1
|
Velazquez C, Herrero Y, Prost K, Bordaquievich M, Neira M, Parborell F, Abramovich D. Reproductive outcomes in female mice offspring due to maternal metformin treatment. Life Sci 2025; 363:123416. [PMID: 39864616 DOI: 10.1016/j.lfs.2025.123416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
AIMS Metformin has shown beneficial effects on reproduction in women. However, its use during pregnancy remains controversial, as metformin can cross the placenta. Most studies have focused on the metabolic impact on the offspring of treated mothers, with limited information regarding its reproductive effects. The aim of this study was to evaluate potential alterations in ovarian function and fertility in female offspring of mothers treated with metformin during pregnancy and lactation. MATERIALS AND METHODS C57BL/6 female mice were treated with metformin four weeks before mating, and the treatment was maintained during gestation and lactation. Seven weeks after weaning, metabolic parameters as well as ovarian and reproductive function of the offspring were analyzed. KEY FINDINGS The offspring of treated mothers were lighter at birth and, in adulthood, they had more gonadal adipose tissue with no alterations in body weight. No changes in glucose metabolism were observed. Their follicular development was modified, with more early antral and atretic follicles and less primary and late antral follicles. Anti-Müllerian hormone expression and ovarian angiogenesis were increased. The estrous cycle, hormonal production and fertility were not affected by metformin exposure, however, the F2 generation showed higher body weight at birth. SIGNIFICANCE Metformin can induce fetal programming in animals exposed to it during development, impacting metabolism and ovarian functionality in adulthood. Under physiological conditions, these alterations do not result in reduced fertility or endocrine disruptions. Our data warrant studies in women to make informed decisions regarding metformin administration during critical developmental periods in clinical settings.
Collapse
Affiliation(s)
- Candela Velazquez
- Studies of the Physiopathology of the ovary laboratory, Institute of Biology and Experimental Medicine (IBYME) - National Scientific and Technical Research Council (CONICET), Vuelta de Obligado 2490, C1428ADL Buenos Aires, Argentina
| | - Yamila Herrero
- Studies of the Physiopathology of the ovary laboratory, Institute of Biology and Experimental Medicine (IBYME) - National Scientific and Technical Research Council (CONICET), Vuelta de Obligado 2490, C1428ADL Buenos Aires, Argentina
| | - Katherine Prost
- Pedro Fiorito Hospital, Endocrinology area, Av. Manuel Belgrano 827, B1870 Avellaneda, Buenos Aires Province, Argentina
| | - Mayra Bordaquievich
- Studies of the Physiopathology of the ovary laboratory, Institute of Biology and Experimental Medicine (IBYME) - National Scientific and Technical Research Council (CONICET), Vuelta de Obligado 2490, C1428ADL Buenos Aires, Argentina
| | - Melanie Neira
- Studies of the Physiopathology of the ovary laboratory, Institute of Biology and Experimental Medicine (IBYME) - National Scientific and Technical Research Council (CONICET), Vuelta de Obligado 2490, C1428ADL Buenos Aires, Argentina
| | - Fernanda Parborell
- Studies of the Physiopathology of the ovary laboratory, Institute of Biology and Experimental Medicine (IBYME) - National Scientific and Technical Research Council (CONICET), Vuelta de Obligado 2490, C1428ADL Buenos Aires, Argentina
| | - Dalhia Abramovich
- Studies of the Physiopathology of the ovary laboratory, Institute of Biology and Experimental Medicine (IBYME) - National Scientific and Technical Research Council (CONICET), Vuelta de Obligado 2490, C1428ADL Buenos Aires, Argentina.
| |
Collapse
|
2
|
Belani MA, Shah P, Banker M, Gupta SS. Investigating the potential role of swertiamarin on insulin resistant and non-insulin resistant granulosa cells of poly cystic ovarian syndrome patients. J Ovarian Res 2023; 16:55. [PMID: 36932437 PMCID: PMC10024427 DOI: 10.1186/s13048-023-01126-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND AND AIM Conventional drugs have limitations due to prevalence of contraindications in PCOS patients. To explore the potential effects of swertiamarin, on abrupted insulin and steroidogenic signaling in human luteinized granulosa cells from PCOS patients with or without insulin resistance. EXPERIMENTAL PROCEDURE hLGCs from 8 controls and 16 PCOS patients were classified for insulin resistance based on down regulation of protein expression of insulin receptor-β (INSR- β) as shown in our previous paper. Cells were grouped as control, PCOS-IR and PCOS-NIR, treated with swertiamarin (66 µM) and metformin (1 mM). Expression of key molecules involved in insulin signaling, fat metabolism, IGF system and steroidogenesis were compared between groups. RESULTS Swertiamarin significantly (P < 0.05) reversed the expression of INSR-β, PI(3)K, p-Akt, PKC-ζ, PPARγ, (P < 0.01) IRS (Ser 307) and IGF system in PCOS-IR group and was equally potent to metformin. In the same group, candidate genes viz SREBP1c, FAS, ACC-1 and CPT-1 were down regulated by swertiamarin (P < 0.001) and metformin (P < 0.001). Significant upregulation was demonstrated in expression of StAR, CYP19A1, 17β-HSD and 3β-HSD when treated with swertiamarin (P < 0.01) and metformin (P < 0.01) in PCOS-IR followed by increase in 17β-HSD and 3β-HSD enzyme activity along with estradiol and progesterone secretions. However, swertiamarin did not reveal any effect on PCOS-NIR group as compared to metformin that significantly (P < 0.01) reversed all the parameters related to steroidogenesis and down regulated basal expression of insulin signaling genes. CONCLUSION Swertiamarin, presents itself as a potential fertility drug in hLGCs from PCOS-IR patients.
Collapse
Affiliation(s)
- Muskaan A. Belani
- Dr. Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390 002 India
| | - Preeti Shah
- Nova IVI Fertility, Behind Xavier’s Ladies Hostel, 108, Swastik Society Rd, Navrangpura, Ahmedabad, 390009 Gujarat India
| | - Manish Banker
- Nova IVI Fertility, Behind Xavier’s Ladies Hostel, 108, Swastik Society Rd, Navrangpura, Ahmedabad, 390009 Gujarat India
| | - Sarita S. Gupta
- Dr. Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390 002 India
| |
Collapse
|
3
|
Impact of Musashi-1 and Musashi-2 Double Knockdown on Notch Signaling and the Pathogenesis of Endometriosis. Int J Mol Sci 2022; 23:ijms23052851. [PMID: 35269992 PMCID: PMC8911246 DOI: 10.3390/ijms23052851] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
The stem cell marker and RNA-binding protein Musashi-1 is overexpressed in endometriosis. Musashi-1-siRNA knockdown in Ishikawa cells altered the expression of stem cell related genes, such as OCT-4. To investigate the role of both human Musashi homologues (MSI-1 and MSI-2) in the pathogenesis of endometriosis, immortalized endometriotic 12-Z cells and primary endometriotic stroma cells were treated with Musashi-1- and Musashi-2-siRNA. Subsequently, the impact on cell proliferation, cell apoptosis, cell necrosis, spheroid formation, stem cell phenotype and the Notch signaling pathway was studied in vitro. Using the ENDOMET Turku Endometriosis database, the gene expression of stem cell markers and Notch signaling pathway constituents were analyzed according to localization of the endometriosis lesions. The database analysis demonstrated that expression of Musashi and Notch pathway-related genes are dysregulated in patients with endometriosis. Musashi-1/2-double-knockdown increased apoptosis and necrosis and reduced stem cell gene expression, cell proliferation, and the formation of spheroids. Musashi silencing increased the expression of the anti-proliferation mediator p21. Our findings suggest the therapeutic potential of targeting the Musashi–Notch axis. We conclude that the Musashi genes have an impact on Notch signaling and the pathogenesis of endometriosis through the downregulation of proliferation, stemness characteristics and the upregulation of apoptosis, necrosis and of the cell cycle regulator p21.
Collapse
|
4
|
Notaro ALG, Neto FTL. The use of metformin in women with polycystic ovary syndrome: an updated review. J Assist Reprod Genet 2022; 39:573-579. [PMID: 35156149 PMCID: PMC8995234 DOI: 10.1007/s10815-022-02429-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 02/04/2022] [Indexed: 10/19/2022] Open
Abstract
PURPOSES Polycystic ovary syndrome (PCOS) is a major cause of female infertility, being present in up to 20% of women of childbearing age. Insulin resistance (IR) plays an important role in the pathophysiology of PCOS; therefore, its treatment may benefit women with the syndrome. The main drug used for IR management is metformin (MT). We aim to review the literature on the use of metformin in women with PCOS. METHODS Using the terms "metformin" and "polycystic ovary syndrome," we conducted a search the PubMed, EMBASE, and Google Scholar databases. The research was restricted to articles published in English. Initially, only published meta-analyses were included, in the absence of meta-analyzes, RCT and well-designed prospective studies were used. RESULTS Metformin increases success rates and decreases complication rates when used as an adjunctive medication for ovulation induction during low complexity assisted reproduction treatments and during ovarian stimulation for in vitro fertilization in women with PCOS. Evidence about the effect of metformin on fetal and obstetric complication rates is conflicting. Metformin is associated with high incidence of gastrointestinal symptoms; however, serious adverse effects are rare and there is no evidence of teratogenicity. CONCLUSION For women with PCOS, metformin is a good adjunctive medication for ovulation induction/stimulation for high and low complexity assisted reproduction therapies. The adverse effects are mostly mild, and there is no risk of teratogenicity, but the risk of long-term complications for the offspring is not yet defined. High heterogeneity of the studies limits extrapolation of findings, and further research is needed to determine which women will benefit most from the medication.
Collapse
Affiliation(s)
- Adriana Leal Griz Notaro
- Amare Clinic – Brazil, Av. República do Líbano, n 251, torre 1, sala 504, Pina, Recife, PE CEP 51110-160 Brazil
| | - Filipe Tenorio Lira Neto
- Andros Recife Clinic - Brazil, Av. Boa Viagem, n 179, apt 901, Pina, Recife, PE, CEP 51011-000, Brazil.
| |
Collapse
|
5
|
Soldat-Stanković V, Popović-Pejičić S, Stanković S, Prtina A, Malešević G, Bjekić-Macut J, Livadas S, Ognjanović S, Mastorakos G, Micić D, Macut D. The effect of metformin and myoinositol on metabolic outcomes in women with polycystic ovary syndrome: role of body mass and adiponectin in a randomized controlled trial. J Endocrinol Invest 2022; 45:583-595. [PMID: 34665453 DOI: 10.1007/s40618-021-01691-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 10/08/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE To compare the effects of insulin sensitizers metformin (MET) and myo-inositol (MI) on adiponectin levels and metabolic characteristics in women with polycystic ovary syndrome (PCOS) with respect to their body mass index (BMI). METHODS In this open label, parallel randomized clinical trial, 66 women with PCOS (33 normal-weight and 33 overweight/obese) were randomized to either MI (4 g/day) or MET (1500 mg/day) for a period of 6 months. Serum concentration of adiponectin, hormonal and metabolic laboratory outcomes and clinical assessment of BMI, body composition and Ferriman-Gallwey score (FG score) were evaluated before and after treatment. RESULTS After the 6-month intervention, comparison between MET and MI in time to treatment analysis showed no significant differences between the two treatments for all analyzed parameters. Only borderline significantly lower AUC glucose was found in the MET group in comparison to the MI group (p = 0.071). The main effect of treatment was shown for glucose concentration at 120 min OGTT (p = 0.032) and testosterone (p = 0.002). The main effect of time was shown for body mass (p = 0.004), waist circumference (p < 0.001), BMI (p = 0.003), body fat mass (p = 0.001), adiponectin (p = 0.020), fasting glucose (p = 0.001), testosterone (p = 0.015), SHBG (p = 0.013), 17OH progesterone (p = 0.008), LH (p = 0.004) and estradiol (p = 0.014). CONCLUSION Our study showed similar effects of MET and MI on BMI, body composition, hormonal profile, metabolism of glucose and insulin, and adiponectin level. The two insulin sensitizers, MET and MI, were useful in reducing BMI and improving body composition without significant differences between the two treatments in PCOS women. TRIAL REGISTRATION ISRCTN13199265. Trial registration date: 14.04.2021. (ISRCTN Registry), retrospectively registered.
Collapse
Affiliation(s)
- V Soldat-Stanković
- University Clinical Center of the Republic of Srpska, Internal Medicine Clinic, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - S Popović-Pejičić
- University Clinical Center of the Republic of Srpska, Internal Medicine Clinic, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - S Stanković
- Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - A Prtina
- Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - G Malešević
- University Clinical Center of the Republic of Srpska, Internal Medicine Clinic, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - J Bjekić-Macut
- Department of Endocrinology, CHC Bežanijska Kosa, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - S Livadas
- Endocrine Unit, Metropolitan Hospital, Athens, Greece
| | - S Ognjanović
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, dr Subotica 13, 11000, Belgrade, Serbia
| | - G Mastorakos
- Unit of Endocrine Diseases, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - D Micić
- Department of Medical Sciences, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - D Macut
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, dr Subotica 13, 11000, Belgrade, Serbia.
| |
Collapse
|
6
|
Jensterle M, Kravos NA, Dolžan V, Goričar K, Herman R, Rizzo M, Janež A. Glucose transporter 4 mRNA expression in subcutaneous adipose tissue of women with PCOS remains unchanged despite metformin withdrawal: is there a cellular metabolic treatment legacy effect? Endocrine 2022; 75:804-813. [PMID: 34761355 DOI: 10.1007/s12020-021-02934-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE Metformin induces GLUT-4 mRNA expression in insulin target tissues in PCOS. It is unclear how long this impact is sustained after withdrawal of metformin. We aimed to compare the effect of metformin withdrawal on GLUT-4 mRNA expression in subcutaneous adipose tissue after prior short (ST, 1 year, N = 11) and long term (LT, at least 3 years, N = 13) treatment in obese PCOS women. METHODS At baseline and 6 months after withdrawal, biopsy of subcutaneous adipose tissue followed by quantitative PCR analysis was performed to determine GLUT-4 mRNA expression. RESULTS We found no time/effect differences in GLUT-4 mRNA expression in ST (2-dCt at baseline 0.42 (0.16-0.48) vs 2-dCt after 6 months 0.31 (0.22-0.56), p = 0.594) and no time/effect difference in LT group (2-dCt at baseline 0.24 (0.14-0.39) vs 2-dCt after 6 months 0.25 (0.20-0.38), p = 0.382). There was also no difference in GLUT-4 mRNA expression between both groups at baseline and after 6 months. CONCLUSIONS In summary, 6 months after metformin withdrawal, GLUT-4 mRNA expression in subcutaneous adipose tissue remained stable, regardless of the prior treatment duration.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, 1000, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Nika Aleksandra Kravos
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, 1000, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Katja Goričar
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Rok Herman
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, 1000, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, Palermo, 90133, Italy
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, 1000, Slovenia.
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia.
| |
Collapse
|
7
|
Metformin and Insulin Resistance: A Review of the Underlying Mechanisms behind Changes in GLUT4-Mediated Glucose Transport. Int J Mol Sci 2022; 23:ijms23031264. [PMID: 35163187 PMCID: PMC8836112 DOI: 10.3390/ijms23031264] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Metformin is the most commonly used treatment to increase insulin sensitivity in insulin-resistant (IR) conditions such as diabetes, prediabetes, polycystic ovary syndrome, and obesity. There is a well-documented correlation between glucose transporter 4 (GLUT4) expression and the level of IR. Therefore, the observed increase in peripheral glucose utilization after metformin treatment most likely comes from the induction of GLUT4 expression and its increased translocation to the plasma membrane. However, the mechanisms behind this effect and the critical metformin targets are still largely undefined. The present review explores the evidence for the crucial role of changes in the expression and activation of insulin signaling pathway mediators, AMPK, several GLUT4 translocation mediators, and the effect of posttranscriptional modifications based on previously published preclinical and clinical models of metformin’s mode of action in animal and human studies. Our aim is to provide a comprehensive review of the studies in this field in order to shed some light on the complex interactions between metformin action, GLUT4 expression, GLUT4 translocation, and the observed increase in peripheral insulin sensitivity.
Collapse
|
8
|
Di Pietro M, Velazquez C, Matzkin ME, Frungieri MB, Peña MG, de Zúñiga I, Pascuali N, Irusta G, Bianchi MS, Parborell F, Abramovich D. Metformin has a direct effect on ovarian cells that is dependent on organic cation transporters. Mol Cell Endocrinol 2020; 499:110591. [PMID: 31546019 DOI: 10.1016/j.mce.2019.110591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
Metformin (MET) is the most widely prescribed hypoglycemic drug in type 2 diabetes and Polycystic Ovary Syndrome. Besides its effects on glucose metabolism, MET exerts beneficial effects on these patients' fertility. However, the exact mechanisms of action of MET on female fertility are still unclear. In this work, we analyzed a possible direct effect of MET on ovarian cells. We found expression of the organic cation transporters OCT1, OCT2 and OCT3, responsible for MET uptake into the cells, in rat granulosa cells and human cumulus cells. Furthermore, MET increased pAMPK and decreased VEGF levels both in vivo and in rat granulosa cells in culture. These last effects were reversed when OCTs were inhibited. Our results suggest that MET acts directly on ovarian cells regulating cell metabolism and VEGF expression. Our findings are relevant to optimize PCOS fertility treatment and to explore ovarian MET actions in other female pathologies.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Candela Velazquez
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - María Eugenia Matzkin
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Mónica Beatriz Frungieri
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina; Cátedra de Química, Ciclo Básico Común, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Mariana Gómez Peña
- Centro Médico Pregna Medicina Reproductiva. Ciudad Autónoma de Buenos Aires, Argentina
| | - Ignacio de Zúñiga
- Centro Médico Pregna Medicina Reproductiva. Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - María Silvia Bianchi
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
9
|
Insulin signalling and glucose transport in the ovary and ovarian function during the ovarian cycle. Biochem J 2017; 473:1483-501. [PMID: 27234585 PMCID: PMC4888492 DOI: 10.1042/bcj20160124] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/03/2016] [Indexed: 12/16/2022]
Abstract
Data derived principally from peripheral tissues (fat, muscle and liver) show that insulin signals via diverse interconnecting intracellular pathways and that some of the major intersecting points (known as critical nodes) are the IRSs (insulin receptor substrates), PI3K (phosphoinositide kinase)/Akt and MAPK (mitogen-activated protein kinase). Most of these insulin pathways are probably also active in the ovary and their ability to interact with each other and also with follicle-stimulating hormone (FSH) and luteinizing hormone (LH) signalling pathways enables insulin to exert direct modulating influences on ovarian function. The present paper reviews the intracellular actions of insulin and the uptake of glucose by ovarian tissues (granulosa, theca and oocyte) during the oestrous/menstrual cycle of some rodent, primate and ruminant species. Insulin signals through diverse pathways and these are discussed with specific reference to follicular cell types (granulosa, theca and oocyte). The signalling pathways for FSH in granulosa cells and LH in granulosa and theca cells are summarized. The roles of glucose and of insulin-mediated uptake of glucose in folliculogenesis are discussed. It is suggested that glucose in addition to its well-established role of providing energy for cellular function may also have insulin-mediated signalling functions in ovarian cells, involving AMPK (AMP-dependent protein kinase) and/or hexosamine. Potential interactions of insulin signalling with FSH or LH signalling at critical nodes are identified and the available evidence for such interactions in ovarian cells is discussed. Finally the action of the insulin-sensitizing drugs metformin and the thiazolidinedione rosiglitazone on follicular cells is reviewed.
Collapse
|
10
|
Eggers JC, Martino V, Reinbold R, Schäfer SD, Kiesel L, Starzinski-Powitz A, Schüring AN, Kemper B, Greve B, Götte M. microRNA miR-200b affects proliferation, invasiveness and stemness of endometriotic cells by targeting ZEB1, ZEB2 and KLF4. Reprod Biomed Online 2016; 32:434-45. [PMID: 26854065 DOI: 10.1016/j.rbmo.2015.12.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 02/06/2023]
Abstract
Endometriosis is characterized by growth of endometrial tissue at ectopic locations. Down-regulation of microRNA miR-200b is observed in endometriosis and malignant disease, driving tumour cells towards an invasive state by enhancing epithelial-to-mesenchymal transition (EMT). miR-200b up-regulation may inhibit EMT and invasive growth in endometriosis. To study its functional impact on the immortalized endometriotic cell line 12Z, the stromal cell line ST-T1b, and primary endometriotic stroma cells, a transient transfection approach with microRNA precursors was employed. Expression of bioinformatically predicted targets of miR-200b was analysed by qPCR. The cellular phenotype was monitored by Matrigel invasion assays, digital-holographic video microscopy and flow cytometry. qPCR revealed significant down-regulation of ZEB1 (P < 0.05) and ZEB2 (P < 0.01) and an increase in E-cadherin (P < 0.01). miR-200b overexpression decreased invasiveness (P < 0.0001) and cell motility (P < 0.05). In contrast, cell proliferation (P < 0.0001) and the stemness-associated side population phenotype (P < 0.01) were enhanced following miR-200b transfection. These properties were possibly due to up-regulation of the pluripotency-associated transcription factor KLF4 (P < 0.05) and require attention when considering therapeutic strategies. In conclusion, up-regulation of miR-200b reverts EMT, emerging as a potential therapeutic approach to inhibit endometriotic cell motility and invasiveness.
Collapse
Affiliation(s)
- Julia C Eggers
- Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany
| | - Valentina Martino
- Institute of Biomedical Technologies, National Research Council, 20090 Segrate-Milan, Italy
| | - Rolland Reinbold
- Institute of Biomedical Technologies, National Research Council, 20090 Segrate-Milan, Italy
| | - Sebastian D Schäfer
- Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany
| | - Anna Starzinski-Powitz
- Institute of Anthropology and Human Genetics for Biologists, Johann-Wolfgang-Goethe University of Frankfurt, Frankfurt, Germany
| | - Andreas N Schüring
- Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany
| | - Björn Kemper
- Biomedical Technology Center, University of Muenster, 48149 Muenster, Germany
| | - Burkhard Greve
- Klinik für Strahlentherapie - Radioonkologie, Universitätsklinikum Münster, D-48149 Münster, Germany.
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany.
| |
Collapse
|
11
|
Modulation of the IGF system and proliferation in human endometrial stromal cells by metformin: a dose-dependent effect. Arch Gynecol Obstet 2015; 292:465-72. [PMID: 25687657 DOI: 10.1007/s00404-015-3650-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 02/06/2015] [Indexed: 12/22/2022]
Abstract
PURPOSE To assess the metformin effect on endometrial stromal cell decidualization, proliferation, gene and protein expression of IGFBPs, IGFs and their receptors. METHODS Human endometrial stromal cells (hESCs) were cultured from endometrial biopsies of 11 women undergoing surgery for benign reasons. hESCs were decidualized with and without metformin in increasing doses. Supernatant and cells were harvested after decidualization for 12-14 days, followed by real-time PCR of IGFBP 1-6, IGF I, IGF II and their receptors. Prolactin, and IGFBP-1, -3, and -6 were additionally analyzed in supernatant by ELISA. Proliferation of hESCs and decidualization of hESCs were assessed under the influence of metformin. Data were analyzed using the paired t test with p < 0.05 considered significant. RESULTS While lower concentrations of metformin (10(-4), 10(-5 )M) did not influence the decidualization and proliferation capacity of hESCs, higher concentrations (10(-3), 10(-2 )M metformin) significantly (p < 0.05) diminished decidualization, as well as stromal cell proliferation in a dose-dependent manner. Higher concentrations of metformin lead to a significant (p < 0.05) dose-dependent attenuation of the progesterone effect with regard to IGFBP-1, -3, -5, -6, as well as IGF I receptor, while it did not change the expression of IGFBP-2 and -4, IGF I and II and the IGF II receptor. This was confirmed on the protein level for IGFBP-1, -3, and -6. CONCLUSION We were able to demonstrate for the first time a dose-dependent local effect of metformin within hESCs. Metformin might therefore influence locally the endometrial proliferation and maturation, and could open up new treatment options for gynecological diseases by vaginal application of metformin.
Collapse
|
12
|
Maruthini D, Harris SE, Barth JH, Balen AH, Campbell BK, Picton HM. The effect of metformin treatment in vivo on acute and long-term energy metabolism and progesterone production in vitro by granulosa cells from women with polycystic ovary syndrome. Hum Reprod 2014; 29:2302-16. [PMID: 25139174 PMCID: PMC4164147 DOI: 10.1093/humrep/deu187] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
STUDY QUESTION What are the consequences of polycystic ovary syndrome (PCOS) pathology and metformin-pretreatment in vivo in women with PCOS on the metabolism and steroid production of follicular phenotype- and long-term cultured-granulosa cells (GC)? SUMMARY ANSWER PCOS pathology significantly compromised glucose metabolism and the progesterone synthetic capacity of follicular- and long-term cultured-GCs and the metabolic impact of PCOS on GC function was alleviated by metformin-pretreatment in vivo. WHAT IS KNOWN ALREADY Granulosa cells from women with PCOS have been shown to have an impaired insulin-stimulated glucose uptake and lactate production in vitro. However, these results were obtained by placing GCs in unphysiological conditions in culture medium containing high glucose and insulin concentrations. Moreover, existing data on insulin-responsive steroid production in vitro by PCOS GCs vary. STUDY DESIGN, SIZE AND DURATION Case-control experimental research comparing glucose uptake, pyruvate and lactate production and progesterone production in vitro by GCs from three aetiological groups, all undergoing IVF; healthy control women (Control, n = 12), women with PCOS treated with metformin in vivo (Metformin, n = 8) and women with PCOS not exposed to metformin (PCOS, n = 8). The study was conducted over a period of 3 years between 2007 and 2010. PARTICIPANTS/MATERIALS, SETTING, METHODS Rotterdam criteria were used for the diagnosis of PCOS; all subjects were matched for age, BMI and baseline FSH. Individual patient cultures were undertaken with cells incubated in a validated, physiological, serum-free culture medium containing doses of 0–6 mM glucose and 0–100 ng/ml insulin for 6 h and 144 h to quantify the impact of treatments on acute and long-term metabolism, respectively, and progesterone production. The metabolite content of spent media was measured using spectrophotometric plate reader assay. The progesterone content of spent media was measured by enzyme-linked immunosorbent assay. Viable GC number was quantified after 144 h of culture by the vital dye Neutral Red uptake assay. MAIN RESULTS AND THE ROLE OF CHANCE Granulosa cells from women with PCOS pathology revealed reduced pyruvate production and preferential lactate production in addition to their reduced glucose uptake during cultures (P < 0.05). Metformin pretreatment alleviated this metabolic lesion (P < 0.05) and enhanced cell proliferation in vitro (P < 0.05), but cells retained a significantly reduced capacity for progesterone synthesis compared with controls (P < 0.05). LIMITATIONS, REASONS FOR CAUTION Although significant treatment effects were detected in this small cohort, further studies are required to underpin the molecular mechanisms of the effect of metformin on GCs. WIDER IMPLICATIONS OF THE FINDINGS The individual patient culture strategy combined with multifactorial experimental design strengthens the biological interpretation of the data. Collectively, these results support the notion that there is an inherent impairment in progesterone biosynthetic capacity of the GCs from women with PCOS. The positive, acute metabolic effect and the negative long-term steroidogenic effect on GCs following metformin exposure in vivo may have important implications for follicular development and luteinized GC function when the drug is used in clinical practice. STUDY FUNDING/COMPETING INTEREST(S) No competing interests. This work was supported by the UK Medical Research Council Grant Reference number G0800250.
Collapse
Affiliation(s)
- D Maruthini
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK
| | - S E Harris
- Division of Reproduction and Early Development, Leeds Institute for Genetics, Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - J H Barth
- Department of Clinical Biochemistry, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | - A H Balen
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK
| | - B K Campbell
- Division of Human Development at Nottingham University Hospitals NHS Trust, Department of Obstetrics and Gynaecology, Queen's Medical Centre Campus, Nottingham NG7 2UH, UK
| | - H M Picton
- Division of Reproduction and Early Development, Leeds Institute for Genetics, Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| |
Collapse
|
13
|
Codner E, Iñíguez G, López P, Mujica V, Eyzaguirre FC, Asenjo S, Torrealba I, Cassorla F. Metformin for the treatment of hyperandrogenism in adolescents with type 1 diabetes mellitus. Horm Res Paediatr 2014; 80:343-9. [PMID: 24280743 DOI: 10.1159/000355513] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 09/03/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A high prevalence of hyperandrogenism has been reported in women with type 1 diabetes (T1D). Metformin has been used as a therapeutic agent in patients with polycystic ovarian syndrome and in T1D patients without hyperandrogenism. This study sought to determine the effect of metformin on hyperandrogenism and ovarian function in adolescents with T1D. METHODS We recruited 24 girls with T1D. The participants had hyperandrogenism and displayed suboptimal metabolic control. The patients were enrolled in a randomized, double-blind, placebo-controlled trial. One group received metformin (850 mg bid) and the other group received a placebo. Treatment was administered for 9 months. Ovulation, steroids and gonadotropin levels were evaluated. RESULTS Metformin treatment was associated with decreases in testosterone, free androgen index, androstenedione, 17-OH progesterone and estradiol levels. The girls who were treated with placebo showed stable steroid, gonadotropin and sex hormone-binding globulin levels during the analysis. No differences were observed in the Ferriman-Gallwey scores, ovulation rates, HbA1c levels or daily insulin doses of the girls treated with metformin compared with the placebo group. CONCLUSION Treating hyperandrogenic T1D adolescents with metformin significantly decreased the serum androgens compared to the placebo, but metformin therapy did not significantly affect clinical parameters, such as hirsutism, ovulation and metabolic control.
Collapse
Affiliation(s)
- Ethel Codner
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Ferreira GD, Germeyer A, de Barros Machado A, do Nascimento TL, Strowitzki T, Brum IS, von Eye Corleta H, Capp E. Metformin modulates PI3K and GLUT4 expression and Akt/PKB phosphorylation in human endometrial stromal cells after stimulation with androgen and insulin. Eur J Obstet Gynecol Reprod Biol 2014; 175:157-62. [DOI: 10.1016/j.ejogrb.2014.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 12/17/2013] [Accepted: 01/04/2014] [Indexed: 11/27/2022]
|
15
|
Ferreira GD, Germeyer A, de Barros Machado A, do Nascimento TL, Brum IS, Strowitzki T, von Eye Corleta H, Capp E. Are growth factor receptors modulated by metformin in human endometrial stromal cells after stimulation with androgen and insulin? Arch Gynecol Obstet 2014; 290:361-7. [PMID: 24627035 DOI: 10.1007/s00404-014-3197-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 02/27/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To assess the effect of metformin on gene and protein expression of insulin receptor (IR) and IGF-1 (IGF-1R) receptor in human endometrial stromal cells after stimulation with androgen and insulin. STUDY DESIGN Primary culture of endometrial stromal cells stimulated with estrogen, progesterone with or without androgen or insulin, and treated with metformin for 24 and 48 h, followed by RNA (qRT-PCR) and protein (Western blot) extraction and analysis. RESULTS IR gene expression was increased after treatment with insulin (2.9-fold change, p = 0.027) and further after metformin treatment (4.7-fold change, p < 0.001), and in IGF-1R, the group treated with insulin (1.83-fold change) and metformin (1.78-fold change) showed more expression, than control group (p < 0.001). Similarly, IR protein expression was increased after addition of metformin and insulin (249,869 ± 15,878) in relation to the other groups (p < 0.001). Furthermore, cells treated with insulin (153,634 ± 29,123) and androgen plus insulin (162,854 ± 86,258) had a higher IR protein expression compared to control (104,654 ± 5,634) and androgen group (71,595 ± 3,439, (p = 0.045 and 0.021). In groups treated with insulin (127,711 ± 4,591) and androgen plus insulin (151,098 ± 5,194) the protein IGF-1R was increased compared to control (79,355 ± 3,470) and the androgen-only group (79,326 ± 3,114) (p < 0.001). CONCLUSION Metformin in combination with insulin increased IR protein and gene expressions, while it had no influence on the protein expression of IGF-1R in endometrial stromal cells.
Collapse
Affiliation(s)
- Gustavo Dias Ferreira
- Laboratório de Biologia Molecular Endócrina e Tumoral, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Fuhrmeister IP, Branchini G, Pimentel AM, Ferreira GD, Capp E, Brum IS, von Eye Corleta H. Human granulosa cells: insulin and insulin-like growth factor-1 receptors and aromatase expression modulation by metformin. Gynecol Obstet Invest 2014; 77:156-62. [PMID: 24603137 DOI: 10.1159/000358829] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 01/18/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIM Granulosa cells are the source of the most important ovarian steroids. Even in patients without significant improvement in metabolic parameters, metformin has apparently an important effect on the ovary. The aim of this study was to evaluate gene and protein expression of an insulin receptor (IR), insulin-like growth factor-1 (IGF1) receptor (IGF1R) and aromatase in granulosa cells treated with metformin and insulin. METHODS Luteinized granulosa cells were collected from 27 patients during in vitro fertilization procedures. Cells were isolated, stored in culture for 24 h and divided into four groups: control; metformin for 30 min, and metformin for 30 min plus insulin for 30 or 60 min. RESULTS IR and IGF1R mRNA expression was significantly enhanced by metformin but was not affected by insulin. Aromatase mRNA expression was significantly reduced in metformin-incubated cells following stimulation with insulin for 30 min. No statistical differences were found in IGF1R and aromatase protein expression, and IR expression was not detected. CONCLUSION A direct effect of metformin on the gene expression of IGF1R, IR and aromatase was observed. Further studies should investigate the role of IGF1R, IR and aromatase in ovarian physiology for a better understanding of the effect of metformin.
Collapse
|
17
|
Palomba S, Falbo A, La Sala GB. Metformin and gonadotropins for ovulation induction in patients with polycystic ovary syndrome: a systematic review with meta-analysis of randomized controlled trials. Reprod Biol Endocrinol 2014; 12:3. [PMID: 24387273 PMCID: PMC3895676 DOI: 10.1186/1477-7827-12-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 12/18/2013] [Indexed: 11/10/2022] Open
Abstract
The current systematic review with meta-analysis of randomized controlled trials (RCTs) was aimed to evaluate the effects of metformin on reproductive outcomes in patients with polycystic ovary syndrome (PCOS) who receive gonadotropins for ovulation induction. After systematic review of electronic databases and websites for registration of RCTs, a total of 7 RCTs reporting data on 1023 cycles were included in the final analysis. Descriptive data showed an overall low studies' quality due to unclear sequence generation and allocation concealment, lack of blinding procedure, incomplete outcome data and several biases and/or confounders. Data synthesis showed that metformin improved live-birth (odds ratio [OR] = 1.94, 95% confidence interval [CI] 1.10 to 3.44; P = 0.020) and pregnancy (OR = 2.25, 95% CI 1.50 to 3.38; P < 0.0001) rates, without significant heterogeneity across the studies (P = 0.230, estimation of inconsistency = 30%; and P = 0.710, estimation of inconsistency = 0%, respectively, for live-birth and pregnancy rates). A significant reduction of cancellation rate was observed after metformin administration (OR = 0.41, 95% CI 0.24 to 0.72, P = 0.002) without significant heterogeneity across the studies (P = 0.500, estimation of inconsistency = 0%). Metformin administration influenced or did not influence other secondary endpoints assessed with a significant heterogeneity. In conclusion, metformin administration increases the live-birth and pregnancy rate in PCOS patients who receive gonadotropins for ovulation induction. Further well designed, blinded, placebo-controlled, and adequately powered RCTs are need to confirm that metanalytic results.
Collapse
Affiliation(s)
- Stefano Palomba
- Obstetrics and Gynecology Unit, Department of Obstetrics, Gynecology and Pediatrics, Azienda Ospedaliera ASMN, Istituto di Ricovero e Cura a Carattere Scientifico, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Angela Falbo
- Obstetrics and Gynecology Unit, Department of Obstetrics, Gynecology and Pediatrics, Azienda Ospedaliera ASMN, Istituto di Ricovero e Cura a Carattere Scientifico, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Giovanni B La Sala
- Obstetrics and Gynecology Unit, Department of Obstetrics, Gynecology and Pediatrics, Azienda Ospedaliera ASMN, Istituto di Ricovero e Cura a Carattere Scientifico, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
- University of Modena and Reggio Emilia, Via Università 4, 41100 Modena, Italy
| |
Collapse
|
18
|
Palomba S, Falbo A, La Sala GB. Effects of metformin in women with polycystic ovary syndrome treated with gonadotrophins for in vitrofertilisation and intracytoplasmic sperm injection cycles: a systematic review and meta-analysis of randomised controlled trials. BJOG 2013; 120:267-276. [DOI: 10.1111/1471-0528.12070] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- S Palomba
- Obstetrics and Gynaecology Unit; Department of Obstetrics, Gynaecology and Paediatrics; Azienda Ospedaliera ASMN; Istituto di Ricovero e Cura a Carattere Scientifico; University of Modena and Reggio Emilia; Italy
| | - A Falbo
- Obstetrics and Gynaecology Unit; Department of Obstetrics, Gynaecology and Paediatrics; Azienda Ospedaliera ASMN; Istituto di Ricovero e Cura a Carattere Scientifico; University of Modena and Reggio Emilia; Italy
| | - GB La Sala
- Obstetrics and Gynaecology Unit; Department of Obstetrics, Gynaecology and Paediatrics; Azienda Ospedaliera ASMN; Istituto di Ricovero e Cura a Carattere Scientifico; University of Modena and Reggio Emilia; Italy
| |
Collapse
|
19
|
Metformin enhances the action of insulin on porcine granulosa-lutein cells in vitro. Anim Reprod Sci 2012; 136:100-7. [DOI: 10.1016/j.anireprosci.2012.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 09/17/2012] [Accepted: 10/11/2012] [Indexed: 11/22/2022]
|
20
|
FERLA RITA, HASPINGER EVA, SURMACZ EVA. Metformin inhibits leptin-induced growth and migration of glioblastoma cells. Oncol Lett 2012; 4:1077-1081. [PMID: 23162655 PMCID: PMC3499461 DOI: 10.3892/ol.2012.843] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/18/2012] [Indexed: 11/06/2022] Open
Abstract
Metformin, a derivative of biguanide, is a first-line therapy for type 2 diabetes mellitus. Since the drug has been shown to significantly reduce the risk of various cancers and cancer mortality in diabetic patients, it is being considered as a potential anticancer therapeutic or preventive agent. In cellular models, metformin inhibits the growth of many types of cancer cells; however, its effects on glioblastoma multi-forme (GBM) are not well characterized. Here, we analyzed the effects of metformin on the growth and migration of LN18 and LN229 GBM cells cultured under basal conditions or exposed to leptin, a cytokine that has recently been implicated in GBM development. We found that 2-16 mM metformin reduced basal and leptin-stimulated growth of GBM cells in a dose-dependent manner. Furthermore, the drug significantly inhibited the migration of GBM cells. The action of metformin was mediated through the upregulation of its main signaling molecule, the adenosine monophosphate-activated protein kinase (AMPK), as well as through the downregulation of the signal transducer and activator of transcription 3 (STAT3) and the Akt/PKB serine/threonine protein kinase. In leptin-treated cells, the drug reversed the effects of the cytokine on the AMPK and STAT3 pathways, but modulated Akt activity in a cell-dependent manner. Our results suggest that metformin or similar AMPK-targeting agents with optimized blood-brain-barrier penetrability could be developed as potential treatments of GBM and could be used in conjunction with other target drugs such as leptin receptor antagonists.
Collapse
Affiliation(s)
- RITA FERLA
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122,
USA
| | - EVA HASPINGER
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122,
USA
- Department of Medical Oncology, University of Verona, 37189 Verona,
Italy
| | - EVA SURMACZ
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122,
USA
| |
Collapse
|
21
|
NYGGF4 (PID1) effects on insulin resistance are reversed by metformin in 3T3-L1 adipocytes. J Bioenerg Biomembr 2012; 44:665-71. [PMID: 22968630 DOI: 10.1007/s10863-012-9472-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 08/17/2012] [Indexed: 01/14/2023]
Abstract
NYGGF4 (also called PID1) is a recently discovered gene that is involved in obesity-related insulin resistance (IR). We aimed in the present study to further elucidate the effects of NYGGF4 on IR and the underlying mechanisms through using metformin treatment in 3T3-L1 adipocytes. Our data showed that the metformin pretreatment strikingly enhanced insulin-stimulated glucose uptake through increasing GLUT4 translocation to the PM in NYGGF4 overexpression adipocytes. NYGGF4 overexpression resulted in significant inhibition of tyrosine phosphorylation of IRS-1 and serine phosphorylation of Akt, whereas incubation with metformin strongly activated IRS-1 and Akt phosphorylation in NYGGF4 overexpression adipocytes. The reactive oxygen species (ROS) levels in NYGGF4 overexpression adipocytes were strikingly enhanced, which could be decreased by the metformin pretreatment. Our data also showed that metformin increased the expressions of PGC1-α, NRF-1, and TFAM, which were reduced in the NYGGF4 overexpression adipocytes. These results suggest that NYGGF4 plays a role in IR and its effects on IR could be reversed by metformin through activating IRS-1/PI3K/Akt and AMPK-PGC1-α pathways.
Collapse
|
22
|
Makker A, Goel MM, Das V, Agarwal A. PI3K-Akt-mTOR and MAPK signaling pathways in polycystic ovarian syndrome, uterine leiomyomas and endometriosis: an update. Gynecol Endocrinol 2012; 28:175-81. [PMID: 21916800 DOI: 10.3109/09513590.2011.583955] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PI3K-Akt-mTOR and MAP kinase are two important cell signaling pathways that are activated by steroid hormones and growth factors leading to cellular events including gene expression, cell proliferation and survival. These pathways are considered as an attractive target for the development of novel anticancer molecules, and selective inhibitors specifically targeting different components of these cascades have been developed. This review summarizes the current available knowledge on the PI3K-Akt-mTOR and MAPK pathways and their targeting in estrogen-dependent benign gynecological disorders viz. polycystic ovarian syndrome, uterine leiomyomas and endometriosis, which are a significant cause of high morbidity in women of reproductive age group. Increasing knowledge about the role of the two growth regulatory pathways in the pathogenesis of these disorders may give the opportunity to use specific signal transduction inhibitors for management of these patients in future.
Collapse
Affiliation(s)
- Annu Makker
- Post-Graduate Department of Pathology, CSM Medical University, Lucknow, India.
| | | | | | | |
Collapse
|
23
|
Abstract
AIMS To review the relationship between insulin resistance and polycystic ovary syndrome. METHODS A literature review. RESULTS Insulin resistance likely plays a central pathogenic role in polycystic ovary syndrome and may explain the pleiotropic presentation and involvement of multiple organ systems. Insulin resistance in the skeletal muscle of women with polycystic ovary syndrome involves both intrinsic and acquired defects in insulin signalling. The cellular insulin resistance in polycystic ovary syndrome has been further shown to involve a novel post-binding defect in insulin signal transduction. Treatment of insulin resistance through lifestyle therapy or with a diabetes drug has become mainstream therapy in women with polycystic ovary syndrome. However, effects with current pharmacologic treatment with metformin tend to be modest, with limited benefit as an agent to treat infertility. Insulin resistance contributes to increased risk for pregnancy complications, diabetes and cardiovascular disease risk profile in polycystic ovary syndrome, which is further exacerbated by obesity. While numerous studies demonstrate increased prevalence of cardiovascular disease risk factors in women with polycystic ovary syndrome, there are limited data showing that women with polycystic ovary syndrome are at increased risk for cardiovascular disease events. CONCLUSIONS Insulin resistance is linked to polycystic ovary syndrome. Further study of lifestyle and pharmacologic interventions that reduce insulin resistance, such as metformin, are needed to demonstrate that they are effective in reducing the risk of diabetes, endometrial abnormalities and cardiovascular disease events in women with polycystic ovary syndrome.
Collapse
Affiliation(s)
- J M Pauli
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
24
|
Palomba S, Falbo A, Di Cello A, Cappiello F, Tolino A, Zullo F. Does metformin affect the ovarian response to gonadotropins for in vitro fertilization treatment in patients with polycystic ovary syndrome and reduced ovarian reserve? A randomized controlled trial. Fertil Steril 2011; 96:1128-33. [PMID: 21917254 DOI: 10.1016/j.fertnstert.2011.08.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 06/19/2011] [Accepted: 08/12/2011] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To evaluate the effects of metformin on the ovarian response to gonadotropins given for in vitro fertilization (IVF) programs in patients with polycystic ovary syndrome (PCOS) and reduced ovarian reserve. DESIGN Prospective, parallel, randomized, double-blind, placebo-controlled clinical trial. SETTING Academic departments of obstetrics and gynecology, and a private IVF center. PATIENT(S) Primary infertile patients with PCOS older than 35 years and/or with a basal follicle-stimulating hormone (FSH) level higher than 10 IU/L who were scheduled for IVF cycles. INTERVENTION(S) Gonadotropin-releasing hormone agonist flare-up protocol and high starting doses of recombinant FSH plus metformin or placebo tablets. MAIN OUTCOME MEASURE(S) Primary end point: cancellation rate for low ovarian response. Secondary end-points: other clinical, biochemical, and reproductive data. RESULT(S) Enrollment was stopped after 88 participants had been randomized and analyzed due to an unacceptable increased risk of poor ovarian response in the metformin arm. Statistically significant differences between the metformin and placebo groups were observed in the dose of gonadotropins used, peak estradiol levels, and the number of dominant follicles, retrieved oocytes, and metaphase II oocytes. CONCLUSION(S) In patients with PCOS and reduced ovarian reserve, metformin worsened the response to gonadotropins, and its administration should be stopped before the start of controlled ovarian hyperstimulation for IVF programs. CLINICAL TRIALS IDENTIFICATION NUMBER: NCT01208740.
Collapse
Affiliation(s)
- Stefano Palomba
- Department of Obstetrics and Gynecology, University Magna Graecia of Catanzaro, Catanzaro, Italy.
| | | | | | | | | | | |
Collapse
|
25
|
Capp E, Jauckus J, von Eye Corleta H, Toth B, Strowitzki T, Germeyer A. Does metformin influence the insulin-, IGF I- and IGF II-receptor gene expression and Akt phosphorylation in human decidualized endometrial stromal cells? Eur J Obstet Gynecol Reprod Biol 2011; 158:248-53. [PMID: 21664031 DOI: 10.1016/j.ejogrb.2011.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 05/04/2011] [Accepted: 05/13/2011] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To assess the effects of metformin on insulin-, IGF I-, and IGF II-receptor gene expression and Akt phosphorylation in decidualized human endometrial stromal cells (ESC) after stimulation with insulin, IGF I and II. STUDY DESIGN ESC were isolated from healthy, regularly cycling women and after two passages decidualized with estrogen/progesterone±metformin. Cells were incubated with insulin, IGF I or IGF II for 1, 5, and 10 min to assess Akt phosphorylation by Western blot. To investigate the insulin-, IGF I- and IGF II-receptor gene expression ESC were incubated with insulin, IGF I or IGF II for 6 and 24h. RESULTS Insulin- and IGF I-receptor gene expression in ESC changed significantly after incubation with insulin, IGF I or IGF II. This was further augmented in metformin pretreated cells, while IGF II-receptor gene expression changed particularly after pretreatment with metformin. Akt phosphorylation peaked after 5 min insulin, IGF I and IGF II stimulation in ESC in both control (control 0.08 ± 0.03 vs. insulin 0.74 ± 0.19, IGF I 0.68 ± 0.22, IGF II 0.53 ± 0.13, p<0.05) and metformin pretreated cells (control 0.03 ± 0.01 vs. insulin 0.75 ± 0.11, IGF I 0.74 ± 0.15, IGF II 0.67 ± 0.09, p<0.005). However, there was no significant difference between the control and metformin pretreated group. CONCLUSION Insulin, IGF I and IGF II lead to changes in their receptor gene expression and induced Akt phosphorylation in ESC. These effects were further highlighted in the presence of metformin.
Collapse
Affiliation(s)
- Edison Capp
- Department of Gynecological Endocrinology and Reproductive Medicine, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Nordhoff V, Sonntag B, von Tils D, Götte M, Schüring AN, Gromoll J, Redmann K, Casarini L, Simoni M. Effects of the FSH receptor gene polymorphism p.N680S on cAMP and steroid production in cultured primary human granulosa cells. Reprod Biomed Online 2011; 23:196-203. [PMID: 21680247 DOI: 10.1016/j.rbmo.2011.04.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 04/28/2011] [Accepted: 04/28/2011] [Indexed: 10/18/2022]
Abstract
The study was designed to evaluate in vitro the cellular mechanisms of the single nucleotide polymorphism (SNP) p.N680S of the FSH receptor gene (FSHR) in human granulosa cells (GC) and included patients homozygous for the FSHR SNP (NN/SS) undergoing ovarian stimulation. GC were isolated during oocyte retrieval and cultured for 1–7 days. Basal oestradiol and progesterone concentrations were measured after short-term culture. The kinetics of cAMP, oestradiol and progesterone concentrations in response to various amounts of FSH were analysed in a 6–7 day culture. Basal oestradiol, but not progesterone, concentrations on day 1 of GC culture, were significantly higher in NN compared with SS (P = 0.045), but non-responsive to FSH stimulation. Immunofluorescence microscopy demonstrated the re-appearance of FSHR expression with increasing days in culture. Upon stimulation with FSH, GC cultured for 6–7 days displayed a dose-dependent increase of cAMP, oestradiol and progesterone but no difference in the EC50 values between both variants. Primary long-term GC cultures are a suitable system to study the effects of FSH in vitro. However, the experiments suggest that factors down-stream of progesterone production or external to GC might be involved in the clinically observed differences in an FSHR variant-mediated response to FSH.
Collapse
Affiliation(s)
- Verena Nordhoff
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Domagkstrasse 11, D-48149 Münster, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Rice S, Pellatt LJ, Bryan SJ, Whitehead SA, Mason HD. Action of metformin on the insulin-signaling pathway and on glucose transport in human granulosa cells. J Clin Endocrinol Metab 2011; 96:E427-35. [PMID: 21209036 DOI: 10.1210/jc.2010-2060] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT Hyperinsulinemia in polycystic ovary syndrome is widely treated with the insulin sensitizer metformin, which, in addition to its systemic effects, directly affects the ovarian insulin-stimulated steroidogenesis pathway. OBJECTIVE Our aim was to investigate the interaction of metformin with the other insulin-stimulated ovarian pathway, namely that leading to glucose uptake. DESIGN Human granulosa-luteal cells were cultured with metformin (10(-7) M), insulin (10 ng/ml) or metformin and insulin (met + ins) combined. Insulin receptor (IR) involvement was assessed by culture with an (anti)-insulin receptor (IR) antibody. MAIN OUTCOME MEASURES The effect of metformin on insulin-receptor substrate proteins 1 and 2 (IRS-1 and -2) mRNA and protein expression was determined. The KGN granulosa-cell line was used to investigate the effect of insulin and metformin on Akt activation and glucose transporter-4 (Glut-4) expression. Glut-4 translocation from the cytosol to the membrane was determined in cytoplasmic and membrane-enriched fractions of protein lysates. RESULTS IRS-1 mRNA and protein increased with all treatments. In contrast, basal IRS-2 mRNA levels were barely detectable, but transcription was up-regulated by metformin. The anti-IR antibody reduced treatment-stimulated IRS-1 to basal levels and IRS-2 expression to an even greater extent than IRS-1, showing greater dependence on the IR than IRS-1. Metformin in the presence of insulin activated Akt and this was dependent on phosphoinositide-3 kinase, as was translocation of Glut-4 to the membrane. Metformin was able to substantially enhance the insulin-stimulated translocation of Glut-4 transporters from the cytosol to the membrane. CONCLUSION This net increase in Glut-4 transporters in the plasma membrane has the potential to increase glucose uptake and metabolism by granulosa cells of the insulin-resistant polycystic ovary, thereby facilitating follicle growth.
Collapse
Affiliation(s)
- Suman Rice
- Division of Basic Medical Sciences, St. George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE United Kingdom.
| | | | | | | | | |
Collapse
|
28
|
Palomba S, Falbo A, Russo T, Orio F, Tolino A, Zullo F. Systemic and local effects of metformin administration in patients with polycystic ovary syndrome (PCOS): relationship to the ovulatory response. Hum Reprod 2010; 25:1005-1013. [DOI: 10.1093/humrep/dep466] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
29
|
Romualdi D, Giuliani M, Cristello F, Fulghesu AM, Selvaggi L, Lanzone A, Guido M. Metformin effects on ovarian ultrasound appearance and steroidogenic function in normal-weight normoinsulinemic women with polycystic ovary syndrome: a randomized double-blind placebo-controlled clinical trial. Fertil Steril 2009; 93:2303-10. [PMID: 19268929 DOI: 10.1016/j.fertnstert.2009.01.114] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 01/14/2009] [Accepted: 01/19/2009] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To investigate metformin effects on the endocrine-metabolic parameters and ovarian morphology in normoinsulinemic women with polycystic ovary syndrome (PCOS). DESIGN Randomized double-blind study. SETTING Operative Division of Endocrinological Gynecology, Università Cattolica del Sacro Cuore. PATIENT(S) Twenty-eight normal-weight normoinsulinemic PCOS women. INTERVENTION(S) Patients were randomized to receive metformin 500 mg twice a day (group A, 15 subjects) or placebo (group B, 13 subjects) for 6 months. Ultrasonographic pelvic exams, hormonal and lipid features, and oral glucose tolerance test were performed at baseline and after 3 and 6 months of treatment. MAIN OUTCOME MEASURE(S) Hormonal and glycoinsulinemic assessment, ovarian ultrasound appearance. RESULT(S) Glycoinsulinemic assessment remained unvaried in both groups. About 70% of patients in group A experienced a restoration of menstrual cyclicity. Metformin significantly decreased testosterone levels at 3 and 6 months) and 17-hydroxyprogesterone levels at 6 months, and improved hirsutism score at 6 months. No clinical or hormonal modifications occurred in group B. Metformin, but not placebo, reduced ovarian volume and stromal/total area ratio at 3 and 6 months. CONCLUSION(S) Metformin seems to improve the menstrual pattern and ultrasonographic ovarian features in normoinsulinemic PCOS women. These effects seem to be, at least in part, independent of the insulin-lowering properties of the drug.
Collapse
Affiliation(s)
- Daniela Romualdi
- Department of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
Palomba S, Falbo A, Zullo F, Orio F. Evidence-based and potential benefits of metformin in the polycystic ovary syndrome: a comprehensive review. Endocr Rev 2009; 30:1-50. [PMID: 19056992 DOI: 10.1210/er.2008-0030] [Citation(s) in RCA: 268] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metformin is an insulin sensitizer widely used for the treatment of patients affected by type 2 diabetes mellitus. Because many women with polycystic ovary syndrome (PCOS) are insulin resistant, metformin was introduced in clinical practice to treat these patients also. Moreover, metformin's effect has other targets beside its insulin-sensitizing action. The present review was aimed at describing all evidence-based and potential uses of metformin in PCOS patients. In particular, we will analyze the uses of metformin not only for the treatment of all PCOS-related disturbances such as menstrual disorders, anovulatory infertility, increased abortion, or complicated pregnancy risk, hyperandrogenism, endometrial, metabolic and cardiovascular abnormalities, but also for the prevention of the syndrome.
Collapse
Affiliation(s)
- Stefano Palomba
- Department of Gynecology and Obstetrics, University "Magna Graecia" of Catanzaro, Via Pio X, 88100 Catanzaro, Italy.
| | | | | | | |
Collapse
|
31
|
Nikolova V, Koo CY, Ibrahim SA, Wang Z, Spillmann D, Dreier R, Kelsch R, Fischgräbe J, Smollich M, Rossi LH, Sibrowski W, Wülfing P, Kiesel L, Yip GW, Götte M. Differential roles for membrane-bound and soluble syndecan-1 (CD138) in breast cancer progression. Carcinogenesis 2009; 30:397-407. [PMID: 19126645 DOI: 10.1093/carcin/bgp001] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The heparan sulfate proteoglycan syndecan-1 (Sdc1) modulates cell proliferation, adhesion, migration and angiogenesis. Proteinase-mediated shedding converts Sdc1 from a membrane-bound coreceptor into a soluble effector capable of binding the same ligands. In breast carcinomas, Sdc1 overexpression correlates with poor prognosis and an aggressive phenotype. To distinguish between the roles of membrane-bound and shed forms of Sdc1 in breast cancer progression, human MCF-7 breast cancer cells were stably transfected with plasmids overexpressing wild-type (WT), constitutively shed and uncleavable forms of Sdc1. Overexpression of WT Sdc1 increased cell proliferation, whereas overexpression of constitutively shed Sdc1 decreased proliferation. Fibroblast growth factor-2-mediated mitogen-activated protein kinase signaling was reduced following small-interfering RNA (siRNA)-mediated knockdown of Sdc1 expression. Constitutively, membrane-bound Sdc1 inhibited invasiveness, whereas soluble Sdc1 promoted invasion of MCF-7 cells into matrigel matrices. The latter effect was reversed by the matrix metalloproteinase inhibitors N-isobutyl-N-(4-methoxyphenylsufonyl) glycyl hydroxamic acid and tissue inhibitor of metalloproteinase (TIMP)-1. Affymetrix microarray analysis identified TIMP-1, Furin and urokinase-type plasminogen activator receptor as genes differentially regulated in soluble Sdc1-overexpressing cells. Endogenous TIMP-1 expression was reduced in cells overexpressing soluble Sdc1 and increased in those overexpressing the constitutively membrane-bound Sdc1. Moreover, E-cadherin protein expression was downregulated in cells overexpressing soluble Sdc1. Our results suggest that the soluble and membrane-bound forms of Sdc1 play different roles at different stages of breast cancer progression. Proteolytic conversion of Sdc1 from a membrane-bound into a soluble molecule marks a switch from a proliferative to an invasive phenotype, with implications for breast cancer diagnostics and potential glycosaminoglycan-based therapies.
Collapse
Affiliation(s)
- Viktoriya Nikolova
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mathur R, Alexander CJ, Yano J, Trivax B, Azziz R. Use of metformin in polycystic ovary syndrome. Am J Obstet Gynecol 2008; 199:596-609. [PMID: 19084097 DOI: 10.1016/j.ajog.2008.09.010] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 07/29/2008] [Accepted: 09/03/2008] [Indexed: 12/25/2022]
Abstract
Women with polycystic ovary syndrome (PCOS) have a myriad of phenotypic and clinical features that may guide therapeutic options for metabolic protection and ovulation induction. The use of metformin may prove beneficial in a subset of the population of women with PCOS. Hyperinsulinemia, as demonstrated by elevated insulin levels on a 2-hour 75-g load glucose tolerance test, is an important parameter in deciding whether or not to initiate metformin therapy to women with PCOS with the hope of preventing or delaying the onset of type 2 diabetes mellitus (DM). Cardiovascular risk factors including markers of subclinical inflammation, and dyslipidemia may also be improved by metformin therapy. For ovulation induction, metformin is not as effective as clomiphene citrate as first-line therapy for women with PCOS. There are no clear data to suggest that metformin reduces pregnancy loss or improves pregnancy outcome in PCOS, and it is currently recommended that metformin be discontinued with the first positive pregnancy test result, unless there are other medical indications (eg, type 2 DM). This review addresses practical management guidelines for the uses of metformin in women with PCOS.
Collapse
|
33
|
Mason H, Colao A, Blume-Peytavi U, Rice S, Qureshi A, Pellatt L, Orio F, Atkin SL. Polycystic ovary syndrome (PCOS) trilogy: a translational and clinical review. Clin Endocrinol (Oxf) 2008; 69:831-44. [PMID: 18616705 DOI: 10.1111/j.1365-2265.2008.03329.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Helen Mason
- Basic Medical Sciences, St George's University of London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Seibel SA, Chou KH, Capp E, Spritzer PM, von Eye Corleta H. Effect of metformin on IGF-1 and IGFBP-1 levels in obese patients with polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol 2008; 138:122-4. [PMID: 17350746 DOI: 10.1016/j.ejogrb.2007.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 10/31/2006] [Accepted: 02/05/2007] [Indexed: 11/22/2022]
|
35
|
Stute P, Nisslein T, Götte M, Kamischke A, Kiesel L, Klockenbusch W. Effects of black cohosh on estrogen biosynthesis in normal breast tissue in vitro. Maturitas 2007; 57:382-91. [PMID: 17548177 DOI: 10.1016/j.maturitas.2007.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 04/16/2007] [Accepted: 04/20/2007] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To investigate the effect of black cohosh on the estrogen biosynthesis in the breast in vitro. METHODS Steroid sulfatase (STS) activity was studied in normal breast tissue obtained from pre- and postmenopausal women undergoing reduction mammoplasty. STS protein expression was studied by immunohistochemistry and western blotting. Breast tissue was incubated in vitro without or with black cohosh (iCR) at concentrations ranging from 0.1mg/ml to 1 ng/ml. STS activity was evaluated by incubating homogenized breast tissue with [3H]-estrone sulfate, separating the formed products, estrone (E1) and estradiol (E2), by thin layer chromatography and measuring the amounts of E1 and E2 by scintillation counting. RESULTS STS protein expression and enzymatic activity were detected in all specimens investigated. In all groups, significantly more E1 than E2 was produced. Local estrogen formation was decreased in premenopausal breast tissue by treatment with iCR at 0.1mg/ml (p<or=0.05). CONCLUSIONS iCR decreases local estrogen formation in normal human breast tissue in vitro. This may contribute to the lack of hormonal effects of black cohosh in breast tissue observed in previous studies.
Collapse
Affiliation(s)
- Petra Stute
- Department of Obstetrics and Gynecology, University Clinic of Muenster, 48149 Muenster, Germany.
| | | | | | | | | | | |
Collapse
|
36
|
Marcondes JAM, Yamashita SAY, Maciel GAR, Baracat EC, Halpern A. Metformin in normal-weight hirsute women with polycystic ovary syndrome with normal insulin sensitivity. Gynecol Endocrinol 2007; 23:273-8. [PMID: 17558685 DOI: 10.1080/09513590701192529] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Fifteen normal-weight (body mass index (BMI) 21.50 +/- 1.65 kg/m(2)) hirsute women with polycystic ovary syndrome and normal insulin sensitivity were treated with 850 mg metformin orally, three times daily, for 4 months. Before and at the end of the treatment, clinical data as well as serum concentrations of sex steroid hormones, gonadotropins, fasting plasma glucose and insulin, insulin resistance - homeostasis model assessment (HOMA-IR), carbohydrate tolerance and the area under the curve for insulin (AUC(insulin)) were analyzed. Three patients withdrew from the study. Seven of the remaining 12 patients presented menstrual pattern improvement, followed by ovulatory cycles at the end of the treatment period. There were no changes in BMI and hirsutism score. A significant (p < 0.05) decrease in luteinizing hormone (LH) (from 8.18 +/- 4.34 to 5.05 +/- 1.53 IU/ml), testosterone (from 104.66 +/- 27.54 to 82.00 +/- 23.05 ng/dl), fasting insulin (from 9.66 +/- 4.79 to 7.83 +/- 3.06 microIU/ml), AUC(insulin) (from 9239 +/- 3285 to 7660 +/- 2565 microUI/ml x min) and HOMA-IR (from 2.15 +/- 1.2 to 1.67 +/- 0.74), and a significant increase in follicle-stimulating hormone (FSH) (from 4.05 +/- 1.53 to 5.96 +/- 2.13 IU/ml), were observed at the end of the treatment period. A higher LH and a lower FSH predicted clinical improvement, while basal insulin and AUC(insulin) showed lower predictive value.
Collapse
Affiliation(s)
- José A M Marcondes
- Endocrinology Unit, Hospital das Clínicas, University of São Paulo Medical School of Medicine, São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
37
|
Radke I, Götte M, Kersting C, Mattsson B, Kiesel L, Wülfing P. Expression and prognostic impact of the protein tyrosine phosphatases PRL-1, PRL-2, and PRL-3 in breast cancer. Br J Cancer 2006; 95:347-54. [PMID: 16832410 PMCID: PMC2360632 DOI: 10.1038/sj.bjc.6603261] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The aim of this study was to investigate the expression of the protein tyrosine phosphatases (PTP) PRL-1, PRL-2, and PRL-3 in human breast cancer and to evaluate its clinical and prognostic significance. PRL-PTP mRNA expression was examined in malignant (n=7) and nonmalignant (n=7) cryoconserved breast tissue samples as well as in eight breast cancer cell lines by RT–PCR. Furthermore, protein expression of PRL-3 was analysed semiquantitatively by immunohistochemistry in ductal breast carcinoma in situ (n=135) and invasive breast cancer (n=147) by use of tissue microarray technology (TMA). In 24 lymph node-positive patients we selected the corresponding lymph node metastases for analysis of PRL-3 expression, and a validation set (n=99) of invasive breast cancer samples was examined. Staining results were correlated with clinicopathological parameters and long-term follow-up. PRL-3 mRNA expression was significantly higher in malignant compared to benign breast tissue. For PRL-1 and PRL-2 expression no significant differences were observed. Staining of TMAs showed PRL-3 expression in 85.9% ductal carcinoma in situ and 75.5% invasive breast carcinomas. Analysis of survival parameters revealed a shorter disease-free survival (DFS) in patients with PRL-3-positive carcinomas, and in particular a significantly shorter DFS in nodal-positive patients with PRL-3 overexpressing tumours as compared to PRL-3-negative breast carcinomas (66±7 months (95% CI, 52–80) vs 97±9 months (95% CI, 79–115); P=0.032). Moreover, we found a more frequent expression of PRL-3 in lymph node metastases as compared to the primary tumours (91.7 vs 66.7%; P=0.033). Our results suggest that PRL-3 might serve as a novel prognostic factor in breast cancer, which may help to predict an adverse disease outcome.
Collapse
Affiliation(s)
- I Radke
- Department of Obstetrics and Gynaecology, University of Münster, Albert-Schweitzer-Str. 33, D-48149 Münster, Germany.
| | | | | | | | | | | |
Collapse
|
38
|
Tosca L, Solnais P, Ferré P, Foufelle F, Dupont J. Metformin-induced stimulation of adenosine 5' monophosphate-activated protein kinase (PRKA) impairs progesterone secretion in rat granulosa cells. Biol Reprod 2006; 75:342-51. [PMID: 16760380 DOI: 10.1095/biolreprod.106.050831] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Metformin is an anti-diabetic drug commonly used to treat cycle disorders and anovulation in women with polycystic ovary syndrome. However, the effects and molecular mechanism of metformin in the ovary are not entirely understood. We investigated the effects of this drug on steroidogenesis and proliferation in rat granulosa cells. Metformin (10 mM) treatment for 48 h reduced progesterone and estradiol (E2) production in both basal conditions and under FSH stimulation. It also decreased the levels of the HSD3B, CYP11A1, STAR, and CYP19A1 proteins in response to FSH (10(-8) M) and of HSD3B in the basal state only. Metformin treatment (10 mM, 24 h) also reduced cell proliferation and the levels of CCND2 and CCNE proteins without affecting cell viability, both in the basal state and in response to FSH. Furthermore, metformin treatment for 1 h simultaneously increased the Thr172 phosphorylation of PRKAA (adenosine 5' monophosphate-activated protein kinase alpha) and the Ser79 phosphorylation of ACACA (acetyl-Coenzyme A carboxylase alpha). The adenovirus-mediated production of dominant-negative PRKAA totally abolished the effects of metformin on progesterone secretion, HSD3B and STAR protein production, and MAPK3/1 phosphorylation. Conversely, total inhibition of PRKAA Thr172 phosphorylation with the dominant-negative PRKAA adenovirus did not restore the decrease in E2 production and cell proliferation induced by metformin. Our results therefore strongly suggest that metformin reduces progesterone production via a PRKAA-dependent mechanism, whereas PRKAA activation is not essential for the decrease in E2 production and cell growth induced by metformin in rat granulosa cells.
Collapse
Affiliation(s)
- Lucie Tosca
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, 37380 Nouzilly, France
| | | | | | | | | |
Collapse
|
39
|
King TD, Song L, Jope RS. AMP-activated protein kinase (AMPK) activating agents cause dephosphorylation of Akt and glycogen synthase kinase-3. Biochem Pharmacol 2006; 71:1637-47. [PMID: 16620785 PMCID: PMC1618797 DOI: 10.1016/j.bcp.2006.03.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2006] [Revised: 02/28/2006] [Accepted: 03/06/2006] [Indexed: 12/18/2022]
Abstract
AMP-activated protein kinase (AMPK) is a key cellular sensor of reduced energy supply that is activated by increases in the cellular ratio of AMP/ATP. Phenformin and 5-aminoimidazole-4-carboxamide riboside (AICAR) are two drugs widely used to activate AMPK experimentally. In both differentiated hippocampal neurons and neuroblastoma SH-SY5Y cells we found that these two agents not only activated AMPK, but conversely greatly reduced the activating Ser/Thr phosphorylation of Akt. This blockade of Akt activity consequently lowered the inhibitory serine-phosphorylation of its substrates, glycogen synthase kinase-3alpha/beta (GSK3alpha/beta). An inhibitor of AMPK (Compound C) did not block dephosphorylation of Akt and GSK3. Thus, both drugs widely used to activate AMPK also caused dephosphorylation of Akt and of GSK3. The mechanism for Akt dephosphorylation caused by phenformin, but not AICAR, was due to inhibition of growth factor-induced signaling that leads to Akt phosphorylation. Stimulation of muscarinic receptors with carbachol in SH-SY5Y cells also activated AMPK and transiently caused dephosphorylation of Akt. These findings show that Akt dephosphorylation often occurs concomitantly with AMPK activation when cells are treated with phenformin or AICAR, indicating that these drugs do not only affect AMPK but also cause a coordinated inverse regulation of AMPK and Akt.
Collapse
Affiliation(s)
- Taj D King
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294-0017, USA
| | | | | |
Collapse
|