1
|
Zhang Q, Yang D, Han X, Ren Y, Fan Y, Zhang C, Sun L, Ye T, Wang Q, Ban Y, Cao Y, Zou H, Zhang Z. Alarmins and their pivotal role in the pathogenesis of spontaneous abortion: insights for therapeutic intervention. Eur J Med Res 2024; 29:640. [PMID: 39741354 DOI: 10.1186/s40001-024-02236-1] [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: 09/06/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
Alarmins are a class of molecules released when affected cells damaged or undergo apoptosis. They contain various chemotactic and immunomodulatory proteins or peptides. These molecules regulate the immune response by interacting with pattern recognition receptors (PRRs) and play important roles in inflammatory response, tissue repair, infection defense, and cancer treatment. Spontaneous abortion (SA) is a common pregnancy-related disease, and its pathogenesis has been puzzling clinicians, so it needs to be further studied. In this paper, we first reviewed the research status of various alarmins and SA, focusing on the role of high mobility box 1 (HMGB1), interleukin33 (IL-33), interleukin1β (IL-1β) and S-100 protein (S100 protein) in immune response, inflammation, embryonic development and abortion. Subsequently, this paper summarized the effect of alarmins on pregnancy outcome by influencing angiogenesis-related factors. Finally, from the perspective of aseptic inflammation, the pro-inflammatory signaling pathways involved in various alarmins and their targeted drugs were reviewed. By focusing on specific molecules in alarmins and their receptors and signaling pathways, we can more accurately conduct drug research and development. The purpose of this review is to explore the role of alarmins in SA, and provide important references for early detection of abortion risk, revealing the disease mechanism, developing new therapies and improving the prognosis of patients.
Collapse
Affiliation(s)
- Qiqi Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China
| | - Dandan Yang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China
| | - Xingxing Han
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Yu Ren
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, Anhui, China
| | - Yongqi Fan
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Chao Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, China
| | - Lei Sun
- Department of Clinical Medical, The First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Tingting Ye
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Qiushuang Wang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Youhao Ban
- Hefei Anhua Trauma Rehabilitation Hospital, Hefei, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Huijuan Zou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China.
| | - Zhiguo Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China.
| |
Collapse
|
2
|
Moka MK, Sriram DK, George M. Recent Advances in Individualized Clinical Strategies for Polycystic Ovary Syndrome: Evidence From Clinical Trials and Emerging Pharmacotherapies. Clin Ther 2024:S0149-2918(24)00360-6. [PMID: 39709252 DOI: 10.1016/j.clinthera.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE Clinical trials are advancing the treatment of polycystic ovary syndrome (PCOS), an endocrine disorder affecting 8-13% of women. Lifestyle interventions, including nutritional plans, physical activity, and stress management, can improve reproductive hormones and metabolic health. Novel pharmacotherapies targeting hormonal, metabolic, and reproductive abnormalities are being explored for individualized treatment. Combination therapies and lifestyle interventions like acupuncture, high-intensity interval training, and vitamin D3 supplementation are also being explored. METHODS We conducted a narrative review by searching English-language studies across electronic databases such as PubMed, Science direct, and Google Scholar for articles related to the topics of PCOS and novel drug therapies such as metformin, LIK-066, elagolix, saxenda, exenatide, clomiphene, letrozole, and other diagnostic interventions. Our review excluded preclinical studies and articles not in english. FINDINGS In addition to pharmacological treatments, lifestyle interventions such as Tung's acupuncture, high-intensity interval training (HIIT), and vitamin D3 supplementation have proven effective in managing symptoms of PCOS and enhancing overall health outcomes. These interventions offer a complementary approach to traditional medical therapies, emphasizing the importance of integrating lifestyle modifications into the treatment plan for women with PCOS. IMPLICATIONS This comprehensive approach underscores the importance of tailored treatments in optimizing clinical outcomes and quality of life for women with PCOS. The aim of this review is to highlight recent advancements in the treatment of PCOS through clinical trials and emerging pharmacotherapies, emphasizing the need for individualized and multifaceted treatment approaches.
Collapse
Affiliation(s)
- Murali Krishna Moka
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, Tamil Nadu, India
| | - Damal Kandadai Sriram
- Department of Diabetology and Endocrinology, Hindu Mission Hospital, Tambaram, Chennai, Tamil Nadu, India
| | - Melvin George
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, Tamil Nadu, India.
| |
Collapse
|
3
|
Vachalova V, Kumnova F, Synova T, Anandam KY, Abad C, Karahoda R, Staud F. Metformin inhibits OCT3-mediated serotonin transport in the placenta. Biomed Pharmacother 2024; 179:117399. [PMID: 39243433 DOI: 10.1016/j.biopha.2024.117399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
Proper fetal development requires tight regulation of serotonin concentrations within the fetoplacental unit. This homeostasis is partly maintained by the placental transporter OCT3/SLC22A3, which takes up serotonin from the fetal circulation. Metformin, an antidiabetic drug commonly used to treat gestational diabetes mellitus, was shown to inhibit OCT3. We, therefore, hypothesized that its use during pregnancy could disrupt placental serotonin homeostasis. This hypothesis was tested using three experimental model systems: primary trophoblast cells isolated from the human term placenta, fresh villous human term placenta fragments, and rat term placenta perfusions. Inhibition of serotonin transport by metformin at three concentrations (1 μM, 10 μM, and 100 μM) was assessed in all three models. The OCT3 inhibitor decynium-22 (100 μM) and paroxetine (100 μM), a dual inhibitor of SERT and OCT3, were used as controls. In primary trophoblasts, paroxetine exhibited the strongest inhibition of serotonin uptake, followed by decynium-22. Metformin showed a concentration-dependent effect, reducing serotonin uptake by up to 57 % at the highest concentration. Its inhibitory effect was less pronounced in fresh villous fragments but remained statistically significant at all concentrations. In the perfused rat placenta, metformin demonstrated a concentration-dependent effect, reducing placental serotonin uptake by 44 % at the highest concentration tested. Our findings across all experimental models show inhibition of placental OCT3 by metformin, resulting in reduced serotonin uptake by the trophoblast. This sheds light on mechanisms that may underpin metformin-mediated effects on fetal development.
Collapse
Affiliation(s)
- Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Fiona Kumnova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tetiana Synova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Kasin Yadunandam Anandam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
4
|
Chung CS, Park SE, Hsiao JL, Lee KH. A Review of Hidradenitis Suppurativa in Special Populations: Considerations in Children, Pregnant and Breastfeeding Women, and the Elderly. Dermatol Ther (Heidelb) 2024; 14:2407-2425. [PMID: 39230800 PMCID: PMC11393272 DOI: 10.1007/s13555-024-01249-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic skin condition that significantly impacts patients' quality of life. HS is often challenging to treat. In this review, we discuss the unique characteristics of HS in four special populations: children, the elderly, pregnant individuals, and breastfeeding mothers. In children, diagnosis may be delayed due to atypical and early HS disease presentations. HS management plans must take into consideration the lack of rigorous efficacy and safety data of HS treatments in this population. However, it is important to weigh the risk of treatments against the risk of untreated HS and the morbidity and mortality risk that having HS confers. Pregnancy poses unique challenges for women with HS, with their condition possibly worsening during pregnancy and increased risk of fetal death. Management strategies during pregnancy must consider both maternal and fetal safety. Similarly, breastfeeding mothers require thoughtful medication selection to balance symptom management with infant safety. In the elderly, HS may present more severely and is often complicated by comorbidities. Treating HS in this population should safely accommodate patients' additional health conditions. Furthermore, this review highlights the overall paucity of primary literature addressing management in these populations, underscoring the need for further research to optimize HS care across all stages of life.
Collapse
Affiliation(s)
- Claire S Chung
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sarah E Park
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer L Hsiao
- Department of Dermatology, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, Ezralow Tower, Suite 5301, Los Angeles, CA, 90033-9174, USA
| | - Katrina H Lee
- Department of Dermatology, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, Ezralow Tower, Suite 5301, Los Angeles, CA, 90033-9174, USA.
| |
Collapse
|
5
|
Molin J, Domellöf M, Häggström C, Vanky E, Zamir I, Östlund E, Bixo M. Neonatal outcome following metformin-treated gestational diabetes mellitus: A population-based cohort study. Acta Obstet Gynecol Scand 2024; 103:992-1007. [PMID: 38288656 PMCID: PMC11019529 DOI: 10.1111/aogs.14787] [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: 10/10/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Neonatal hypoglycemia is a common complication associated with gestational diabetes and therefore relevant to consider in evaluations of maternal treatment. We aimed to investigate the risk of neonatal hypoglycemia in offspring exposed to metformin treatment alone (MT) or combined with insulin (MIT) in comparison with nutrition therapy alone (NT), and insulin treatment alone (IT). In addition, we investigated MT in comparison with MIT. Secondary outcomes included neonatal anthropometrics, respiratory morbidity, hyperbilirubinemia, 5-min Apgar score, and preterm birth. MATERIAL AND METHODS This Swedish population-based cohort included 16 181 women diagnosed with gestational diabetes, and their singleton offspring born in 2019-2021. We estimated risk as adjusted odds ratio (aOR) with 95% confidence interval (CI), using individual-level, linkage register-data in multivariable logistic regression models. RESULTS In the main analysis, MT was associated with a lower risk of neonatal hypoglycemia vs NT (aOR 0.85, 95% CI: 0.74-0.96), vs MIT (0.74 [0.64-0.87]), and vs IT (0.47 [0.40-0.55]), whereas MIT was associated with a similar risk of neonatal hypoglycemia vs NT (1.14 [0.99-1.30]) and with lower risk vs IT (0.63 [0.53-0.75]). However, supplemental feeding rates were lower for NT vs pharmacological treatments (p < 0.001). In post hoc subgroup analyses including only exclusively breastfed offspring, the risk of neonatal hypoglycemia was modified and similar among MT and NT, and higher in MIT vs NT. Insulin exposure, alone or combined with metformin, was associated with increased risk of being large for gestational age. Compared with NT, exposure to any pharmacological treatment was associated with significantly lower risk of 5-min Apgar score < 4. All other secondary outcomes were comparable among the treatment categories. CONCLUSIONS The risk of neonatal hypoglycemia appears to be comparable among offspring exposed to single metformin treatment and nutrition therapy alone, and the lower risk that we observed in favor of metformin is probably explained by a difference in supplemental feeding practices rather than metformin per se. By contrast, the lower risk favoring metformin exposure over insulin exposure was not explained by supplemental feeding. However, further investigations are required to determine whether the difference is an effect of metformin per se or mediated by other external factors.
Collapse
Affiliation(s)
- Johanna Molin
- Department of Clinical SciencesUmeå UniversityUmeåSweden
| | | | - Christel Häggström
- Northern Registry Center, Department of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health SciencesNorwegian University of Science and TechnologyTrondheimNorway
- Department of Obstetrics and GynecologySt. Olav's Hospital, Trondheim University HospitalTrondheimNorway
| | - Itay Zamir
- Department of Clinical SciencesUmeå UniversityUmeåSweden
| | - Eva Östlund
- Department of Clinical Sciences and EducationSödersjukhuset, Karolinska InstituteStockholmSweden
| | - Marie Bixo
- Department of Clinical SciencesUmeå UniversityUmeåSweden
| |
Collapse
|
6
|
Velazquez-Cervantes MA, López-Ortega O, Cruz-Holguín VJ, Herrera Moro-Huitron L, Flores-Pliego A, Lara-Hernandez I, Comas-García M, Villavicencio-Carrisoza O, Helguera-Reppeto AC, Arévalo-Romero H, Vázquez-Martínez ER, León-Juárez M. Metformin Inhibits Zika Virus Infection in Trophoblast Cell Line. Curr Microbiol 2024; 81:133. [PMID: 38592489 DOI: 10.1007/s00284-024-03651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/26/2024] [Indexed: 04/10/2024]
Abstract
Zika virus (ZIKV) infections have been associated with severe clinical outcomes, which may include neurological manifestations, especially in newborns with intrauterine infection. However, licensed vaccines and specific antiviral agents are not yet available. Therefore, a safe and low-cost therapy is required, especially for pregnant women. In this regard, metformin, an FDA-approved drug used to treat gestational diabetes, has previously exhibited an anti-ZIKA effect in vitro in HUVEC cells by activating AMPK. In this study, we evaluated metformin treatment during ZIKV infection in vitro in a JEG3-permissive trophoblast cell line. Our results demonstrate that metformin affects viral replication and protein synthesis and reverses cytoskeletal changes promoted by ZIKV infection. In addition, it reduces lipid droplet formation, which is associated with lipogenic activation of infection. Taken together, our results indicate that metformin has potential as an antiviral agent against ZIKV infection in vitro in trophoblast cells.
Collapse
Affiliation(s)
- Manuel Adrían Velazquez-Cervantes
- Laboratorio de Virología Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, 11000, Mexico City, Mexico
| | - Orestes López-Ortega
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, Institut Necker Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Victor Javier Cruz-Holguín
- Laboratorio de Virología Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, 11000, Mexico City, Mexico
| | - Luis Herrera Moro-Huitron
- Laboratorio de Virología Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, 11000, Mexico City, Mexico
| | - Arturo Flores-Pliego
- Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, 11000, Mexico City, Mexico
| | - Ignacio Lara-Hernandez
- Sección de Microscopía de Alta Resolución, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Atunóma de San Luis Potrosí, San Luis Potosí, SLP, Mexico
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, Mexico
| | - Mauricio Comas-García
- Sección de Microscopía de Alta Resolución, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Atunóma de San Luis Potrosí, San Luis Potosí, SLP, Mexico
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, Mexico
| | | | - Addy Cecilia Helguera-Reppeto
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, Institut Necker Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Haruki Arévalo-Romero
- Laboratorio de Inmunología y Microbiología Molecular, División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Méndez, Mexico
| | - Edgar Ricardo Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Universidad Nacional Autónoma de México, 11000, Mexico City, Mexico
| | - Moises León-Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, 11000, Mexico City, Mexico.
| |
Collapse
|
7
|
Du Y, Zhu J, Guo Z, Wang Z, Wang Y, Hu M, Zhang L, Yang Y, Wang J, Huang Y, Huang P, Chen M, Chen B, Yang C. Metformin adverse event profile: a pharmacovigilance study based on the FDA Adverse Event Reporting System (FAERS) from 2004 to 2022. Expert Rev Clin Pharmacol 2024; 17:189-201. [PMID: 38269492 DOI: 10.1080/17512433.2024.2306223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Metformin has the potential for treating numerous diseases, but there are still many unrecognized and unreported adverse events (AEs). METHODS We selected data from the United States FDA Adverse Event Reporting System (FAERS) database from the first quarter (Q1) of 2004 to the fourth quarter (Q4) of 2022 for disproportionality analysis to assess the association between metformin and related adverse events. RESULTS In this study 10,500,295 case reports were collected from the FAERS database, of which 56,674 adverse events related to metformin were reported. A total of 643 preferred terms (PTs) and 27 system organ classes (SOCs) that were significant disproportionality conforming to the four algorithms simultaneously were included. The SOCs included metabolic and nutritional disorders (p = 0.00E + 00), gastrointestinal disorders (p = 0.00E + 00) and others. PT levels were screened for adverse drug reaction (ADR) signals such as acute pancreatitis (p = 0.00E + 00), melas syndrome, pemphigoid (p = 0.00E + 00), skin eruption (p = 0.00E + 00) and drug exposure during pregnancy (p = 0.00E + 00). CONCLUSION Most of our results were consistent with the specification, but some new signals of adverse reactions such as acute pancreatitis were not included. Therefore, further studies are needed to validate unlabeled adverse reactions and provide important support for clinical monitoring and risk identification of metformin.
Collapse
Affiliation(s)
- Yikuan Du
- Central Laboratory, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, People's Republic of China
| | - Jinfeng Zhu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Zhuoming Guo
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Zhenjie Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Yuni Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Mianda Hu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Lingzhi Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Yurong Yang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Jinjin Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Yixing Huang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Peiying Huang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Mianhai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Bo Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Chun Yang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| |
Collapse
|
8
|
Mnatzaganian G, Taylor M, He F, Yuen N, McIntyre HD, Woodward M, Ma L, Huxley RR. Differences in neonatal adverse outcomes among women with gestational diabetes mellitus managed by diet or medication: a propensity score matched analysis of a population-based sample. Gynecol Endocrinol 2023; 39:2250005. [PMID: 37608764 DOI: 10.1080/09513590.2023.2250005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
OBJECTIVE Neonatal outcomes in women with and without medically managed gestational diabetes mellitus (GDM) were compared after accounting for differences in maternal baseline characteristics using a propensity score (PS) analysis. METHODS Women without preexisting diabetes, delivering singletons during 2010-2017 in a large hospital, were eligible for inclusion. Using nearest-neighbour PS matching, women with non-pharmacological managed GDM were matched with women whose GDM was medically managed. A conditional logistic regression consequently compared the neonatal adverse outcomes between the groups after adjusting for gestational age, induction of labor, birth type, and number of ultrasounds conducted during the pregnancy. RESULTS Of the overall 10028 births, GDM was diagnosed in 930 (9.3%), of whom 710 (76.3%) were successfully matched. The conditional regressions found higher risk of neonatal adverse outcomes in neonates of women with non-pharmacological managed GDM compared to neonates of women with medically managed GDM. These included a higher risk of hypoglycemia (odds ratio (OR) 1.56, 95% confidence interval (CI) 1.03-2.38, p = 0.037), hypothermia (OR 2.29, 95%CI 1.05-5.00, p = 0.037), and birth injuries (OR 3.50, 95%CI 1.62-7.58, p = 0.001), and a higher risk of being small for gestational age (OR 2.06, 95%CI 1.01-4.18, p = 0.046) and being admitted to a special care unit (OR 2.04, 95%CI 1.29-3.21, p = 0.002). CONCLUSIONS The increased neonatal morbidity associated with non-medicated GDM identified in our study may indicate that diet and lifestyle changes alone are not sufficient to achieve glycaemic control in some women with GDM. Our findings indicate that gestational diabetes management approach is independently associated with neonatal outcomes.
Collapse
Affiliation(s)
- George Mnatzaganian
- Rural Allied Health, La Trobe Rural Health School, La Trobe University, Bendigo, Victoria, Australia
| | - Marietta Taylor
- Rural Allied Health, La Trobe Rural Health School, La Trobe University, Bendigo, Victoria, Australia
| | - Fan He
- Rural Allied Health, La Trobe Rural Health School, La Trobe University, Bendigo, Victoria, Australia
| | - Nicola Yuen
- Department of Women's & Children's Services, Bendigo Health, Bendigo, Victoria, Australia
| | - H David McIntyre
- Obstetric Medicine, Mater Health Services, University of Queensland, Brisbane, Queensland, Australia
| | - Mark Woodward
- The George Institute for Global Health, Imperial College London, London, UK
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Liangkun Ma
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Rachel R Huxley
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
- Faculty of Health, Deakin University, Melbourne, Australia
| |
Collapse
|
9
|
Tosti G, Barberio A, Tartaglione L, Rizzi A, Di Leo M, Viti L, Sirico A, De Carolis S, Pontecorvi A, Lanzone A, Pitocco D. Lights and shadows on the use of metformin in pregnancy: from the preconception phase to breastfeeding and beyond. Front Endocrinol (Lausanne) 2023; 14:1176623. [PMID: 37409227 PMCID: PMC10319127 DOI: 10.3389/fendo.2023.1176623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
During pregnancy, the complex hormonal changes lead to a progressive decrease of insulin sensitivity that can drive the onset of gestational diabetes (GDM) or worsen an already-known condition of insulin resistance like type 2 diabetes, polycystic ovarian syndrome (PCOS), and obesity, with complications for the mother and the fetus. Metformin during pregnancy is proving to be safe in a growing number of studies, although it freely crosses the placenta, leading to a fetal level similar to maternal concentration. The aim of this literature review is to analyze the main available evidence on the use of metformin during, throughout, and beyond pregnancy, including fertilization, lactation, and medium-term effects on offspring. Analyzed studies support the safety and efficacy of metformin during pregnancy. In pregnant women with GDM and type 2 diabetes, metformin improves obstetric and perinatal outcomes. There is no evidence that it prevents GDM in women with pregestational insulin resistance or improves lipid profile and risk of GDM in pregnant women with PCOS or obesity. Metformin could have a role in reducing the risk of preeclampsia in pregnant women with severe obesity, the risk of late miscarriages and preterm delivery in women with PCOS, and the risk of ovarian hyperstimulation syndrome, increasing the clinical pregnancy rate in women with PCOS undergoing in vitro fertilization (IVF/FIVET). Offspring of mothers with GDM exposed to metformin have no significant differences in body composition compared with insulin treatment, while it appears to be protective for metabolic and cardiovascular risk.
Collapse
Affiliation(s)
- Giulia Tosti
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Annarita Barberio
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Linda Tartaglione
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Alessandro Rizzi
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Mauro Di Leo
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Luca Viti
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Angelo Sirico
- Catholic University School of Medicine, Rome, Italy
- Department of Woman and Child Health, Woman Health Area Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Sara De Carolis
- Catholic University School of Medicine, Rome, Italy
- Department of Woman and Child Health, Woman Health Area Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Alfredo Pontecorvi
- Catholic University School of Medicine, Rome, Italy
- Department of Endocrinology, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonio Lanzone
- Catholic University School of Medicine, Rome, Italy
- Department of Woman and Child Health, Woman Health Area Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Dario Pitocco
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| |
Collapse
|
10
|
Kattini R, Kelly L, Hummelen R. Systematic review of the use of metformin compared to insulin for the management of gestational diabetes: Implications for low-resource settings. CANADIAN JOURNAL OF RURAL MEDICINE 2023; 28:59-65. [PMID: 37005989 DOI: 10.4103/cjrm.cjrm_40_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Introduction This systematic review examines the effectiveness of metformin treatment compared to insulin treatment for gestational diabetes within the context of a low-resource environment. Methods Electronic data searches of Medline, EMBASE, Scopus and Google scholar databases from 1 January, 2005 to 30 June, 2021 were performed using medical subject headings: 'gestational diabetes or pregnancy diabetes mellitus' AND 'Pregnancy or pregnancy outcomes' AND 'Insulin' AND 'Metformin Hydrochloride Drug Combination/or Metformin/or Hypoglycemic Agents' AND 'Glycemic control or blood glucose'. Randomized controlled trials were included if: participants were pregnant women with gestational diabetes mellitus (GDM); the interventions were metformin and/or insulin. Studies among women with pre-gestational diabetes, non-randomised control trials or studies with a limited description of the methodology were excluded. Outcomes included adverse maternal outcomes: weight gain, C-section, pre-eclampsia and glycaemic control and adverse neonatal outcomes: birth weight, macrosomia, pre-term birth and neonatal hypoglycaemia. The revised Cochrane Risk of Bias Assessment for randomised trials was used for the evaluation of bias. Results We screened 164 abstracts and 36 full-text articles. Fourteen studies met the inclusion criteria. The studies provide moderate to high-quality evidence demonstrating the effectiveness of metformin as an alternative therapy to insulin. Risk of bias was low; multiple countries and robust sample sizes improved external validity. All studies were from urban centres with no rural data. Conclusion These recent high quality studies comparing metformin to insulin for the treatment of GDM generally found either improved or equivalent pregnancy outcome and good glycaemic control for most patients, although many required insulin supplementation. Its ease of use, safety and efficacy suggest metformin may simplify the management of gestational diabetes, particularly in rural and other low-resource environments.
Collapse
Affiliation(s)
- Ribal Kattini
- Sioux Lookout Local Education Group, Sioux Lookout, Ontario, Canada; Medical Student, Monash University, Melbourne, Australia
| | - Len Kelly
- Sioux Lookout Meno Ya Win Health Centre, Sioux Lookout, Ontario, Canada
| | - Ruben Hummelen
- Division of Clinical Sciences, Northern Ontario School of Medicine, Sioux Lookout, Ontario, Canada
| |
Collapse
|
11
|
Yakout SM, Hussein S, Al-Attas OS, Hussain SD, Saadawy GM, Al-Daghri NM. Hepatokines fetuin A and fetuin B status in women with/without gestational diabetes mellitus. Am J Transl Res 2023; 15:1291-1299. [PMID: 36915725 PMCID: PMC10006815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 01/03/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVES To assess circulating fetuin A and fetuin B levels in participants with and without Gestational Diabetes Mellitus (GDM) and to find out their correlations with other different parameters relating to gestational diabetes in Saudi women. METHODS A total of 123 Saudi pregnant women (N: 46 GDM and N: 77 healthy control) were included in this observational study. Fasting blood samples were collected to assess serum lipids, insulin and fetuin A and fetuin B. Serum fetuin A and fetuin B were quantified by commercially available kits. RESULTS The median value of fetuin A was slight lower in GDM patients [2003 pg/ml (866-3369)] than in the control group [2015 pg/ml (1060-2951)] without significant difference (P=0.95). The median value of fetuin B was also slight lower in GDM patients [3292 ng/ml (782-6740)] than the control group [3514 ng/ml (364-14854)] but without significant difference (P=0.564). There was a significant inverse correlation between fetuin B and total cholesterol in control group. CONCLUSIONS The present study did not find a significant association between fetuins A and B with GDM or insulin resistance, but there was a significant inverse correlation between fetuin B and total cholesterol in the control group, reflecting good glucose control and adequate use of lipids in the nutrition of the fetus. Further research is required in the future to understand fetuin's role in the progression of GDM in Saudi women.
Collapse
Affiliation(s)
- Sobhy M Yakout
- Department of Biochemistry, College of Science, King Saud University Riyadh 11451, Kingdom of Saudi Arabia
| | - Samira Hussein
- Department of Biochemistry, College of Science, King Saud University Riyadh 11451, Kingdom of Saudi Arabia
| | - Omar S Al-Attas
- Department of Biochemistry, College of Science, King Saud University Riyadh 11451, Kingdom of Saudi Arabia
| | - Syed D Hussain
- Department of Biochemistry, College of Science, King Saud University Riyadh 11451, Kingdom of Saudi Arabia
| | - Gamal M Saadawy
- Department of Biochemistry, College of Science, King Saud University Riyadh 11451, Kingdom of Saudi Arabia
| | - Nasser M Al-Daghri
- Department of Biochemistry, College of Science, King Saud University Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Bramante CT, Buse JB, Liebovitz D, Nicklas J, Puskarich MA, Cohen K, Belani H, Anderson B, Huling JD, Tignanelli C, Thompson J, Pullen M, Siegel L, Proper J, Odde DJ, Klatt N, Sherwood N, Lindberg S, Wirtz EL, Karger A, Beckman K, Erickson S, Fenno S, Hartman K, Rose M, Patel B, Griffiths G, Bhat N, Murray TA, Boulware DR. Outpatient treatment of Covid-19 with metformin, ivermectin, and fluvoxamine and the development of Long Covid over 10-month follow-up. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.12.21.22283753. [PMID: 36597543 PMCID: PMC9810227 DOI: 10.1101/2022.12.21.22283753] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Long Covid is an emerging chronic illness potentially affecting millions, sometimes preventing the ability to work or participate in normal daily activities. COVID-OUT was an investigator-initiated, multi-site, phase 3, randomized, quadruple-blinded placebo-controlled clinical trial (NCT04510194). The design simultaneously assessed three oral medications (metformin, ivermectin, fluvoxamine) using two by three parallel treatment factorial assignment to efficiently share placebo controls and assessed Long Covid outcomes for 10 months to understand whether early outpatient treatment of SARS-CoV-2 with metformin, ivermectin, or fluvoxamine prevents Long Covid. Methods This was a decentralized, remotely delivered trial in the US of 1,125 adults age 30 to 85 with overweight or obesity, fewer than 7 days of symptoms, and enrolled within three days of a documented SARS-CoV-2 infection. Immediate release metformin titrated over 6 days to 1,500mg per day 14 days total; ivermectin 430mcg/kg/day for 3 days; fluvoxamine, 50mg on day one then 50mg twice daily through 14 days. Medical-provider diagnosis of Long Covid, reported by participant by day 300 after randomization was a pre-specified secondary outcome; the primary outcome of the trial was severe Covid by day 14. Result The median age was 45 years (IQR 37 to 54), 56% female of whom 7% were pregnant. Two percent identified as Native American; 3.7% as Asian; 7.4% as Black/African American; 82.8% as white; and 12.7% as Hispanic/Latino. The median BMI was 29.8 kg/m2 (IQR 27 to 34); 51% had a BMI >30kg/m2. Overall, 8.4% reported having received a diagnosis of Long Covid from a medical provider: 6.3% in the metformin group and 10.6% in the metformin control; 8.0% in the ivermectin group and 8.1% in the ivermectin control; and 10.1% in the fluvoxamine group and 7.5% in the fluvoxamine control. The Hazard Ratio (HR) for Long Covid in the metformin group versus control was 0.58 (95% CI 0.38 to 0.88); 0.99 (95% CI 0.592 to 1.643) in the ivermectin group; and 1.36 in the fluvoxamine group (95% CI 0.785 to 2.385). Conclusions There was a 42% relative decrease in the incidence of Long Covid in the metformin group compared to its blinded control in a secondary outcome of this randomized phase 3 trial.
Collapse
Affiliation(s)
| | - John B Buse
- Endocrinology, University of North Carolina, Chapel Hill, ND
| | - David Liebovitz
- General Internal Medicine, Northwestern University, Chicago, IL
| | - Jacinda Nicklas
- General Internal Medicine, University of Colorado, Denver, CO
| | | | - Ken Cohen
- UnitedHealth Group, Optum Labs, Minnetonka, MN
| | - Hrishikesh Belani
- Department of Medicine, Olive View - University of California, Los Angeles, CA
| | - Blake Anderson
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia; Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Jared D Huling
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | | | - Jennifer Thompson
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew Pullen
- Infectious Diseases, University of Minnesota Medical School, Minneapolis, MN
| | - Lianne Siegel
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Jennifer Proper
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - David J Odde
- Department of Biomedical Engineering University of Minnesota, Minneapolis, MN
| | - Nichole Klatt
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, MN
| | - Nancy Sherwood
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sarah Lindberg
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Esteban Lemus Wirtz
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Amy Karger
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Kenny Beckman
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Spencer Erickson
- General Internal Medicine, University of Minnesota, Minneapolis, MN
| | - Sarah Fenno
- General Internal Medicine, University of Minnesota, Minneapolis, MN
| | - Katrina Hartman
- General Internal Medicine, University of Minnesota, Minneapolis, MN
| | - Michael Rose
- General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Barkha Patel
- General Internal Medicine, University of Minnesota, Minneapolis, MN
| | | | - Neeta Bhat
- General Internal Medicine, University of Minnesota, Minneapolis, MN
| | - Thomas A Murray
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - David R Boulware
- Infectious Diseases, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
13
|
Dodd S, Sominsky L, Siskind D, Bortolasci CC, Carvalho AF, Maes M, Walker AJ, Walder K, Yung AR, Williams LJ, Myles H, Watson T, Berk M. The role of metformin as a treatment for neuropsychiatric illness. Eur Neuropsychopharmacol 2022; 64:32-43. [PMID: 36191545 DOI: 10.1016/j.euroneuro.2022.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 12/12/2022]
Abstract
Advances in psychopharmacology have been significantly slower to evolve than in other disciplines of medicine and therefore investigation into novel therapeutic approaches is required. Additionally, concurrent metabolic conditions are prevalent among people with mental disorders. Metformin is a widely used hypoglycaemic agent that is now being studied for use beyond diabetes management. Evidence is emerging that metformin has multiple effects on diverse neurobiological pathways and consequently may be repurposed for treating mental illness. Metformin may have beneficial neuroimmunological, neuroplastic, neuro-oxidative and neuro-nitrosative effects across a range of psychiatric and neurodegenerative illnesses. Mechanisms include glucose lowering effects and effects on AMP-activated protein kinase (AMPK) signalling, however the best evidence for clinical benefit is through the glucose lowering effects, with other mechanisms less supported by the current evidence base. This narrative review aims to draw together the existing evidence for use of metformin as a psychopharmaceutical and present the role of metformin in the context of physical and psychiatric ill health, including metabolic, endocrinological and cancer domains. It not only has therapeutic potential in medical comorbidity but may have potential in core illness domains.
Collapse
Affiliation(s)
- Seetal Dodd
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, the University of Melbourne, Parkville, VIC, Australia; Centre for Youth Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Luba Sominsky
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Barwon Health Laboratory, University Hospital Geelong, Barwon Health, VIC, Australia
| | - Dan Siskind
- Metro South Addiction and Mental Health Service, MIRT, Level 2, 228 Logan Rd, Woolloongabba, Brisbane, Qld 4102, Australia University of Queensland School of Clinical Medicine, Brisbane, Australia Queensland Centre for Mental Health Research, Brisbane, Australia; Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Andre F Carvalho
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adam J Walker
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Alison R Yung
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Centre for Youth Mental Health, University of Melbourne, Parkville, VIC, Australia; School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Lana J Williams
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Hannah Myles
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia; Northern Adelaide Mental Health Service, Salisbury, SA, Australia
| | - Tayler Watson
- Mental Health, Drugs and Alcohol Service, Barwon Health, Geelong VIC, Australia
| | - Michael Berk
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, the University of Melbourne, Parkville, VIC, Australia; Centre for Youth Mental Health, University of Melbourne, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| |
Collapse
|
14
|
Verma V, Mehendale AM. A Review on the Use of Metformin in Pregnancy and Its Associated Fetal Outcomes. Cureus 2022; 14:e30039. [PMID: 36381747 PMCID: PMC9637404 DOI: 10.7759/cureus.30039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022] Open
Abstract
A commonly used first-line anti-diabetic medication, metformin, has been used in pregnancy. The drug is known to have specific effects on different organs around the body. One of these organs includes the ovaries. Therefore, for more than 40 years, it has often been prescribed for maternal obesity along with gestational diabetes mellitus. Untreated pregnancies like these frequently result in complications for both the mother and the fetus, like macrosomia, pregnancy-induced hypertension, obstructed labor, stillbirths, and perinatal deaths. In addition, there is also evidence that these mothers tend to develop type II diabetes mellitus during their pregnancy and even a few years post-delivery. These complications can be controlled or even reduced with the help of metformin, sometimes combining it with insulin or clomiphene citrate if required. There is still a need to cautiously prescribe the drug by outweighing its benefits against the risk associated with it. The current research on the subject leans more towards the benefits offered to the mother during pregnancy. Only a few randomized, controlled trials have been conducted on the fetal condition after the mother has been administered metformin. Furthermore, these studies lack the appropriate sample size and long-term follow-up on these metformin-exposed offspring. As a result, there are no reliable data available to clinicians and physicians about the drug. Owing to its benefits in certain pregnancies, it is less likely that the drug will cease to be prescribed. Therefore, it becomes increasingly imperative to conduct more research on this topic to ensure the drug is safe for the mother and the offspring.
Collapse
|
15
|
Maternal Treatment with Metformin Persistently Ameliorates High-Fat Diet-Induced Metabolic Symptoms and Modulates Gut Microbiota in Rat Offspring. Nutrients 2022; 14:nu14173612. [PMID: 36079869 PMCID: PMC9460832 DOI: 10.3390/nu14173612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
A maternal high-fat (HF) diet has long-term deleterious effect on offspring. This study aims to evaluate whether maternal metformin (MT) treatment ameliorates the adverse effects of maternal HF diet on offspring and the role of gut microbiota in it. Pregnant Sprague-Dawley rats were randomly assigned to a HF diet (60% fat) or a standard chow diet (11.8% fat) group, and part of the HF diet group rats were co-treated with MT via drinking water (300 mg/kg/day), resulting in three groups according to maternal diet and MT treatment during gestation and lactation. All offspring were weaned on a chow diet. A maternal HF diet showed a significant deleterious effect on offspring’s metabolic phenotype and induced colonic inflammation and gut-barrier disruption through the reshaped gut microbiota. The daily oral administration of MT to HF-fed dams during gestation and lactation reversed the dysbiosis of gut microbiota in both dams and adult offspring. The hypothalamic TGR5 expression and plasma bile acids composition in adult male offspring was restored by maternal MT treatment, which could regulate hypothalamic appetite-related peptides expression and alleviate inflammation, thereby improving male offspring’s metabolic phenotype. The present study indicates that targeting the gut–brain axis through the mother may be an effective strategy to control the metabolic phenotype of offspring.
Collapse
|
16
|
Pan ML, Chen LR, Chen KH. Prepregnancy Polycystic Ovary Syndrome as a Risk Factor of Subsequent Preterm Labor: A National Population-Based Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19095470. [PMID: 35564864 PMCID: PMC9103656 DOI: 10.3390/ijerph19095470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023]
Abstract
Background: Preterm labor and the following preterm births, which account for most of the perinatal deaths, are an important issue in public health. The study aims to assess the risk of subsequent preterm labor in pregnant females who have prepregnancy polycystic ovary syndrome (PCOS). Methods: This study has enrolled 1,000,000 randomly sampled females retrieved from the Taiwan National Health Insurance Research Database (NHIRD) during 1998−2012. The study excluded prepregnancy PCOS females who were initially diagnosed at age <15 or >45, and those who had inconsistent diagnoses. Moreover, the medical records of blood hormone tests, gynecologic ultrasonography, pelvic examinations, and tocometers were verified to confirm the accuracy of both diagnoses of PCOS and preterm labor. Among the prepregnancy PCOS females who became pregnant (the case group), each was age-matched to four females without prepregnancy PCOS (the control group). Results: Pregnant females in the case group (n = 1959) had a higher incidence of preterm labor than those in the control group (n = 7836) (42.98% vs. 21.99%, p < 0.0001). Analyzed by using logistic regression, the risk of preterm labor was significantly higher in the case group compared with the control group (crude OR: 2.674; 95% CI: 2.410−2.968, p < 0.0001). After adjustment with covariates, further analysis revealed a similar trend (adjusted OR: 2.405; 95% CI: 2.158−2.680, p < 0.0001). Among 1959 PCOS females in the case group, 196 had undergone metformin treatment. Compared with females without metformin treatment (the non-metformin subgroup), the metformin users (metformin subgroup) presented a reduced risk for preterm labor (adjusted OR: 2.238; 95% CI: 1.657−3.023). The risk of subsequent preterm labor was reduced by about 10% for the metformin subgroup compared with the non-metformin subgroup. Conclusions: Prepregnancy PCOS is an independent and significant risk factor of subsequent preterm labor. Among prepregnancy PCOS females, the risk of preterm labor is lowered by about 10% in metformin users compared with non-metformin females.
Collapse
Affiliation(s)
- Mei-Lien Pan
- Information Technology Service Center, National YangMing ChiaoTung University, Taipei 112, Taiwan;
| | - Li-Ru Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan;
- Department of Mechanical Engineering, National YangMing ChiaoTung University, Hsinchu 300, Taiwan
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, Buddhist Tzu-Chi Medical Foundation, Taipei 231, Taiwan
- School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
- Correspondence: ; Tel.: +886-2-6628-9779; Fax: +886-2-6628-9009
| |
Collapse
|
17
|
Chen X, Li X, Kang S, Duan W. Correlation of Serum IGF-1R, VEGF, and ET Levels with Bone Mineral Density in Type 2 Diabetic Mellitus Patients Treated with Metformin Plus α-Glucosidase Inhibitors. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:7707875. [PMID: 35497918 PMCID: PMC9050299 DOI: 10.1155/2022/7707875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/14/2022] [Accepted: 03/28/2022] [Indexed: 11/21/2022]
Abstract
Diabetes mellitus is a common chronic disease. This study aimed to investigate the correlation between serum insulin-like growth factor 1 receptor (IGF-1R), vascular endothelial growth factor (VEGF), endothelin (ET) levels, and bone mineral density (BMD) in type 2 diabetic mellitus (T2DM) patients treated with metformin plus α-glucosidase inhibitors and evaluate the predictive value of serum factors in the prognosis of osteoporosis in these patients. It was a prospective study that enrolled 142 patients with T2DM treated in Dinghu District People's Hospital from March 2019 to May 2020. All enrollments were randomized (1 : 1) to receive either metformin (control group) or metformin plus α-glucosidase inhibitors (study group). After 12 weeks of treatment, metformin plus α-glucosidase inhibitors were associated with significantly lower levels of 2 hPG, FPG, HbA1c, and HOMA-IR versus metformin alone (P < 0.05). After treatment, the BMD was positively correlated with IGF-1R and negatively correlated with VEGF and ET. Alpha-glucosidase inhibitors plus metformin for primary T2DM can effectively manage blood glucose and reduce insulin resistance in patients, but the prediction of osteoporosis development remains to be further explored in large sample studies.
Collapse
Affiliation(s)
- Xue Chen
- Department of Pathology and Pathophysiology, Zhaoqing Medical College, Zhaoqing City, Guangdong Province 526020, China
| | - Xiaosheng Li
- Emergency Department, Dinghu District People's Hospital, Zhaoqing City, Guangdong Province 526070, China
| | - Sheng Kang
- Department of Endocrinology, Lanling County People's Hospital, Linyi City, Shandong Province 277700, China
| | - Wenbiao Duan
- School Office, Zhaoqing Medical College, Zhaoqing City, Guangdong Province 526020, China
| |
Collapse
|
18
|
Athar F, Templeman NM. C. elegans as a model organism to study female reproductive health. Comp Biochem Physiol A Mol Integr Physiol 2022; 266:111152. [PMID: 35032657 DOI: 10.1016/j.cbpa.2022.111152] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/17/2022]
Abstract
Female reproductive health has been historically understudied and underfunded. Here, we present the advantages of using a free-living nematode, Caenorhabditis elegans, as an animal system to study fundamental aspects of female reproductive health. C. elegans is a powerful high-throughput model organism that shares key genetic and physiological similarities with humans. In this review, we highlight areas of pressing medical and biological importance in the 21st century within the context of female reproductive health. These include the decline in female reproductive capacity with increasing chronological age, reproductive dysfunction arising from toxic environmental insults, and cancers of the reproductive system. C. elegans has been instrumental in uncovering mechanistic insights underlying these processes, and has been valuable for developing and testing therapeutics to combat them. Adopting a convenient model organism such as C. elegans for studying reproductive health will encourage further research into this field, and broaden opportunities for making advancements into evolutionarily conserved mechanisms that control reproductive function.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.
| |
Collapse
|
19
|
Jerzak M, Szafarowska M. Preliminary Results for Personalized Therapy in Pregnant Women with Polycystic Ovary Syndrome During the COVID-19 Pandemic. Arch Immunol Ther Exp (Warsz) 2022; 70:13. [PMID: 35325391 PMCID: PMC8943102 DOI: 10.1007/s00005-022-00650-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 01/12/2022] [Indexed: 12/11/2022]
Abstract
Increased androgen level, hyperinsulinemia, diabetes, impaired fibrinolysis, obesity, hypertension, chronic inflammation, abnormal immune response to infections and hyperhomocysteinemia are the most common abnormalities related to polycystic ovary syndrome (PCOS) women and are the factors predisposing to the severe course of COVID-19. The SARS-Cov-2 infection during pregnancy is associated with an increased risk of complications (spontaneous abortion), similar to those in PCOS. The treatment of PCOS pregnant women with a history of fertility failures raises many doubts, especially during the COVID pandemic. However, due to the increasing incidence of infections among reproductive people and the potentially more serious course in pregnant women, numerous questions about the safety and effectiveness of the treatment are still very current. In our study we presented a series of cases of recurrent miscarriages or recurrent implantation failure PCOS pregnant women with confirmed COVID-19. The diagnosis of infertility confirmed the presence of plasminogen activator inhibitor type 1 and/or 5,10-methylenetetrahydrofolate reductase polymorphisms in each of them. Moreover, some of the women presented immune dysfunction associated with infertility. We have described the personalized treatments of each pregnant patient included: metformin, enoxaparin and tacrolimus. The treatment applied had the expected effect, supporting the implantation processes. Furthermore, despite the ambiguous data according to immunological therapy of infertile women during the COVID pandemic, we observed a mild or asymptomatic COVID-19 course and we noticed no pregnancy complications.
Collapse
Affiliation(s)
| | - Monika Szafarowska
- Department of Gynecology and Oncological Gynecology, Military Medical Institute, Warsaw, Poland.
| |
Collapse
|
20
|
Subarto CB, Matsuzaki M, Estrade M, Sundemeir S, Hakimi M, Isnaeni Y, Hadi H, Kurniasari Y, Fatimah S, Yulyani L, Wijayanti I. A Qualitative Study: Mothers’ Experience in the Management of Gestational Diabetes Mellitus during and after Pregnancy in Yogyakarta, Indonesia. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Gestational Diabetes Mellitus (GDM) incidence is related to maternal and child health risks, such as pre-eclampsia, premature births, and the development of type 2 diabetes at 5-10 years after pregnancy. The adaptation process of women with GDM is complicated and requires a series of treatments and behavior changes over a short period. The psychological support of families and healthcare professionals is indispensable in achieving GDM management success.
Objectives: This research aimed to explore Indonesian mothers' experiences in GDM management and support during and after pregnancy
Methods: The design of research implemented qualitative approaches, phenomenology, and the homogenous purposive technique to recruit 12 participants. In-depth interviews were carried out with each participant using face to face. Interviews were recorded, verbatim transcribed, and analyzed thematically by Collaizi.
Results: Five themes emerged: responses to GDM diagnosis; GDM management during pregnancy; obstacles in GDM management; support for GDM management; and post-natal self-management. Adherence to diet and exercise recommendations is difficult for some participants, although they have a good understanding of GDM. Feeling the fetus's movement of and surrendering to God become forms of psychological support and motivation in the management of gestational diabetes mellitus.
Conclusions: The barriers and facilitators for GDM management identified here are multidimensional and may help facilitate health workers to more effectively support women with GDM in overcoming the perceived obstacles so that they can still feel a safe and comfortable during pregnancy with minimum risk.
Collapse
|
21
|
Intensive Medical Nutrition Therapy Alone or with Added Metformin to Prevent Gestational Diabetes Mellitus among High-Risk Mexican Women: A Randomized Clinical Trial. Nutrients 2021; 14:nu14010062. [PMID: 35010938 PMCID: PMC8746971 DOI: 10.3390/nu14010062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to examine the efficacy of intensive medical nutrition therapy (MNT) plus metformin in preventing gestational diabetes mellitus (GDM) among high-risk Mexican women. An open-label randomized clinical trial was conducted. Inclusion criteria were pregnant women with three or more GDM risk factors: Latino ethnic group, maternal age >35 years, body mass index >25 kg/m2, insulin resistance, and a history of previous GDM, prediabetes, a macrosomic neonate, polycystic ovarian syndrome, or a first-degree relative with type 2 diabetes. Women before 15 weeks of gestation were assigned to group 1 (n = 45): intensive MNT-plus metformin (850 mg twice/day) or group 2 (n = 45): intensive MNT without metformin. Intensive MNT included individual dietary counseling, with ≤50% of total energy from high carbohydrates. The primary outcome was the GDM incidence according to the International Association of Diabetes Pregnancy Study Groups criteria. There were no significant differences in baseline characteristics and adverse perinatal outcomes between the groups. The GDM incidence was n = 11 (24.4%) in the MNT plus metformin group versus n = 7 (15.5%) in the MNT without metformin group: p = 0.42 (RR: 1.57 [95% CI: 0.67–3.68]). There is no benefit in adding metformin to intensive MNT to prevent GDM among high-risk Mexican women. Clinical trials registration: NCT01675310.
Collapse
|
22
|
Bakhtyukov AA, Derkach KV, Sorokoumov VN, Stepochkina AM, Romanova IV, Morina IY, Zakharova IO, Bayunova LV, Shpakov AO. The Effects of Separate and Combined Treatment of Male Rats with Type 2 Diabetes with Metformin and Orthosteric and Allosteric Agonists of Luteinizing Hormone Receptor on Steroidogenesis and Spermatogenesis. Int J Mol Sci 2021; 23:198. [PMID: 35008624 PMCID: PMC8745465 DOI: 10.3390/ijms23010198] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
In men with type 2 diabetes mellitus (T2DM), steroidogenesis and spermatogenesis are impaired. Metformin and the agonists of luteinizing hormone/human chorionic gonadotropin(hCG)-receptor (LH/hCG-R) (hCG, low-molecular-weight allosteric LH/hCG-R-agonists) can be used to restore them. The aim was to study effectiveness of separate and combined administration of metformin, hCG and 5-amino-N-tert-butyl-2-(methylsulfanyl)-4-(3-(nicotinamido)phenyl)thieno[2,3-d]pyrimidine-6-carboxamide (TP3) on steroidogenesis and spermatogenesis in male rats with T2DM. hCG (15 IU/rat/day) and TP3 (15 mg/kg/day) were injected in the last five days of five-week metformin treatment (120 mg/kg/day). Metformin improved testicular steroidogenesis and spermatogenesis and restored LH/hCG-R-expression. Compared to control, in T2DM, hCG stimulated steroidogenesis and StAR-gene expression less effectively and, after five-day administration, reduced LH/hCG-R-expression, while TP3 effects changed weaker. In co-administration of metformin and LH/hCG-R-agonists, on the first day, stimulating effects of LH/hCG-R-agonists on testosterone levels and hCG-stimulated expression of StAR- and CYP17A1-genes were increased, but on the 3-5th day, they disappeared. This was due to reduced LH/hCG-R-gene expression and increased aromatase-catalyzed estradiol production. With co-administration, LH/hCG-R-agonists did not contribute to improving spermatogenesis, induced by metformin. Thus, in T2DM, metformin and LH/hCG-R-agonists restore steroidogenesis and spermatogenesis, with metformin being more effective in restoring spermatogenesis, and their co-administration improves LH/hCG-R-agonist-stimulating testicular steroidogenesis in acute but not chronic administration.
Collapse
Affiliation(s)
- Andrey A. Bakhtyukov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| | - Kira V. Derkach
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| | - Viktor N. Sorokoumov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
- Institute of Chemistry, Saint Petersburg State University, 198504 St. Petersburg, Russia
| | - Anna M. Stepochkina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| | - Irina V. Romanova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| | - Irina Yu. Morina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| | - Irina O. Zakharova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| | - Liubov V. Bayunova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| | - Alexander O. Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 St. Petersburg, Russia; (A.A.B.); (K.V.D.); (V.N.S.); (A.M.S.); (I.V.R.); (I.Y.M.); (I.O.Z.); (L.V.B.)
| |
Collapse
|
23
|
Pascual-Morena C, Cavero-Redondo I, Álvarez-Bueno C, Martínez-Hortelano JA, Reina-Gutiérrez S, Saz-Lara A, Núñez de Arenas-Arroyo S, Martínez-Vizcaíno V. Physical Exercise vs. Metformin to Improve Delivery- and Newborn-Related Outcomes Among Pregnant Women With Overweight: A Network Meta-Analysis. Front Med (Lausanne) 2021; 8:796009. [PMID: 34957166 PMCID: PMC8696129 DOI: 10.3389/fmed.2021.796009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Overweight/obesity is associated with the risk of delivery- and newborn-related complications in pregnancy. Interventions such as exercise or metformin could reduce the risk of these complications. Objective: To estimate and compare the effects of different types of exercise interventions (i.e., aerobic, resistance, combined exercise) and metformin on delivery- and newborn-related outcomes among pregnant women with overweight/obesity. Methods: MEDLINE, Scopus, Web of Science, Cochrane Library databases and the gray literature were searched from inception to September 2021. This systematic review was registered in PROSPERO (CDR: 42019121715). Randomized controlled trials (RCTs) of metformin or an exercise intervention aimed at preventing cesarean section, preterm birth, macrosomia, or birth weight among pregnant women with overweight/obesity were included. Random effects meta-analyses and frequentist network meta-analyses (NMA) were conducted for each outcome. Results: Fifteen RCTs were included. In the NMA, metformin reduced the risk of cesarean section (RR = 0.66, 95% CI: 0.46, 0.95), combined exercise reduced the risk of macrosomia (RR = 0.37, 95% CI: 0.14, 0.95), and aerobic exercise reduced birth weight (mean difference = -96.66 g, 95% CI: -192.45, -0.88). In the subgroup among pregnant women with obesity, metformin reduced the risk of cesarean section (RR = 0.66, 95% CI: 0.45, 0.97). Conclusions: Combined exercise could reduce the risk of macrosomia in pregnant women with overweight, whereas metformin could reduce the risk of cesarean section in pregnant women with obesity. However, previous evidence suggests a larger effect of physical exercise in other outcomes for this population group. Therefore, the medicalization of healthy pregnant women with obesity is not justified by the current evidence. Systematic Review Registration: PROSPERO: CRD42019121715; https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42019121715.
Collapse
Affiliation(s)
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad de Castilla—La Mancha, Cuenca, Spain
- Rehabilitation in Health Research Center (CIRES), Universidad de las Américas, Santiago, Chile
| | - Celia Álvarez-Bueno
- Health and Social Research Center, Universidad de Castilla—La Mancha, Cuenca, Spain
- Universidad Politécnica y Artística del Paraguay, Asunción, Paraguay
| | - José Alberto Martínez-Hortelano
- Health and Social Research Center, Universidad de Castilla—La Mancha, Cuenca, Spain
- Guadalajara University Hospital, Health Service of Castilla-La Mancha (SESCAM), Guadalajara, Spain
| | - Sara Reina-Gutiérrez
- Health and Social Research Center, Universidad de Castilla—La Mancha, Cuenca, Spain
| | - Alicia Saz-Lara
- Health and Social Research Center, Universidad de Castilla—La Mancha, Cuenca, Spain
| | | | - Vicente Martínez-Vizcaíno
- Health and Social Research Center, Universidad de Castilla—La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
24
|
Molin J, Vanky E, Løvvik TS, Dehlin E, Bixo M. Gestational weight gain, appetite regulating hormones, and metformin treatment in polycystic ovary syndrome: A longitudinal, placebo-controlled study. BJOG 2021; 129:1112-1121. [PMID: 34865304 DOI: 10.1111/1471-0528.17042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To explore mechanisms that modulate gestational weight gain (GWG) in women with polycystic ovary syndrome (PCOS) and healthy controls. DESIGN Sub-sample of randomised controlled trials (PCOS) combined with a prospective cohort (controls). SETTING Eleven Norwegian, Swedish, and Icelandic hospitals. POPULATION Pregnant women with PCOS treated with metformin (PCOS-M, n = 36) or placebo (PCOS-P, n = 37), and healthy pregnant women (HC, n = 15). METHODS Serum levels of the appetite regulating hormones leptin, ghrelin, allopregnanolone, and soluble leptin receptor (sOB-R) were determined in the first and third trimesters. MAIN OUTCOME MEASURES Excessive GWG (eGWG) relative to body mass index according to Institute of Medicine (IOM) guideline. Serum leptin/sOB-R ratio, or free-leptin-index (FLI), as biomarker of leptin sensitivity. Serum ghrelin and allopregnanolone levels. RESULTS The overall prevalence of eGWG was 44% (38/86). Women with eGWG had higher first and third trimester FLI (P < 0.001), and lower third trimester allopregnanolone levels (P = 0.003) versus women with non-eGWG. The prevalence of eGWG was lower in PCOS-M versus PCOS-P (28% versus 62%, odds ratio = 0.4, 95% CI 0.2-0.8, P = 0.005). FLI decreased during pregnancy in PCOS-M (P = 0.01), but remained unaltered in PCOS-P and HC. Ghrelin and allopregnanolone levels were comparable in PCOS-M, PCOS-P and HC throughout pregnancy. CONCLUSION Excessive GWG is associated with enhanced leptin resistance, and attenuated physiological increase in serum allopregnanolone levels during pregnancy. Metformin reduces the risk for eGWG and improves leptin sensitivity in pregnant women with PCOS.
Collapse
Affiliation(s)
- Johanna Molin
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University, Umeå, Sweden
| | - 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
| | - 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 Gynaecology, St Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eva Dehlin
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University, Umeå, Sweden
| | - Marie Bixo
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University, Umeå, Sweden
| |
Collapse
|
25
|
Mercado-Méndez S, González-Sepúlveda L, Romaguera J, González-Rodríguez LA. The Use of Oral Hypoglycemic Agents during Pregnancy: An Alternative to Insulin? PUERTO RICO HEALTH SCIENCES JOURNAL 2021; 40:162-167. [PMID: 35077074 PMCID: PMC9048127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Gestational Diabetes Mellitus (GDM) and Type 2 Diabetes Mellitus (DM2) are metabolic disorders characterized by increased insulin resistance. Although insulin is the treatment of choice in pregnant patients with DM, the prescription of oral hypoglycemic agents (OHA) has been increasing among practitioners. This study aimed to evaluate the maternal and neonatal outcomes when oral hypoglycemic agents were used in diabetic pregnant women. METHODS Medical records from the Maternal-Infant Care Unit Clinics SoM-UPR (n=149) were reviewed. Patients that were treated with metformin, sulfonylurea or insulin were included. Maternal and neonatal outcomes were compared between groups. RESULTS Patient's mean age was 28 ± 6 years. The majority had GDM (91%). The most common comorbidity was hypertension (9.9%). Lifestyle modification was used as treatment in 77% of patients during the second trimester, but its use decreased to 33% during the third trimester. Insulin was the treatment of choice. Among the OHA, sulfonylurea was preferred. Postprandial glucose levels were lower in patients who used insulin as compared to those without medications. CONCLUSION No significant differences were found in maternal outcomes such as C-section, induction of labor, episiotomy or preterm labor, or neonatal outcomes such as macrosomia, neonatal hypoglycemia or congenital abnormalities among treatment groups. OHA can be considered as an alternative to insulin for the treatment of DM during pregnancy in selected cases.
Collapse
Affiliation(s)
- Sheila Mercado-Méndez
- University of Puerto Rico School of Medicine – Department of Medicine - Endocrinology, Diabetes and Metabolism Division
| | - Lorena González-Sepúlveda
- Puerto Rico Clinical and Translational Research Consortium, University of Puerto Rico Medical Sciences Campus
| | - Josefina Romaguera
- University of Puerto Rico School of Medicine – Department of Obstetrics and Gynecology
| | - Loida A. González-Rodríguez
- University of Puerto Rico School of Medicine – Department of Medicine - Endocrinology, Diabetes and Metabolism Division
| |
Collapse
|
26
|
Quadir H. Current Therapeutic Use of Metformin During Pregnancy: Maternal Changes, Postnatal Effects and Safety. Cureus 2021; 13:e18818. [PMID: 34804675 PMCID: PMC8592788 DOI: 10.7759/cureus.18818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/16/2021] [Indexed: 11/28/2022] Open
Abstract
Metformin is one of the most easily available medications for diabetes and has a relatively low cost. It is not only used in diabetes but is also effective in polycystic ovarian syndrome (PCOS) and obesity. Although insulin is the first choice when it comes to treating pregnant women with gestational diabetes mellitus (GDM), metformin has also been debated as a good choice after modification of diet. As metformin passes through the placenta, it is essential to know its consequence of leading to insulin resistance in the fetus as well as the impact on postnatal development. The use of metformin during GDM has raised many trials demonstrating that outcomes from the use of metformin are similar to those achieved with insulin. Follow-up studies were also conducted that assessed the impact on children exposed to metformin in utero. This review highlights the experimental evidence relating to the use of metformin during pregnancy for different conditions, and its impact on the growth and development of offspring.
Collapse
Affiliation(s)
- Huma Quadir
- Internal Medicine/Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
27
|
Dinicola S, Unfer V, Facchinetti F, Soulage CO, Greene ND, Bizzarri M, Laganà AS, Chan SY, Bevilacqua A, Pkhaladze L, Benvenga S, Stringaro A, Barbaro D, Appetecchia M, Aragona C, Bezerra Espinola MS, Cantelmi T, Cavalli P, Chiu TT, Copp AJ, D’Anna R, Dewailly D, Di Lorenzo C, Diamanti-Kandarakis E, Hernández Marín I, Hod M, Kamenov Z, Kandaraki E, Monastra G, Montanino Oliva M, Nestler JE, Nordio M, Ozay AC, Papalou O, Porcaro G, Prapas N, Roseff S, Vazquez-Levin M, Vucenik I, Wdowiak A. Inositols: From Established Knowledge to Novel Approaches. Int J Mol Sci 2021; 22:10575. [PMID: 34638926 PMCID: PMC8508595 DOI: 10.3390/ijms221910575] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/13/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022] Open
Abstract
Myo-inositol (myo-Ins) and D-chiro-inositol (D-chiro-Ins) are natural compounds involved in many biological pathways. Since the discovery of their involvement in endocrine signal transduction, myo-Ins and D-chiro-Ins supplementation has contributed to clinical approaches in ameliorating many gynecological and endocrinological diseases. Currently both myo-Ins and D-chiro-Ins are well-tolerated, effective alternative candidates to the classical insulin sensitizers, and are useful treatments in preventing and treating metabolic and reproductive disorders such as polycystic ovary syndrome (PCOS), gestational diabetes mellitus (GDM), and male fertility disturbances, like sperm abnormalities. Moreover, besides metabolic activity, myo-Ins and D-chiro-Ins deeply influence steroidogenesis, regulating the pools of androgens and estrogens, likely in opposite ways. Given the complexity of inositol-related mechanisms of action, many of their beneficial effects are still under scrutiny. Therefore, continuing research aims to discover new emerging roles and mechanisms that can allow clinicians to tailor inositol therapy and to use it in other medical areas, hitherto unexplored. The present paper outlines the established evidence on inositols and updates on recent research, namely concerning D-chiro-Ins involvement into steroidogenesis. In particular, D-chiro-Ins mediates insulin-induced testosterone biosynthesis from ovarian thecal cells and directly affects synthesis of estrogens by modulating the expression of the aromatase enzyme. Ovaries, as well as other organs and tissues, are characterized by a specific ratio of myo-Ins to D-chiro-Ins, which ensures their healthy state and proper functionality. Altered inositol ratios may account for pathological conditions, causing an imbalance in sex hormones. Such situations usually occur in association with medical conditions, such as PCOS, or as a consequence of some pharmacological treatments. Based on the physiological role of inositols and the pathological implications of altered myo-Ins to D-chiro-Ins ratios, inositol therapy may be designed with two different aims: (1) restoring the inositol physiological ratio; (2) altering the ratio in a controlled way to achieve specific effects.
Collapse
Affiliation(s)
- Simona Dinicola
- Systems Biology Group Lab, 00161 Rome, Italy; (S.D.); (V.U.); (M.B.); (C.A.); (M.S.B.E.); (G.M.)
| | - Vittorio Unfer
- Systems Biology Group Lab, 00161 Rome, Italy; (S.D.); (V.U.); (M.B.); (C.A.); (M.S.B.E.); (G.M.)
| | - Fabio Facchinetti
- Obstetrics and Gynecology Unit, Mother-Infant and Adult Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Christophe O. Soulage
- CarMeN Lab, INSA-Lyon, INSERM U1060, INRA, University Claude Bernard Lyon 1, 69100 Villeurbanne, France;
| | - Nicholas D. Greene
- Newlife Birth Defects Research Centre and Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London WC1E 6BT, UK; (N.D.G.); (A.J.C.)
| | - Mariano Bizzarri
- Systems Biology Group Lab, 00161 Rome, Italy; (S.D.); (V.U.); (M.B.); (C.A.); (M.S.B.E.); (G.M.)
- Department of Experimental Medicine, University La Sapienza, 00161 Rome, Italy
| | - Antonio Simone Laganà
- Department of Obstetrics and Gynecology, Hospital “Filippo Del Ponte”, University of Insubria, 21100 Varese, Italy;
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Arturo Bevilacqua
- Department of Dynamic, Clinical Psychology and Health Studies, Sapienza University, 00161 Rome, Italy;
| | - Lali Pkhaladze
- Zhordania and Khomasuridze Institute of Reproductology, Tbilisi 0112, Georgia;
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Annarita Stringaro
- National Center for Drug Research and Evaluation, Italian National Institute of Health, 00161 Rome, Italy;
| | - Daniele Barbaro
- U.O. Endocrinology in Livorno Hospital, USL Nordovest Toscana, 57100 Livorno, Italy;
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, Regina Elena National Cancer Institute, IRCCS, 00161 Rome, Italy;
| | - Cesare Aragona
- Systems Biology Group Lab, 00161 Rome, Italy; (S.D.); (V.U.); (M.B.); (C.A.); (M.S.B.E.); (G.M.)
| | | | - Tonino Cantelmi
- Institute for Interpersonal Cognitive Therapy, 00100 Rome, Italy;
| | - Pietro Cavalli
- Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | | | - Andrew J. Copp
- Newlife Birth Defects Research Centre and Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London WC1E 6BT, UK; (N.D.G.); (A.J.C.)
| | - Rosario D’Anna
- Department of Human Pathology, University of Messina, 98122 Messina, Italy;
| | - Didier Dewailly
- Faculty of Medicine, University of Lille, 59000 Lille, France;
| | - Cherubino Di Lorenzo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome Polo Pontino, 04100 Latina, Italy;
| | - Evanthia Diamanti-Kandarakis
- Department of Endocrinology and Diabetes, HYGEIA Hospital, Marousi, 15123 Athens, Greece; (E.D.-K.); (E.K.); (O.P.)
| | - Imelda Hernández Marín
- Human Reproduction Department, Hospital Juárez de México, Universidad Nacional Autónoma de México (UNAM), Mexico City 07760, Mexico;
| | - Moshe Hod
- Department of Obstetrics and Gynecology Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel;
| | - Zdravko Kamenov
- Department of Internal Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Eleni Kandaraki
- Department of Endocrinology and Diabetes, HYGEIA Hospital, Marousi, 15123 Athens, Greece; (E.D.-K.); (E.K.); (O.P.)
| | - Giovanni Monastra
- Systems Biology Group Lab, 00161 Rome, Italy; (S.D.); (V.U.); (M.B.); (C.A.); (M.S.B.E.); (G.M.)
| | | | - John E. Nestler
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | | | - Ali C. Ozay
- Department of Obstetrics and Gynecology, Near East University Hospital, Nicosia 99138, Cyprus;
| | - Olga Papalou
- Department of Endocrinology and Diabetes, HYGEIA Hospital, Marousi, 15123 Athens, Greece; (E.D.-K.); (E.K.); (O.P.)
| | | | - Nikos Prapas
- IAKENTRO, Infertility Treatment Center, 54250 Thessaloniki, Greece;
| | - Scott Roseff
- Reproductive Endocrinology and Infertility, South Florida Institute for Reproductive Medicine (IVFMD), Boca Raton, FL 33458, USA;
| | - Monica Vazquez-Levin
- Instituto de Biología y Medicina Experimental (IBYME, CONICET-FIBYME), Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina (CONICET), Buenos Aires 2490, Argentina;
| | - Ivana Vucenik
- Department of Medical & Research Technology and Pathology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
| | - Artur Wdowiak
- Diagnostic Techniques Unit, Medical University of Lublin, 20-081 Lublin, Poland;
| |
Collapse
|
28
|
Zhao L, Huang L. Predictive Value of Homeostasis Model Assessment of Insulin Resistance, Visceral Fat Index, and Prepregnancy Body Mass Index in Gestational Metabolic Syndrome. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6709725. [PMID: 34603475 PMCID: PMC8486542 DOI: 10.1155/2021/6709725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/03/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To explore the diagnostic value of homeostasis model assessment of insulin resistance (HOMA-IR), visceral fat index (VAI), and prepregnancy body mass index (BMI) in gestational metabolic syndrome (GMS). METHODS From December 2019 to March 2021,122 GMS high-risk pregnant women who received routine antenatal clinic visits and planned to give birth in our hospital were selected as the research objects. Pregnant women were divided into the GMS group (n = 79) and the control group (n = 43) according to GMS diagnostic criteria during the gestation period of 32-36 weeks. The general information such as age and gestational week of pregnant women as well as HOMA-IR, VAI, and BMI before pregnancy were compared between the two groups. The glucose and lipid metabolism indexes of pregnant women in two groups were analyzed, the detection rates of HOMA-IR, VAI, and prepregnancy BMI in GMS between the two groups were compared. Logistic regression was used to analyze the risk factors for GMS in pregnant women, and receiver operating characteristic curve (ROC) was used to analyze the diagnostic value of HOMA-IR, VAI, prepregnancy BMI, and the combination of the three for GMS. RESULTS The body weight, systolic blood pressure, and diastolic blood pressure of the GMS group were higher than those of the control group, and the differences were statistically significant (P < 0.05). There was no significant difference in age, gestational week, and height between the two groups (P > 0.05). The levels of FPG, FINS, TC, TG, LDL-C, and FFA in the GMS group were higher than the control group, the level of HDL-C in the GMS group was lower than the control group, and the difference was statistically significant (P < 0.05). The levels of HOMA-IR, VAI, and prepregnancy BMI in the GMS group were higher than those in the control group, and the differences were statistically significant (P < 0.05). The positive detection rates of HOMA-IR, VAI, and prepregnancy BMI in the GMS group were 83.54%, 86.07%, and 81.01%, respectively. There was no significant difference in the positive detection rates of HOMA-IR, VAI, and prepregnancy BMI between the two groups (P > 0.05). High levels of HOMA-IR, VAI, and prepregnancy BMI were risk factors for GMS in pregnant women (P < 0.05). ROC curve showed area under the curve for HOMA-IR was 0.810, area under the curve for VAI was 0.771, and area under the curve for prepregnancy BMI was 0.749. The AUC for the combination of HOMA-IR, VAI, and prepregnancy BMI was 0.918. CONCLUSION HOMA-IR, VAI, and prepregnancy BMI in GMS have a high detection rate and certain diagnostic value, and the combination of the three has higher clinical value.
Collapse
Affiliation(s)
- Laiping Zhao
- The Affiliated Nanhua Hospital, Department of Obstetrics and Gynecology Division 1, Hengyang Medical School, University of South China, Hengyang 421002, Hunan, China
| | - Lichun Huang
- The Affiliated Nanhua Hospital, Department of Joint Sports, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
29
|
Benham JL, Donovan LE, Yamamoto JM. Metformin in Pregnancy for Women with Type 2 Diabetes: a Review. Curr Diab Rep 2021; 21:36. [PMID: 34495393 DOI: 10.1007/s11892-021-01409-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/21/2021] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW To review the current evidence for the use of metformin in pregnancy for women with type 2 diabetes. RECENT FINDINGS A large, multicenter, double-blind randomized controlled trial found that women with type 2 diabetes in pregnancy treated with metformin as an adjunct to insulin therapy had less gestational weight gain, insulin requirements, caesarian sections, macrosomia, and neonatal adiposity, but more neonates were small for gestational age (SGA) compared with insulin alone. It is unclear if the higher number of SGA infants are a direct result of metformin exposure or mediated through other effects such as less gestational weight gain and improved glycemic control. Additional follow-up studies of offspring exposed to metformin in utero are required. Metformin may be a useful adjunctive treatment for women with type 2 diabetes in pregnancy to help meet glycemic targets if there are no concerns for or indications of SGA.
Collapse
Affiliation(s)
- Jamie L Benham
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lois E Donovan
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Obstetrics and Gynecology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Jennifer M Yamamoto
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Internal Medicine, Children's Hospital Research Institute of Manitoba, University of Manitoba, Room 836, 715 McDermot Ave, Winnipeg, MB, R3E3P4, Canada.
| |
Collapse
|
30
|
Li X, Celotto S, Pizzol D, Gasevic D, Ji MM, Barnini T, Solmi M, Stubbs B, Smith L, López Sánchez GF, Pesolillo G, Yu Z, Tzoulaki I, Theodoratou E, Ioannidis JPA, Veronese N, Demurtas J. Metformin and health outcomes: An umbrella review of systematic reviews with meta-analyses. Eur J Clin Invest 2021; 51:e13536. [PMID: 33709434 DOI: 10.1111/eci.13536] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The objective was to capture the breadth of outcomes that have been associated with metformin use and to systematically assess the quality, strength and credibility of these associations using the umbrella review methodology. METHODS Four major databases were searched until 31 May 2020. Meta-analyses of observational studies and meta-analyses of randomized controlled trials (RCTs) (including active and placebo control arms) were included. RESULTS From 175 eligible publications, we identified 427 different meta-analyses, including 167 meta-analyses of observational studies, 147 meta-analyses of RCTs for metformin vs placebo/no treatment and 113 meta-analyses of RCTs for metformin vs active medications. There was no association classified as convincing or highly suggestive from meta-analyses of observational studies, but some suggestive/weak associations of metformin use with a lower mortality risk of CVD and cancer. In meta-analyses of RCTs, metformin was associated with a lower incidence of diabetes in people with prediabetes or no diabetes at baseline; lower ovarian hyperstimulation syndrome incidence (in women in controlled ovarian stimulation); higher success for clinical pregnancy rate in poly-cystic ovary syndrome (PCOS); and significant reduction in body mass index in people with type 1 diabetes mellitus, in women who have obesity/overweight with PCOS and in obese/overweight women. Of 175 publications, 166 scored as low or critically low quality per AMSTAR 2 criteria. CONCLUSIONS Observational evidence on metformin seems largely unreliable. Randomized evidence shows benefits for preventing diabetes and in some gynaecological and obstetrical settings. However, almost all meta-analyses are of low or critically low quality according to AMSTAR 2 criteria.
Collapse
Affiliation(s)
- Xue Li
- School of Public Health, Zhejiang University, Hangzhou, China
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Stefano Celotto
- MD, Primary Care Department, AAS3 Alto Friuli e Collinare e Medio Friuli, Udine, Italy
| | - Damiano Pizzol
- Italian Agency for Development Cooperation, Khartoum, Sudan
| | - Danijela Gasevic
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Meng-Meng Ji
- School of Public Health, Nanjing Medical University, Nanjing, China
| | | | - Marco Solmi
- Neurosciences Department, University of Padua, Padua, Italy
- Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Brendon Stubbs
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Lee Smith
- The Cambridge Centre for Sport and Exercise Sciences, Anglia Ruskin University, Cambridge, UK
| | - Guillermo F López Sánchez
- Faculty of Health, Education, Medicine and Social Care, School of Medicine, Vision and Eye Research Institute, Anglia Ruskin University-Cambridge Campus, Cambridge, UK
| | | | - Zengli Yu
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - John P A Ioannidis
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Nicola Veronese
- Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Jacopo Demurtas
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
- Primary Care Department USL Toscana Sud-Est, Grosseto, Italy
| |
Collapse
|
31
|
Carvajal JA, Oporto JI. The Myometrium in Pregnant Women with Obesity. Curr Vasc Pharmacol 2021; 19:193-200. [PMID: 32484103 DOI: 10.2174/1570161118666200525133530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022]
Abstract
Obesity is a worldwide public health problem, affecting at least one-third of pregnant women. One of the main problems of obesity during pregnancy is the resulting high rate of cesarean section. The leading cause of this higher frequency of cesarean sections in obese women, compared with that in nonobese women, is an altered myometrial function that leads to lower frequency and potency of contractions. In this article, the disruptions of myometrial myocytes were reviewed in obese women during pregnancy that may explain the dysfunctional labor. The myometrium of obese women exhibited lower expression of connexin43, a lower function of the oxytocin receptor, and higher activity of the potassium channels. Adipokines, such as leptin, visfatin, and apelin, whose concentrations are higher in obese women, decreased myometrial contractility, perhaps by inhibiting the myometrial RhoA/ROCK pathway. The characteristically higher cholesterol levels of obese women alter myometrial myocyte cell membranes, especially the caveolae, inhibiting oxytocin receptor function, and increasing the K+ channel activity. All these changes in the myometrial cells or their environment decrease myometrial contractility, at least partially explaining the higher rate of cesarean of sections in obese women.
Collapse
Affiliation(s)
- Jorge A Carvajal
- Departamento de Obstetricia, Unidad de Medicina Materno Fetal, Mexico City, Mexico
| | - Joaquín I Oporto
- Estudiante de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
32
|
Cao R, To K, Kachour N, Beever A, Owens J, Sathananthan A, Singh P, Kolloli A, Subbian S, Venketaraman V. Everolimus-induced effector mechanism in macrophages and survivability of Erdman, CDC1551 and HN878 strains of Mycobacterium tuberculosis infection. Biomol Concepts 2021; 12:46-54. [PMID: 34062056 PMCID: PMC8993225 DOI: 10.1515/bmc-2021-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/07/2021] [Indexed: 12/02/2022] Open
Abstract
With a disease as widespread and destructive as tuberculosis, more effective drugs and healthcare strategies, in addition to the current antibiotics regimen, are crucial for the enhanced well-being of millions of people suffering from the disease. Host-directed therapy is a new and emerging concept in treating chronic infectious diseases, such as tuberculosis. Repurposing of anti-cancer drugs, such as everolimus, may be an effective way to supplement the standard antibiotic treatment. Individuals with type 2 diabetes are increasingly susceptible to co-morbidities and co-infections including Mycobacterium tuberculosis, the causative agent of tuberculosis. We demonstrated in this study that in vitro everolimus treatment of granulomas from individuals with type 2 diabetes caused significant reduction in the viability of Mycobacterium tuberculosis.Further investigations revealed the effects of everolimus in targeting foamy macrophages, a macrophage phenotype that forms around granulomas, and is characterized by a higher lipid accumulation inside the cells. These foamy macrophages are thought to harbor dormant bacilli, which are potential sources of disease reactivation. Therefore, blocking foamy macrophage formation would help better killing of intracellular bacteria. Here, we report the potential of everolimus treatment to downregulate lipid content within the foamy macrophages of in vitro granulomas, thus leading to a potential decrease in the number of foamy macrophages and a more robust response to Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Ruoqiong Cao
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Kimberly To
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Nala Kachour
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Abrianna Beever
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - James Owens
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Airani Sathananthan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Pooja Singh
- Department of Pulmonary, Allergy, and Critical Care Medicine, The University of Alabama at Birmingham, Birmingham, AL35294, USA; The Public Health Research Institute at New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Afsal Kolloli
- The Public Health Research Institute at New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Selvakumar Subbian
- The Public Health Research Institute at New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
33
|
Interaction between Metformin, Folate and Vitamin B 12 and the Potential Impact on Fetal Growth and Long-Term Metabolic Health in Diabetic Pregnancies. Int J Mol Sci 2021; 22:ijms22115759. [PMID: 34071182 PMCID: PMC8198407 DOI: 10.3390/ijms22115759] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Metformin is the first-line treatment for many people with type 2 diabetes mellitus (T2DM) and gestational diabetes mellitus (GDM) to maintain glycaemic control. Recent evidence suggests metformin can cross the placenta during pregnancy, thereby exposing the fetus to high concentrations of metformin and potentially restricting placental and fetal growth. Offspring exposed to metformin during gestation are at increased risk of being born small for gestational age (SGA) and show signs of ‘catch up’ growth and obesity during childhood which increases their risk of future cardiometabolic diseases. The mechanisms by which metformin impacts on the fetal growth and long-term health of the offspring remain to be established. Metformin is associated with maternal vitamin B12 deficiency and antifolate like activity. Vitamin B12 and folate balance is vital for one carbon metabolism, which is essential for DNA methylation and purine/pyrimidine synthesis of nucleic acids. Folate:vitamin B12 imbalance induced by metformin may lead to genomic instability and aberrant gene expression, thus promoting fetal programming. Mitochondrial aerobic respiration may also be affected, thereby inhibiting placental and fetal growth, and suppressing mammalian target of rapamycin (mTOR) activity for cellular nutrient transport. Vitamin supplementation, before or during metformin treatment in pregnancy, could be a promising strategy to improve maternal vitamin B12 and folate levels and reduce the incidence of SGA births and childhood obesity. Heterogeneous diagnostic and screening criteria for GDM and the transient nature of nutrient biomarkers have led to inconsistencies in clinical study designs to investigate the effects of metformin on folate:vitamin B12 balance and child development. As rates of diabetes in pregnancy continue to escalate, more women are likely to be prescribed metformin; thus, it is of paramount importance to improve our understanding of metformin’s transgenerational effects to develop prophylactic strategies for the prevention of adverse fetal outcomes.
Collapse
|
34
|
Kasper P, Breuer S, Hoffmann T, Vohlen C, Janoschek R, Schmitz L, Appel S, Fink G, Hünseler C, Quaas A, Demir M, Lang S, Steffen HM, Martin A, Schramm C, Bürger M, Mahabir E, Goeser T, Dötsch J, Hucklenbruch-Rother E, Bae-Gartz I. Maternal Exercise Mediates Hepatic Metabolic Programming via Activation of AMPK-PGC1α Axis in the Offspring of Obese Mothers. Cells 2021; 10:1247. [PMID: 34069390 PMCID: PMC8158724 DOI: 10.3390/cells10051247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity is associated with an increased risk of hepatic metabolic dysfunction for both mother and offspring and targeted interventions to address this growing metabolic disease burden are urgently needed. This study investigates whether maternal exercise (ME) could reverse the detrimental effects of hepatic metabolic dysfunction in obese dams and their offspring while focusing on the AMP-activated protein kinase (AMPK), representing a key regulator of hepatic metabolism. In a mouse model of maternal western-style-diet (WSD)-induced obesity, we established an exercise intervention of voluntary wheel-running before and during pregnancy and analyzed its effects on hepatic energy metabolism during developmental organ programming. ME prevented WSD-induced hepatic steatosis in obese dams by alterations of key hepatic metabolic processes, including activation of hepatic ß-oxidation and inhibition of lipogenesis following increased AMPK and peroxisome-proliferator-activated-receptor-γ-coactivator-1α (PGC-1α)-signaling. Offspring of exercised dams exhibited a comparable hepatic metabolic signature to their mothers with increased AMPK-PGC1α-activity and beneficial changes in hepatic lipid metabolism and were protected from WSD-induced adipose tissue accumulation and hepatic steatosis in later life. In conclusion, this study demonstrates that ME provides a promising strategy to improve the metabolic health of both obese mothers and their offspring and highlights AMPK as a potential metabolic target for therapeutic interventions.
Collapse
Affiliation(s)
- Philipp Kasper
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Saida Breuer
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Thorben Hoffmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Lisa Schmitz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Gregor Fink
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christoph Hünseler
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Alexander Quaas
- Department of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany;
| | - Münevver Demir
- Charité Campus Mitte and Campus Virchow Clinic, Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, D-13353 Berlin, Germany;
| | - Sonja Lang
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Hans-Michael Steffen
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Anna Martin
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Christoph Schramm
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Martin Bürger
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, D-50937 Cologne, Germany;
| | - Tobias Goeser
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| |
Collapse
|
35
|
Albaghdadi AJH, Kan FWK. Therapeutic Potentials of Low-Dose Tacrolimus for Aberrant Endometrial Features in Polycystic Ovary Syndrome. Int J Mol Sci 2021; 22:2872. [PMID: 33808965 PMCID: PMC7998611 DOI: 10.3390/ijms22062872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/24/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a major anovulatory infertility affecting a great proportion of women of childbearing age and is associated with obesity, insulin resistance and chronic inflammation. Poor endometrial receptivity and recurrent implantation failure are major hurdles to the establishment of pregnancy in women with PCOS. The accumulating body of evidence obtained from experimental and clinical studies suggests a link between inherent adaptive and innate immune irregularities and aberrant endometrial features in PCOS. The use of conventional therapeutic interventions such as lifestyle modification, metformin and ovarian stimulation has achieved limited clinical success in restoring ovulation and endometrial receptivity in women with PCOS. Unlike other immunosuppressive drugs prescribed in the clinical management of autoimmune and inflammatory disorders that may have deleterious effects on fertility and fetal development, preclinical studies in mice and in women without PCOS but with repeated implantation failure revealed potential therapeutic benefits for the use of low-dose tacrolimus in treating female infertility. Improved systemic and ovarian immune functions, endometrial progesterone receptor and coreceptor expressions and uterine vascular adaptation to pregnancy were among features of enhanced progesterone-receptor sensitivity in the low-dose tacrolimus-treated mouse model of the disease. In this review, we have compiled available experimental and clinical data in literature on endometrial progesterone resistance and current therapeutic options, as well as mechanisms of actions and reported outcomes relevant to the potential therapeutic benefits for the use of low-dose tacrolimus in treating PCOS-associated female infertility.
Collapse
Affiliation(s)
| | - Frederick W. K. Kan
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| |
Collapse
|
36
|
Hong JGS, Tan PC, Kamarudin M, Omar SZ. Prophylactic metformin after antenatal corticosteroids (PROMAC): a double blind randomized controlled trial. BMC Pregnancy Childbirth 2021; 21:138. [PMID: 33588801 PMCID: PMC7885598 DOI: 10.1186/s12884-021-03628-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background Antenatal corticosteroids (ACS) are increasingly used to improve prematurity-related neonatal outcome. A recognized and common adverse effect from administration of antenatal corticosteroid is maternal hyperglycemia. Even normal pregnancy is characterized by relative insulin resistance and glucose intolerance. Treatment of maternal hyperglycemia after ACS might be indicated due to the higher risk of neonatal acidosis which may coincide with premature birth. Metformin is increasingly used to manage diabetes mellitus during pregnancy as it is effective and more patient friendly. There is no data on prophylactic metformin to maintain euglycemia following antenatal corticosteroids administration. Methods A double blind randomized trial. 103 women scheduled to receive two doses of 12-mg intramuscular dexamethasone 12-hour apart were separately randomized to take prophylactic metformin or placebo after stratification according to their gestational diabetes (GDM) status. First oral dose of allocated study drug was taken at enrolment and continued 500 mg twice daily for 72 hours if not delivered. Six-point blood sugar profiles were obtained each day (pre- and two-hour post breakfast, lunch and dinner) for up to three consecutive days. A hyperglycemic episode is defined as capillary glucose fasting/pre-meal ≥ 5.3 mmol/L or two-hour post prandial/meal ≥ 6.7 mmol/L. Primary outcome was hyperglycemic episodes on Day-1 (first six blood sugar profile points) following antenatal corticosteroids. Results Number of hyperglycemic episodes on the first day were not significantly different (mean ± standard deviation) 3.9 ± 1.4 (metformin) vs. 4.1 ± 1.6 (placebo) p = 0.64. Hyperglycemic episodes markedly reduced on second day in both arms to 0.9 ± 1.0 (metformin) vs. 1.2 ± 1.0 (placebo) p = 0.15 and further reduced to 0.6 ± 1.0 (metformin) vs. 0.7 ± 1.0 (placebo) p = 0.67 on third day. Hypoglycemic episodes during the 3-day study period were few and all other secondary outcomes were not significantly different. Conclusions In euglycemic and diet controllable gestational diabetes mellitus women, antenatal corticosteroids cause sustained maternal hyperglycemia only on Day-1. The magnitude of Day-1 hyperglycemia is generally low. Prophylactic metformin does not reduce antenatal corticosteroids’ hyperglycemic effect. Trial registration The trial is registered in the ISRCTN registry on May 4 2017 with trial identifier 10.1186/ISRCTN10156101.
Collapse
Affiliation(s)
- Jesrine Gek Shan Hong
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603, Kuala Lumpur, Malaysia.
| | - Peng Chiong Tan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603, Kuala Lumpur, Malaysia
| | - Maherah Kamarudin
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603, Kuala Lumpur, Malaysia
| | - Siti Zawiah Omar
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Shpakov AO. Improvement Effect of Metformin on Female and Male Reproduction in Endocrine Pathologies and Its Mechanisms. Pharmaceuticals (Basel) 2021; 14:ph14010042. [PMID: 33429918 PMCID: PMC7826885 DOI: 10.3390/ph14010042] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Metformin (MF), a first-line drug to treat type 2 diabetes mellitus (T2DM), alone and in combination with other drugs, restores the ovarian function in women with polycystic ovary syndrome (PCOS) and improves fetal development, pregnancy outcomes and offspring health in gestational diabetes mellitus (GDM) and T2DM. MF treatment is demonstrated to improve the efficiency of in vitro fertilization and is considered a supplementary drug in assisted reproductive technologies. MF administration shows positive effect on steroidogenesis and spermatogenesis in men with metabolic disorders, thus MF treatment indicates prospective use for improvement of male reproductive functions and fertility. MF lacks teratogenic effects and has positive health effect in newborns. The review is focused on use of MF therapy for restoration of female and male reproductive functions and improvement of pregnancy outcomes in metabolic and endocrine disorders. The mechanisms of MF action are discussed, including normalization of metabolic and hormonal status in PCOS, GDM, T2DM and metabolic syndrome and restoration of functional activity and hormonal regulation of the gonadal axis.
Collapse
Affiliation(s)
- Alexander O Shpakov
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 Saint Petersburg, Russia
| |
Collapse
|
38
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
39
|
The Effects of Maternal Metformin Treatment on Late Prenatal and Early Postnatal Development of the Offspring Are Modulated by Sex. Pharmaceuticals (Basel) 2020; 13:ph13110363. [PMID: 33158193 PMCID: PMC7694275 DOI: 10.3390/ph13110363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 11/17/2022] Open
Abstract
Metformin is currently used to improve pregnancy outcome in women affected by polycystic ovary syndrome (PCOS) or diabetes. However, metformin may also be useful in pregnancies at risk of intrauterine growth restriction (IUGR) since it improves placental efficiency and the fetuses' developmental competence. There is no data on the duration of the effect of this treatment from the prenatal up to the postnatal stages. Therefore, the present trial aimed at determining the impact of metformin treatment on the offspring neonatal traits and early postnatal development (i.e., during lactation) using an in vivo swine model. The results support that maternal metformin treatment during pregnancy induces protective changes in body shape and composition of the progeny (i.e., larger head size and body length at birth and higher total viscera weight at weaning). However, there were also major effects of the offspring sex (smaller corpulence in females and lower relative weight of main viscerae in males), which should be considered for further preclinical studies and when even the current clinical application in women affected by PCOS or diabetes is implemented.
Collapse
|
40
|
Sun CC, Lai YN, Wang WH, Xu XM, Li XQ, Wang H, Zheng JY, Zheng JQ. Metformin Ameliorates Gestational Diabetes Mellitus-Induced Endothelial Dysfunction via Downregulation of p65 and Upregulation of Nrf2. Front Pharmacol 2020; 11:575390. [PMID: 33162888 PMCID: PMC7581851 DOI: 10.3389/fphar.2020.575390] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/07/2020] [Indexed: 01/14/2023] Open
Abstract
Gestational diabetes mellitus (GDM) causes oxidative stress in mothers and infants and causes vascular endothelial dysfunction, which is a key factor for maternal and fetal cardiovascular diseases in the later stage of GDM, seriously threatening the life and health of mothers and infants. Nowadays, metformin (MET) has been discovered to improve endothelial function, but studies regarding the mechanism of MET improving endothelial cell function and alleviating endothelial function under hyperglycemia are still extremely limited. We aimed to investigate whether MET exerts its protective role against hyperglycemia-induced endothelial dysfunction through p65 and Nrf2. In our studies, applying cell migration assay and tube formation assay, we observed an obvious improvement of endothelial function under MET-treated, as characterized by that MET accelerated GDM-attenuated migration and angiogenesis of HUVECs. And ELISA assay results uncovered that Nrf2 expression level was decreased in GDM placenta, HVUECs and maternal serum comparing with normal group, however activation Nrf2 largely ameliorated tube formation under hyperglycemic condition. Furthermore, MET elevated the Nrf2 expression level and the level of nuclear Nrf2 accumulation in hyperglycemic HUVECs. Besides, preliminary evidence predicted that Nrf2 expression was modulated by transcription factor p65, which was increased in GDM peripheral blood, placenta and HUVECs, and suppression of p65 could recover GDM-induced suppression of angiogenesis. In addition, we also confirmed MET restores the GDM-induced angiogenesis impairment may via downregulation of p65 and upregulation of Nrf2. Taken together, the endothelial protective effect of MET under GDM (HG) conditions could be partly attributed to its role in downregulating p65 and upregulating Nrf2.
Collapse
Affiliation(s)
- Cong Cong Sun
- Wenzhou Key Laboratory of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Ya Nan Lai
- Wenzhou Key Laboratory of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Wen Huan Wang
- Wenzhou Key Laboratory of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Xiao Min Xu
- Wenzhou Key Laboratory of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Xiao Qing Li
- Wenzhou Key Laboratory of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Hai Wang
- Wenzhou Key Laboratory of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Jia Yong Zheng
- Wenzhou Key Laboratory of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Jian Qiong Zheng
- Department of Obstetrics and Gynecology, The Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| |
Collapse
|
41
|
Mokkala K, Vahlberg T, Houttu N, Koivuniemi E, Laitinen K. Distinct Metabolomic Profile Because of Gestational Diabetes and its Treatment Mode in Women with Overweight and Obesity. Obesity (Silver Spring) 2020; 28:1637-1644. [PMID: 32705820 DOI: 10.1002/oby.22882] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/18/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Whether the presence of gestational diabetes (GDM) and its treatment mode influence the serum metabolic profile in women with overweight or obesity was studied. METHODS The serum metabolic profiles of 352 women with overweight or obesity participating in a mother-infant clinical study were analyzed with a targeted NMR approach (at 35.1 median gestational weeks). GDM was diagnosed with a 2-hour 75-g oral glucose tolerance test. RESULTS The metabolomic profile of the women with GDM (n = 100) deviated from that of women without GDM (n = 252). Differences were seen in 70 lipid variables, particularly higher concentrations of very low-density lipoprotein particles and serum triglycerides were related to GDM. Furthermore, levels of branched-chain amino acids and glycoprotein acetylation, a marker of low-grade inflammation, were higher in women with GDM. Compared with women with GDM treated with diet only, the women treated with medication (n = 19) had higher concentrations of severalizes of VLDL particles and their components, leucine, and isoleucine, as well as glycoprotein acetylation. CONCLUSIONS A clearly distinct metabolic profile was detected in GDM, which deviated even more if the patient was receiving medical treatment. This suggests a need for more intense follow-up and therapy for women with GDM during pregnancy and postpartum to reduce their long-term adverse health risks.
Collapse
Affiliation(s)
- Kati Mokkala
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Tero Vahlberg
- Department of Clinical Medicine, Biostatistics, University of Turku, Turku, Finland
| | - Noora Houttu
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Ella Koivuniemi
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Kirsi Laitinen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Turku University Hospital, Turku, Finland
| |
Collapse
|
42
|
Jorquera G, Echiburú B, Crisosto N, Sotomayor-Zárate R, Maliqueo M, Cruz G. Metformin during Pregnancy: Effects on Offspring Development and Metabolic Function. Front Pharmacol 2020; 11:653. [PMID: 32625081 PMCID: PMC7311748 DOI: 10.3389/fphar.2020.00653] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Maternal obesity during pregnancy and gestational diabetes mellitus (GDM) are both associated with of several postnatal diseases in the offspring, including obesity, early onset hypertension, diabetes mellitus, and reproductive alterations. Metformin is an oral drug that is being evaluated to treat GDM, obesity-associated insulin resistance, and polycystic ovary syndrome (PCOS) during pregnancy. The beneficial effects of metformin on glycemia and pregnancy outcomes place it as a good alternative for its use during pregnancy. In this line of thought, improving the metabolic status of the pregnant mother by using metformin should avoid the consequences of insulin resistance on the offspring's fetal and postnatal development. However, some human and animal studies have shown that metformin during pregnancy could amplify these alterations and be associated with excessive postnatal weight gain and obesity. In this minireview, we discuss not only the clinical and experimental evidence that supports the benefits of using metformin during pregnancy but also the evidence showing a possible negative impact of this drug on the offspring's development.
Collapse
Affiliation(s)
- Gonzalo Jorquera
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valpararaíso, Chile
| | - Bárbara Echiburú
- Laboratory of Endocrinology and Metabolism, West Division, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nicolás Crisosto
- Laboratory of Endocrinology and Metabolism, West Division, Faculty of Medicine, University of Chile, Santiago, Chile.,Unit of Endocrinology, Clínica Las Condes, Santiago, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valpararaíso, Chile
| | - Manuel Maliqueo
- Laboratory of Endocrinology and Metabolism, West Division, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valpararaíso, Chile
| |
Collapse
|
43
|
Lane SL, Doyle AS, Bales ES, Lorca RA, Julian CG, Moore LG. Increased uterine artery blood flow in hypoxic murine pregnancy is not sufficient to prevent fetal growth restriction†. Biol Reprod 2020; 102:660-670. [PMID: 31711123 PMCID: PMC7068112 DOI: 10.1093/biolre/ioz208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/19/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Incomplete maternal vascular responses to pregnancy contribute to pregnancy complications including intrauterine growth restriction (IUGR) and preeclampsia. We aimed to characterize maternal vascular dysfunction in a murine model of fetal growth restriction as an approach toward identifying targetable pathways for improving pregnancy outcomes. We utilized a murine model of late-gestation hypoxia-induced IUGR that reduced E18.5 fetal weight by 34%. Contrary to our hypothesis, uterine artery blood flow as measured in vivo by Doppler ultrasound was increased in mice housed under hypobaric hypoxia (385 mmHg; 5500 m) vs normoxia (760 mmHg; 0 m). Using wire myography, uterine arteries isolated from hypoxic mice had similar vasodilator responses to the two activators A769662 and acetylcholine as those from normoxic mice, although the contribution of an increase in nitric oxide production to uterine artery vasodilation was reduced in the hypoxic vs normoxic groups. Vasoconstrictor responses to phenylephrine and potassium chloride were unaltered by hypoxia. The levels of activated adenosine monophosphate-activated protein kinase (AMPK) were reduced with hypoxia in both the uterine artery and placenta as measured by western blot and immunohistochemistry. We concluded that the rise in uterine artery blood flow may be compensatory to hypoxia but was not sufficient to prevent fetal growth restriction. Although AMPK signaling was reduced by hypoxia, AMPK was still receptive to pharmacologic activation in the uterine arteries in which it was a potent vasodilator. Thus, AMPK activation may represent a new therapy for pregnancy complications involving reduced uteroplacental perfusion.
Collapse
Affiliation(s)
- Sydney L Lane
- Integrated Physiology PhD Program, University of Colorado Denver Graduate School, Aurora, CO, USA
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Alexandrea S Doyle
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Elise S Bales
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Colleen G Julian
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
44
|
The impact of prenatal environment on postnatal life and performance: Future perspectives for prevention and treatment. Theriogenology 2020; 150:15-19. [PMID: 31983467 DOI: 10.1016/j.theriogenology.2020.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/16/2022]
Abstract
The present review aims to offer a non-comprehensive outline of the current state-of-the-art and future perspectives on management and therapeutic tools for intrauterine growth restriction (IUGR) and associated prenatal programming in both human and animal species. Animals are used as models for the study of phenomena related to IUGR, but also for research on prenatal therapies with the main objective of designing and developing preventive and therapeutic strategies. The research is currently paying attention on maternal-focused pharmacological treatments and nutritional strategies but also on fetal-focused treatments. Fetal-focused treatments, administered either directly at the fetus or by using infusion of umbilical cord, amniotic sac or placenta, which avoids the administration of substances at high doses to the mother for allowing their availability at the fetoplacental level. The results obtained in this area of research using large animals (rabbits, pigs and ruminants) have a dual interest, for translational biomedicine and for veterinary medicine and animal production.
Collapse
|
45
|
Doi SAR, Furuya-Kanamori L, Toft E, Musa OAH, Islam N, Clark J, Thalib L. Metformin in pregnancy to avert gestational diabetes in women at high risk: Meta-analysis of randomized controlled trials. Obes Rev 2020; 21:e12964. [PMID: 31667980 DOI: 10.1111/obr.12964] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 01/08/2023]
Abstract
Previous randomized and observational studies on the efficacy of metformin in pregnancy to reduce incident gestational diabetes mellitus (GDM) in women at high risk (obesity, polycystic ovary syndrome [PCOS], or pregestational insulin resistance) have been conflicting and several groups are planning further randomized controlled trials (RCTs) to answer this question conclusively. This work assesses the efficacy of metformin in pregnancy to avert one outcome-incident GDM in women at high risk. We included RCTs comparing metformin with usual care or placebo controls in terms of incident GDM and recruiting women at high risk during early pregnancy. Eleven eligible trials enrolled 2370 adult women whose intervention arm consisted of metformin started at conception or before 20 weeks of gestation. Risk of GDM was similar in intervention compared with controls (risk ratio [RR] 1.03; 95% confidence interval [CI], 0.85-1.24). The data were of sufficient quality meeting the criteria for consistency and directness. We conclude that metformin does not contribute to averting the GDM outcome in women at high risk when initiated in pregnancy. The evidence provided by this synthesis affirms that further broad clinical trials investigating this question are no longer needed.
Collapse
Affiliation(s)
- Suhail A R Doi
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Luis Furuya-Kanamori
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar.,Research School of Population Health, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Egon Toft
- Deans Office, College of Medicine, Qatar University, Doha, Qatar
| | - Omran A H Musa
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Nazmul Islam
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Justin Clark
- The Centre for Research into Evidence Based Practice, Bond University, Gold Coast, Queensland, Australia
| | - Lukman Thalib
- Department of Public Health, College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
46
|
Wang J, Wu D, Guo H, Li M. Hyperandrogenemia and insulin resistance: The chief culprit of polycystic ovary syndrome. Life Sci 2019; 236:116940. [PMID: 31604107 DOI: 10.1016/j.lfs.2019.116940] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/23/2019] [Accepted: 10/06/2019] [Indexed: 02/07/2023]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common systemic reproductive endocrine diseases, which has a variety of effects on a woman's health. Because of the involvement of multiple pathways and the lack of common clues, PCOS demonstrates multifactorial properties and heterogeneity of symptoms. Recent studies have demonstrated that the core etiology and primary endocrine characteristics of PCOS are hyperandrogenemia (HA) and insulin resistance (IR). HA and IR are the main causes of PCOS and they can interplay each other in the occurrence and development of PCOS. Just because of this, the study about the effects of HA and IR on pathophysiology of various related symptoms of PCOS is very important to understand the pathogenesis of PCOS. This paper reviews the main symptoms of PCOS, including neuroendocrine disorders, reproductive processes, dyslipidemia, obesity, hypertension, nonalcoholic fatty liver disease (NAFLD), and sleep disordered breathing, which seriously affect the physical and mental health of PCOS women. The increasing knowledge of the development pattern of HA and IR in PCOS suggests that changes in diet and lifestyle, and the discovery of potential therapeutic agents may improve PCOS. However, further studies are needed to clarify the mutual influence and relation of HA and IR in development of PCOS. This review provides an overview of the current knowledge about the effects of HA and IR on PCOS.
Collapse
Affiliation(s)
- Juan Wang
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Daichao Wu
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Hui Guo
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Meixiang Li
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China.
| |
Collapse
|
47
|
Maple-Brown LJ, Lindenmayer G, Barzi F, Whitbread C, Connors C, Moore E, Boyle J, Kirkwood M, Lee IL, Longmore D, van Dokkum P, Wicks M, Dowden M, Inglis C, Cotter M, Kirkham R, Corpus S, Chitturi S, Thomas S, O'Dea K, Zimmet P, Oats J, McIntyre HD, Brown A, Shaw JE. Real-world experience of metformin use in pregnancy: Observational data from the Northern Territory Diabetes in Pregnancy Clinical Register. J Diabetes 2019; 11:761-770. [PMID: 30680949 DOI: 10.1111/1753-0407.12905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/11/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND In Australia's Northern Territory, Indigenous mothers account for 33% of births and have high rates of hyperglycemia in pregnancy. The prevalence of type 2 diabetes (T2D) in pregnancy is up to 10-fold higher in Indigenous than non-Indigenous Australian mothers, and the use of metformin is common. We assessed birth outcomes in relation to metformin use during pregnancy from a clinical register. METHODS The study included women with gestational diabetes (GDM), newly diagnosed diabetes in pregnancy (DIP), or pre-existing T2D from 2012 to 2016. Data were analyzed for metformin use in the third trimester. Regression models were adjusted for maternal age, body mass index, parity, and insulin use. RESULTS Of 1649 pregnancies, 814 (49.4%) were to Indigenous women, of whom 234 (28.7%) had T2D (vs 4.6% non-Indigenous women; P < 0.001). Metformin use was high in Indigenous women (84%-90% T2D, 42%-48% GDM/DIP) and increased over time in non-Indigenous women (43%-100% T2D, 14%-35% GDM/DIP). Among Indigenous women with GDM/DIP, there were no significant differences between groups with and without metformin in cesarean section (51% vs 39%; adjusted odds ratio [aOR] 1.25, 95% confidence interval [CI] 0.87-1.81), large for gestational age (24% vs 13%; aOR 1.5, 95% CI 0.9-2.5), or serious neonatal adverse events (9.4% vs 5.9%; aOR 1.32, 95% CI 0.68-2.57). Metformin use was independently associated with earlier gestational age (37.7 vs 38.5 weeks), but the risk did not remain independently higher after exclusion of women managed with medical nutrition therapy alone, and the increase in births <37 weeks was not significant on multivariate analysis. CONCLUSIONS We found no clear evidence of any adverse outcomes related to the use of metformin for the treatment of hyperglycemia in pregnancy.
Collapse
Affiliation(s)
- Louise J Maple-Brown
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Greta Lindenmayer
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Federica Barzi
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Cherie Whitbread
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Christine Connors
- Northern Territory Department of Health, Darwin, Northern Territory, Australia
| | - Elizabeth Moore
- Aboriginal Medical Services Alliance Northern Territory, Darwin, Northern Territory, Australia
| | - Jacqueline Boyle
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marie Kirkwood
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - I-Lynn Lee
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Danielle Longmore
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Paula van Dokkum
- Department of Medicine, Alice Springs Hospital, Alice Springs, Northern Territory, Australia
- Population Health Research, Baker Heart and Diabetes Institute, Alice Springs, Northern Territory, Australia
| | - Mary Wicks
- Department of Medicine, Alice Springs Hospital, Alice Springs, Northern Territory, Australia
| | | | | | - Margaret Cotter
- Aboriginal Medical Services Alliance Northern Territory, Darwin, Northern Territory, Australia
| | - Renae Kirkham
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Sumaria Corpus
- Danila Dilba Health Service, Darwin, Northern Territory, Australia
| | - Sridhar Chitturi
- Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Sujatha Thomas
- Obstetrics and Gynaecology Department, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Kerin O'Dea
- Population Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Paul Zimmet
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jeremy Oats
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Harold D McIntyre
- Mater Medical Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Alex Brown
- Obstetrics and Gynaecology Department, Royal Darwin Hospital, Darwin, Northern Territory, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jonathan E Shaw
- Population Health Research, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
48
|
Maternal Metformin Treatment Improves Developmental and Metabolic Traits of IUGR Fetuses. Biomolecules 2019; 9:biom9050166. [PMID: 31035702 PMCID: PMC6572102 DOI: 10.3390/biom9050166] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022] Open
Abstract
Metformin is an anti-hyperglycemic drug widely used for the treatment of insulin resistance and glucose intolerance and is currently considered for preventing large-for-gestational-age (LGA) offspring in pregnant women affected by obesity or diabetes. Our hypothesis was the opposite—metformin may be used for improving the development of offspring affected by intrauterine growth restriction (IUGR) and preventing the appearance of small-for-gestational-age (SGA) neonates in non-obese and non-diabetic but malnourished pregnancies. The current study, performed in a swine preclinical model of IUGR by undernutrition, showed that fetuses in the treated group showed no significant increases in body-weight, but showed a significantly higher weight of the brain, the total thoracic and abdominal viscera, the liver, the kidneys, the spleen, and the adrenal glands. Maternal metformin treatment was also related to significant increases in the fetal plasma concentration of parameters indicative of glycemic (glucose and fructosamine) and lipid profiles (triglycerides). Overall, these results suggest a protective effect of the treatment on the developmental competence of the fetuses. These findings may be of high value for human medicine in case of maternal malnutrition, since metformin is a cheap drug easily available, but also in case of placental deficiency, since metformin seems to improve placental development and function.
Collapse
|