1
|
Bolte E, Dean T, Garcia B, Seferovic MD, Sauter K, Hummel G, Bucher M, Li F, Hicks J, Qin X, Suter MA, Barrozo ER, Jochum M, Shope C, Friedman JE, Gannon M, Wesolowski SR, McCurdy CE, Kievit P, Aagaard KM. Initiation of metformin in early pregnancy results in fetal bioaccumulation, growth restriction, and renal dysmorphology in a primate model. Am J Obstet Gynecol 2024; 231:352.e1-352.e16. [PMID: 38871238 PMCID: PMC11344684 DOI: 10.1016/j.ajog.2024.06.002] [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: 01/17/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND In recent years, pragmatic metformin use in pregnancy has stretched to include prediabetes mellitus, type 2 diabetes mellitus, gestational diabetes mellitus, and (most recently) preeclampsia. However, with its expanded use, concerns of unintended harm have been raised. OBJECTIVE This study developed an experimental primate model and applied ultrahigh performance liquid chromatography coupled to triple-quadrupole mass spectrometry for direct quantitation of maternal and fetal tissue metformin levels with detailed fetal biometry and histopathology. STUDY DESIGN Within 30 days of confirmed conception (defined as early pregnancy), 13 time-bred (timed-mated breeding) Rhesus dams with pregnancies designated for fetal necropsy were initiated on twice-daily human dose-equivalent 10 mg/kg metformin or vehicle control. Pregnant dams were maintained as pairs and fed either a control chow or 36% fat Western-style diet. Metformin or placebo vehicle control was delivered in various treats while the animals were separated via a slide. A cesarean delivery was performed at gestational day 145, and amniotic fluid and blood were collected, and the fetus and placenta were delivered. The fetus was immediately necropsied by trained primate center personnel. All fetal organs were dissected, measured, sectioned, and processed per clinical standards. Fluid and tissue metformin levels were assayed using validated ultrahigh performance liquid chromatography coupled to triple-quadrupole mass spectrometry in selected reaction monitoring against standard curves. RESULTS Among 13 pregnancies at gestational day 145 with fetal necropsy, 1 dam and its fetal tissues had detectable metformin levels despite being allocated to the vehicle control group (>1 μmol metformin/kg maternal weight or fetal or placental tissue), whereas a second fetus allocated to the vehicle control group had severe fetal growth restriction (birthweight of 248.32 g [<1%]) and was suspected of having a fetal congenital condition. After excluding these 2 fetal pregnancies from further analyses, 11 fetuses from dams initiated on either vehicle control (n=4: 3 female and 1 male fetuses) or 10 mg/kg metformin (n=7: 5 female and 2 male fetuses) were available for analyses. Among dams initiated on metformin at gestational day 30 (regardless of maternal diet), significant bioaccumulation within the fetal kidney (0.78-6.06 μmol/kg; mean of 2.48 μmol/kg), liver (0.16-0.73 μmol/kg; mean of 0.38 μmol/kg), fetal gut (0.28-1.22 μmol/kg; mean of 0.70 μmol/kg), amniotic fluid (0.43-3.33 μmol/L; mean of 1.88 μmol/L), placenta (0.16-1.00 μmol/kg; mean of 0.50 μmol/kg), fetal serum (0.00-0.66 μmol/L; mean of 0.23 μmol/L), and fetal urine (4.10-174.10 μmol/L; mean of 38.5 μmol/L) was observed, with fetal levels near biomolar equivalent to maternal levels (maternal serum: 0.18-0.86 μmol/L [mean of 0.46 μmol/L]; maternal urine: 42.60-254.00 μmol/L [mean of 149.30 μmol/L]). Western-style diet feeding neither accelerated nor reduced metformin bioaccumulations in maternal or fetal serum, urine, amniotic fluid, placenta, or fetal tissues. In these 11 animals, fetal bioaccumulation of metformin was associated with less fetal skeletal muscle (57% lower cross-sectional area of gastrocnemius) and decreased liver, heart, and retroperitoneal fat masses (P<.05), collectively driving lower delivery weight (P<.0001) without changing the crown-rump length. Sagittal sections of fetal kidneys demonstrated delayed maturation, with disorganized glomerular generations and increased cortical thickness. This renal dysmorphology was not accompanied by structural or functional changes indicative of renal insufficiency. CONCLUSION Our study demonstrates fetal bioaccumulation of metformin with associated fetal growth restriction and renal dysmorphology after maternal initiation of the drug within 30 days of conception in primates. Given these results and the prevalence of metformin use during pregnancy, additional investigation of any potential immediate and enduring effects of prenatal metformin use is warranted.
Collapse
Affiliation(s)
- Erin Bolte
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Tyler Dean
- Oregon National Primate Research Center, Beaverton, OR
| | - Brandon Garcia
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Maxim D Seferovic
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | | | - Gwendolynn Hummel
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Matthew Bucher
- Department of Human Physiology, University of Oregon, Eugene OR
| | - Feng Li
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - John Hicks
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Xuan Qin
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Melissa A Suter
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Enrico R Barrozo
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Michael Jochum
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Cynthia Shope
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Maureen Gannon
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | | | | | - Paul Kievit
- Oregon National Primate Research Center, Beaverton, OR
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Oregon National Primate Research Center, Beaverton, OR.
| |
Collapse
|
2
|
Talmo MSA, Fløysand IS, Nilsen GØ, Løvvik TS, Ødegård R, Juliusson PB, Vanky E, Simpson MR. Growth Restriction in the Offspring of Mothers With Polycystic Ovary Syndrome. JAMA Netw Open 2024; 7:e2430543. [PMID: 39190302 PMCID: PMC11350484 DOI: 10.1001/jamanetworkopen.2024.30543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/03/2024] [Indexed: 08/28/2024] Open
Abstract
Importance Polycystic ovary syndrome (PCOS) is a common endocrine disorder, characterized by subfertility, increased risk of metabolic diseases, and pregnancy complications. Previous studies diverge regarding the association between maternal PCOS and newborn anthropometrics. Objective To explore the association between maternal PCOS and newborn anthropometrics and the modifying effects of maternal body mass index, PCOS phenotype, and gestational diabetes. Design, Setting, and Participants This cohort study followed up women from the first half of pregnancy to birth and combined data from 3 clinical trials of pregnant women with PCOS and a reference population consisting of participants in the Norwegian Mother, Father, and Child Cohort (MoBa) Study, with data from the Medical Birth Registry of Norway. The recruitment period for the clinical trials was between October 1, 2000, and August 31, 2017, and for MoBa, between July 1, 1999, and December 31, 2008. Participants included women with singleton pregnancies and live-born children. Data were analyzed from January 1 to June 15, 2023. Exposure Maternal PCOS status. Main Outcomes and Measures Newborn birth weight, birth length, and head circumference as continuous variables and z scores, and ponderal index (calculated as the birth weight in grams × 100 divided by the birth length in centimeters cubed), placenta weight, and ratio of birth weight to placenta weight (BWPW). Results The cohort included 390 pregnant women with PCOS (mean [SD] age, 29.6 [4.2] years) and 68 708 women in the reference group (mean [SD] age, 30.4 [4.5] years). Offspring in the PCOS group had lower birth weight, birth length, and head circumference than in the reference group offspring. The estimated mean differences in z scores were -0.26 (95% CI, -0.38 to -0.14) for birth weight, -0.19 (95% CI, -0.33 to -0.05) for birth length, and -0.13 (95% CI, -0.26 to -0.01) for head circumference. The PCOS group also had a lower ponderal index (-0.04 [95% CI, -0.07 to -0.004] g × 100/cm3) and placenta weight (-24 [95% CI, -43 to -5)] g), and higher BWPW ratio (0.4 [95% CI, 0.3 to 0.5]). The association between growth restriction and PCOS was more apparent when additionally adjusting for body mass index. Neither PCOS phenotype nor gestational diabetes diagnosis was associated with neonatal anthropometry in women with PCOS. Conclusions and Relevance In this cohort of mother-infant pairs, maternal PCOS status was associated with lower birth weight, shorter birth length, and smaller head circumference in the offspring. This growth restriction was more pronounced when adjusting for BMI, providing insight into the association between PCOS and body mass index. The study contributed to the understanding of how PCOS affects the offspring.
Collapse
Affiliation(s)
- Maren Sophie Aaserud Talmo
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingvild Skogedal Fløysand
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Tone S. Løvvik
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Obstetrics and Gynecology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Rønnaug Ødegård
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Centre for Obesity Research, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Petur Benedikt Juliusson
- Department of Health Registry Research and Development, National Institute of Public Health, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Paediatric and Adolescent Medicine, Haukeland University Hospital, Bergen, Norway
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Obstetrics and Gynecology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
3
|
Nilsen GØ, Simpson MR, Hanem LGE, Løvvik T, Ødegård R, Stokkeland LMT, Andersen M, Juliusson PB, Vanky E. Anthropometrics of neonates born to mothers with PCOS with metformin or placebo exposure in utero. Acta Obstet Gynecol Scand 2024; 103:176-187. [PMID: 37488743 PMCID: PMC10755130 DOI: 10.1111/aogs.14637] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/26/2023]
Abstract
INTRODUCTION Fetal growth may be affected by both maternal polycystic ovary syndrome (PCOS) and metformin therapy. Here, we explore the effect of intrauterine metformin exposure on birth anthropometrics of infants born to women with PCOS. We also investigated whether the effect of metformin on birth anthropometrics is modified by maternal pre-pregnancy body mass index, PCOS hyperandrogenic phenotype, serum androgen levels, preconception use of metformin and offspring sex. Additionally, we assessed newborn anthropometrics in relation to a national reference population. MATERIAL AND METHODS Individual data from three randomized controlled triasl were pooled. The randomized controlled trials investigated the effects of metformin in pregnant women with PCOS. In all, 397 and 403 were randomized to the metformin and placebo groups, respectively. A Scandinavian growth reference was used to calculate sex and gestational age adjusted z-scores. Linear regression models were used to estimate the effect of metformin on offspring z-scores of head circumference, birth length, birthweight, placental weight, body mass index, ponderal index and birthweight:placental weight ratio. S-testosterone, s-androstenedione, and s-sex-hormone binding globulin from four timepoints in pregnancy were analyzed. RESULTS Compared with the PCOS-placebo group, newborns in the PCOS-metformin group had larger head circumference (head circumference z-score: mean difference = 0.25, 95% CI = 0.11- 0.40). This effect of metformin on head circumference z-score was particularly observed among offspring of overweight/obese mothers and mothers with hyperandrogenic PCOS-phenotype. We observed no difference in other anthropometric measures between the metformin and placebo groups or any clear interaction between maternal androgen levels and metformin. Newborns in the PCOS-placebo group were shorter than in the reference population (birth length z-score: mean = -0.04, 95% CI = -0.05 to -0.03), but head circumference and birthweight were similar. CONCLUSIONS Larger head circumference was observed at birth in metformin-exposed offspring of mothers with PCOS. PCOS-offspring were also shorter, with a similar birthweight to the reference population, indirectly indicating higher weight-to-height ratio at birth.
Collapse
Affiliation(s)
- Guro Ørndal Nilsen
- Faculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Melanie Rae Simpson
- Department of Public Health and NursingNorwegian University of Science and TechnologyTrondheimNorway
| | - Liv Guro Engen Hanem
- Children's Clinic, St. Olav's HospitalTrondheim University HospitalTrondheimNorway
| | - Tone Shetelig Løvvik
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and TechnologyTrondheimNorway
- Department of Obstetrics and Gynecology, St. Olav's HospitalTrondheim University HospitalTrondheimNorway
| | - Rønnaug Ødegård
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and TechnologyTrondheimNorway
- Center for Obesity Research, St. Olav's HospitalTrondheim University HospitalTrondheimNorway
| | - Live Marie T. Stokkeland
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and TechnologyTrondheimNorway
- Center of Molecular Inflammation Research (CEMIR)Norwegian University of Science and Technology (NTNU)TrondheimNorway
| | | | - Petur Benedikt Juliusson
- Department of Health Registry Research and DevelopmentNational Institute of Public HealthBergenNorway
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Eszter Vanky
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and TechnologyTrondheimNorway
- Department of Obstetrics and Gynecology, St. Olav's HospitalTrondheim University HospitalTrondheimNorway
| |
Collapse
|
4
|
Palm CVB, Dreyer AF, Boye H, Jørgensen JS, Wu C, Højsager FD, Jensen TK, Glintborg D, Andersen MS. Higher free testosterone in the third trimester was associated with lower abdominal circumference at birth in boys: Odense child cohort. BJOG 2024; 131:36-45. [PMID: 37443462 DOI: 10.1111/1471-0528.17595] [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: 03/07/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVE To investigate associations between maternal testosterone status and offspring birth anthropometrics. DESIGN Population-based prospective cohort study. SETTING University Hospital. POPULATION 1486 mother-child dyads from Odense Child Cohort. METHODS Maternal blood samples were collected at gestational weeks 27-30 and free testosterone (FT) levels were calculated using the Vermeulen equation from total testosterone (TT) analysed by mass spectrometry and sex hormone binding globulin. Associations between FT or TT levels and birth anthropometrics were analysed with multiple linear regression models according to offspring sex with adjustment for maternal age, parity, smoking and educational level. Analyses were repeated with polycystic ovary syndrome as exposure for offspring birth anthropometrics. MAIN OUTCOME MEASURES Offspring birth weight (BW), birth length, abdominal and head circumferences. RESULTS Maternal mean (SD) age was 30.2 (4.5) years and pre-pregnancy body mass index was 23.5 (5.3) kg/m2 . In boys (n = 787), higher FT was associated with lower birth weight (adjusted doubling constant = -65.53, P = 0.010), shorter birth length (adjusted doubling constant = -0.43, P < 0.001), and lower abdominal circumference (adjusted doubling constant = -0.39, P < 0.001); Higher TT was associated with lower abdominal circumference (adjusted doubling constant = -0.25, P = 0.028). In girls, no associations were found between maternal FT or TT and offspring anthropometrics. CONCLUSIONS Higher maternal free testosterone exposure was linked to reduced birth weight, length and abdominal circumference in boys, whereas girls were not susceptible to maternal testosterone exposure.
Collapse
Affiliation(s)
- Camilla V B Palm
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Anja F Dreyer
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Henriette Boye
- Odense Child Cohort, Hans Christian Andersen Hospital for Children and Adolescents, Odense University Hospital, Odense, Denmark
- OPEN Patient data Explorative Network (OPEN), University of Southern Denmark, Odense, Denmark
| | - Jan S Jørgensen
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Gynaecology and Obstetrics, Odense University Hospital, Odense, Denmark
| | - Chunsen Wu
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Frederik D Højsager
- Odense Child Cohort, Hans Christian Andersen Hospital for Children and Adolescents, Odense University Hospital, Odense, Denmark
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Tina K Jensen
- Odense Child Cohort, Hans Christian Andersen Hospital for Children and Adolescents, Odense University Hospital, Odense, Denmark
- OPEN Patient data Explorative Network (OPEN), University of Southern Denmark, Odense, Denmark
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Dorte Glintborg
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Marianne S Andersen
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Swenson KS, Wang D, Jones AK, Nash MJ, O’Rourke R, Takahashi DL, Kievit P, Hennebold JD, Aagaard KM, Friedman JE, Jones KL, Rozance PJ, Brown LD, Wesolowski SR. Metformin Disrupts Signaling and Metabolism in Fetal Hepatocytes. Diabetes 2023; 72:1214-1227. [PMID: 37347736 PMCID: PMC10450827 DOI: 10.2337/db23-0089] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/20/2023] [Indexed: 06/24/2023]
Abstract
Metformin is used by women during pregnancy to manage diabetes and crosses the placenta, yet its effects on the fetus are unclear. We show that the liver is a site of metformin action in fetal sheep and macaques, given relatively abundant OCT1 transporter expression and hepatic uptake following metformin infusion into fetal sheep. To determine the effects of metformin action, we performed studies in primary hepatocytes from fetal sheep, fetal macaques, and juvenile macaques. Metformin increases AMP-activated protein kinase (AMPK) signaling, decreases mammalian target of rapamycin (mTOR) signaling, and decreases glucose production in fetal and juvenile hepatocytes. Metformin also decreases oxygen consumption in fetal hepatocytes. Unique to fetal hepatocytes, metformin activates stress pathways (e.g., increased PGC1A gene expression, NRF-2 protein abundance, and phosphorylation of eIF2α and CREB proteins) alongside perturbations in hepatokine expression (e.g., increased growth/differentiation factor 15 [GDF15] and fibroblast growth factor 21 [FGF21] expression and decreased insulin-like growth factor 2 [IGF2] expression). Similarly, in liver tissue from sheep fetuses infused with metformin in vivo, AMPK phosphorylation, NRF-2 protein, and PGC1A expression are increased. These results demonstrate disruption of signaling and metabolism, induction of stress, and alterations in hepatokine expression in association with metformin exposure in fetal hepatocytes. ARTICLE HIGHLIGHTS The major metformin uptake transporter OCT1 is expressed in the fetal liver, and fetal hepatic uptake of metformin is observed in vivo. Metformin activates AMPK, reduces glucose production, and decreases oxygen consumption in fetal hepatocytes, demonstrating similar effects as in juvenile hepatocytes. Unique to fetal hepatocytes, metformin activates metabolic stress pathways and alters the expression of secreted growth factors and hepatokines. Disruption of signaling and metabolism with increased stress pathways and reduced anabolic pathways by metformin in the fetal liver may underlie reduced growth in fetuses exposed to metformin.
Collapse
Affiliation(s)
- Karli S. Swenson
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Dong Wang
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Amanda K. Jones
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Michael J. Nash
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Rebecca O’Rourke
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Diana L. Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jon D. Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Kjersti M. Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine & Texas Children’s Hospital, Houston, TX
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Kenneth L. Jones
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Paul J. Rozance
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Laura D. Brown
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | | |
Collapse
|
6
|
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: 19] [Impact Index Per Article: 9.5] [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
|
7
|
Grindheim S, Ebbing C, Karlsen HO, Skulstad SM, Real FG, Lønnebotn M, Løvvik T, Vanky E, Kessler J. Metformin exposure, maternal PCOS status and fetal venous liver circulation: A randomized, placebo-controlled study. PLoS One 2022; 17:e0262987. [PMID: 35089960 PMCID: PMC8797196 DOI: 10.1371/journal.pone.0262987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 11/16/2021] [Indexed: 11/19/2022] Open
Abstract
Background Metformin is prescribed to women with polycystic ovary syndrome (PCOS) to prevent pregnancy complications. Children exposed to metformin vs. placebo in utero, have increased head circumference at birth and are more overweight and obese at 8 years of age. Also, maternal PCOS-status seems to alter the long-term cardio-metabolic health of offspring. We hypothesized that the long-term effects of metformin-exposure and/or maternal PCOS may be mediated by circulatory adaptations during fetal life. Material and methods This is a sub-study of a larger double-blinded, placebo-controlled trial, where women with PCOS were randomized to metformin (2g/day) or placebo in pregnancy, a total of 487 women. A sub-group of participants (N = 58) took part in this sub-study and had an extended ultrasound examination at gestational week 32, including blood flow velocity and diameter measurements of the umbilical vein (UV), the ductus venosus (DV) and the portal vein (PV). Blood flow volume was calculated and adjusted for estimated fetal weight (EFW) (normalized flow). Metformin exposed fetuses were compared to placebo exposed fetuses. Fetuses of mothers with PCOS (metformin [n = 30] and placebo [n = 28]) were compared to a low-risk reference population (N = 160) by z-score statistics. Results There was no difference in fetal liver flow between metformin vs. placebo-exposed fetuses. Fetuses of mothers with PCOS had higher EFW (0.63 [95% CI 0.44–0.83] p<0.001), lower normalized UV, DV, PV, and lower total venous liver blood flows than the reference population. Conclusion Metformin during pregnancy did not affect fetal liver blood-flow. In our population, maternal PCOS-status was associated with reduced total venous liver blood-flow, which may explain altered growth and metabolism later in life.
Collapse
Affiliation(s)
- Sindre Grindheim
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| | - Cathrine Ebbing
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Svein Magne Skulstad
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Francisco Gómez Real
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marianne Lønnebotn
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Tone Løvvik
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Obstetrics and Gynaecology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Obstetrics and Gynaecology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jørg Kessler
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Islam H, Masud J, Islam YN, Haque FKM. An update on polycystic ovary syndrome: A review of the current state of knowledge in diagnosis, genetic etiology, and emerging treatment options. WOMEN'S HEALTH 2022; 18:17455057221117966. [PMID: 35972046 PMCID: PMC9386861 DOI: 10.1177/17455057221117966] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Polycystic ovary syndrome is the most common endocrine disorder in women of reproductive age, which is still incurable. However, the symptoms can be successfully managed with proper medication and lifestyle interventions. Despite its prevalence, little is known about its etiology. In this review article, the up-to-date diagnostic features and parameters recommended on the grounds of evidence-based data and different guidelines are explored. The ambiguity and insufficiency of data when diagnosing adolescent women have been put under special focus. We look at some of the most recent research done to establish relationships between different gene polymorphisms with polycystic ovary syndrome in various populations along with the underestimated impact of environmental factors like endocrine-disrupting chemicals on the reproductive health of these women. Furthermore, the article concludes with existing treatments options and the scopes for advancement in the near future. Various therapies have been considered as potential treatment through multiple randomized controlled studies, and clinical trials conducted over the years are described in this article. Standard therapies ranging from metformin to newly found alternatives based on vitamin D and gut microbiota could shine some light and guidance toward a permanent cure for this female reproductive health issue in the future.
Collapse
Affiliation(s)
- Hiya Islam
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Jaasia Masud
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Yushe Nazrul Islam
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Fahim Kabir Monjurul Haque
- Microbiology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| |
Collapse
|
9
|
Cao Q, Hu Y, Fu J, Huang X, Wu L, Zhang J, Huang W. Gestational metformin administration in women with polycystic ovary syndrome: A systematic review and meta-analysis of randomized control studies. J Obstet Gynaecol Res 2021; 47:4148-4157. [PMID: 34571574 DOI: 10.1111/jog.15044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/26/2021] [Accepted: 09/15/2021] [Indexed: 01/03/2023]
Abstract
AIMS To evaluate metformin's effects on pregnancy outcomes in women with polycystic ovary syndrome. METHODS A literature search was conducted using PubMed, EMBASE, Web of Science, MEDLINE, and the Cochrane Library. All randomized controlled trials comparing metformin administration during pregnancy versus placebo or blank in PCOS women were selected. The primary outcomes were the incidence of gestational diabetes mellitus (GDM), preterm delivery, and miscarriage. We combined data with the Review Manager. Bayesian meta-analysis was employed for further verification with the R software. RESULTS Six randomized control trial studies involving 1229 participants were included. Metformin use was associated with reduced risk of preterm delivery (Risk ratios [RR], 0.45; 95% CI, 0.25-0.80; p, 0.007) and higher larger neonatal head circumference (Mean difference (MD), 0.47; 95% CI, 0.20-0.74; p, 0.0006] but had no effect on the incidence of GDM (RR 1.87; 95% CI, 0.58-1.87; p, 0.89), miscarriage (RR, 0.85; 95% CI, 0.45-1.60; p, 0.62), pre-eclampsia (RR, 1.18; 95% CI, 0.43-3.21; p, 0.75), neonatal length (MD, 0.33; 95% CI, -0.12-0.78; p, 0.15) and birthweight (MD, 73.78; 95% CI, -52.98-200.53; p, 0.17). CONCLUSIONS Metformin administration in PCOS pregnancies was associated with reduced preterm delivery risk and larger neonatal head circumference.
Collapse
Affiliation(s)
- Qi Cao
- Department of Obstetrics and Gynaecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuanyuan Hu
- Department of Obstetrics and Gynaecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jing Fu
- Department of Obstetrics and Gynaecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xin Huang
- Department of Obstetrics and Gynaecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lukanxuan Wu
- Department of Obstetrics and Gynaecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jiani Zhang
- Department of Obstetrics and Gynaecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wei Huang
- Department of Obstetrics and Gynaecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Bao Z, Zhang Q, Pan M, Xi X, Wang Y, Zhang F, Wang F, Zou Y, Qu F. Alterations of brain metrics in fetuses of women with polycystic ovary syndrome : a retrospective study based on fetal magnetic resonance imaging. BMC Pregnancy Childbirth 2021; 21:557. [PMID: 34391385 PMCID: PMC8364105 DOI: 10.1186/s12884-021-04015-w] [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: 02/18/2021] [Accepted: 07/20/2021] [Indexed: 11/12/2022] Open
Abstract
Background Maternal polycystic ovary syndrome (PCOS) has potential detrimental effects on the neurodevelopment of offspring. This study aimed to evaluate the brain metrics in fetuses of women with PCOS based on fetal magnetic resonance imaging (MRI). Methods This retrospective study included 60 pregnant women with PCOS (PCOS group) and 120 pregnant non-PCOS women (control group). Fetal MRI was performed followed an ultrasound and for numerous clinical indications including known or suspected fetal pathology, history of fetal abnormality in previous pregnancy or in a family member. Fetal brain biometry and apparent diffusion coefficient (ADC) value were analysed. Results After adjusting for potential confounders, fetuses in the PCOS group showed the following characteristics compared to fetuses in the control group: (1) smaller cerebral fronto-occipital diameter (FOD), vermian height (VH) and anteroposterior diameter of the pons (APDP) (evident before 32 weeks; P = 0.042, P = 0.002 and P = 0.016, respectively); (2) larger left and right biparietal index (evident before 32 weeks; P = 0.048 and P = 0.025, respectively); (3) smaller left lateral ventricle (LV) (evident after 32 weeks; P = 0.005); (4) larger anteroposterior diameter of the vermis (APDV) and hippocampal infolding angle (HIA) (evident after 32 weeks; P = 0.003 and P < 0.001, respectively); (5) higher ADC value in frontal white matter (FWM) and in basal ganglia (BG) (evident before and after 32 weeks; all P < 0.05). Conclusions There exist a different pattern of brain metrics in PCOS offspring in utero.
Collapse
Affiliation(s)
- Zhongkun Bao
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China
| | - Manman Pan
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China
| | - Xi Xi
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China
| | - Yuanlin Wang
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China
| | - Fangfang Zhang
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China
| | - Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China
| | - Yu Zou
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China.
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Zhejiang, 310006, Hangzhou, China.
| |
Collapse
|
11
|
Zhang FF, Zhang Q, Wang YL, Wang FF, Hardiman PJ, Qu F. Intergenerational Influences between Maternal Polycystic Ovary Syndrome and Offspring: An Updated Overview. J Pediatr 2021; 232:272-281. [PMID: 33482217 DOI: 10.1016/j.jpeds.2021.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Fang-Fang Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan-Lin Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang-Fang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
12
|
Cranial molding on neonates in Ghana: mothers' perspective and their knowledge on potential harm to babies' brain. Childs Nerv Syst 2021; 37:1703-1711. [PMID: 33409614 DOI: 10.1007/s00381-020-05001-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Traditional cranial molding is an ancient practice prevalent in Ghana. In this work, we aimed at assessing mothers'/caregivers' perspective and their knowledge on potential harm of cranial molding on neonates. METHODS Two hundred and one (201) nursing mothers with babies aged 1-12 months were sampled in a cross-sectional study using questionnaires. We assessed the mothers'/caregivers' reasons for cranial molding, their perceived benefits of this practice, and their knowledge about the potential harm this practice pose to babies. RESULTS Sixty four percent (64%) of mothers confirmed they practice cranial molding on their babies either on their own or through the assistance of a caretaker. However, 72% of all mothers/caregivers did not know this practice has the potential to harm the baby in any way. Mothers'/caregivers' reasons for this practice included the following: to achieve a more "beautiful" head shape, hasten the healing of the fontanelle, and limit head growth. There was a significant association between the mothers'/caregivers' level of education and the practice of cranial molding (p value < 0.05). However, there was no association between head symmetry and cranial molding (p value > 0.05). CONCLUSIONS AND IMPLICATIONS Majority of mothers/caregivers were actively engaged in cranial molding on neonates but remain ignorant about the potential harm this practice could have on their babies. Mothers/caregivers therefore need to be educated about the potential harm posed by traditional cranial molding on neonates.
Collapse
|
13
|
D'Alterio MN, Sigilli M, Succu AG, Ghisu V, Laganà AS, Sorrentino F, Nappi L, Tinelli R, Angioni S. Pregnancy outcomes in women with polycystic ovarian syndrome (PCOS). Minerva Obstet Gynecol 2021; 74:45-59. [PMID: 33876903 DOI: 10.23736/s2724-606x.21.04758-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrinological disease of reproductive-aged women, with an estimated incidence ranging from 5% to 15%. The clinical manifestations of PCOS are heterogeneous and vary according to the age of the patient. Insulin resistance (IR), hyperandrogenism, and obesity are widely assumed to play a pivotal role in the pathophysiological mechanism of PCOS. As previously stated by many conducted meta-analyses, PCOS can cause a rising risk of pregnancy complications, including maternal, fetal, and neonatal complications. Pregnancy-induced hypertension (PIH), preeclampsia (PE), gestational diabetes mellitus (GDM), spontaneous preterm birth (PTB), and an increased necessity for a cesarean section (CS) are the most documented maternal implications. Regarding fetal outcomes, PCOS has also been correlated with elevated neonatal morbidity, prematurity, fetal growth restriction (FGR), birth weight variations (large for gestational age (LGA) and small for gestational age (SGA), and transfer to the neonatal intensive care unit (NICU). Owing to the variability of the studies performed, the association of PCOS with an elevated risk of adverse pregnancy outcomes is still controversial. This variability is found in the diagnosis and clinical presentations of PCOS, and can be influenced by pre-pregnancy circumstances and therapies as well as particular population and environmental features. The Amsterdam consensus guidelines confirm that obesity and IR can worsen maternal and fetal complications; thus, a closer follow-up should be offered to PCOS women during pregnancy.
Collapse
Affiliation(s)
- Maurizio N D'Alterio
- Division of Obstetrics and Gynecology, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy -
| | - Marco Sigilli
- Division of Obstetrics and Gynecology, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonio G Succu
- Division of Obstetrics and Gynecology, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Valeria Ghisu
- Division of Obstetrics and Gynecology, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonio S Laganà
- Department of Obstetrics and Gynecology, Filippo del Ponte Hospital, University of Insubria, Varese, Italy
| | - Felice Sorrentino
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynaecology, University of Foggia, Foggia, Italy
| | - Luigi Nappi
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynaecology, University of Foggia, Foggia, Italy
| | - Raffaele Tinelli
- Department of Obstetrics and Gynecology, Valle d'Itria Hospital, Martina Franca, Taranto, Italy
| | - Stefano Angioni
- Division of Obstetrics and Gynecology, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
14
|
Raperport C, Chronopoulou E, Homburg R. Effects of metformin treatment on pregnancy outcomes in patients with polycystic ovary syndrome. Expert Rev Endocrinol Metab 2021; 16:37-47. [PMID: 33634727 DOI: 10.1080/17446651.2021.1889366] [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: 11/27/2020] [Accepted: 02/09/2021] [Indexed: 01/28/2023]
Abstract
INTRODUCTION This review covers the current evidence regarding the use of metformin as a therapeutic intervention for optimizing pregnancy outcomes in women with polycystic ovary syndrome (PCOS). AREAS COVERED After searching Medline, Embase and CINAHL, all important large clinical trials and observational studies plus systematic reviews, meta-analyses and Cochrane reviews have been summarized here. The results have been compared to culminate in a thorough review and discussion on the use of metformin in relation to reproductive outcomes for women with PCOS. The role of metformin in PCOS is explored both in terms of achieving conception and during pregnancy. The existing evidence around metformin use is summarized both during the preconceptual period and during pregnancy, in relation to reproductive outcomes. EXPERT OPINION Metformin is a widely used medication, often prescribed to improve reproductive outcomes for women with PCOS. However, the evidence remains equivocal regarding its efficacy both in optimizing fertility and pregnancy outcomes. More research is required with special emphasis on metformin use within different populations, including ethnic groups and women with varying BMI ranges.
Collapse
Affiliation(s)
- Claudia Raperport
- Fertility Unit, Homerton University Hospital, London, UK
- Blizard Institute, Queen Mary University of London, London, UK
| | | | - Roy Homburg
- Fertility Unit, Homerton University Hospital, London, UK
| |
Collapse
|
15
|
Hegerty CK. The new gestational diabetes: Treatment, evidence and consent. Aust N Z J Obstet Gynaecol 2020; 60:482-485. [PMID: 32506466 PMCID: PMC7317553 DOI: 10.1111/ajo.13116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/07/2019] [Indexed: 12/27/2022]
|
16
|
Zhang Q, Bao ZK, Deng MX, Xu Q, Ding DD, Pan MM, Xi X, Wang FF, Zou Y, Qu F. Fetal growth, fetal development, and placental features in women with polycystic ovary syndrome: analysis based on fetal and placental magnetic resonance imaging. J Zhejiang Univ Sci B 2020; 21:977-989. [PMID: 33843163 PMCID: PMC7759450 DOI: 10.1631/jzus.b2000350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/09/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS), a common endocrine-metabolic dysfunction in reproductive-aged women, may be involved in compromised pregnancy and offspring outcomes. This study aimed to investigate whether maternal PCOS affects fetal growth, fetal development, and placental features. METHODS This retrospective case-control study included 60 pregnant women with PCOS (PCOS group) and 120 healthy pregnant women without PCOS (control group). Fetal magnetic resonance imaging (MRI) was performed followed by an ultrasound examination and indications for imaging, including known or suspected fetal pathology, history of fetal abnormality in previous pregnancy or in a family member, and concern for placenta accreta. Fetal MRI images were analyzed for head circumference (HC), abdomen circumference (AC), lung-to-liver signal intensity ratio (LLSIR, a prenatal marker of fetal lung maturity), lengths of liver and kidney diameters in fetuses, and placental relative signal intensity on T2-weighted single-shot fast spin echo (SSFSE) imaging (rSISSFSE), and placental relative apparent diffusion coefficient value (rADC). Data on height and weight of offspring were collected through telephone follow-up. RESULTS Compared to the control group, the PCOS group showed the following characteristics: (1) smaller biparietal diameter and femur length in fetuses (P=0.026 and P=0.005, respectively), (2) smaller HC in fetuses (evident after 32 weeks; P=0.044), (3) lower LLSIR and smaller dorsoventral length of liver in fetuses (evident before 32 weeks; P=0.005 and P=0.019, respectively), and (4) smaller placental thickness (evident before 32 weeks; P=0.017). No significant differences in placental rSISSFSE or rADC were observed between the groups (all P>0.05). No significant differences in height and weight of offspring during childhood existed between the groups (all P>0.05). CONCLUSIONS There exist alterations of fetal growth, fetal development, and placental features from women with PCOS.
Collapse
Affiliation(s)
- Qing Zhang
- School of Obstetrics and Gynecology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Zhong-kun Bao
- Department of Radiology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Mei-xiang Deng
- Department of Radiology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Qiong Xu
- Department of Radiology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Dan-dan Ding
- Department of Radiology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Man-man Pan
- School of Obstetrics and Gynecology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xi Xi
- School of Obstetrics and Gynecology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Fang-fang Wang
- Department of Chinese Integrative Medicine, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yu Zou
- Department of Radiology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Fan Qu
- Department of Chinese Integrative Medicine, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
17
|
Mokou M, Yang S, Zhan B, Geng S, Li K, Yang M, Yang G, Deng W, Liu H, Liu D, Zhu Z, Li L. Elevated Circulating Fetuin-B Levels Are Associated with Insulin Resistance and Reduced by GLP-1RA in Newly Diagnosed PCOS Women. Mediators Inflamm 2020; 2020:2483435. [PMID: 33061822 PMCID: PMC7545451 DOI: 10.1155/2020/2483435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Previous studies have suggested that Fetuin-B seems to be a secreted adipokine related to metabolic diseases. However, the results have been inconsistent. Here, our objective is to investigate the changes in circulating Fetuin-B levels in women with polycystic ovary syndrome (PCOS) and analyze the association of Fetuin-B and insulin resistance (IR). METHODS The current study is comprised of a cross-sectional study and a series of interventional studies. Oral glucose tolerance test (OGTT) and euglycemic-hyperinsulinemic clamp (EHC) were engaged to assess glucose tolerance and insulin sensitivity. Serum Fetuin-B levels were determined by ELISA. RESULTS Serum Fetuin-B and TNF-α levels were markedly increased in women with PCOS compared to healthy women. Circulating Fetuin-B was positively associated with body mass index, waist-to-hip ratio, the percentage of body fat (FAT%), systolic blood pressure, triglyceride, low-density lipoprotein cholesterol, fasting blood glucose, 2 h blood glucose after glucose overload, fasting insulin, 2 h insulin after glucose overload, HOMA-insulin resistance index (HOMA-IR), the area under the curve for insulin (AUCi), AUCg, and TNF-α, while negatively associated with M value and follicular stimulating hormone (FSH). During the EHC, Fetuin-B levels were found to be significantly increased in PCOS women. After a glucose challenge, serum Fetuin-B levels in healthy women were significantly increased. Lipid infusion reduced serum Fetuin-B levels in 30 healthy subjects. After six months of glucagon-like peptide-1 receptor agonist (GLP-1RA) intervention, serum Fetuin-B concentrations in PCOS women markedly decreased following ameliorated IR. CONCLUSION Our results indicate that Fetuin-B may be a biomarker of IR in individuals with PCOS. This trial is registered with ChiCTR-IIR-16007901.
Collapse
Affiliation(s)
- Mani Mokou
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, 400016 Chongqing, China
| | - Shan Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Bin Zhan
- The Thirteenth People's Hospital of Chongqing, Chongqing 400016, China
| | - Shan Geng
- The Thirteenth People's Hospital of Chongqing, Chongqing 400016, China
| | - Kejia Li
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Mengliu Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Wuquan Deng
- Department of Endocrinology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, China
| | - Hua Liu
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA
| | - Dongfang Liu
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing 400010, China
| | - Ling Li
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, 400016 Chongqing, China
| |
Collapse
|
18
|
Poprzeczny AJ, Louise J, Deussen AR, Dodd JM. Effect of metformin in addition to an antenatal diet and lifestyle intervention on fetal growth and adiposity: the GRoW randomised trial. BMC Endocr Disord 2020; 20:139. [PMID: 32928167 PMCID: PMC7488668 DOI: 10.1186/s12902-020-00618-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 08/31/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The infants born to women who are overweight or obese in pregnancy are at an increased risk of being born macrosomic or large for gestational age. Antenatal dietary and lifestyle interventions have been shown to be ineffective at reducing this risk. Our aim was to examine the effects of metformin in addition to a diet and lifestyle intervention on fetal growth and adiposity among women with a BMI above the healthy range. METHODS Women who had a body mass index ≥25 kg/m2 in early pregnancy, and a singleton gestation, were enrolled in the GRoW trial from three public maternity hospitals in metropolitan Adelaide. Women were invited to have a research ultrasounds at 28 and 36 weeks' gestation at which ultrasound measures of fetal biometry and adiposity were obtained. Fetal biometry z-scores and trajectories were calculated. Measurements and calculations were compared between treatment groups. This secondary analysis was pre-specified. RESULTS Ultrasound data from 511 women were included in this analysis. The difference in femur length at 36 weeks' gestation was (0.07 cm, 95% CI 0.01-0.14 cm, p = 0.019) and this was was statistically significant, however the magnitude of effect was small. Differences between treatment groups for all other fetal biometry measures, z-scores, estimated fetal weight, and adiposity measures at 28 and 36 weeks' gestation were similar. CONCLUSIONS The addition of metformin to dietary and lifestyle advice in pregnancy for overweight and obese women has no clinically relevant effect on ultrasound measures of fetal biometry or adiposity. TRIAL REGISTRATION Australian and New Zealand Clinical Trials Registry ( ACTRN12612001277831 ).
Collapse
Affiliation(s)
- Amanda J Poprzeczny
- The University of Adelaide, The Robinson Research Institute, and Discipline of Obstetrics and Gynaecology, Adelaide, South Australia, Australia.
- Department of Perinatal Medicine, The Women's and Children's Hospital, Women's and Babies Division, Adelaide, South Australia, Australia.
- The University of Adelaide, Women's and Children's Hospital, 72 King William Road, North Adelaide, South Australia, 5006, Australia.
| | - Jennie Louise
- The University of Adelaide, The Robinson Research Institute, and Discipline of Obstetrics and Gynaecology, Adelaide, South Australia, Australia
- The University of Adelaide, School of Public Health, Adelaide, South Australia, Australia
| | - Andrea R Deussen
- The University of Adelaide, The Robinson Research Institute, and Discipline of Obstetrics and Gynaecology, Adelaide, South Australia, Australia
| | - Jodie M Dodd
- The University of Adelaide, The Robinson Research Institute, and Discipline of Obstetrics and Gynaecology, Adelaide, South Australia, Australia
- Department of Perinatal Medicine, The Women's and Children's Hospital, Women's and Babies Division, Adelaide, South Australia, Australia
| |
Collapse
|
19
|
Gonzalez CD, Alvariñas J, Bagnes MFG, Di Girolamo G. Metformin and Pregnancy Outcomes: Evidence Gaps and Unanswered Questions. ACTA ACUST UNITED AC 2020; 14:54-60. [PMID: 30585549 DOI: 10.2174/1574884714666181224151116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/26/2018] [Accepted: 12/14/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Metformin is sometimes used as an alternative to insulin in gestational diabetes mellitus (GDM). It is also used to achieve ovulation in polycystic ovary syndrome (PCOS). Pre-natal exposure to metformin results from its continuation after a successful ovulation in women with PCOS, its maintenance in women with pre-gestational diabetes or the installation of metformin in GDM. Little is known about the potential consequences of metformin exposure on pregnancy outcomes and offspring development. The aim of this review is to summarize the metformin effects on pregnancy outcomes and offspring development. Gaps in the available evidence and unanswered questions are also discussed. METHODS A comprehensive literature search was carried out to identify eligible studies from MEDLINE/PubMed, EMBASE and SCIELO databases through 1995 first semester. RESULTS Several factors limit the effect of metformin on embryos. In contrast, placental transport of metformin is effective allowing for a higher fetal exposure; the impact of this finding remains unclear. It seems that the interruption of metformin after a pregnancy diagnosis in women with PCOS is not associated with a higher miscarriage risk and it continuation does not seem to impair the maternal metabolic prognosis or prevent emerging GDM. CONCLUSIONS It seems to have no sense to prolong the use of metformin after a pregnancy diagnosis in women with PCOS. Patients with GDM may be treated with metformin under on judicious basis, and a careful attachment to clinical guidelines and regulations is recommended. The long-term effects of pre-natal exposure to metformin on the offspring remain uncertain.
Collapse
Affiliation(s)
- Claudio D Gonzalez
- Universidad de Buenos Aires, Facultad de Medicina, Centro de Vigilancia y Seguridad de Medicamentos, Departamento de Toxicologia y Farmacologia, Buenos Aires, Argentina.,Departamento de Farmacologia, Instituto Universitario CEMIC, Buenos Aires, Argentina
| | - Jorge Alvariñas
- Sociedad Argentina de Diabetes, Comite de Farmacologia, Buenos Aires, Argentina
| | - Maria F G Bagnes
- Departamento de Farmacologia, Instituto Universitario CEMIC, Buenos Aires, Argentina
| | - Guillermo Di Girolamo
- Universidad de Buenos Aires, Facultad de Medicina, Centro de Vigilancia y Seguridad de Medicamentos, Departamento de Toxicologia y Farmacologia, Buenos Aires, Argentina.,Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini", Buenos Aires, Argentina.,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Toxicologia y Farmacologia, Tercera Cátedra de Farmacología, Buenos Aires, Argentina
| |
Collapse
|
20
|
Polycystic Ovary Syndrome: Impact of Lipotoxicity on Metabolic and Reproductive Health. Obstet Gynecol Surv 2020; 74:223-231. [PMID: 31344250 DOI: 10.1097/ogx.0000000000000661] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Importance Polycystic ovary syndrome (PCOS) is the most common endocrinopathy of reproductive-aged women. Women with PCOS are at increased risk of developing several metabolic and reproductive abnormalities, including metabolic syndrome. Underlying the combined metabolic and reproductive dysfunction is lipotoxicity, defined as the ectopic deposition of lipid in nonadipose tissue where it induces oxidative stress linked with insulin resistance and inflammation. Objective To examine what metabolic components underlie insulin resistance in PCOS, how lipotoxicity through insulin resistance impairs metabolism and reproduction in these women, and why evidence-based, individualized management is essential for their care. Evidence Acquisition PubMed search was performed using relevant terms to identify journal articles related to the subject. Relevant textbook chapters were also used. Results Polycystic ovary syndrome by Rotterdam criteria represents a complex syndrome of heterogeneous expression with variable adverse metabolic and reproductive implications. Women with classic PCOS are often insulin resistant and at greatest risk of developing metabolic syndrome with preferential fat accumulation and weight gain. Moreover, PCOS women may also have an altered capacity to properly store fat, causing ectopic lipid accumulation in nonadipose tissue, including the ovaries, where it can perpetuate insulin resistance and inflammation and harm the oocyte. Conclusions and Relevance A personalized approach to managing PCOS is essential to improve the health of all PCOS women through cost-effective prevention and/or treatment, to minimize the risk of pregnancy complications in those individuals wishing to conceive, and to optimize the long-term health of PCOS women and their offspring.
Collapse
|
21
|
Greger HK, Hanem LGE, Østgård HF, Vanky E. Cognitive function in metformin exposed children, born to mothers with PCOS - follow-up of an RCT. BMC Pediatr 2020; 20:60. [PMID: 32039724 PMCID: PMC7008569 DOI: 10.1186/s12887-020-1960-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/05/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metformin is widely used in pregnancy to treat gestational diabetes mellitus and polycystic ovary syndrome (PCOS). Association between PCOS and developmental delay in offspring, and larger head circumference of metformin-exposed newborns has been reported. The objective of this study was to explore whether metformin exposure in utero had any effect on offspring cognitive function. METHOD The current study is a follow-up of two randomized, placebo-controlled studies which were conducted at 11 public hospitals in Norway In the baseline studies (conducted in 2000-2003, and 2005-2009), participants were randomized to metformin 1700 and 2000 mg/d or placebo from first trimester to delivery. There was no intervention in the current study. We invited parents of 292 children to give permission for their children to participate; 93 children were included (mean age 7.7 years). The follow-up study was conducted in 2014-2016. The Wechsler Preschool and Primary Scale of Intelligence version III and the Wechsler Intelligence Scale for Children version IV were applied for cognitive assessment. Androstenedione and testosterone were measured in maternal blood samples at four time-points in pregnancy. RESULTS We found no difference in mean, full scale IQ in metformin (100.0 (SD 13.2)) vs. placebo-exposed (100.9 (SD 10.1)) children. There was an association between metformin exposure in utero and borderline intellectual function of children (full scale IQ between 70 and 85). Free testosterone index in gestational week 19, and androstenedione in gestational week 36 correlated positively to full scale IQ. CONCLUSIONS We found no evidence of long-term effect of metformin on average child cognitive function. The increase of borderline intellectual functioning in metformin-exposed children must be interpreted with caution due to small sample size. TRIAL REGISTRATION The baseline study was registered on 12 September 2005 at the US National Institute of Health (ClinicalTrials.gov) # NCT00159536.
Collapse
Affiliation(s)
- Hanne Klæboe Greger
- Departement of Mental Health, Faculty of Medicine and Health Sciences, NTNU -Norwegian University of Science and Technology, Trondheim, Norway. .,St.Olavs Hospital, Trondheim, Norway.
| | - Liv Guro Engen Hanem
- St.Olavs Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU -Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Eszter Vanky
- St.Olavs Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU -Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
22
|
Abbott DH, Rogers J, Dumesic DA, Levine JE. Naturally Occurring and Experimentally Induced Rhesus Macaque Models for Polycystic Ovary Syndrome: Translational Gateways to Clinical Application. Med Sci (Basel) 2019; 7:medsci7120107. [PMID: 31783681 PMCID: PMC6950671 DOI: 10.3390/medsci7120107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/16/2019] [Accepted: 11/16/2019] [Indexed: 12/19/2022] Open
Abstract
Indian rhesus macaque nonhuman primate models for polycystic ovary syndrome (PCOS) implicate both female hyperandrogenism and developmental molecular origins as core components of PCOS etiopathogenesis. Establishing and exploiting macaque models for translational impact into the clinic, however, has required multi-year, integrated basic-clinical science collaborations. Paradigm shifting insight has accrued from such concerted investment, leading to novel mechanistic understanding of PCOS, including hyperandrogenic fetal and peripubertal origins, epigenetic programming, altered neural function, defective oocytes and embryos, adipogenic constraint enhancing progression to insulin resistance, pancreatic decompensation and type 2 diabetes, together with placental compromise, all contributing to transgenerational transmission of traits likely to manifest in adult PCOS phenotypes. Our recent demonstration of PCOS-related traits in naturally hyperandrogenic (High T) female macaques additionally creates opportunities to employ whole genome sequencing to enable exploration of gene variants within human PCOS candidate genes contributing to PCOS-related traits in macaque models. This review will therefore consider Indian macaque model contributions to various aspects of PCOS-related pathophysiology, as well as the benefits of using macaque models with compellingly close homologies to the human genome, phenotype, development and aging.
Collapse
Affiliation(s)
- David H. Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
- Correspondence: ; Tel.: +1-608-698-1953
| | - Jeffrey Rogers
- Department of Molecular and Human Genetics and Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Jon E. Levine
- Department of Neuroscience, Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA;
| |
Collapse
|
23
|
Prentice PM, Olga L, Petry CJ, Simmons D, Murphy HR, Hughes IA, Acerini CL, Ong KK, Dunger DB. Reduced size at birth and persisting reductions in adiposity in recent, compared with earlier, cohorts of infants born to mothers with gestational diabetes mellitus. Diabetologia 2019; 62:1977-1987. [PMID: 31396660 PMCID: PMC6805804 DOI: 10.1007/s00125-019-4970-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS This study aimed to explore the infancy growth trajectories of 'recent' and 'earlier' offspring of mothers with gestational diabetes mellitus (OGDM), each compared with the same control infants, and investigate whether 'recent' OGDM still exhibit a classical phenotype, with macrosomia and increased adiposity. METHODS Within a prospective observational birth cohort, 98 'earlier' OGDM born between 2001 and 2009 were identified using 75 g oral glucose tolerance testing at 28 weeks gestation, 122 recent OGDM born between 2011 and 2013 were recruited postnatally through antenatal diabetes clinics, and 876 normal birthweight infants of mothers with no history of diabetes were recruited across the full study period as the control group. All infants followed the same study protocol (measurements at birth, 3, 12 and 24 months, including weight, length and skinfold thickness indicating adiposity, and detailed demographic data). In all cases, GDM was defined using the International Association of Diabetes and Pregnancy Study Group criteria. RESULTS Earlier OGDM had higher birthweight SD scores (SDS) than control infants. Conversely, recent OGDM had similar birthweight- and length SDS to control infants (mean ± SD, 0.1 ± 1.0 and- 0.1 ± 0.9, respectively), but lower mean skinfold thickness SDS (-0.4 ± 0.6 vs 0.0 ± 0.9; p < 0.001). After birth, earlier OGDM showed reduced gains in weight and length between 3 and 12 months. In contrast, recent OGDM had increased weight and skinfold thickness gains until 3 months, followed by reduced gains in those variables from 3 to 12 months, compared with control infants. At 24 months, recent OGDM had lower adiposity than control infants (mean skinfold thickness SDS -0.3 ± 0.7 vs 0.0 ± 0.8; p < 0.001). At all time points recent OGDM had lower growth measurements than earlier OGDM. CONCLUSIONS/INTERPRETATION Recent OGDM showed different growth trajectories to the earlier group, namely normalisation of birthweight and reduced adiposity at birth, followed by initial rapid weight gain but subsequent reduced adiposity postnatally. While avoidance of macrosomia at birth may be advantageous, the longer-term health implications of these changing growth trajectories are uncertain.
Collapse
Affiliation(s)
- Philippa M Prentice
- Department of Paediatrics, University of Cambridge, Box 116, Level 8, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Department of Paediatrics, North Middlesex University Hospital NHS Trust, London, UK
| | - Laurentya Olga
- Department of Paediatrics, University of Cambridge, Box 116, Level 8, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Clive J Petry
- Department of Paediatrics, University of Cambridge, Box 116, Level 8, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - David Simmons
- Wolfson Diabetes and Endocrine Clinic, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Macarthur Clinical School, Western Sydney University, Sydney, NSW, Australia
| | - Helen R Murphy
- Wolfson Diabetes and Endocrine Clinic, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Diabetes and Endocrinology, Norfolk and Norwich University Hospital, Norwich, UK
- Women's Health Academic Centre, Division of Women's and Children's Health, King's College London, London, UK
| | - Ieuan A Hughes
- Department of Paediatrics, University of Cambridge, Box 116, Level 8, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Carlo L Acerini
- Department of Paediatrics, University of Cambridge, Box 116, Level 8, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Ken K Ong
- Department of Paediatrics, University of Cambridge, Box 116, Level 8, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, UK
- Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Box 116, Level 8, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
- Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
24
|
Vanky E, Engen Hanem LG, Abbott DH. Children born to women with polycystic ovary syndrome-short- and long-term impacts on health and development. Fertil Steril 2019; 111:1065-1075. [PMID: 31056313 DOI: 10.1016/j.fertnstert.2019.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/30/2022]
Abstract
Maternal PCOS status may negatively influence offspring infant and childhood growth, cardiometabolic health, reproductive health, and neurodevelopment. Current findings across studies are divergent, often because of small numbers of subjects, as well as heterogeneous selection criteria, ethnicities, and definitions of control groups. Coexisting maternal obesity, pregnancy complications, and comorbidity make it difficult to identify the contribution of maternal PCOS. Large, prospective, international, multiethnic studies with standardized investigation protocols and questionnaires on PCOS offspring health and development are needed.
Collapse
Affiliation(s)
- Eszter Vanky
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynecology and Obstetrics, St. Olav's Hospital, Trondheim, Norway.
| | - Liv Guro Engen Hanem
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - David H Abbott
- Department of Obstetrics and Gynecology and Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
25
|
Løvvik TS, Carlsen SM, Salvesen Ø, Steffensen B, Bixo M, Gómez-Real F, Lønnebotn M, Hestvold KV, Zabielska R, Hirschberg AL, Trouva A, Thorarinsdottir S, Hjelle S, Berg AH, Andræ F, Poromaa IS, Mohlin J, Underdal M, Vanky E. Use of metformin to treat pregnant women with polycystic ovary syndrome (PregMet2): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2019; 7:256-266. [PMID: 30792154 DOI: 10.1016/s2213-8587(19)30002-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/15/2018] [Accepted: 01/02/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND Women with polycystic ovary syndrome (PCOS) have an increased risk of pregnancy complications. Epi-analysis of two previous randomised controlled trials that compared metformin with placebo during pregnancy in women with PCOS showed a significant reduction in late miscarriages and preterm births in the metformin group. The aim of this third randomised trial (PregMet2) was to test the hypothesis that metformin prevents late miscarriage and preterm birth in women with PCOS. METHODS PregMet2 was a randomised, placebo-controlled, double-blind, multicentre trial done at 14 hospitals in Norway, Sweden, and Iceland. Singleton pregnant women with PCOS aged 18-45 years were eligible for inclusion. After receiving information about the study at their first antenatal visit or from the internet, women signed up individually to participate in the study. Participants were randomly assigned (1:1) to receive metformin or placebo by computer-generated random numbers. Randomisation was in blocks of ten for each country and centre; the first block had a random size between one and ten to assure masking. Participants were assigned to receive oral metformin 500 mg twice daily or placebo during the first week of treatment, which increased to 1000 mg twice daily or placebo from week 2 until delivery. Placebo tablets and metformin tablets were identical and participants and study personnel were masked to treatment allocation. The primary outcome was the composite incidence of late miscarriage (between week 13 and week 22 and 6 days) and preterm birth (between week 23 and week 36 and 6 days), analysed in the intention-to-treat population. Secondary endpoints included the incidence of gestational diabetes, preeclampsia, pregnancy-induced hypertension, and admission of the neonate to the neonatal intensive care unit. We also did a post-hoc individual participant data analysis of pregnancy outcomes, pooling data from the two previous trials with the present study. The study was registered with ClinicalTrials.gov, number NCT01587378, and EudraCT, number 2011-002203-15. FINDINGS The study took place between Oct 19, 2012, and Sept 1, 2017. We randomly assigned 487 women to metformin (n=244) or placebo (n=243). In the intention-to-treat analysis, our composite primary outcome of late miscarriage and preterm birth occurred in 12 (5%) of 238 women in the metformin group and 23 (10%) of 240 women in the placebo group (odds ratio [OR] 0·50, 95% CI 0·22-1·08; p=0·08). We found no significant differences for our secondary endpoints, including incidence of gestational diabetes (60 [25%] of 238 women in the metformin group vs 57 [24%] of 240 women in the placebo group; OR 1·09, 95% CI 0·69-1·66; p=0·75). We noted no substantial between-group differences in serious adverse events in either mothers or offspring, and no serious adverse events were considered drug-related by principal investigators. In the post-hoc pooled analysis of individual participant data from the present trial and two previous trials, 18 (5%) of 397 women had late miscarriage or preterm delivery in the metformin group compared with 40 (10%) of 399 women in the placebo group (OR 0·43, 95% CI 0·23-0·79; p=0·004). INTERPRETATION In pregnant women with PCOS, metformin treatment from the late first trimester until delivery might reduce the risk of late miscarriage and preterm birth, but does not prevent gestational diabetes. FUNDING Research Council of Norway, Novo Nordisk Foundation, St Olav's University Hospital, and Norwegian University of Science and Technology.
Collapse
Affiliation(s)
- Tone S Løvvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynaecology and Obstetrics, St Olav's University Hospital, Trondheim, Norway.
| | - Sven M Carlsen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Endocrinology, St Olav's University Hospital, Trondheim, Norway
| | - Øyvind Salvesen
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Berglind Steffensen
- Department of Obstetrics and Gynaecology, University Hospital of Iceland, Reykjavik, Iceland
| | - Marie Bixo
- Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Francisco Gómez-Real
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marianne Lønnebotn
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
| | | | | | - Angelica L Hirschberg
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Anastasia Trouva
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden; Centre for Clinical Research Sörmland, Uppsala University, Uppsala, Sweden
| | | | | | - Ann Hilde Berg
- Women's Clinic, Innlandet Hospital Trust, Lillehammer, Norway
| | - Frida Andræ
- Women's Clinic, Nordlands Hospital Trust, Bodø, Norway
| | - Inger S Poromaa
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Johanna Mohlin
- Department of Clinical Sciences, Umeå University, Umeå, Sweden; Södersjukhuset, Stockholm, Sweden
| | - Maria Underdal
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynaecology and Obstetrics, St Olav's University Hospital, Trondheim, Norway
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynaecology and Obstetrics, St Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
26
|
Abbott DH, Dumesic DA, Levine JE. Hyperandrogenic origins of polycystic ovary syndrome - implications for pathophysiology and therapy. Expert Rev Endocrinol Metab 2019; 14:131-143. [PMID: 30767580 PMCID: PMC6992448 DOI: 10.1080/17446651.2019.1576522] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) diagnosis comprises combinations of female hyperandrogenism, menstrual irregularity and polycystic ovaries. While it is a familial and highly prevalent endocrine disorder, progress towards a cure is hindered by absence of a definitive pathogenic mechanism and lack of an animal model of naturally occurring PCOS. AREAS COVERED These include an overview of PCOS and its potential etiology, and an examination of insights gained into its pathogenic origins. Animal models derived from experimentally-induced hyperandrogenism during gestation, or from naturally-occurring PCOS-like traits, most reliably demonstrate reproductive, neuroendocrine and metabolic pathogenesis. EXPERT OPINION Genetic studies, while identifying at least 17 PCOS risk genes, account for <10% of women with PCOS. A number of PCOS risk genes involve regulation of gonadotropin secretion or action, suggesting a reproductive neuroendocrine basis for PCOS pathogenesis. Consistent with this notion, a number of animal models employing fetal androgen excess demonstrate epigenetic induction of PCOS-like traits, including reproductive neuroendocrine and metabolic dysfunction. Monkey models are most comprehensive, while mouse models provide molecular insight, including identifying the androgen receptor, particularly in neurons, as mediating androgen-induced PCOS-like programming. Naturally-occurring female hyperandrogenism is also demonstrated in monkeys. Animal models are poised to delineate molecular gateways to PCOS pathogenesis.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jon E Levine
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
27
|
Hanem LGE, Salvesen Ø, Juliusson PB, Carlsen SM, Nossum MCF, Vaage MØ, Ødegård R, Vanky E. Intrauterine metformin exposure and offspring cardiometabolic risk factors (PedMet study): a 5-10 year follow-up of the PregMet randomised controlled trial. THE LANCET CHILD & ADOLESCENT HEALTH 2019; 3:166-174. [PMID: 30704873 DOI: 10.1016/s2352-4642(18)30385-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Metformin is increasingly used to treat gestational diabetes and type 2 diabetes in pregnancy, and in attempts to improve pregnancy outcomes in polycystic ovary syndrome and obesity. It passes across the placenta with possible long-term consequences for the offspring. We previously explored the effect of metformin, given to women with polycystic ovary syndrome during pregnancy, on children's growth up to 4 years of age. In this 5-10 year follow-up, we examined the cardiometabolic risk factors in these children. METHODS This is a follow-up of children from the PregMet study, a double-blind, randomised controlled trial comparing metformin with placebo in polycystic ovary syndrome pregnancies. In the PregMet study, between Feb 4, 2005, and Jan 27, 2009, 257 pregnant women aged 18-45 years with polycystic ovary syndrome according to the Rotterdam criteria were included with 274 singleton pregnancies at 5-12 weeks of gestation at 11 study centres in Norway. 17 women participated twice. Pregnant women were randomised to metformin (2000 mg/day) or placebo from inclusion in the first trimester to birth. Randomisation was stratified according to metformin use at conception. In this follow-up, the primary endpoint was body-mass index (BMI) in the offspring at 5-10 years of age assessed by the standard deviation score (Z score). The primary endpoint was analysed with independent sample t tests. ClinicalTrials.gov number NCT00159536. FINDINGS Of the 255 invited children from the PregMet study, 141 (55%) consented to participate and were included between April 29, 2014, and July 12, 2016. Maternal baseline characteristics in the first trimester were similar between groups. Children in the metformin group had a higher BMI Z score than those in the placebo group (difference in means=0·41, 95% CI 0·03-0·78, p=0·03). INTERPRETATION The increased BMI in metformin-exposed children might indicate a potential risk of inferior cardiometabolic health. Implications for adult health cannot be excluded. FUNDING The Research Council of Norway, Novo Nordisk Foundation, St Olavs University Hospital, and the Norwegian University of Science and Technology.
Collapse
Affiliation(s)
- Liv Guro Engen Hanem
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Øyvind Salvesen
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Petur B Juliusson
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Registries, Norwegian Institute of Public Health, Sentrum, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Sven M Carlsen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Torgarden, Trondheim, Norway
| | - Marit Cecilie Fonn Nossum
- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marte Øye Vaage
- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rønnaug Ødegård
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Centre for Obesity Research, St. Olavs Hospital, Trondheim University Hospital, Torgarden, Trondheim, Norway
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Obstetrics and Gynecology, St. Olavs Hospital, Trondheim University Hospital, Torgarden, Trondheim, Norway
| |
Collapse
|
28
|
Faure M, Bertoldo MJ, Khoueiry R, Bongrani A, Brion F, Giulivi C, Dupont J, Froment P. Metformin in Reproductive Biology. Front Endocrinol (Lausanne) 2018; 9:675. [PMID: 30524372 PMCID: PMC6262031 DOI: 10.3389/fendo.2018.00675] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Initially produced in Europe in 1958, metformin is still one of the most widely prescribed drugs to treat type II diabetes and other comorbidities associated with insulin resistance. Metformin has been shown to improve fertility outcomes in females with insulin resistance associated with polycystic ovary syndrome (PCOS) and in obese males with reduced fertility. Metformin treatment reinstates menstrual cyclicity, decreases the incidence of cesareans, and limits the number of premature births. Notably, metformin reduces steroid levels in conditions associated with hyperandrogenism (e.g., PCOS and precocious puberty) in females and improves fertility of adult men with metabolic syndrome through increased testosterone production. While the therapeutical use of metformin is considered to be safe, in the last 10 years some epidemiological studies have described phenotypic differences after prenatal exposure to metformin. The goals of this review are to briefly summarize the current knowledge on metformin focusing on its effects on the female and male reproductive organs, safety concerns, including the potential for modulating fetal imprinting via epigenetics.
Collapse
Affiliation(s)
- Melanie Faure
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| | - Michael J Bertoldo
- Discipline of Obstetrics and Gynaecology, School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Rita Khoueiry
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Alice Bongrani
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| | - François Brion
- INERIS, Direction des Risques Chroniques, Pole VIVA, Unite d'ecotoxicologie in vitro et in vivo, BP2, Verneuil-en-Halatte, France
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Medical Investigations of Neurodevelopmental Disorders Institute, University of California, Davis, Davis, CA, United States
| | - Joelle Dupont
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| | - Pascal Froment
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| |
Collapse
|
29
|
Prenatal metformin exposure or organic cation transporter 3 knock-out curbs social interaction preference in male mice. Pharmacol Res 2018; 140:21-32. [PMID: 30423430 DOI: 10.1016/j.phrs.2018.11.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 09/21/2018] [Accepted: 11/07/2018] [Indexed: 12/15/2022]
Abstract
Poorly managed gestational diabetes can lead to severe complications for mother and child including fetal overgrowth, neonatal hypoglycemia and increased autism risk. Use of metformin to control it is relatively new and promising. Yet safety concerns regarding gestational metformin use remain, as its long-term effects in offspring are unclear. In light of beneficial findings with metformin for adult mouse social behavior, we hypothesized gestational metformin treatment might also promote offspring sociability. To test this, metformin was administered to non-diabetic, lean C57BL/6 J female mice at mating, with treatment discontinued at birth or wean. Male offspring exposed to metformin through birth lost social interaction preference relative to controls by time in chambers, but not by sniffing measures. Further, prenatal metformin exposure appeared to enhance social novelty preference only in females. However due to unbalanced litters and lack of statistical power, firm establishment of any sex-dependency of metformin's effects on sociability was not possible. Since organic cation transporter 3 (OCT3) transports metformin and is dense in placenta, social preferences of OCT3 knock-out males were measured. Relative to wild-type, OCT3 knock-outs had reduced interaction preference. Our data indicate gestational metformin exposure under non-diabetic conditions, or lack of OCT3, can impair social behavior in male C57BL6/J mice. Since OCT3 transports serotonin and tryptophan, impaired placental OCT3 function is one common mechanism that could persistently impact central serotonin systems and social behavior. Yet no gross alterations in serotonergic function were evident by measure of serotonin transporter density in OCT3, or serotonin turnover in metformin-exposed offspring brains. Mechanisms underlying the behavioral outcomes, and if with gestational diabetes the same would occur, remain unclear. Metformin's impacts on placental transporters and serotonin metabolism or AMPK activity in fetal brain need further investigation to clarify benefits and risks to offspring sociability from use of metformin to treat gestational diabetes.
Collapse
|
30
|
Palomba S, Falbo A, Daolio J, Battaglia FA, La Sala GB. Pregnancy complications in infertile patients with polycystic ovary syndrome: updated evidence. MINERVA GINECOLOGICA 2018; 70. [DOI: 10.23736/s0026-4784.18.04230-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
31
|
Andersen M, Glintborg D. Diagnosis and follow-up of type 2 diabetes in women with PCOS: a role for OGTT? Eur J Endocrinol 2018; 179:D1-D14. [PMID: 29921567 DOI: 10.1530/eje-18-0237] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is common in premenopausal women. The majority of women with PCOS have insulin resistance and the risk of type 2 diabetes mellitus (T2D) is higher in women with PCOS compared to controls. In non-pregnant women with PCOS, glycemic status may be assessed by oral glucose tolerance test (OGTT), fasting plasma glucose (FPG) or HbA1c. OGTT has been reckoned gold standard test for diagnosing T2D, but OGTT is rarely used for diagnostic purpose in other non-pregnant individuals at risk of T2D, apart from PCOS. OGTT has questionable reproducibility, and high sensitivity of the 2-h glucose value is at the expense of relatively low specificity, especially regarding impaired glucose tolerance (IGT). Furthermore, lean women with PCOS are rarely diagnosed with T2D and only few percent of normal-weight women have prediabetes. Glycemic status is necessary at diagnosis and during follow-up of PCOS, especially in women with high risk of T2D (obesity, previous gestational diabetes (GDM)). We suggest that OGTT should be used in the same situations in PCOS as in other patient groups at risk of T2D. OGTT is indicated for diagnosing GDM; however, OGTT during pregnancy may not be indicated in lean women with PCOS without other risk factors for GDM.
Collapse
Affiliation(s)
- Marianne Andersen
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Dorte Glintborg
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| |
Collapse
|