;
| | - Cinzia Di Pietro
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
2
|
Garcia-Beltran C, Malpique R, Andersen MS, Bas F, Bassols J, Darendeliler F, Díaz M, Dieris B, Fanelli F, Fröhlich-Reiterer E, Gambineri A, Glintborg D, López-Bermejo A, Mann C, Marin S, Obermayer-Pietsch B, Ødegård R, Ravn P, Reinehr T, Renzulli M, Salvador C, Singer V, Vanky E, Torres JV, Yildiz M, de Zegher F, Ibáñez L. SPIOMET4HEALTH-efficacy, tolerability and safety of lifestyle intervention plus a fixed dose combination of spironolactone, pioglitazone and metformin (SPIOMET) for adolescent girls and young women with polycystic ovary syndrome: study protocol for a multicentre, randomised, double-blind, placebo-controlled, four-arm, parallel-group, phase II clinical trial. Trials 2023; 24:589. [PMID: 37715279 PMCID: PMC10503102 DOI: 10.1186/s13063-023-07593-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most prevalent, chronic endocrine-metabolic disorder of adolescents and young women (AYAs), affecting 5-10% of AYAs worldwide. There is no approved pharmacological therapy for PCOS. Standard off-label treatment with oral contraceptives (OCs) reverts neither the underlying pathophysiology nor the associated co-morbidities. Pilot studies have generated new insights into the pathogenesis of PCOS, leading to the development of a new treatment consisting of a fixed, low-dose combination of two so-called insulin sensitisers [pioglitazone (PIO), metformin (MET)] and one mixed anti-androgen and anti-mineralocorticoid also acting as an activator of brown adipose tissue [spironolactone (SPI)], within a single tablet (SPIOMET). The present trial will evaluate the efficacy, tolerability and safety of SPIOMET, on top of lifestyle measures, for the treatment of PCOS in AYAs. METHODS In this multicentre, randomised, double-blind, placebo-controlled, four-arm, parallel-group, phase II clinical trial, AYAs with PCOS will be recruited from 7 clinical centres across Europe. Intention is to randomise a total of 364 eligible patients into four arms (1:1:1:1): Placebo, PIO, SPI + PIO (SPIO) and SPI + PIO + MET (SPIOMET). Active treatment over 12 months will consist of lifestyle guidance plus the ingestion of one tablet daily (at dinner time); post-treatment follow-up will span 6 months. Primary endpoint is on- and post-treatment ovulation rate. Secondary endpoints are clinical features (hirsutism, menstrual regularity); endocrine-metabolic variables (androgens, lipids, insulin, inflammatory markers); epigenetic markers; imaging data (carotid intima-media thickness, body composition, abdominal fat partitioning, hepatic fat); safety profile; adherence, tolerability and acceptability of the medication; and quality of life in the study participants. Superiority (in this order) of SPIOMET, SPIO and PIO will be tested over placebo, and if present, subsequently the superiority of SPIOMET versus PIO, and if still present, finally versus SPIO. DISCUSSION The present study will be the first to evaluate-in a randomised, double-blind, placebo-controlled way-the efficacy, tolerability and safety of SPIOMET treatment for early PCOS, on top of a lifestyle intervention. TRIAL REGISTRATION EudraCT 2021-003177-58. Registered on 22 December 2021. https://www.clinicaltrialsregister.eu/ctr-search/search?query=%092021-003177-58 .
Collapse
Affiliation(s)
- Cristina Garcia-Beltran
- Paediatric Endocrinology, Paediatric Research Institute Sant Joan de Déu, University of Barcelona, 08950, Esplugues, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, 28029, Spain
| | - Rita Malpique
- Paediatric Endocrinology, Paediatric Research Institute Sant Joan de Déu, University of Barcelona, 08950, Esplugues, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, 28029, Spain
| | - Marianne S Andersen
- Department of Gynaecology and Obstetrics and Department of Endocrinology, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Firdevs Bas
- Pediatric Endocrinology Unit, Istanbul University, Istanbul, Turkey
| | - Judit Bassols
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), Girona, Spain
| | | | - Marta Díaz
- Paediatric Endocrinology, Paediatric Research Institute Sant Joan de Déu, University of Barcelona, 08950, Esplugues, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, 28029, Spain
| | - Barbara Dieris
- Department of Paediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten-Herdecke, Datteln, Germany
| | - Flaminia Fanelli
- Department of Medical and Surgical Science-DIMEC, Division of Endocrinology and Diabetes Prevention and Care, University of Bologna - S. Orsola-Hospital, Bologna, Italy
| | - Elke Fröhlich-Reiterer
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Alessandra Gambineri
- Department of Medical and Surgical Science-DIMEC, Division of Endocrinology and Diabetes Prevention and Care, University of Bologna - S. Orsola-Hospital, Bologna, Italy
| | - Dorte Glintborg
- Department of Gynaecology and Obstetrics and Department of Endocrinology, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Abel López-Bermejo
- Paediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), Paediatrics, Dr. Josep Trueta Hospital, Department of Medical Sciences, University of Girona, Girona, Spain
| | | | - Silvia Marin
- Paediatric Endocrinology, Paediatric Research Institute Sant Joan de Déu, University of Barcelona, 08950, Esplugues, Barcelona, Spain
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Rønnaug Ødegård
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Centre for Obesity Research, St. Olavs Hospital, Trondheim University Hospital, Torgarden, Trondheim, Norway
| | - Pernille Ravn
- Department of Gynaecology and Obstetrics and Department of Endocrinology, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Thomas Reinehr
- Department of Paediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten-Herdecke, Datteln, Germany
| | - Matteo Renzulli
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Cristina Salvador
- Paediatric Endocrinology, Paediatric Research Institute Sant Joan de Déu, University of Barcelona, 08950, Esplugues, Barcelona, Spain
| | - Viola Singer
- Department of Paediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten-Herdecke, Datteln, Germany
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Obstetrics and Gynaecology, St. Olavs Hospital, Trondheim University Hospital, 7006, Trondheim, Norway
| | | | - Melek Yildiz
- Pediatric Endocrinology Unit, Istanbul University, Istanbul, Turkey
| | - Francis de Zegher
- Leuven Research & Development, University of Leuven, 3000, Louvain, Belgium
| | - Lourdes Ibáñez
- Paediatric Endocrinology, Paediatric Research Institute Sant Joan de Déu, University of Barcelona, 08950, Esplugues, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, 28029, Spain.
| |
Collapse
|
3
|
Ibáñez L, de Zegher F. Adolescent PCOS: a postpubertal central obesity syndrome. Trends Mol Med 2023; 29:354-363. [PMID: 36964058 DOI: 10.1016/j.molmed.2023.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/26/2023]
Abstract
Adolescent polycystic ovary syndrome (PCOS) is a highly prevalent, reversible, endocrine-metabolic mode essentially driven by ectopic fat, which, in turn, often results from a mismatch between early adipogenesis and later lipogenesis, or between prenatal and postnatal weight gain. The key features of adolescent PCOS are menstrual irregularity and androgen excess (hirsutism, acne, and/or high testosterone). Adolescent PCOS is frequently preceded by rapid maturation (early variants of adrenarche/pubarche and puberty/menarche, also accelerated by ectopic fat) and is diagnosed between 2 and 8 years after menarche, thus during late adolescence or early adulthood. Treatment of adolescent PCOS should not only focus on symptoms, but also reduce the amount of ectopic fat, thereby aiming for an overall state of preconception health.
Collapse
Affiliation(s)
- Lourdes Ibáñez
- Endocrinology Department, Research Institute Sant Joan de Déu, University of Barcelona, 08950 Esplugues, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Francis de Zegher
- Leuven Research and Development, University of Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
4
|
Xu C, Luo M, Liu X, Wei T, Zhou Z, Li C, He Z, Sui H. MicroRNA-1298-5p in granulosa cells facilitates cell autophagy in polycystic ovary syndrome by suppressing glutathione-disulfide reductase. Cell Tissue Res 2023:10.1007/s00441-023-03747-9. [PMID: 36781484 DOI: 10.1007/s00441-023-03747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/23/2023] [Indexed: 02/15/2023]
Abstract
The aim of this study was to investigate the effect and mechanism of action of miR-1298-5p in polycystic ovary syndrome (PCOS). Granulosa cells were isolated from follicular fluid of patients with PCOS and healthy women, and the expression of miR-1298-5p and glutathione-disulfide reductase (GSR) mRNA in these cells was evaluated using reverse transcription-quantitative polymerase chain reaction (qRT-PCR). Clinical data were obtained from all subjects, and reproductive hormones and endocrine indices were assayed to analyze the correlation between miR-1298-5p and clinicopathological characteristics of patients with PCOS. Following transfection with the miR-1298-5p mimic or inhibitor and/or pcDNA3.1-GSR, LC3 immunofluorescence and transmission electron microscopy were used to evaluate autophagy in the COV434 human granulosa cell line. Additionally, western blotting was performed to detect LC3-II, Beclin 1, and p62 protein levels in COV434 cells. The interaction between miR-1298-5p and GSR was also examined. A PCOS rat model was established and injected with the miR-1298-5p antagomir, followed by measurement of body and ovary weights, histological examination, and autophagosome observation. The protein expression levels of GSR, LC3-II, Beclin 1, and p62 were determined in rat ovaries. miR-1298-5p was expressed at a high level, and GSR was downregulated in granulosa cells from patients with PCOS. In COV434 cells, miR-1298-5p inversely mediated GSR expression, and miR-1298-5p mimic transfection promoted autophagy, whereas GSR overexpression blocked miR-1298-5p mimic-promoted autophagy. In PCOS rats, miR-1298-5p inhibition reduced autophagy and alleviated abnormalities in follicular development. Overall, miR-1298-5p enhances autophagy in granulosa cells by downregulating GSR, thereby affecting PCOS development.
Collapse
Affiliation(s)
- Changlong Xu
- The Reproductive Medical Center, Nanning Second People's Hospital, No.13 Dancun Road, Nanning, Guangxi 530031, People's Republic of China
| | - Mingjiu Luo
- College of Animal Science and Veterinary, Shandong Agricultural University, Tai'an, Shandong 271018, People's Republic of China
| | - Xiaodong Liu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699, Qingdao Road, Huaiyin District, Jinan, Shandong, 250000, People's Republic of China
| | - Tao Wei
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699, Qingdao Road, Huaiyin District, Jinan, Shandong, 250000, People's Republic of China
| | - Zheng Zhou
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699, Qingdao Road, Huaiyin District, Jinan, Shandong, 250000, People's Republic of China
| | - Changze Li
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699, Qingdao Road, Huaiyin District, Jinan, Shandong, 250000, People's Republic of China
| | - Zilin He
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699, Qingdao Road, Huaiyin District, Jinan, Shandong, 250000, People's Republic of China
| | - Hongshu Sui
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699, Qingdao Road, Huaiyin District, Jinan, Shandong, 250000, People's Republic of China.
| |
Collapse
|
5
|
Garcia-Beltran C, Bassols J, Carreras-Badosa G, López Bermejo A, Ibáñez L, de Zegher F. Raised Thyroid-Stimulating Hormone in Girls with Polycystic Ovary Syndrome: Effects of Randomized Interventions. Horm Res Paediatr 2023; 96:458-464. [PMID: 36649684 DOI: 10.1159/000529183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) in women associates with raised levels of circulating thyroid-stimulating hormone (TSH) and with high rates of gestational complications. A low range of preconception TSH is followed by low rates of gestational complications. It is unknown whether TSH levels are elevated in adolescents with PCOS and, if so, whether traditional or exploratory treatments can lower them into safe preconception range. We investigated TSH in nonobese adolescents with PCOS, including the effects of randomized interventions. METHODS Morning TSH was a safety marker in randomized pilot studies comparing the effects of an oral contraceptive (OC) versus those of a low-dose combination of spironolactone-pioglitazone-metformin (SPIOMET) in nonobese adolescents with PCOS. A post hoc analysis compared TSH levels in PCOS (N = 62) versus controls, TSH changes on treatment (for 1 year), and TSH levels posttreatment (for 1 year). RESULTS Mean TSH levels were higher in PCOS patients than in control girls (p < 0.01). On-treatment TSH levels diverged (p < 0.001), remaining elevated on OC, and descending swiftly on SPIOMET, well into safe preconception range. Posttreatment TSH levels were stable in both subgroups. On-treatment changes of circulating TSH associated to those of liver fat (R = 0.307, p = 0.017). CONCLUSION The endocrine signature of early PCOS is herewith extended to include modestly raised levels of circulating TSH; the normalizing effects of SPIOMET intervention in nonobese adolescents with PCOS are herewith extended to include on- and posttreatment TSH.
Collapse
Affiliation(s)
- Cristina Garcia-Beltran
- Endocrinology Department, Research Institute Sant Joan de Déu, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Judit Bassols
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), Girona, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), Girona, Spain
| | - Abel López Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), Girona, Spain
- Department of Pediatrics, Dr. Josep Trueta Hospital, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Lourdes Ibáñez
- Endocrinology Department, Research Institute Sant Joan de Déu, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Francis de Zegher
- Leuven Research and Development, University of Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Huang CC, Yang PK, Huang YS, Chen SU, Yang YS, Chen MJ. The role of circulating miRNAs in mechanism of action and prediction of therapeutic responses of metformin in polycystic ovarian syndrome. Fertil Steril 2023; 119:858-868. [PMID: 36627014 DOI: 10.1016/j.fertnstert.2022.12.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/30/2022] [Accepted: 12/31/2022] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To study the involvement of microribonucleic acids (miRNAs) in the pathogenesis of chronic anovulation and mechanism of metformin treatment in polycystic ovary syndrome (PCOS). DESIGN Case-control and prospective validation cohort study. SETTING Tertiary university hospital. PATIENT(S) A total of 146 patients with PCOS and chronic anovulation and 20 non-PCOS controls were enrolled. Patients who resumed ovulation after metformin treatment (MET-OV) and remained anovulatory after metformin treatment (MET-AO) were assigned to MET-OV and MET-AO groups, respectively. INTERVENTION(S) All patients with PCOS received metformin treatment for 6 months. MAIN OUTCOME MEASURE(S) Baseline and chronological changes in the plasma levels of 14 miRNAs (miR-21, 93, 132, 193b, 221, 222, 223, 27a, 125b, 200b, 212, 320a, 429, and 483) selected by literature review, anthropometric data, and hormonal as well as metabolic profiles were measured. Predictive modeling based on baseline circulatory miRNA levels and clinical parameters was performed to predict ovulation recovery after metformin treatment. RESULT(S) No significant differences were observed in the baseline hormonal and metabolic profiles between the MET-OV and MET-AO groups. However, the expression of miR-27a, miR-93, and miR-222 was significantly higher in the MET-OV group than that for the MET-AO and control groups. After 6 months of metformin treatment, the levels of insulin, luteinizing hormone, and 6 circulating miRNAs (miR-21, 27a, 93, 221, 222, and 223) and homeostatic model assessment for insulin resistance decreased significantly in the MET-OV group, but remained unchanged in the MET-AO group. The area under curve, sensitivity, and specificity of the adjusted prediction model, based on miRNA levels and clinical parameters using logistic regression analysis for predicting ovulatory response after metformin treatment, were 0.807, 0.892, and 0.632, respectively. CONCLUSION(S) The present study demonstrated a distinct pattern of baseline expression and chronological changes in the levels of several circulatory miRNAs between the MET-OV and MET-AO groups, suggesting that aberrantly overexpressed diabetogenic miRNAs are involved in the pathophysiology of chronic anovulation in PCOS, and their down-regulation might contribute toward the therapeutic effects of metformin. This could provide new insights into the mechanism of action and applicability of individualized metformin therapy in women with PCOS.
Collapse
Affiliation(s)
- Chu-Chun Huang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Kai Yang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Shuang Huang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shee-Uan Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Shih Yang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Obstetrics and Gynecology, Fu Jen Catholic University Hospital, New Taipei, Taiwan
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan; Livia Shangyu Wan Chair Professor of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
7
|
microRNAs in newborns with low birth weight: relation to birth size and body composition. Pediatr Res 2022; 92:829-837. [PMID: 34799665 DOI: 10.1038/s41390-021-01845-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Children with low birth weight (LBW) have a higher risk of developing endocrine-metabolic disorders later in life. Deregulation of specific microRNAs (miRNAs) could underscore the programming of adult pathologies. We analyzed the miRNA expression pattern in both umbilical cord serum samples from LBW and appropriate-for-gestational-age (AGA) newborns and maternal serum samples in the 3rd trimester of gestation, and delineated the relationships with fetal growth, body composition, and markers of metabolic risk. METHODS Serum samples of 12 selected mother-newborn pairs, including 6 LBW and 6 AGA newborns, were used for assessing miRNA profile by RNA-sequencing. The miRNAs with differential expression were validated in a larger cohort [49 maternal samples and 49 umbilical cord samples (24 LBW, 25 AGA)] by RT-qPCR. Anthropometric, endocrine-metabolic markers and body composition (by DXA) in infants were determined longitudinally over 12 months. RESULTS LBW newborns presented reduced circulating concentrations of miR-191-3p (P = 0.015). miR-191-3p levels reliably differentiated LBW from AGA individuals (ROC AUC = 0.76) and were positively associated with anthropometric and body composition measures at birth and weight Z-score at 12 months (P < 0.05). CONCLUSIONS miR-191-3p was reliably different in LBW individuals, and could be a new player in the epigenetic mechanisms linking LBW and future endocrine-metabolic adverse outcomes. IMPACT Children with low birth weight (LBW) have a higher risk of developing endocrine-metabolic disorders. Deregulation of specific microRNAs (miRNAs) could underscore the programming of those pathologies. miR-191-3p is downregulated in serum of LBW newborns, and its concentrations associate positively with neonatal anthropometric measures, with lean mass and bone accretion at age 15 days and with weight Z-score at age 12 months. miR-191-3p was reliably different in individuals with LBW, and could be a new player in the epigenetic mechanisms connecting LBW and future endocrine-metabolic adverse outcomes.
Collapse
|
8
|
Motahari Rad H, Mowla SJ, Ramazanali F, Rezazadeh Valojerdi M. Characterization of altered microRNAs related to different phenotypes of polycystic ovarian syndrome (PCOS) in serum, follicular fluid, and cumulus cells. Taiwan J Obstet Gynecol 2022; 61:768-779. [PMID: 36088043 DOI: 10.1016/j.tjog.2022.05.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 10/14/2022] Open
Abstract
OBJECTIVE Polycystic ovarian syndrome (PCOS) is a metabolic syndrome in which steroidogenesis, folliculogenesis, and cellular adhesion play crucial roles in its prognosis. These pathways are controlled and regulated by some small non-coding RNAs called microRNAs (miRs). Several miRs have differential expression in PCOS compared to healthy women, and their dysregulation suggests important roles of miRs in PCOS pathophysiology. However, the role of miRs is still unclear, especially in various phenotypes of PCOS. MATERIALS AND METHODS This study was conducted to evaluate the diagnostic potential of miR-212-3p, miR-490-5p, miR-647, and miR-4643 in different subtypes of PCOS. Accordingly, nineteen PCOS patients with different subtypes based on Rotterdam criteria (A: 8, B: not detected in this study, C: 5, and D: 6 patients) and six control age and BMI matched women under ICSI treatment were selected. The relative expression of miRs was then measured in blood serum before hormonal treatment (S1) and before ovum pickup (S2), follicular fluid (FF), and cumulus cells (CC) in all subjects. Also, the expression of miRs predicted target genes (AMH, AR, CYP11A1, CYP17A1, CYP19A1, GDF9, and HSD17B12) were done in the CC of understudy groups. RESULTS In general, the results indicated that PCOS significantly increased the expression of miR-212-3p, miR-490-5p, and miR-4643 in FF and CCs compared to control. Although these miRs tend to increase in serum 1 of the PCOS patients, the differences were insignificant. However, there was a significant reduction in the expression of miR-647 in FF and CCs between PCOS vs. control. In addition, the miRs had significantly different expressions in various phenotypes of PCOS. For example, high levels of miR-647 in S2 and low levels of miR-490 in FF and miR-212 in CC can differentiate phenotype A from the other. Also, upregulation of miR-212 in FF and miR-4643 in S1 and low levels of this miR in FF can specifically differentiate subtype A from D. On the other hand, high levels of miR-4643 in FF and miR-490 in CC and lower titter of miR-647 can distinguish subtype C from the other. On the other hand, high levels of AMH, AR, CYP11, CYP17, and HSD17 in the hyperandrogenic PCOS and upregulation of CYP19A1 in the hypoandrogenic group can validate the role of selected miRs in the prognosis of PCOS. CONCLUSION Characterization of altered microRNAs in serum, FF, and CCs and their targets in CC showed that the miRs might play critical roles in steroidogenesis and folliculogenesis. These miRs may be used for molecular classification of PCOS subtypes and as biomarkers for PCOS diagnosis.
Collapse
Affiliation(s)
- Hanieh Motahari Rad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fariba Ramazanali
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Tamaddon M, Azimzadeh M, Tavangar SM. microRNAs and long non-coding RNAs as biomarkers for polycystic ovary syndrome. J Cell Mol Med 2022; 26:654-670. [PMID: 34989136 PMCID: PMC8817139 DOI: 10.1111/jcmm.17139] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/05/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is known as the most common metabolic/endocrine disorder among women of reproductive age. Its complicated causality assessment and diagnostic emphasized the role of non‐coding regulatory RNAs as molecular biomarkers in studying, diagnosing and even as therapeutics of PCOS. This review discusses a comparative summary of research into microRNAs (miRNAs) and long non‐coding RNAs (lncRNAs) that are molecularly or statistically related to PCOS. We categorize the literature in terms of centering on either miRNAs or lncRNAs and discuss the combinatory studies and promising ideas as well. Additionally, we compare the pros and cons of the prominent research methodologies used for each of the abovementioned research themes and discuss how errors can be stopped from propagation by selecting correct methodologies for future research. Finally, it can be concluded that research into miRNAs and lncRNAs has the potential for identifying functional networks of regulation with multiple mRNAs (and hence, functional proteins). This new understanding may eventually afford clinicians to control the molecular course of the pathogenesis better. With further research, RNA (with statistical significance and present in the blood) may be used as biomarkers for the disease, and more possibilities for RNA therapy agents can be identified.
Collapse
Affiliation(s)
- Mona Tamaddon
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Azimzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Mir FA, Mall R, Iskandarani A, Ullah E, Samra TA, Cyprian F, Parray A, Alkasem M, Abdalhakam I, Farooq F, Abou-Samra AB. Characteristic MicroRNAs Linked to Dysregulated Metabolic Pathways in Qatari Adult Subjects With Obesity and Metabolic Syndrome. Front Endocrinol (Lausanne) 2022; 13:937089. [PMID: 35937842 PMCID: PMC9352892 DOI: 10.3389/fendo.2022.937089] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/24/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Obesity-associated dysglycemia is associated with metabolic disorders. MicroRNAs (miRNAs) are known regulators of metabolic homeostasis. We aimed to assess the relationship of circulating miRNAs with clinical features in obese Qatari individuals. METHODS We analyzed a dataset of 39 age-matched patients that includes 18 subjects with obesity only (OBO) and 21 subjects with obesity and metabolic syndrome (OBM). We measured 754 well-characterized human microRNAs (miRNAs) and identified differentially expressed miRNAs along with their significant associations with clinical markers in these patients. RESULTS A total of 64 miRNAs were differentially expressed between metabolically healthy obese (OBO) versus metabolically unhealthy obese (OBM) patients. Thirteen out of 64 miRNAs significantly correlated with at least one clinical trait of the metabolic syndrome. Six out of the thirteen demonstrated significant association with HbA1c levels; miR-331-3p, miR-452-3p, and miR-485-5p were over-expressed, whereas miR-153-3p, miR-182-5p, and miR-433-3p were under-expressed in the OBM patients with elevated HbA1c levels. We also identified, miR-106b-3p, miR-652-3p, and miR-93-5p that showed a significant association with creatinine; miR-130b-5p, miR-363-3p, and miR-636 were significantly associated with cholesterol, whereas miR-130a-3p was significantly associated with LDL. Additionally, miR-652-3p's differential expression correlated significantly with HDL and creatinine. CONCLUSIONS MicroRNAs associated with metabolic syndrome in obese subjects may have a pathophysiologic role and can serve as markers for obese individuals predisposed to various metabolic diseases like diabetes.
Collapse
Affiliation(s)
- Fayaz Ahmad Mir
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Raghvendra Mall
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Ahmad Iskandarani
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ehsan Ullah
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
| | - Tareq A Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Farhan Cyprian
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar
| | - Aijaz Parray
- Qatar Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Meis Alkasem
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ibrahem Abdalhakam
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Faisal Farooq
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
11
|
Bahmyari S, Jamali Z, Khatami SH, Vakili O, Roozitalab M, Savardashtaki A, Solati A, Mousavi P, Shabaninejad Z, Vakili S, Behrouj H, Ghasemi H, Movahedpour A. microRNAs in female infertility: An overview. Cell Biochem Funct 2021; 39:955-969. [PMID: 34708430 DOI: 10.1002/cbf.3671] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022]
Abstract
Infertility impacts a considerable number of women worldwide, and it affects different aspects of family life and society. Although female infertility is known as a multifactorial disorder, there are strong genetic and epigenetic bases. Studies revealed that miRNAs play critical roles in initiation and development of female infertility related disorders. Early diagnosis and control of these diseases is an essential key for improving disease prognosis and reducing the possibility of infertility and other side effects. Investigating the possible use of miRNAs as biomarkers and therapeutic options is valuable, and it merits attention. Thus, in this article, we reviewed research associated with female diseases and highlighted microRNAs that are related to the polycystic ovary syndrome (up to 30 miRNAs), premature ovarian failure (10 miRNAs), endometriosis (up to 15 miRNAs), uterine fibroids (up to 15 miRNAs), endometrial polyp (3 miRNAs), and pelvic inflammatory (6 miRNAs), which are involved in one or more ovarian or uterine disease-causing processes.
Collapse
Affiliation(s)
- Sedigheh Bahmyari
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Jamali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahin Roozitalab
- Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arezoo Solati
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pegah Mousavi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Behrouj
- Department of Clinical Biochemistry, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Hassan Ghasemi
- Department of Clinical Biochemistry, Abadan University of Medical Sciences, Abadan, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Ding R, Kang W, Wu D, Wang L. Protective effect of propofol via the regulation of ovarian granulosa cell proliferation and apoptosis. Exp Ther Med 2021; 22:988. [PMID: 34345270 PMCID: PMC8311282 DOI: 10.3892/etm.2021.10420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/15/2021] [Indexed: 12/05/2022] Open
Abstract
Propofol is an anesthetic frequently used in surgery. Accumulating evidence suggests that propofol exhibits an effect on cell viability, apoptosis and invasion in several types of cancer cells. MicroRNAs (miRNAs) have been reported to play pivotal roles in the development of polycystic ovary syndrome (PCOS). However, the diagnostic applications of miR-451a in PCOS remain unknown. The present study aimed to elucidate the effects of propofol on ovarian granulosa cell proliferation and apoptosis and illustrate the specific mechanisms associated with this process. Human ovarian granulosa cell-like KGN cells, which were used as a representative of granulosa cells in the present study, were treated with different concentrations (0, 1, 5 and 10 µg/ml) of propofol for 48 h and cell proliferation and apoptosis were assessed using MTT and flow cytometry assays, respectively. Propofol treatment resulted in significant inhibition of cell viability and induction of apoptosis in KGN cells, which was accompanied with increased cleaved caspase 3 and suppressed pro-caspase 3 expression levels. Furthermore, propofol reduced Wnt3a and β-catenin protein and mRNA expression levels. miR-451a expression in KGN cells was evaluated by reverse transcription-quantitative PCR (RT-qPCR). miR-451a expression was upregulated in propofol-stimulated KGN cells. The data further demonstrated that miR-451a mimics suppressed cell proliferation and increased apoptosis of KGN cells compared with cells transfected with control mimics. Furthermore, the association between miR-451a and propofol was investigated. Rescue experiments were performed to investigate the anti-proliferative mechanism of propofol in ovarian granulosa cells. KGN cells were transfected with miR-451a inhibitor or inhibitor control sequences for 6 h and treated with 10 µg/ml propofol for an additional 48 h. The results from the MTT, RT-qPCR and western blot assays indicated that 10 µg/ml propofol inhibited cell viability, induced apoptosis, enhanced cleaved caspase 3 expression, reduced pro-caspase 3 levels and inhibited the protein and mRNA expression of Wnt3a and β-catenin. However, inhibition of miR-451a demonstrated the opposite effects. In conclusion, the results of the present study revealed that propofol exerted an anti-proliferative and apoptosis-inducing role in ovarian granulosa cells through mediation of miR-451a expression. In addition, the data indicated that miR-451a may be used as an effective therapeutic target for PCOS treatment.
Collapse
Affiliation(s)
- Rong Ding
- Department of Anesthesiology, Hainan General Hospital (Hainan Hospital Affiliated to Hainan Medical University), Haikou, Hainan 570311, P.R. China
| | - Wenyue Kang
- Department of Anesthesiology, Hainan General Hospital (Hainan Hospital Affiliated to Hainan Medical University), Haikou, Hainan 570311, P.R. China
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General Hospital (Hainan Hospital Affiliated to Hainan Medical University), Haikou, Hainan 570311, P.R. China
| | - Lin Wang
- Department of Anesthesiology, Hainan General Hospital (Hainan Hospital Affiliated to Hainan Medical University), Haikou, Hainan 570311, P.R. China
| |
Collapse
|
13
|
Zhang Z, Sang M, Liu S, Shao J, Cai Y. Differential expression of long non-coding RNA Regulator of reprogramming and its molecular mechanisms in polycystic ovary syndrome. J Ovarian Res 2021; 14:79. [PMID: 34148561 PMCID: PMC8215827 DOI: 10.1186/s13048-021-00829-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a common endocrine disease in women of reproductive age. Multiple studies have shown that long non-coding RNAs (lncRNA) and microRNAs (miRNA) play a role in PCOS. This study aimed to explore the role and molecular mechanism of lncRNA -Regulator of reprogramming (lncROR) in PCOS. Results Expression level of lncROR in PCOS patients was up-regulated, while level of miR-206 was down-regulated in comparison with control group (P < 0.001). Logistics regression analysis showed that lncROR and miR-206 were independent predictors of PCOS. The ROC curve showed that lncROR had a high diagnostic value for PCOS with an AUC value of 0.893. Pearson correlation coefficient indicated that the expression level of miR-206 was negatively correlated with the level of lncROR. CCK-8 assay and apoptosis assay revealed that downregulation of lncROR up-regulated the expression of miR-206, thereby inhibiting cell proliferation and promoting cell apoptosis. However, silencing the expression of miR-206 reversed the above effects caused by down-regulation of lncROR expression. Luciferase reporter gene assay suggested that there was a target relationship between lncROR and miR-206. VEGF was proved to be the target gene of miR-206. Conclusions Highly expressed lncROR indirectly up-regulated the expression of VEGF by down-regulating the expression of miR-206, thereby promoting the proliferation of KGN cells and inhibiting apoptosis, and further promoting the development of PCOS.
Collapse
Affiliation(s)
- Zhihong Zhang
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oilfield, Daqing, 163000, Heilongjiang, China
| | - Min Sang
- Gynecology Clinic, The First Hospital of Harbin, No.151 Diduan Street, Heilongjiang, 150010, Harbin, China.
| | - Siqin Liu
- Laboratory Department, General Hospital of Daqing Oilfield, Daqing, 163000, Heilongjiang, China
| | - Jing Shao
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oilfield, Daqing, 163000, Heilongjiang, China
| | - Yunjiang Cai
- Department of Psychology, Harbin Medical Univercity (Daqing), Daqing, 163319, Heilongjiang, China
| |
Collapse
|
14
|
Yang T, Wang L, Zhang Y, Zheng J, Liu L. MicroRNA-451a plays a role in polycystic ovary syndrome by regulating ovarian granulosa cell proliferation and apoptosis. Exp Ther Med 2021; 21:583. [PMID: 33850555 PMCID: PMC8027723 DOI: 10.3892/etm.2021.10015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/15/2020] [Indexed: 11/28/2022] Open
Abstract
The present study aimed to investigate whether microRNA (miR)-451a plays a role in polycystic ovary syndrome by regulating the biological function of ovarian granulosa cells and investigate the underlying molecular mechanism. In the present study, reverse transcription-quantitative PCR (RT-qPCR) analysis detected markedly low expression of miR-451a in KGN cells. TargetScan predicted that cyclic AMP-dependent transcription factor ATF-2 (ATF2) was a potential target gene of miR-451a, which was confirmed by a Dual-Luciferase reporter gene assay. Moreover, western blotting and RT-qPCR experiments indicated that ATF2 was significantly overexpressed in KGN cells. In addition, western blotting and RT-qPCR experiments were utilized to assess cell transfection efficiency, and it was found that miR-451a mimic significantly increased miR-451a expression in KGN cells. Subsequently, MTT assay was performed to detect cell proliferation and flow cytometry was utilized to detect cell apoptosis. Western blot and RT-qPCR assays were utilized to assess the protein and mRNA expression of ATF2 and cyclin D1. The results confirmed that miR-451a mimic significantly decreased ATF2 protein and mRNA expression in KGN cells, and this decrease was reversed by ATF2-plasmid co-transfection. Moreover, miR-451a mimic inhibited cell proliferation, enhanced cell apoptosis, reduced cyclin D1 expression, increased caspase-3 activity and cleaved caspase-3 protein levels, while it reduced pro-caspase 3 protein levels in KGN cells, and these effects were significantly reversed by ATF2-plasmid. The present preliminary results demonstrated that miR-451a regulated the proliferation and apoptosis of ovarian granulosa cells by targeting ATF2. Thus, the miR-451a/ATF2 axis may be a new potential target for the treatment of polycystic ovary syndrome.
Collapse
Affiliation(s)
- Tianjin Yang
- Department of Obstetrics and Gynecology, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Lie Wang
- Reproductive Center, Qingdao Women and Children's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Yun Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jindan Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Lili Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
15
|
de Zegher F, Díaz M, Villarroya J, Cairó M, López-Bermejo A, Villarroya F, Ibáñez L. The relative deficit of GDF15 in adolescent girls with PCOS can be changed into an abundance that reduces liver fat. Sci Rep 2021; 11:7018. [PMID: 33782413 PMCID: PMC8007831 DOI: 10.1038/s41598-021-86317-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
A prime concern of young patients with Polycystic Ovary Syndrome (PCOS) is the control of body adiposity, given their tendency to gain weight and/or their difficulty to lose weight. Circulating growth-and-differentiation factor-15 (GDF15) facilitates the control of body weight via receptors in the brainstem. C-reactive protein (CRP) and insulin are endogenous GDF15 secretagogues. We hypothesised that PCOS in non-obese adolescents is characterised by low concentrations of circulating GDF15, when judged by the degree of CRP and insulin drive. GDF15 was added as a post-hoc endpoint of two previously reported, randomised studies in non-obese adolescent girls with PCOS (N = 58; 60% normal weight; 40% overweight) who received either an oral oestroprogestogen contraceptive (OC), or a low-dose combination of spironolactone-pioglitazone-metformin (SPIOMET) for 1 year; subsequently, all girls remained untreated for 1 year. Adolescent girls with regular menses (N = 20) served as healthy controls. Circulating GDF15, CRP and fasting insulin were assessed prior to treatment, and halfway the on- and post-treatment years. Pre-treatment, the absolute GDF15 concentrations were normal in PCOS girls, but their relative levels were markedly low, in view of the augmented CRP and insulin drives. OC treatment was accompanied by a near-doubling of circulating GDF15 (on average, from 296 to 507 pg/mL) and CRP, so that the relative GDF15 levels remained low. SPIOMET treatment was accompanied by a 3.4-fold rise of circulating GDF15 (on average, from 308 to 1045 pg/mL) and by a concomitant lowering of CRP and insulin concentrations towards normal, so that the relative GDF15 levels became markedly abundant. Post-OC, the relatively low GDF15 levels persisted; post-SPIOMET, the circulating concentrations of GDF15, CRP and insulin were all normal. BMI remained stable in both treatment groups. Only SPIOMET was accompanied by a reduction of hepato-visceral fat (by MRI) towards normal. In conclusion, early PCOS was found to be characterised by a relative GDF15 deficit that may partly explain the difficulties that young patients experience to control their body adiposity. This relative GDF15 deficit persisted during and after OC treatment. In contrast, SPIOMET treatment was accompanied by an absolute and a relative abundance of GDF15, and followed by normal GDF15, CRP and insulin concentrations. The present findings strengthen the rationale to raise the concentrations of circulating GDF15 in early PCOS, for example with a SPIOMET-like intervention that attenuates low-grade inflammation, insulin resistance and ectopic adiposity, without necessarily lowering body weight.Clinical trial registries: ISRCTN29234515 and ISRCTN11062950.
Collapse
Affiliation(s)
- Francis de Zegher
- Department of Development and Regeneration, University of Leuven, 3000, Leuven, Belgium
| | - Marta Díaz
- Endocrinology Department, Institut de Recerca Pediàtric, Hospital Sant Joan de Déu, University of Barcelona, Passeig de Sant Joan de Déu, 2, Esplugues, 08950, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Joan Villarroya
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), ISCIII, Madrid, Spain
| | - Montserrat Cairó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), ISCIII, Madrid, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), Dr. Josep Trueta Hospital, 17007, Girona, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), ISCIII, Madrid, Spain
| | - Lourdes Ibáñez
- Endocrinology Department, Institut de Recerca Pediàtric, Hospital Sant Joan de Déu, University of Barcelona, Passeig de Sant Joan de Déu, 2, Esplugues, 08950, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain.
| |
Collapse
|
16
|
Luo Y, Cui C, Han X, Wang Q, Zhang C. The role of miRNAs in polycystic ovary syndrome with insulin resistance. J Assist Reprod Genet 2021; 38:289-304. [PMID: 33405004 PMCID: PMC7884539 DOI: 10.1007/s10815-020-02019-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023] Open
Abstract
PURPOSE This review aims to summarize the key findings of several miRNAs and their roles in polycystic ovary syndrome with insulin resistance, characterize the disease pathogenesis, and establish a new theoretical basis for diagnosing, treating, and preventing polycystic ovary syndrome. METHODS Relevant scientific literature was covered from 1992 to 2020 by searching the PubMed database with search terms: insulin/insulin resistance, polycystic ovary syndrome, microRNAs, and metabolic diseases. References of relevant studies were cross-checked. RESULTS The related miRNAs (including differentially expressed miRNAs) and their roles in pathogenesis, and possible therapeutic targets and pathways, are discussed, highlighting controversies and offering thoughts for future directions. CONCLUSION We found abundant evidence on the role of differentially expressed miRNAs with its related phenotypes in PCOS. Considering the essential role of insulin resistance in the pathogenesis of PCOS, the alterations of associated miRNAs need more research attention. We speculate that race/ethnicity or PCOS phenotype and differences in methodological differences might lead to inconsistencies in research findings; thus, several miRNA profiles need to be investigated further to qualify for the potential therapeutic targets for PCOS-IR.
Collapse
Affiliation(s)
- Yingliu Luo
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Chenchen Cui
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Xiao Han
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Qian Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Cuilian Zhang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China.
| |
Collapse
|
17
|
Zhou J, Jin X, Sheng Z, Zhang Z. miR-206 serves an important role in polycystic ovary syndrome through modulating ovarian granulosa cell proliferation and apoptosis. Exp Ther Med 2021; 21:179. [PMID: 33500693 PMCID: PMC7818533 DOI: 10.3892/etm.2021.9610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022] Open
Abstract
An increasing number of studies have reported that microRNAs (miRNAs) have an important role in polycystic ovary syndrome (PCOS). Downregulation of miR-206 in patients with PCOS has been found, however, its specific role remains unclear. The present study aimed to investigate the roles of miR-206 in (PCOS) and to determine the underlying molecular mechanisms. Reverse transcription-quantitative PCR (RT-qPCR) was performed to analyze the expression levels of miR-206 in normal ovarian surface epithelial IOSE80 cells and human ovarian granulosa cell-like KGN cells. TargetScan was used to predict the target gene of miR-206, which was subsequently verified using a dual-luciferase reporter gene assay. The mRNA expression levels of cyclin D2 (CCND2) and the transfection efficiencies of the miR-206 mimic and CCDN2 overexpression plasmid were determined using RT-qPCR analysis. The protein expression levels of CCND2, cleaved-caspase-3 and pro-caspase-3 were analyzed using western blotting, and an MTT assay and flow cytometric analysis were used to evaluate the cell viability and levels of apoptosis, respectively, in the cells following transfection. Finally, the activity of caspase-3 was analyzed using a caspase-3 activity assay kit. The results of the present study revealed that the expression levels of miR-206 were downregulated in KGN cells compared with IOSE80 cells. CCND2 was predicted and verified to be a direct target gene of miR-206, and the mRNA and protein expression levels of CCND2 were discovered to be upregulated in KGN cells compared with IOSE80 cells. The miR-206 mimic and CCND2 overexpression plasmid significantly upregulated the expression levels of miR-206 and CCND2, respectively, in KGN cells. The miR-206 mimic also downregulated the expression levels of CCND2 in KGN cells, while this effect was reversed following the transfection with the CCND2 overexpression plasmid. Compared with the mimic control group, the miR-206 mimic significantly decreased the cell viability, induced the levels of apoptosis, increased the activity of caspase-3, upregulated cleaved-caspase-3 protein expression levels and downregulated pro-caspase-3 protein expression levels in KGN cells following transfection; these effects were reversed following the overexpression of CCND2. In conclusion, the findings of the present study suggested that miR-206 may serve an important role in PCOS through modulating ovarian granulosa cell viability and apoptosis.
Collapse
Affiliation(s)
- Jie Zhou
- Reproductive Endocrine Center, Hangzhou Women's Hospital, Hangzhou, Zhejiang 310008, P.R. China
| | - Xuejing Jin
- Reproductive Endocrine Center, Hangzhou Women's Hospital, Hangzhou, Zhejiang 310008, P.R. China
| | - Zhumei Sheng
- Reproductive Endocrine Center, Hangzhou Women's Hospital, Hangzhou, Zhejiang 310008, P.R. China
| | - Zhifen Zhang
- Reproductive Endocrine Center, Hangzhou Women's Hospital, Hangzhou, Zhejiang 310008, P.R. China
| |
Collapse
|
18
|
Ibáñez L, Díaz M, García-Beltrán C, Malpique R, Garde E, López-Bermejo A, de Zegher F. Toward a Treatment Normalizing Ovulation Rate in Adolescent Girls With Polycystic Ovary Syndrome. J Endocr Soc 2020; 4:bvaa032. [PMID: 32342022 PMCID: PMC7182125 DOI: 10.1210/jendso/bvaa032] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Adolescent polycystic ovary syndrome (PCOS) is characterized by androgen excess and oligomenorrhea, and commonly driven by hepato-visceral fat excess ("central obesity") ensuing from a mismatch between prenatal and postnatal nutrition, on a background of genetic susceptibility. There is no approved treatment for adolescent PCOS. We report the pooled results of 2 pilot studies in nonobese girls with PCOS (N = 62, age 15.8 years) that compared the effects of randomized treatment for 1 year, either with an oral estro-progestogen contraceptive (OC), or with a low-dose combination of spironolactone-pioglitazone-metformin (SPIOMET, targeting the excess of ectopic fat). Auxological and endocrine-metabolic variables (including fasting insulin, androgens, high-molecular-weight adiponectin [HMW-adiponectin], and microRNA [miR]-451a), body composition (dual x-ray absorptiometry) and hepato-visceral fat (magnetic resonance imaging) were assessed on- and posttreatment. Data from menstrual diaries were combined with weekly salivary progesterone measurements to infer ovulation rates during the second and fourth quarter of the posttreatment year. OC and SPIOMET treatment reduced the androgen excess comparably, and had no differential effects on total-body lean or fat mass. However, SPIOMET was accompanied by more broadly normalizing effects, including on hepato-visceral fat and on circulating insulin, HMW-adiponectin, and miR-451a. On average, there were 3-fold more ovulations post-SPIOMET than post-OC; normovulation was only observed after SPIOMET; anovulation was >10-fold more prevalent post-OC. Pooled results of randomized studies in nonobese adolescent girls with PCOS indicate that SPIOMET treatment leads to an overall healthier, more insulin-sensitive condition-with less ectopic fat-than OC treatment, and to a more normal posttreatment ovulation rate.
Collapse
Affiliation(s)
- Lourdes Ibáñez
- Institut de Recerca Pediàtric Hospital Sant Joan de Déu, University of Barcelona, Esplugues, Barcelona, Spain,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain,Correspondence: Lourdes Ibáñez, MD, PhD, Endocrinology Department, Pediatric Research Institute, Hospital Sant Joan de Déu, University of Barcelona, Passeig de Sant Joan de Déu, 2, 08950 Esplugues, Barcelona, Spain, Phone: +34 93 2804000, ext. 4424, Fax: +34 93 2033959, e-mail:
| | - Marta Díaz
- Institut de Recerca Pediàtric Hospital Sant Joan de Déu, University of Barcelona, Esplugues, Barcelona, Spain,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Cristina García-Beltrán
- Institut de Recerca Pediàtric Hospital Sant Joan de Déu, University of Barcelona, Esplugues, Barcelona, Spain,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Rita Malpique
- Institut de Recerca Pediàtric Hospital Sant Joan de Déu, University of Barcelona, Esplugues, Barcelona, Spain,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Edurne Garde
- Institut de Recerca Pediàtric Hospital Sant Joan de Déu, University of Barcelona, Esplugues, Barcelona, Spain,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI) and Dr. Josep Trueta Hospital, Girona, Spain
| | - Francis de Zegher
- Department of Development & Regeneration, University of Leuven, Leuven, Belgium
| |
Collapse
|