1
|
Ningombam P, Kundu RD, Ghosh S, Ghosh SK, Wangkheimayum VD. Impact of metformin on gene expression in Burmese loach (Lepidocephalichthys berdmorei) from Manipur, India. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 978:179458. [PMID: 40253858 DOI: 10.1016/j.scitotenv.2025.179458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/28/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Pharmaceutical contaminants like metformin, a widely used antidiabetic drug, pose emerging threats to aquatic ecosystems. This study investigates the endocrine-disrupting effects of metformin on Burmese loach (Lepidocephalichthys berdmorei), a bio-indicator species in Manipur's freshwater systems. Fish were exposed to 40 μg/L, 120 μg/L, and 360 μg/L of metformin for 28 days, and effects on gene expression, gonadal histology, and molecular interactions were assessed. Gene expression analysis revealed significant upregulation of AR, SULT2A1, CYP19A1, and 17β-HSD in metformin-treated males (p < 0.05). 3β-HSD was notably elevated at 360 μg/L. Despite these molecular changes, no histological differences were observed between treated and control groups. Molecular docking showed that SULT2A1 had the strongest interaction with metformin (-5.2 kcal/mol), followed by CYP19A1 (-5.0 kcal/mol) and AR (-4.9 kcal/mol). Molecular dynamics (MD) simulations confirmed the stability of the SULT2A1-metformin complex, with reduced RMSD, compactness, and residue fluctuations at the active site. The binding free energy (∆G) of -5.24 kcal/mol further supports this stable interaction. Additionally, structural deviations were observed in SULT2A1 upon metformin binding, suggesting potential functional alterations. These findings suggest that metformin alters endocrine function in L. berdmorei by modulating gene expression and interacting with key endocrine proteins, particularly SULT2A1. As L. berdmorei plays a crucial role in freshwater ecosystems, such disruptions may impact aquatic biodiversity. This study provides novel insights into metformin's molecular toxicity and highlights L. berdmorei as a potential model for xenoestrogen detection in freshwater environments.
Collapse
Affiliation(s)
- Pratima Ningombam
- Department of Zoology, Manipur University, Canchipur 795003, Manipur, India.
| | - Raima Das Kundu
- Indian Barcode of Life (InBOL) Healthcare Pvt. Ltd. (www.inbol.org), 28/2G, Nakuleswar Bhattacharjee Lane, Kolkata 700026, India.
| | - Semanti Ghosh
- Department of Biotechnology, School of Life Sciences, Swami Vivekananda University, Barrackpore, Kolkata 700121, India.
| | | | | |
Collapse
|
2
|
Ursavas S, Kuyucu Y, Yenilmez ED, Tuli A, Tekayev M, Mete UO. The evaluation of the effects of metformin on the rat testes at a light and electron microscopic level. Biochem Biophys Res Commun 2025; 753:151507. [PMID: 39983548 DOI: 10.1016/j.bbrc.2025.151507] [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: 10/27/2024] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
The present study aimed to assess the histological and ultrastructural changes in the testes induced by metformin. Eighteen adult male Wistar rats were divided into three groups: a control group, a low-dose metformin group, and a high-dose metformin group. Following thirty days of metformin administration, blood samples, and testes tissues were collected and subjected to biochemical, histological, and ultrastructural analyses. In the groups treated with metformin, degenerative changes were observed, including irregular seminiferous tubules, disruption of epithelial integrity, a decrease in spermatogenic cells, and dilated intercellular spaces. These changes were evident in both the light and electron microscopic evaluations, and the severity varied depending on the metformin dosage. The findings showed that the diameter of the seminiferous epithelium and epithelium height decreased significantly in the high-dose group. Also, Follicle-stimulating hormone (FSH), Luteinizing hormone (LH), and testosterone levels altered significantly, with increased levels of all hormones observed in the high-dose metformin-administered group than in the control group. For the markers of oxidative stress and antioxidant status respectively there were no significant differences observed in the levels of Malondialdehyde (MDA) between the groups, however, Superoxide dismutase (SOD) activity significantly decreased in the high-dose metformin-administered group compared to the control group.
Collapse
Affiliation(s)
- Selin Ursavas
- Department of Histology and Embryology, Faculty of Medicine, Istinye University, Istanbul, Turkey; Department of Histology and Embryology, Faculty of Medicine, Cukurova University, Adana, Turkey; Department of Histology and Embryology, Hamidiye Faculty of Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, 34668, Turkey.
| | - Yurdun Kuyucu
- Department of Histology and Embryology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ebru Dundar Yenilmez
- Department of Biochemistry, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Abdullah Tuli
- Department of Biochemistry, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Muhammetnur Tekayev
- Department of Histology and Embryology, Hamidiye Faculty of Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, 34668, Turkey
| | - Ufuk O Mete
- Department of Histology and Embryology, Faculty of Medicine, Cukurova University, Adana, Turkey
| |
Collapse
|
3
|
Mason T, Alesi S, Fernando M, Vanky E, Teede HJ, Mousa A. Metformin in gestational diabetes: physiological actions and clinical applications. Nat Rev Endocrinol 2025; 21:77-91. [PMID: 39455749 DOI: 10.1038/s41574-024-01049-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2024] [Indexed: 10/28/2024]
Abstract
Metformin is an effective oral hypoglycaemic agent used in the treatment of type 2 diabetes mellitus; however, its use in pregnancy for the treatment of gestational diabetes mellitus (GDM) remains controversial owing to concerns around safety and efficacy. This comprehensive review outlines the physiological metabolic functions of metformin and synthesizes existing literature and key knowledge gaps pertaining to the use of metformin in pregnancy across various end points in women with GDM. On the basis of current evidence, metformin reduces gestational weight gain, neonatal hypoglycaemia and macrosomia and increases insulin sensitivity. However, considerable heterogeneity between existing studies and the grouping of aggregate and often inharmonious data within meta-analyses has led to disparate findings regarding the efficacy of metformin in treating hyperglycaemia in GDM. Innovative analytical approaches with stratification by individual-level characteristics (for example, obesity, ethnicity, GDM severity and so on) and treatment regimens (diagnostic criteria, treatment timing and follow-up duration) are needed to establish efficacy across a range of end points and to identify which, if any, subgroups might benefit from metformin treatment during pregnancy.
Collapse
Affiliation(s)
- Taitum Mason
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
| | - Simon Alesi
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
| | - Melinda Fernando
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Helena J Teede
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
- Department of Endocrinology and Diabetes, Monash Health, Clayton, Victoria, Melbourne, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia.
| |
Collapse
|
4
|
Newman C, Dunne FP. Treatment of Diabetes in Pregnancy With Metformin. Obstet Gynecol 2024; 144:660-669. [PMID: 39208454 DOI: 10.1097/aog.0000000000005705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 09/04/2024]
Abstract
Metformin is a commonly used drug in the treatment of type 2 diabetes and has been used to treat gestational diabetes since the 1970s. In pregnancy, its proven benefits include reduced gestational weight gain and reduced fetal size; some studies have shown reduced risk of cesarean delivery and lower rates of hypertension. Metformin can reduce the need for insulin therapy but does not eliminate such need in many patients. Despite these benefits, metformin crosses the placenta and has been associated with increases in the risk of giving birth to small-for-gestational-age neonates in some studies of individuals with type 2 diabetes in pregnancy. In addition, higher body mass index (BMI) z-scores have been observed among exposed offspring in some of the long-term follow-up studies. Nevertheless, metformin's low cost, ease of administration, and global reach make it a reasonable intervention in a population affected by rising rates of obesity and diabetes in pregnancy. Further follow-up studies are required to monitor the long-term health of exposed offspring.
Collapse
Affiliation(s)
- Christine Newman
- Institute for Clinical Trials, the HRB-Clinical Research Facility, and the College of Medicine, Nursing and Health Sciences, University of Galway, and Galway University Hospital, Galway, Ireland
| | | |
Collapse
|
5
|
Sharpe RM. Endocrine disruption and male reproductive disorders: unanswered questions. Hum Reprod 2024; 39:1879-1888. [PMID: 38926156 PMCID: PMC11373384 DOI: 10.1093/humrep/deae143] [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: 04/10/2024] [Revised: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Maternal exposure to endocrine-disrupting chemicals (EDCs) in human pregnancy is widely considered as an important cause of adverse changes in male reproductive health due to impaired foetal androgen production/action. However, the epidemiological evidence supporting this view is equivocal, except for certain phthalates, notably diethyl hexyl phthalate (DEHP). Maternal phthalate exposure levels associated with adverse reproductive changes in epidemiological studies are several thousand-fold lower than those needed to suppress foetal androgen production in rats, and direct studies using human foetal testis tissue show no effect of high phthalate exposure on androgen production. This conundrum is unexplained and raises fundamental questions. Human DEHP exposure is predominantly via food with highest exposure associated with consumption of a Western style (unhealthy) diet. This diet is also associated with increased exposure to the most common EDCs, whether persistent (chlorinated or fluorinated chemicals) or non-persistent (phthalates, bisphenols) compounds, which are found at highest levels in fatty and processed foods. Consequently, epidemiological studies associating EDC exposure and male reproductive health disorders are confounded by potential dietary effects, and vice versa. A Western diet/lifestyle in young adulthood is also associated with low sperm counts. Disentangling EDC and dietary effects in epidemiological studies is challenging. In pregnancy, a Western diet, EDC exposure, and maternal living in proximity to industrial sites are all associated with impaired foetal growth/development due to placental dysfunction, which predisposes to congenital male reproductive disorders (cryptorchidism, hypospadias). While the latter are considered to reflect impaired foetal androgen production, effects resulting from foetal growth impairment (FGI) are likely indirect. As FGI has numerous life-long health consequences, and is affected by maternal lifestyle, research into the origins of male reproductive disorders should take more account of this. Additionally, potential effects on foetal growth/foetal testis from the increasing use of medications in pregnancy deserves more research attention.
Collapse
Affiliation(s)
- Richard M Sharpe
- Centre for Reproductive Health, Institute for Regeneration & Repair, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
6
|
McEwen I, Huybrechts KF, Straub L, Hernández-Díaz S. Patterns of paternal medication dispensation around the time of conception. Paediatr Perinat Epidemiol 2024; 38:461-466. [PMID: 38949455 PMCID: PMC11365770 DOI: 10.1111/ppe.13098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Past research on the safety of prenatal exposure to medications has focused on maternal use during gestation, with limited research into the potential effects of paternal use during the spermatogenic period preceding conception. Knowing the most common medications used by fathers around the time of conception can inform research priorities in this field. OBJECTIVES To identify the most common medications dispensed to fathers in the preconception period. METHODS Within the MarketScan research database of commercially insured individuals in the United States from 2011 to 2020, we identified pregnancies, estimated the date of conception, linked each pregnancy to the father using family enrolment information and required minimum enrolment period and prescription benefits. Then, we described the use of prescription medications by the father during the 90 days before conception based on pharmacy dispensation claims. RESULTS Of 4,437,550 pregnancies, 51.6% were linked with a father. Among the 1,413,762 pregnancies connected with a father that also met the inclusion criteria, the most common classes of medications dispensed were psychotropics (8.66%), antibiotics (7.21%), and analgesics (6.82%). The most frequently dispensed medications were amoxicillin (3.75%), azithromycin (3.15%), fluticasone (2.70%) and acetaminophen/hydrocodone (2.70%). Some fathers filled prescriptions for medications associated with foetal embryopathy when used by the mother, including mycophenolate (0.04%), methotrexate (0.03%) and isotretinoin (0.02%). CONCLUSIONS More than a third of fathers filled at least one prescription medication in the preconception period, and several of them are known to be embryotoxic, emphasizing the necessity for further investigation into the potential teratogenicity of paternal exposure.
Collapse
Affiliation(s)
- Isobel McEwen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health
| | - Krista F Huybrechts
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
| | - Loreen Straub
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
| | | |
Collapse
|
7
|
Rotem RS, Weisskopf MG, Huybrechts KF, Hernández-Díaz S. Paternal Use of Metformin During the Sperm Development Period Preceding Conception and Risk for Major Congenital Malformations in Newborns. Ann Intern Med 2024; 177:851-861. [PMID: 38885501 DOI: 10.7326/m23-1405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Metformin is the most used oral antidiabetic medication. Despite its established safety profile, it has known antiandrogenic and epigenetic modifying effects. This raised concerns about possible adverse developmental effects caused by genomic alterations related to paternal use of metformin during the spermatogenesis period preceding conception. OBJECTIVE To assess the potential adverse intergenerational effect of metformin by examining the association between paternal metformin use during spermatogenesis and major congenital malformations (MCMs) in newborns. DESIGN Nationally representative cohort study. SETTING A large Israeli health fund. PARTICIPANTS 383 851 live births linked to fathers and mothers that occurred in 1999 to 2020. MEASUREMENTS MCMs and parental cardiometabolic conditions were ascertained using clinical diagnoses, medication dispensing information, and laboratory test results. The effect of metformin use on MCMs was estimated using general estimating equations, accounting for concurrent use of other antidiabetic medications and parental cardiometabolic morbidity. RESULTS Compared with unexposed fathers, the prevalence of cardiometabolic morbidity was substantially higher among fathers who used metformin during spermatogenesis, and their spouses. Whereas the crude odds ratio (OR) for paternal metformin exposure in all formulations and MCMs was 1.28 (95% CI, 1.01 to 1.64), the adjusted OR was 1.00 (CI, 0.76 to 1.31). Within specific treatment regimens, the adjusted OR was 0.86 (CI, 0.60 to 1.23) for metformin in monotherapy and 1.36 (CI, 1.00 to 1.85) for metformin in polytherapy, a treatment that was more common in patients with more poorly controlled diabetes. LIMITATION Laboratory test results for hemoglobin A1c to assess underlying diabetes severity were available only for a subset of the cohort. CONCLUSION Paternal use of metformin in monotherapy does not increase the risk for MCMs. Association for metformin in polytherapy could potentially be explained by worse underlying parental cardiometabolic risk profile. PRIMARY FUNDING SOURCE None.
Collapse
Affiliation(s)
- Ran S Rotem
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, and Kahn-Sagol-Maccabi Research and Innovation Institute, Maccabi Healthcare Services, Tel Aviv, Israel (R.S.R.)
| | - Marc G Weisskopf
- Department of Environmental Health and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts (M.G.W.)
| | - Krista F Huybrechts
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts (K.F.H.)
| | - Sonia Hernández-Díaz
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts (S.H.)
| |
Collapse
|
8
|
Shenoy MT, Mondal S, Fernandez CJ, Pappachan JM. Management of male obesity-related secondary hypogonadism: A clinical update. World J Exp Med 2024; 14:93689. [PMID: 38948417 PMCID: PMC11212738 DOI: 10.5493/wjem.v14.i2.93689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 06/19/2024] Open
Abstract
The global obesity pandemic has resulted in a rise in the prevalence of male obesity-related secondary hypogonadism (MOSH) with emerging evidence on the role of testosterone therapy. We aim to provide an updated and practical approach towards its management. We did a comprehensive literature search across MEDLINE (via PubMed), Scopus, and Google Scholar databases using the keywords "MOSH" OR "Obesity-related hypogonadism" OR "Testosterone replacement therapy" OR "Selective estrogen receptor modulator" OR "SERM" OR "Guidelines on male hypogonadism" as well as a manual search of references within the articles. A narrative review based on available evidence, recommendations and their practical implications was done. Although weight loss is the ideal therapeutic strategy for patients with MOSH, achievement of significant weight reduction is usually difficult with lifestyle changes alone in real-world practice. Therefore, androgen administration is often necessary in the management of hypogonadism in patients with MOSH which also improves many other comorbidities related to obesity. However, there is conflicting evidence for the appropriate use of testosterone replacement therapy (TRT), and it can also be associated with complications. This evidence-based review updates the available evidence including the very recently published results of the TRAVERSE trial and provides comprehensive clinical practice pearls for the management of patients with MOSH. Before starting testosterone replacement in functional hypogonadism of obesity, it would be desirable to initiate lifestyle modification to ensure weight reduction. TRT should be coupled with the management of other comorbidities related to obesity in MOSH patients. Balancing the risks and benefits of TRT should be considered in every patient before and during long-term management.
Collapse
Affiliation(s)
- Mohan T Shenoy
- Department of Endocrinology, Sree Gokulam Medical College, and Research Foundation, Trivandrum 695607, Kerala, India
| | - Sunetra Mondal
- Department of Endocrinology, NRS Medical College and Hospital, Kolkata 700014, West Bengal, India
| | - Cornelius James Fernandez
- Department of Endocrinology & Metabolism, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston PE21 9QS, United Kingdom
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| |
Collapse
|
9
|
Zhu Y, Engmann M, Medina D, Han X, Das P, Bartke A, Ellsworth BS, Yuan R. Metformin treatment of juvenile mice alters aging-related developmental and metabolic phenotypes in sex-dependent and sex-independent manners. GeroScience 2024; 46:3197-3218. [PMID: 38227136 PMCID: PMC11009201 DOI: 10.1007/s11357-024-01067-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/30/2023] [Indexed: 01/17/2024] Open
Abstract
Metformin has attracted increasing interest for its potential benefits in extending healthspan and longevity. This study examined the effects of early-life metformin treatment on the development and metabolism of C57BL/6 J (B6) mice, with metformin administered to juvenile mice from 15 to 56 days of age. Metformin treatment led to decreased body weight in both sexes (P < 0.05, t-test). At 9 weeks of age, mice were euthanized and organ weights were recorded. The relative weight of retroperitoneal fat was decreased in females, while relative weights of perigonadal and retroperitoneal fat were decreased, and relative liver weight was increased in males (P < 0.05, t-test). Glucose and insulin tolerance tests (GTT and ITT) were conducted at the age of 7 weeks. ANOVA revealed a significant impairment in insulin sensitivity by the treatment, and a significantly interactive effect on glucose tolerance between sex and treatment, underscoring a disparity in GTT between sexes in response to the treatment. Metformin treatment reduced circulating insulin levels in fasting and non-fasting conditions for male mice, with no significant alterations observed in female mice. qRT-PCR analysis of glucose metabolism-related genes (Akt2, Glut2, Glut4, Irs1, Nrip1, Pi3k, Pi3kca, Pkca) in the liver and skeletal muscle reveals metformin-induced sex- and organ-specific effects on gene expression. Comparison with previous studies in heterogeneous UM-HET3 mice receiving the same treatment suggests that genetic differences may contribute to variability in the effects of metformin treatment on development and metabolism. These findings indicate that early-life metformin treatment affects development and metabolism in both sex- and genetics-dependent manners.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Morgan Engmann
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Xiuqi Han
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Pratyusa Das
- Department of Physiology, Southern Illinois University SIU School of Medicine, 1135 Lincoln Drive, Life Science III, Room 2062, Carbondale, IL, 62901, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Buffy S Ellsworth
- Department of Physiology, Southern Illinois University SIU School of Medicine, 1135 Lincoln Drive, Life Science III, Room 2062, Carbondale, IL, 62901, USA
| | - Rong Yuan
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA.
| |
Collapse
|
10
|
Rindone GM, Dasso ME, Centola CL, Sobarzo CM, Galardo MN, Meroni SB, Riera MF. Effect of Metformin on Sertoli Cell Fatty Acid Metabolism and Blood-Testis Barrier Formation. BIOLOGY 2024; 13:330. [PMID: 38785812 PMCID: PMC11117697 DOI: 10.3390/biology13050330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Sertoli cells (SCs) are essential to maintaining germ cell development. Metformin, the main pharmacologic treatment for pediatric type 2 diabetes, is administered to children during SC maturation. The present study aimed to analyze whether metformin affects SC energy metabolism and blood-testis barrier (BTB) integrity. Primary SC cultures were used for the in vitro studies. In vivo effects were studied in Sprague-Dawley rats treated with 200 mg/kg metformin from Pnd14 to Pnd30. Metformin decreased fatty acid oxidation and increased 3-hydroxybutyrate production in vitro. Moreover, it decreased the transepithelial electrical resistance across the monolayer and induced ZO-1 redistribution, suggesting an alteration of cell junctions. In vivo, a mild but significant increase in BTB permeability and ZO-1 expression was observed in the metformin group, without changes in testicular histology and meiosis progression. Additionally, adult rats that received metformin treatment during the juvenile period showed no alteration in BTB permeability or daily sperm production. In conclusion, metformin exposure may affect BTB permeability in juvenile rats, but this seems not to influence spermatogenesis progression. Considering the results obtained in adult animals, it is possible to speculate that metformin treatment during the juvenile period does not affect testicular function in adulthood.
Collapse
Affiliation(s)
- Gustavo Marcelo Rindone
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI–División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina; (G.M.R.); (M.E.D.); (C.L.C.); (M.N.G.); (S.B.M.)
| | - Marina Ercilia Dasso
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI–División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina; (G.M.R.); (M.E.D.); (C.L.C.); (M.N.G.); (S.B.M.)
| | - Cecilia Lucia Centola
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI–División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina; (G.M.R.); (M.E.D.); (C.L.C.); (M.N.G.); (S.B.M.)
| | - Cristian Marcelo Sobarzo
- Instituto de Investigaciones Biomédicas (INBIOMED), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina;
| | - María Noel Galardo
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI–División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina; (G.M.R.); (M.E.D.); (C.L.C.); (M.N.G.); (S.B.M.)
| | - Silvina Beatriz Meroni
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI–División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina; (G.M.R.); (M.E.D.); (C.L.C.); (M.N.G.); (S.B.M.)
| | - María Fernanda Riera
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI–División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina; (G.M.R.); (M.E.D.); (C.L.C.); (M.N.G.); (S.B.M.)
| |
Collapse
|
11
|
Pourkarim F, Entezari-Maleki T. Metformin Plus Insulin for Preexisting Diabetes or Gestational Diabetes in Pregnancy. JAMA 2024; 331:1502. [PMID: 38568677 DOI: 10.1001/jama.2024.1380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Affiliation(s)
- Fariba Pourkarim
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
12
|
Paschou SA, Shalit A, Gerontiti E, Athanasiadou KI, Kalampokas T, Psaltopoulou T, Lambrinoudaki I, Anastasiou E, Wolffenbuttel BHR, Goulis DG. Efficacy and safety of metformin during pregnancy: an update. Endocrine 2024; 83:259-269. [PMID: 37798604 PMCID: PMC10850184 DOI: 10.1007/s12020-023-03550-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023]
Abstract
During the last decades, gestational diabetes mellitus (GDM) prevalence has been on the rise. While insulin remains the gold standard treatment for GDM, metformin use during pregnancy is controversial. This review aimed to comprehensively assess the available data on the efficacy and safety of metformin during pregnancy, both for the mother and the offspring. Metformin has been validated for maternal efficacy and safety, achieving comparable glycemic control with insulin. Additionally, it reduces maternal weight gain and possibly the occurrence of hypertensive disorders. During the early neonatal period, metformin administration does not increase the risk of congenital anomalies or other major adverse effects, including lower APGAR score at 5 min, neonatal intensive care unit admissions, and respiratory distress syndrome. Several studies have demonstrated a reduction in neonatal hypoglycemia. Metformin has been associated with an increase in preterm births and lower birth weight, although this effect is controversial and depends on the indication for which it was administered. Evidence indicates possible altered fetal programming and predisposition to childhood obesity and metabolic syndrome during adulthood after use of metformin in pregnancy. With critical questions still requiring a final verdict, ongoing research on the field must be conducted.
Collapse
Affiliation(s)
- Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Almog Shalit
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Gerontiti
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kleoniki I Athanasiadou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Kalampokas
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Irene Lambrinoudaki
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
13
|
Lu Z, Guo Y, Xu D, Xiao H, Dai Y, Liu K, Chen L, Wang H. Developmental toxicity and programming alterations of multiple organs in offspring induced by medication during pregnancy. Acta Pharm Sin B 2023; 13:460-477. [PMID: 36873163 PMCID: PMC9978644 DOI: 10.1016/j.apsb.2022.05.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/05/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
Abstract
Medication during pregnancy is widespread, but there are few reports on its fetal safety. Recent studies suggest that medication during pregnancy can affect fetal morphological and functional development through multiple pathways, multiple organs, and multiple targets. Its mechanisms involve direct ways such as oxidative stress, epigenetic modification, and metabolic activation, and it may also be indirectly caused by placental dysfunction. Further studies have found that medication during pregnancy may also indirectly lead to multi-organ developmental programming, functional homeostasis changes, and susceptibility to related diseases in offspring by inducing fetal intrauterine exposure to too high or too low levels of maternal-derived glucocorticoids. The organ developmental toxicity and programming alterations caused by medication during pregnancy may also have gender differences and multi-generational genetic effects mediated by abnormal epigenetic modification. Combined with the latest research results of our laboratory, this paper reviews the latest research progress on the developmental toxicity and functional programming alterations of multiple organs in offspring induced by medication during pregnancy, which can provide a theoretical and experimental basis for rational medication during pregnancy and effective prevention and treatment of drug-related multiple fetal-originated diseases.
Collapse
Affiliation(s)
- Zhengjie Lu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Dan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Hao Xiao
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| |
Collapse
|
14
|
Corpuz-Hilsabeck M, Culty M. Impact of endocrine disrupting chemicals and pharmaceuticals on Sertoli cell development and functions. Front Endocrinol (Lausanne) 2023; 14:1095894. [PMID: 36793282 PMCID: PMC9922725 DOI: 10.3389/fendo.2023.1095894] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/04/2023] [Indexed: 02/01/2023] Open
Abstract
Sertoli cells play essential roles in male reproduction, from supporting fetal testis development to nurturing male germ cells from fetal life to adulthood. Dysregulating Sertoli cell functions can have lifelong adverse effects by jeopardizing early processes such as testis organogenesis, and long-lasting processes such as spermatogenesis. Exposure to endocrine disrupting chemicals (EDCs) is recognized as contributing to the rising incidence of male reproductive disorders and decreasing sperm counts and quality in humans. Some drugs also act as endocrine disruptors by exerting off-target effects on endocrine tissues. However, the mechanisms of toxicity of these compounds on male reproduction at doses compatible with human exposure are still not fully resolved, especially in the case of mixtures, which remain understudied. This review presents first an overview of the mechanisms regulating Sertoli cell development, maintenance, and functions, and then surveys what is known on the impact of EDCs and drugs on immature Sertoli cells, including individual compounds and mixtures, and pinpointing at knowledge gaps. Performing more studies on the impact of mixtures of EDCs and drugs at all ages is crucial to fully understand the adverse outcomes these chemicals may induce on the reproductive system.
Collapse
|
15
|
Wen J, Yi Z, Chen Y, Huang J, Mao X, Zhang L, Zeng Y, Cheng Q, Ye W, Liu Z, Liu F, Liu J. Efficacy of metformin therapy in patients with cancer: a meta-analysis of 22 randomised controlled trials. BMC Med 2022; 20:402. [PMID: 36280839 PMCID: PMC9594974 DOI: 10.1186/s12916-022-02599-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/10/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND To investigate whether metformin monotherapy or adjunctive therapy improves the prognosis in patients with any type of cancer compared to non-metformin users (age ≥18). METHODS Databases (Medline, Embase, and the Cochrane Central Register of Controlled Trials) and clinical trial registries ( ClinicalTrials.gov ; the World Health Organization International Clinical Trials Registry Platform) were screened for randomized, controlled trials (RCT) reporting at least progression-free survival (PFS) and/or overall survival (OS). Main outcome measures included hazard ratios (HR), and combined HRs and 95% confidence intervals (CI) were calculated using random-effects models. RESULTS Of the 8419 records screened, 22 RCTs comprising 5943 participants were included. Pooled HRs were not statistically significant in both PFS (HR 0.97, 95% CI 0.82-1.15, I2 = 50%) and OS (HR 0.98, 95% CI 0.86-1.13, I2 = 33%) for patients with cancer between the metformin and control groups. Subgroup analyses demonstrated that metformin treatment was associated with a marginally significant improvement in PFS in reproductive system cancers (HR 0.86, 95% CI 0.74-1.00) and a significantly worse PFS in digestive system cancers (HR 1.45, 95% CI 1.03-2.04). The PFS or OS was observed consistently across maintenance dose, diabetes exclusion, median follow-up, risk of bias, and combined antitumoral therapies. CONCLUSION Metformin treatment was not associated with cancer-related mortality in adults compared with placebo or no treatment. However, metformin implied beneficial effects in the PFS of the patients with reproductive system cancers but was related to a worse PFS in digestive system cancers. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration number CRD42022324672.
Collapse
Affiliation(s)
- Jie Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuyao Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jing Huang
- National Clinical Research Center for Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xueyi Mao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenrui Ye
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Jingfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
16
|
de Oliveira JS, Silva AADN, Dias FCR, de Oliveira EL, de Oliveira Filho EF, Soares PC, Ferreira CMDO, da Silva Junior VA. Histomorphometric and oxidative evaluation of the offspring's testis from type 2 diabetic female rats treated with metformin and pentoxifylline. Int J Exp Pathol 2022; 103:174-189. [PMID: 35734873 PMCID: PMC9482357 DOI: 10.1111/iep.12446] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 11/27/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) during pregnancy is characterized by high levels of reactive oxygen species and pro-inflammatory factors in the placenta. Once these reactive species reach the foetus, they trigger physiological adaptations that allow the foetus to survive, but programme the organism to develop metabolic disorders in adulthood. The male reproductive system is highly susceptible to foetal programming. This study aimed to investigate the effects of intrauterine exposure to T2D on testicular histomorphometry and redox homeostasis of adult rats and evaluate the effects of maternal treatment with metformin and pentoxifylline. Female rats were induced to T2D, then treated with metformin and pentoxifylline, or co-treated with both drugs. The females were mated, the male offspring were sacrificed on postnatal day 90, and the testicles were collected for analysis. Metformin protected the tubular compartment, with the maintenance of the Sertoli cell population and daily sperm production. Pentoxifylline attenuated the effects of diabetes on Leydig cells, in addition to stimulating testosterone production and lowering lipid peroxidation. Intrauterine exposure to T2D results in important testicular alterations that compromise gonadal function, and the co-treatment with metformin and pentoxifylline may represent a promising therapy that attenuates these effects by combining the positive influences in both the tubular and interstitial compartments of the testicular parenchyma.
Collapse
Affiliation(s)
| | | | | | | | | | - Pierre Castro Soares
- Department of Veterinary MedicineFederal Rural University of PernambucoRecifePernambucoBrazil
| | | | | |
Collapse
|
17
|
Zhao J, Yang PC, Yang H, Wang ZB, El-Samahy M, Wang F, Zhang YL. Dietary supplementation with metformin improves testis function and semen quality and increases antioxidants and autophagy capacity in goats. Theriogenology 2022; 188:79-89. [DOI: 10.1016/j.theriogenology.2022.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/10/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022]
|
18
|
Triggle CR, Mohammed I, Bshesh K, Marei I, Ye K, Ding H, MacDonald R, Hollenberg MD, Hill MA. Metformin: Is it a drug for all reasons and diseases? Metabolism 2022; 133:155223. [PMID: 35640743 DOI: 10.1016/j.metabol.2022.155223] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Metformin was first used to treat type 2 diabetes in the late 1950s and in 2022 remains the first-choice drug used daily by approximately 150 million people. An accumulation of positive pre-clinical and clinical data has stimulated interest in re-purposing metformin to treat a variety of diseases including COVID-19. In polycystic ovary syndrome metformin improves insulin sensitivity. In type 1 diabetes metformin may help reduce the insulin dose. Meta-analysis and data from pre-clinical and clinical studies link metformin to a reduction in the incidence of cancer. Clinical trials, including MILES (Metformin In Longevity Study), and TAME (Targeting Aging with Metformin), have been designed to determine if metformin can offset aging and extend lifespan. Pre-clinical and clinical data suggest that metformin, via suppression of pro-inflammatory pathways, protection of mitochondria and vascular function, and direct actions on neuronal stem cells, may protect against neurodegenerative diseases. Metformin has also been studied for its anti-bacterial, -viral, -malaria efficacy. Collectively, these data raise the question: Is metformin a drug for all diseases? It remains unclear as to whether all of these putative beneficial effects are secondary to its actions as an anti-hyperglycemic and insulin-sensitizing drug, or result from other cellular actions, including inhibition of mTOR (mammalian target for rapamycin), or direct anti-viral actions. Clarification is also sought as to whether data from ex vivo studies based on the use of high concentrations of metformin can be translated into clinical benefits, or whether they reflect a 'Paracelsus' effect. The environmental impact of metformin, a drug with no known metabolites, is another emerging issue that has been linked to endocrine disruption in fish, and extensive use in T2D has also raised concerns over effects on human reproduction. The objectives for this review are to: 1) evaluate the putative mechanism(s) of action of metformin; 2) analyze the controversial evidence for metformin's effectiveness in the treatment of diseases other than type 2 diabetes; 3) assess the reproducibility of the data, and finally 4) reach an informed conclusion as to whether metformin is a drug for all diseases and reasons. We conclude that the primary clinical benefits of metformin result from its insulin-sensitizing and antihyperglycaemic effects that secondarily contribute to a reduced risk of a number of diseases and thereby enhancing healthspan. However, benefits like improving vascular endothelial function that are independent of effects on glucose homeostasis add to metformin's therapeutic actions.
Collapse
Affiliation(s)
- Chris R Triggle
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar.
| | - Ibrahim Mohammed
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Khalifa Bshesh
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Isra Marei
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Kevin Ye
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Hong Ding
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Ross MacDonald
- Distribution eLibrary, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, a Cumming School of Medicine, University of Calgary, T2N 4N1, Canada
| | - Michael A Hill
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia 65211, MO, USA
| |
Collapse
|
19
|
Tseng CH. Metformin’s effects on varicocele, erectile dysfunction, infertility and prostate-related diseases: A retrospective cohort study. Front Pharmacol 2022; 13:799290. [PMID: 35935880 PMCID: PMC9355151 DOI: 10.3389/fphar.2022.799290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/30/2022] [Indexed: 12/20/2022] Open
Abstract
Objectives: To investigate the risk of varicocele, erectile dysfunction (ED), infertility, prostatitis, benign prostate hyperplasia (BPH) and prostate cancer associated with metformin use. Materials and methods: A total of 261,838 males, mean age 52.39 years (SD: 11.39), with a new-onset type 2 diabetes mellitus in 1999–2009 were identified from Taiwan’s National Health Insurance. Among them, 175,171 were metformin initiators [metformin (+)] and 86,667 were non-metformin initiators [metformin (−)] in the initial 12-month prescriptions of antidiabetic drugs. Follow-up started after the initial 12-month prescriptions. Outcomes were followed up until 31 December 2011. Intention-to-treat (ITT) and per-protocol (PP) hazard ratios comparing metformin (+) to metformin (−) were estimated by Cox regression incorporated with the inverse probability of treatment-weighting using propensity scores. Results: The median follow-up time ranged 5.55–6.82 years in metformin (−) and 4.36–5.17 years in metformin (+) for different outcomes in ITT analyses. The respective median follow-up time in PP analyses ranged 2.20–2.61 years in metformin (−) and ranged 3.99–4.65 years in metformin (+). In the ITT analyses, for metformin (−), the incidence rates (per 100,000 person-years) of varicocele, ED, infertility, prostatitis, BPH and prostate cancer were 26.42, 455.89, 22.82, 590.23, 4226.19, and 141.69, respectively; and the respective incidence rates for metformin (+) were 25.65, 488.10, 32.60, 510.30, 3685.66, and 116.57. The hazard ratios (95% confidence intervals) comparing metformin (+) to metformin (−) in the ITT analyses were 0.960 (0.784–1.174) for varicocele, 1.077 (1.026–1.130) for ED, 1.368 (1.116–1.676) for infertility, 0.887 (0.849–0.927) for prostatitis, 0.883 (0.868–0.899) for BPH and 0.878 (0.802–0.961) for prostate cancer. The hazard ratios for the respective outcomes in the PP analyses were 0.845 (0.662–1.078), 1.350 (1.264–1.441), 1.396 (1.078–1.808), 0.800 (0.756–0.846), 0.875 (0.855–0.895), and 0.613 (0.548–0.686). Conclusion: Metformin use in patients with type 2 diabetes mellitus is associated with a neutral effect on varicocele, a higher risk of sexual dysfunction (ED and infertility) and a reduced risk of prostate-related health (prostatitis, BPH and prostate cancer).
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Environmental Health and Occupational Medicine of the National Health Research Institutes, Zhunan, Taiwan
- *Correspondence: Chin-Hsiao Tseng,
| |
Collapse
|
20
|
Lundgaard Riis M, Jørgensen A. Deciphering Sex-Specific Differentiation of Human Fetal Gonads: Insight From Experimental Models. Front Cell Dev Biol 2022; 10:902082. [PMID: 35721511 PMCID: PMC9201387 DOI: 10.3389/fcell.2022.902082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Sex-specific gonadal differentiation is initiated by the expression of SRY in male foetuses. This promotes a signalling pathway directing testicular development, while in female foetuses the absence of SRY and expression of pro-ovarian factors promote ovarian development. Importantly, in addition to the initiation of a sex-specific signalling cascade the opposite pathway is simultaneously inhibited. The somatic cell populations within the gonads dictates this differentiation as well as the development of secondary sex characteristics via secretion of endocrine factors and steroid hormones. Opposing pathways SOX9/FGF9 (testis) and WNT4/RSPO1 (ovary) controls the development and differentiation of the bipotential mouse gonad and even though sex-specific gonadal differentiation is largely considered to be conserved between mice and humans, recent studies have identified several differences. Hence, the signalling pathways promoting early mouse gonad differentiation cannot be directly transferred to human development thus highlighting the importance of also examining this signalling in human fetal gonads. This review focus on the current understanding of regulatory mechanisms governing human gonadal sex differentiation by combining knowledge of these processes from studies in mice, information from patients with differences of sex development and insight from manipulation of selected signalling pathways in ex vivo culture models of human fetal gonads.
Collapse
Affiliation(s)
- Malene Lundgaard Riis
- Department of Growth and Reproduction, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Anne Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
21
|
Wensink MJ, Lu Y, Tian L, Shaw GM, Rizzi S, Jensen TK, Mathiesen ER, Skakkebæk NE, Lindahl-Jacobsen R, Eisenberg ML. Preconception Antidiabetic Drugs in Men and Birth Defects in Offspring : A Nationwide Cohort Study. Ann Intern Med 2022; 175:665-673. [PMID: 35344380 PMCID: PMC9844982 DOI: 10.7326/m21-4389] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Diabetes reduces semen quality and increasingly occurs during reproductive years. Diabetes medications, such as metformin, have glucose-independent effects on the male reproductive system. Associations with birth defects in offspring are unknown. OBJECTIVE To evaluate whether the risk for birth defects in offspring varies with preconceptional pharmacologic treatment of fathers with diabetes. DESIGN Nationwide prospective registry-based cohort study. SETTING Denmark from 1997 to 2016. PARTICIPANTS All liveborn singletons from mothers without histories of diabetes or essential hypertension. MEASUREMENTS Offspring were considered exposed if their father filled 1 or more prescriptions for a diabetes drug during the development of fertilizing sperm. Sex and frequencies of major birth defects were compared across drugs, times of exposure, and siblings. RESULTS Of 1 116 779 offspring included, 3.3% had 1 or more major birth defects (reference). Insulin-exposed offspring (n = 5298) had the reference birth defect frequency (adjusted odds ratio [aOR], 0.98 [95% CI, 0.85 to 1.14]). Metformin-exposed offspring (n = 1451) had an elevated birth defect frequency (aOR, 1.40 [CI, 1.08 to 1.82]). For sulfonylurea-exposed offspring (n = 647), the aOR was 1.34 (CI, 0.94 to 1.92). Offspring whose fathers filled a metformin prescription in the year before (n = 1751) or after (n = 2484) sperm development had reference birth defect frequencies (aORs, 0.88 [CI, 0.59 to 1.31] and 0.92 [CI, 0.68 to 1.26], respectively), as did unexposed siblings of exposed offspring (3.2%; exposed vs. unexposed OR, 1.54 [CI, 0.94 to 2.53]). Among metformin-exposed offspring, genital birth defects, all in boys, were more common (aOR, 3.39 [CI, 1.82 to 6.30]), while the proportion of male offspring was lower (49.4% vs. 51.4%, P = 0.073). LIMITATION Information on underlying disease status was limited. CONCLUSION Preconception paternal metformin treatment is associated with major birth defects, particularly genital birth defects in boys. Further research should replicate these findings and clarify the causation. PRIMARY FUNDING SOURCE National Institutes of Health.
Collapse
Affiliation(s)
- Maarten J Wensink
- Department of Epidemiology, Biostatistics and Biodemography, and Interdisciplinary Center on Population Dynamics, University of Southern Denmark, Odense C, Denmark (M.J.W., R.L.)
| | - Ying Lu
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California (Y.L., L.T.)
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California (Y.L., L.T.)
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California (G.M.S.)
| | - Silvia Rizzi
- Interdisciplinary Center on Population Dynamics and Department of Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, Odense M, Denmark (S.R.)
| | - Tina Kold Jensen
- Department of Environmental Medicine, University of Southern Denmark, Odense C, Denmark (T.K.J.)
| | - Elisabeth R Mathiesen
- Centre for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen University, Copenhagen, Denmark (E.R.M.)
| | - Niels E Skakkebæk
- Juliane Marie Centre, Department of Growth and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark (N.E.S.)
| | - Rune Lindahl-Jacobsen
- Department of Epidemiology, Biostatistics and Biodemography, and Interdisciplinary Center on Population Dynamics, University of Southern Denmark, Odense C, Denmark (M.J.W., R.L.)
| | - Michael L Eisenberg
- Male Reproductive Medicine and Surgery, Department of Urology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California (M.L.E.)
| |
Collapse
|
22
|
Tavlo M, Skakkebæk NE, Mathiesen ER, Kristensen DM, Kjær KH, Andersson AM, Lindahl-Jacobsen R. Hypothesis: Metformin is a potential reproductive toxicant. Front Endocrinol (Lausanne) 2022; 13:1000872. [PMID: 36339411 PMCID: PMC9627511 DOI: 10.3389/fendo.2022.1000872] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/05/2022] [Indexed: 11/25/2022] Open
Abstract
Metformin is the first-line oral treatment for type 2 diabetes mellitus and is prescribed to more than 150 million people worldwide. Metformin's effect as a glucose-lowering drug is well documented but the precise mechanism of action is unknown. A recent finding of an association between paternal metformin treatment and increased numbers of genital birth defects in sons and a tendency towards a skewed secondary sex ratio with less male offspring prompted us to focus on other evidence of reproductive side effects of this drug. Metformin in humans is documented to reduce the circulating level of testosterone in both men and women. In experimental animal models, metformin exposure in utero induced sex-specific reproductive changes in adult rat male offspring with reduced fertility manifested as a 30% decrease in litter size and metformin exposure to fish, induced intersex documented in testicular tissue. Metformin is excreted unchanged into urine and feces and is present in wastewater and even in the effluent of wastewater treatment plants from where it spreads to rivers, lakes, and drinking water. It is documented to be present in numerous freshwater samples throughout the world - and even in drinking water. We here present the hypothesis that metformin needs to be considered a potential reproductive toxicant for humans, and probably also for wildlife. There is an urgent need for studies exploring the association between metformin exposure and reproductive outcomes in humans, experimental animals, and aquatic wildlife.
Collapse
Affiliation(s)
- Maja Tavlo
- Faculty of Health Sciences, Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense C, Denmark
- Interdisciplinary Center on Population Dynamics, University of Southern Denmark, Odense C, Denmark
- *Correspondence: Maja Tavlo,
| | - Niels E. Skakkebæk
- Department of Growth and Reproduction, Copenhagen University Hospital — Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth R. Mathiesen
- Centre for Pregnant Women with Diabetes, Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David M. Kristensen
- Department of Neurology, Danish Headache Center, Rigshospitalet - Glostrup, University of Copenhagen, Copenhagen, Denmark
- University of Rennes, Inserm, École des hautes études en santé publique (EHESP), Irset (Institut de recherche en santé environment et travail) UMR_S, Rennes, France
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kurt H. Kjær
- Globe Institute, Section for GeoGenetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction, Copenhagen University Hospital — Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Rune Lindahl-Jacobsen
- Faculty of Health Sciences, Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense C, Denmark
- Interdisciplinary Center on Population Dynamics, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
23
|
Leisegang K, Roychoudhury S, Slama P, Finelli R. The Mechanisms and Management of Age-Related Oxidative Stress in Male Hypogonadism Associated with Non-communicable Chronic Disease. Antioxidants (Basel) 2021; 10:1834. [PMID: 34829704 PMCID: PMC8615233 DOI: 10.3390/antiox10111834] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Androgens have diverse functions in muscle physiology, lean body mass, the regulation of adipose tissue, bone density, neurocognitive regulation, and spermatogenesis, the male reproductive and sexual function. Male hypogonadism, characterized by reduced testosterone, is commonly seen in ageing males, and has a complex relationship as a risk factor and a comorbidity in age-related noncommunicable chronic diseases (NCDs), such as obesity, metabolic syndrome, type 2 diabetes, and malignancy. Oxidative stress, as a significant contributor to the ageing process, is a common feature between ageing and NCDs, and the related comorbidities, including hypertension, dyslipidemia, hyperglycemia, hyperinsulinemia, and chronic inflammation. Oxidative stress may also be a mediator of hypogonadism in males. Consequently, the management of oxidative stress may represent a novel therapeutic approach in this context. Therefore, this narrative review aims to discuss the mechanisms of age-related oxidative stress in male hypogonadism associated with NCDs and discusses current and potential approaches for the clinical management of these patients, which may include conventional hormone replacement therapy, nutrition and lifestyle changes, adherence to the optimal body mass index, and dietary antioxidant supplementation and/or phytomedicines.
Collapse
Affiliation(s)
- Kristian Leisegang
- School of Natural Medicine, Faculty of Community and Health Sciences, Bellville, Cape Town 7535, South Africa
| | | | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, 61300 Brno, Czech Republic
| | | |
Collapse
|
24
|
Faure MC, Khoueiry R, Quanico J, Acloque H, Guerquin MJ, Bertoldo MJ, Chevaleyre C, Ramé C, Fournier I, Salzet M, Dupont J, Froment P. In Utero Exposure to Metformin Reduces the Fertility of Male Offspring in Adulthood. Front Endocrinol (Lausanne) 2021; 12:750145. [PMID: 34745014 PMCID: PMC8565088 DOI: 10.3389/fendo.2021.750145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Metformin is a drug used for the treatment of type 2 diabetes and disorders associated with insulin resistance. Metformin is also used in the treatment of pregnancy disorders such as gestational diabetes. However, the consequences of foetal exposure to metformin on the fertility of exposed offspring remain poorly documented. In this study, we investigated the effect of in utero metformin exposure on the fertility of female and male offspring. We observed that metformin is detectable in the blood of the mother and in amniotic fluid and blood of the umbilical cord. Metformin was not measurable in any tissues of the embryo, including the gonads. The effect of metformin exposure on offspring was sex specific. The adult females that had been exposed to metformin in utero presented no clear reduction in fertility. However, the adult males that had been exposed to metformin during foetal life exhibited a 30% reduction in litter size compared with controls. The lower fertility was not due to a change in sperm production or the motility of sperm. Rather, the phenotype was due to lower sperm head quality - significantly increased spermatozoa head abnormality with greater DNA damage - and hypermethylation of the genomic DNA in the spermatozoa associated with lower expression of the ten-eleven translocation methylcytosine dioxygenase 1 (TET1) protein. In conclusion, while foetal metformin exposure did not dramatically alter gonad development, these results suggest that metabolic modification by metformin during the foetal period could change the expression of epigenetic regulators such as Tet1 and perturb the genomic DNA in germ cells, changes that might contribute to a reduced fertility.
Collapse
Affiliation(s)
- Mélanie C. Faure
- l’Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), UMR85 Physiologie de la Reproduction et des Comportements/Centre national de la Recherche Scientifique (CNRS), UMR7247/Université François Rabelais de Tours/Institut français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Rita Khoueiry
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Jusal Quanico
- Université Lille 1, INSERM U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Villeneuve d’Ascq, France
| | - Hervé Acloque
- Université Paris-Saclay, INRAE, AgroParisTech, Génétique Animale et Biologie Intégrative (GABI), Jouy-en-Josas, France
| | - Marie-Justine Guerquin
- UMR967 INSERM, Commissariat à l'Énergie Atomique (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut de Radiobiologie Cellulaire et Moléculaire (iRCM)/Service Cellules Souches et Radiation (SCSR)/LDG, Université Paris Diderot, Sorbonne Paris Cité, Université Paris-Sud, Université Paris-Saclay, Laboratory of Development of the Gonads, Fontenay aux Roses, France
| | - Michael J. Bertoldo
- Fertility and Research Centre, School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Claire Chevaleyre
- l’Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), UMR85 Physiologie de la Reproduction et des Comportements/Centre national de la Recherche Scientifique (CNRS), UMR7247/Université François Rabelais de Tours/Institut français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Christelle Ramé
- l’Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), UMR85 Physiologie de la Reproduction et des Comportements/Centre national de la Recherche Scientifique (CNRS), UMR7247/Université François Rabelais de Tours/Institut français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Isabelle Fournier
- Université Lille 1, INSERM U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Villeneuve d’Ascq, France
| | - Michel Salzet
- Université Lille 1, INSERM U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Villeneuve d’Ascq, France
| | - Joëlle Dupont
- l’Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), UMR85 Physiologie de la Reproduction et des Comportements/Centre national de la Recherche Scientifique (CNRS), UMR7247/Université François Rabelais de Tours/Institut français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Pascal Froment
- l’Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), UMR85 Physiologie de la Reproduction et des Comportements/Centre national de la Recherche Scientifique (CNRS), UMR7247/Université François Rabelais de Tours/Institut français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| |
Collapse
|
25
|
Vieira HR, Gonçalves GD, Alves VS, de Melo MAB, Borges SC, Klagenberg J, Neves CQ, Previate C, Saavedra LPJ, Siervo GEMDL, Malta A, Prado MAADC, Palma-Rigo K, Buttow NC, Fernandes GSA, Mathias PCDF. Neonatal metformin short exposure inhibits male reproductive dysfunction caused by a high-fat diet in adult rats. Toxicol Appl Pharmacol 2021; 429:115712. [PMID: 34481828 DOI: 10.1016/j.taap.2021.115712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 01/06/2023]
Abstract
Metformin (Met) is widely used to control blood glucose levels and acts on various organs, including reproductive tissues, to improve reproductive and lifespan. This study evaluated whether neonatal Met exposure prevented male reproductive dysfunction caused by being overweight during adulthood. Randomized Wistar rat pups received an intraperitoneal injection from postnatal days (PNDs) 1 to 12of saline (Sal; 0.9% NaCl/day in 2mL/kg) or Met (100 mg/kg/day in 2 mL/kg). From PNDs 60 to 90, the animals received a regular (R; 4.5% fat; Sal R and Met R groups) or a high-fat (HF; 35% fat; Sal HF and Met HF groups) diet. At PND 90, all animals were euthanized to evaluate their biometric and reproductive parameters. The Sal and Met groups with R showed similar body weights, however, the HF diet increased the body weight in both groups. The Sal HF group showed testicular damage regarding in antioxidant status and inflammatory profile in the epididymal cauda. The HF diet reduced Leydig and Sertoli cells numbers, with lower sperm quality. The Met R animals showed positive reproductive programming, due to improved antioxidant defense, inflammatory biomarkers, and sperm morphology. Met HF prevented HF diet damage to reproductive organs and sperm morphology, but not to sperm motility. Early Met exposure positively affected the male reproductive system of adult rats, preventing reproductive HF disorders. STATEMENT OF NOVELTY AND SIGNIFICANCE: Metformin is used to control type 2 diabetes mellitus and can act to improve metabolism and lifespan. Metformin avoidance is recommended during pregnancy, but there is no information regarding its use when breastfeeding. For the first time, we showed in this current study that metformin positively acts in the male reproductive tissues and helps involved in later life. These data showed a better antioxidant defense and anti-inflammatory profile of Metformin animals than Saline animals and might directly improve reproductive organs morphophysiology and sperm morphology. Also, the neonatal Met application programs the male reproduction to counterbalance damages from an obesogenic environment in later life.
Collapse
Affiliation(s)
- Henrique Rodrigues Vieira
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil; Department of Anatomy, Institute of Biomedical Science III, University of São Paulo (USP), Av. Prof. Lineu Prestes, 2415, CEP: 05508-000 São Paulo, São Paulo, Brazil.
| | - Gessica Dutra Gonçalves
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Vander Silva Alves
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Milene Aparecida Bobato de Melo
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Stephanie Carvalho Borges
- Department of Morphological Sciences, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Josana Klagenberg
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Camila Quaglio Neves
- Department of Morphological Sciences, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Carina Previate
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Gláucia Eloisa Munhoz de Lion Siervo
- Department of General Biology, Biological Sciences Center, State University of Londrina (UEL), Rodovia Celso Garcia Cid, PR 445, CEP: 86057-970 Londrina, Paraná, Brazil
| | - Ananda Malta
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Marialba Avezum Alves de Castro Prado
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Kesia Palma-Rigo
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil; Faculdade Adventista Paranaense, PR-317 Km 119 Gleba, R. Paiçandu, Lote 80 - Zona Rural, CEP: 87130-000 Ivatuba - Paraná, Brazil
| | - Nilza Cristina Buttow
- Department of Morphological Sciences, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| | - Glaura Scantamburlo Alves Fernandes
- Department of General Biology, Biological Sciences Center, State University of Londrina (UEL), Rodovia Celso Garcia Cid, PR 445, CEP: 86057-970 Londrina, Paraná, Brazil
| | - Paulo Cezar de Freitas Mathias
- Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá (UEM), Av. Colombo, 5790, CEP: 87020-900 Maringá, Paraná, Brazil
| |
Collapse
|
26
|
Phillips J, Akemann C, Shields JN, Wu CC, Meyer DN, Baker BB, Pitts DK, Baker TR. Developmental phenotypic and transcriptomic effects of exposure to nanomolar levels of metformin in zebrafish. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 87:103716. [PMID: 34311114 PMCID: PMC8446320 DOI: 10.1016/j.etap.2021.103716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 05/06/2023]
Abstract
Metformin is found in the majority of lakes and streams in the United States, leading to widespread environmental exposure. Results of the present study indicate that extended duration metformin exposure at critical developmental periods leads to decreased survival rates in zebrafish (danio rerio), an NIH approved human model. Significant abnormalities are seen with extended duration metformin exposure from 4 h post fertilization up to 5 days post fertilization, although short term metformin exposure for 24 h at 4-5 days post fertilization did not lead to any significant abnormalities. Both extended and short term duration did however have an impact on locomotor activity of zebrafish, and several genes involved in neurological and cardiovascular development were differentially expressed after exposure to metformin. The changes seen in behavior, gene expression and morphological abnormalities caused by metformin exposure should be examined further in future studies in order to assess their potential human health implications as metformin prescriptions continue to increase worldwide.
Collapse
Affiliation(s)
- Jessica Phillips
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Camille Akemann
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Jeremiah N Shields
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Chia-Chen Wu
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Danielle N Meyer
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Bridget B Baker
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - David K Pitts
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Tracie R Baker
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA.
| |
Collapse
|
27
|
Hanem LGE, Salvesen Ø, Madsen A, Sagen JV, Mellgren G, Juliusson PB, Carlsen SM, Vanky E, Ødegård R. Maternal PCOS status and metformin in pregnancy: Steroid hormones in 5-10 years old children from the PregMet randomized controlled study. PLoS One 2021; 16:e0257186. [PMID: 34499672 PMCID: PMC8428669 DOI: 10.1371/journal.pone.0257186] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/20/2021] [Indexed: 11/18/2022] Open
Abstract
Objective Polycystic ovary syndrome (PCOS) is a common endocrine disorder, with potential effects on offspring both genetically and through altered intrauterine environment. Metformin, which ameliorate hormonal disturbances in non-pregnant women with PCOS is increasingly used in pregnancy. It passes the placenta, and the evidence on potential consequences for offspring endocrine development is scarce. We explore the potential effects of maternal PCOS status and intrauterine metformin exposure on offspring steroid hormone levels. Design This is a follow-up study of 5–10 years old children from the PregMet-study–a randomized controlled trial comparing metformin (2000 mg/day) to placebo during PCOS pregnancies. Of the 255 children invited, 117 (46%) were included. Methods There was no intervention in this follow-up study. Outcomes were serum levels of androstenedione, testosterone, SHBG, cortisol, 17-hydroxyprogesterone, 11-deoxycortisol and calculated free testosterone converted to gender-and age adjusted z-scores from a Norwegian reference population. These were compared in i) placebo-exposed children versus children from the reference population (z-score zero) by the deviation in z-score by one-sample t-tests and ii) metformin versus placebo-exposed children by two-sample t-tests. Holm-Bonferroni adjustments were performed to account for multiple endpoints. Results Girls of mothers with PCOS (n = 30) had higher mean z-scores of androstenedione (0.73 (95% confidence interval (CI) 0.41 to 1.06), p<0.0001), testosterone (0.76 (0.51 to 1.00), p<0.0001), and free testosterone (0.99 (0.67 to 1.32), p<0.0001) than the reference population. Metformin-exposed boys (n = 31) tended to have higher 11-deoxycortisol z-score than placebo-exposed boys (n = 24) (mean difference 0.65 (95% CI 0.14–1.17), p = 0.014). Conclusion Maternal PCOS status was associated with elevated androgens in 5- to 10-year-old daughters, which might indicate earlier maturation and increased risk of developing PCOS. An impact of metformin in pregnancy on steroidogenesis in children born to mothers with PCOS cannot be excluded. Our findings need confirmation in studies that include participants that have entered puberty.
Collapse
Affiliation(s)
- Liv Guro Engen Hanem
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Children’s clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- * E-mail:
| | - Øyvind Salvesen
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - André Madsen
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Jørn V. Sagen
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
- KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway
| | - Petur Benedikt Juliusson
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
- Department of Health Registries, Norwegian Institute of Public Health, Bergen, Norway
| | - Sven Magnus Carlsen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Obstetrics and Gynecology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Rønnaug Ødegård
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Children’s clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Centre for Obesity Research, Dept. of Surgery St. Olav University Hospital, Trondheim, Norway
| |
Collapse
|
28
|
Estradiol ameliorates metformin-inhibited Sertoli cell proliferation via AMPK/TSC2/mTOR signaling pathway. Theriogenology 2021; 175:7-22. [PMID: 34481229 DOI: 10.1016/j.theriogenology.2021.08.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/31/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022]
Abstract
Metformin is a commonly used for treating type 2 diabetes and it acts on a variety of organs including the male reproductive system. 17β-estradiol plays an important role in Sertoli cell (SC) proliferation which determines the germ cell development and spermatogenesis. The aim of this study is to investigate the effect of metformin on immature chicken SC proliferation and the potential mechanisms by which 17β-estradiol regulate this process. Results showed that metformin significantly inhibited SC proliferation, whereas 17β-estradiol weakened the inhibitory effects of metformin on SC viability, cell growth, and cell cycle progression. SC proliferation-inhibiting effect of metformin exposure was regulated by decreasing adenosine triphosphate level and respiratory enzyme activity in the mitochondria; this process was possibly mediated by the adenosine monophosphate-activated protein kinase (AMPK)/tuberous sclerosis complex 2 (TSC2)/mammalian target of rapamycin (mTOR) signaling pathway, which was regulated by the down-expressed miR-1764 and by the decreased antioxidant enzyme activity and excessive reactive oxygen species generation. In addition, SCs transfected with the miR-1764 agomir led to an improvement of proliferation capacity through down-regulating AMPKα2 level, which further decreased TSC2 expression and induced mTOR activation. However, the anti-proliferative effect of miR-1764 antagomir can be alleviated by 17β-estradiol treatment via the up-expression of miR-1764 in transfected SCs. Our findings suggest appropriate dose of exogenous 17β-estradiol treatment can ameliorate the inhibitory effect of metformin on SC proliferation via the regulation of AMPK/TSC2/mTOR signaling pathway, this might reduce the risk of poor male fertility caused by the abuse of anti-diabetic agents.
Collapse
|
29
|
Connan-Perrot S, Léger T, Lelandais P, Desdoits-Lethimonier C, David A, Fowler PA, Mazaud-Guittot S. Six Decades of Research on Human Fetal Gonadal Steroids. Int J Mol Sci 2021; 22:ijms22136681. [PMID: 34206462 PMCID: PMC8268622 DOI: 10.3390/ijms22136681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
Human fetal gonads acquire endocrine steroidogenic capabilities early during their differentiation. Genetic studies show that this endocrine function plays a central role in the sexually dimorphic development of the external genitalia during fetal development. When this endocrine function is dysregulated, congenital malformations and pathologies are the result. In this review, we explain how the current knowledge of steroidogenesis in human fetal gonads has benefited from both the technological advances in steroid measurements and the assembly of detailed knowledge of steroidogenesis machinery and its expression in human fetal gonads. We summarise how the conversion of radiolabelled steroid precursors, antibody-based assays, mass spectrometry, ultrastructural studies, and the in situ labelling of proteins and mRNA have all provided complementary information. In this review, our discussion goes beyond the debate on recommendations concerning the best choice between the different available technologies, and their degrees of reproducibility and sensitivity. The available technologies and techniques can be used for different purposes and, as long as all quality controls are rigorously employed, the question is how to maximise the generation of robust, reproducible data on steroid hormones and their crucial roles in human fetal development and subsequent functions.
Collapse
Affiliation(s)
- Stéphane Connan-Perrot
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, 35000 Rennes, France; (S.C.-P.); (P.L.); (C.D.-L.); (A.D.)
| | - Thibaut Léger
- Fougères Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), CEDEX, 35306 Fougères, France;
| | - Pauline Lelandais
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, 35000 Rennes, France; (S.C.-P.); (P.L.); (C.D.-L.); (A.D.)
| | - Christèle Desdoits-Lethimonier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, 35000 Rennes, France; (S.C.-P.); (P.L.); (C.D.-L.); (A.D.)
| | - Arthur David
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, 35000 Rennes, France; (S.C.-P.); (P.L.); (C.D.-L.); (A.D.)
| | - Paul A. Fowler
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK;
| | - Séverine Mazaud-Guittot
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, 35000 Rennes, France; (S.C.-P.); (P.L.); (C.D.-L.); (A.D.)
- Correspondence: ; Tel.: +33-2-23-23-58-86
| |
Collapse
|
30
|
Zhang FF, Zhang Q, Wang YL, Wang FF, Hardiman PJ, Qu F. Intergenerational Influences between Maternal Polycystic Ovary Syndrome and Offspring: An Updated Overview. J Pediatr 2021; 232:272-281. [PMID: 33482217 DOI: 10.1016/j.jpeds.2021.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Fang-Fang Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan-Lin Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang-Fang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
31
|
Rindone GM, Gorga A, Pellizzari EH, Camberos MDC, Galardo MN, Da Ros VG, Buffone MG, Meroni SB, Riera MF. Postnatal metformin treatment alters rat Sertoli cell proliferation and daily sperm production. Andrology 2021; 9:965-976. [PMID: 33305512 DOI: 10.1111/andr.12957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The direct correlation between Sertoli cell number and sperm production capacity highlights the importance of deciphering external factors that modify Sertoli cell proliferation. A growing body of evidence in vitro suggests that metformin, the main pharmacological agent for type 2 diabetes treatment in children, exerts anti-proliferative effects on Sertoli cells. OBJECTIVE The aims of this study were to investigate the effect of metformin administration during postnatal period on Sertoli cell proliferation and on cell cycle regulators expression and to analyze the impact of this treatment on the sperm production capacity in adulthood. MATERIALS AND METHODS Sprague Dawley rat pups were randomly divided into two groups: MET (receiving daily 200 mg/kg metformin, from Pnd3 to Pnd7 inclusive) and control (receiving vehicle). BrdU incorporation was measured to assess proliferation. Gene expression analyses were performed in Sertoli cells isolated from animals of both groups. Daily sperm production and sperm parameters were measured in adult male rats (Pnd90) that received neonatal treatment. RESULTS MET group exhibited a significant decrease in BrdU incorporation in Sertoli cells. Concordantly, MET group showed a reduction in cyclin D1 and E2 expression and an increase in p21 expression in Sertoli cells. In addition, metformin-treated animals displayed lower values of daily sperm production on Pnd90. DISCUSSION AND CONCLUSION These results suggest that metformin treatment may lead to a decrease in Sertoli cell proliferation, a concomitant altered expression of cell cycle regulators and ultimately, a reduction in daily sperm production in adult animals.
Collapse
Affiliation(s)
- Gustavo Marcelo Rindone
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Agostina Gorga
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Eliana Herminia Pellizzari
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - María Del Carmen Camberos
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - María Noel Galardo
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | | | | | - Silvina Beatriz Meroni
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - María Fernanda Riera
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
32
|
Ambrosio-Albuquerque EP, Cusioli LF, Bergamasco R, Sinópolis Gigliolli AA, Lupepsa L, Paupitz BR, Barbieri PA, Borin-Carvalho LA, de Brito Portela-Castro AL. Metformin environmental exposure: A systematic review. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 83:103588. [PMID: 33460803 DOI: 10.1016/j.etap.2021.103588] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/20/2020] [Accepted: 01/11/2021] [Indexed: 05/20/2023]
Abstract
This review discussed the occurrence, ecological impacts, and effects of metformin, a drug used for type 2 diabetes among other diseases. It is one of the most commonly found medicines in aquatic environments owing to its incomplete metabolism in the human body, and is eventually disposed in wastewater. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses were followed as a guide. After searching various databases, 48 eligible studies were selected for the review. Metformin reportedly occurs in different environmental matrices, as measurable concentrations of metformin are found in sewage (urban and hospital), influent/sludge/effluent from wastewater treatment plants, surface water (rivers, lakes, estuaries, oceans, and non-specific sources), tap/drinking water, and sediment (lake and recipient seawaters). Data on metformin detection in aquatic environments in 14 countries were studied, but a consensus on the risk patterns of pharmaceutical products was not determined. Many studies have been conducted on different test organisms, demonstrating that metformin can drive the expression of diverse genes, particularly those responsible for endocrine hormone pathways. Chronic exposure to metformin can be tested using models and other tools to understand this field, which remains largely unexplored. Our results contribute to the current ecotoxicology knowledge related to typically used drugs and provide a basis for further investigations.
Collapse
Affiliation(s)
| | | | - Rosângela Bergamasco
- Department of Chemical Engineering, State University of Maringa, Maringá, Brazil
| | | | - Luara Lupepsa
- Ambiental Biotechnology Pos Graduated Program, State University of Maringá, Maringá, Brazil
| | | | - Pablo Américo Barbieri
- Ambiental Biotechnology Pos Graduated Program, State University of Maringá, Maringá, Brazil
| | | | | |
Collapse
|
33
|
Tseng CH. The Effect of Metformin on Male Reproductive Function and Prostate: An Updated Review. World J Mens Health 2021; 40:11-29. [PMID: 33831975 PMCID: PMC8761231 DOI: 10.5534/wjmh.210001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/02/2022] Open
Abstract
Metformin is the first-line oral antidiabetic drug that shows multiple pleiotropic effects of anti-inflamation, anti-cancer, anti-aging, anti-microbia, anti-atherosclerosis, and immune modulation. Metformin's effects on men's related health are reviewed here, focusing on reproductive health under subtitles of erectile dysfunction (ED), steroidogenesis and spermatogenesis; and on prostate-related health under subtitles of prostate specific antigen (PSA), prostatitis, benign prostate hyperplasia (BPH), and prostate cancer (PCa). Updated literature suggests a potential role of metformin on arteriogenic ED but controversial and contradictory effects (either protective or harmful) on testicular functions of testosterone synthesis and spermatogenesis. With regards to prostate-related health, metformin use may be associated with lower levels of PSA in humans, but its clinical implications require more research. Although there is a lack of research on metform's effect on prostatitis, it may have potential benefits through its anti-microbial and anti-inflammatory properties. Metformin may reduce the risk of BPH by inhibiting the insulin-like growth factor 1 pathway and some but not all studies suggest a protective role of metformin on the risk of PCa. Many clinical trials are being conducted to investigate the use of metformin as an adjuvant therapy for PCa but results currently available are not conclusive. While some trials suggest a benefit in reducing the metastasis and recurrence of PCa, others do not show any benefit. More research works are warranted to illuminate the potential usefulness of metformin in the promotion of men's health.
Collapse
Affiliation(s)
- Chin Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Division of Environmental Health and Occupational Medicine of the National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
34
|
Ebokaiwe AP, Obeten KE, Okori SO, David EE, Olusanya O, Chukwu CJ, Okoro N, Ehiri RC. Co-administration of Selenium Nanoparticles and Metformin Abrogate Testicular Oxidative Injury by Suppressing Redox Imbalance, Augmenting Sperm Quality and Nrf2 Protein Expression in Streptozotocin-Induced Diabetic Rats. Biol Trace Elem Res 2020; 198:544-556. [PMID: 32103411 DOI: 10.1007/s12011-020-02082-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Selenium nanoparticles (SeNPs) and metformin (Met) elicit individually protective effects against testicular oxidative injury in diabetic rats. However, the combined effects of both compounds have not been investigated. We investigated the effects of SeNPs and Met individual/co-treatment on testicular oxidative injury in diabetic rats. Diabetes was induced by a single intraperitoneal administration of streptozotocin (STZ-40 mg/kg bwt). The rats were equally divided into 6 groups: Group one-non-diabetic; group two-diabetic untreated; and group six-non-diabetic received citrate buffer (2 mL/kg bwt), while group three, four, and five received SeNPs (0.1 mg/kg bwt), Met (50 mg/kg bwt), and SeNPs/Met combined respectively, for 42 days. Results revealed that SeNPs, as well as Met treatment significantly (p < 0.001), lowered blood glucose levels and improved relative organ weights in treated rats than those of the untreated group. Moreover, a synergistic effect was observed in the co-administration group. Additionally, combined treatment elicited better effect, in augmenting the pituitary and testicular hormone (LH, FSH, prolactin, and testosterone) levels, marker enzymes/protein associated with steroidogenesis (3-βHSD, 17-βHSD, and StAR protein), and sperm functional parameters than those of individual treatment groups, when compared with control. Furthermore, the combinatorial effects of SeNPs and Met surpassed their influence in attenuating testicular oxidative stress/inflammation and upregulation of Nrf2 protein expression in diabetic rats when compared with control. Overall, normal rats, co-treated with SeNPs and Met, did not reveal any deleterious effect. Therefore, SeNPs and Met combined treatment may better improve testes function in diabetic conditions than an individual regimen.
Collapse
Affiliation(s)
- Azubuike P Ebokaiwe
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria.
| | - Kebe E Obeten
- Department of Anatomy, Cross River University of Technology, Cross River State, Okuku, Nigeria
| | - Stephen O Okori
- Department of Anatomy, Cross River University of Technology, Cross River State, Okuku, Nigeria
| | - Ebuka E David
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | | | - Chukwuma J Chukwu
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | - Nworie Okoro
- Department of Biology/Microbiology/Biotechnology, Alex-Ekwueme Federal University, Ndufu-Alike, Nigeria
| | - Richard C Ehiri
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| |
Collapse
|
35
|
Nna VU, Bakar ABA, Ahmad A, Mohamed M. Diabetes-induced testicular oxidative stress, inflammation, and caspase-dependent apoptosis: the protective role of metformin. Arch Physiol Biochem 2020; 126:377-388. [PMID: 30513216 DOI: 10.1080/13813455.2018.1543329] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Context: Metformin's effect on glycaemic control is well documented, but its effect on diabetes-induced testicular impairment has been scarcely reported.Objective: To investigate the effects of metformin on testicular oxidative stress, inflammation, and apoptosis, which largely contribute to fertility decline in diabetic state.Methods: Male Sprague-Dawley rats were divided into 3 groups (n = 6/group) namely: normal control (NC), diabetic control (DC), and metformin (300 mg/kg b.w./d)-treated diabetic groups. Metformin was administrated for 4 weeks.Results: Decreased mRNA expressions and activities of antioxidant enzymes were seen in the testes of DC group. mRNA and protein expressions of pro-inflammatory and pro-apoptotic markers increased, while interleukin-10 and proliferating cell nuclear antigen (PCNA) decreased in the testes of DC group. Treatment with metformin up-regulated antioxidant enzymes, down-regulated inflammation, and apoptosis and increased PCNA immunoexpression in the testes.Conclusions: Metformin protects the testes from diabetes-induced impairment and may improve male reproductive health in diabetic state.
Collapse
Affiliation(s)
- Victor Udo Nna
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Ainul Bahiyah Abu Bakar
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Azlina Ahmad
- Basic Science and Oral Biology Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- Unit of Integrative Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
36
|
Yang W, Wang L, Wang F, Yuan S. Roles of AMP-Activated Protein Kinase (AMPK) in Mammalian Reproduction. Front Cell Dev Biol 2020; 8:593005. [PMID: 33330475 PMCID: PMC7710906 DOI: 10.3389/fcell.2020.593005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/23/2020] [Indexed: 12/01/2022] Open
Abstract
Reproduction is an energy demanding function and only take place in case of sufficient available energy status in mammals. Metabolic diseases such as anorexia nervosa are clinically associated with reduced fertility. AMP-activated protein kinase (AMPK), as a major regulator of cellular energy homeostasis, is activated in limited energy reserves to ensure the orderly progress of various physiological activities. In recent years, mounting evidence shows that AMPK is involved in the regulation of reproductive function through multiple mechanisms. AMPK is likely to be a metabolic sensor integrating central and peripheral signals. In this review, we aim to explore the preclinical studies published in the last decade that investigate the role of AMP-activated protein kinase in the reproductive field, and its role as a target for drug therapy of reproductive system-related diseases. We also emphasized the emerging roles of AMPK in transcriptional regulation of reproduction processes and metabolisms, which are tightly related to the energy state and fertility of an organism.
Collapse
Affiliation(s)
- Weina Yang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingjuan Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengli Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Effects of short-term exposure to high-fat diet on histology of male and female gonads in rats. Acta Histochem 2020; 122:151558. [PMID: 32622421 DOI: 10.1016/j.acthis.2020.151558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
Abstract
Obesity, which reaches an epidemic, is characterized by alterations in metabolic and hormonal profiles. Moreover, uncontrolled obesity may lead to development of diabetes type 2, which accounts for about 90% of all diabetic cases. In obesity, besides changes in metabolism, numerous co-morbidities are reported, e.g. disruptions of reproductive functions. Additionally, sex differences in development of this disease occur. We hypothesized that short-term exposure to high-fat diet (HFD; containing 50% of total energy from fat) would alter histology of testes and ovaries, and thus contribute to reproductive disruptions in male and female rats. Adult rats were fed ad libitum with HFD for 6-7 weeks and its effects on histology of testes and ovaries (n = 4/sex and treatment group) were studied using hematoxylin-eosin staining followed by microscopic analysis and compared to control (laboratory chow fed) group. We have found that in male rats fed with HFD there were: 1) decrease in diameter of seminiferous tubules due to smaller luminar diameter, and no change in epithelium height; 2) decrease in number of Sertoli cells; 3) no changes in number of spermatogonia and in percentage of semen in seminiferous tubules. In female rats exposed to HFD we have seen: 1) decrease in diameters of corpora lutea; 2) decrease in diameter of ovarian follicles types 7 and 8, but no changes in their number; 3) no changes in number of early primary follicles, primary follicles, and secondary follicles. We concluded that relatively short-term exposure to HFD in rats leads to changes in histology of both testes and ovaries, thus affecting reproductive functions.
Collapse
|
38
|
Meng Y, Xiang R, Yan H, Zhou Y, Hu Y, Yang J, Zhou Y, Cui Q. Transcriptomic landscape profiling of metformin-treated healthy mice: Implication for potential hypertension risk when prophylactically used. J Cell Mol Med 2020; 24:8138-8150. [PMID: 32529766 PMCID: PMC7348147 DOI: 10.1111/jcmm.15472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Recently, the first-line anti-diabetic drug metformin shows versatile protective effects against several diseases and is potentially prescribed to healthy individual for prophylactic use against ageing or other pathophysiological processes. However, for healthy individuals, it remains unclear what effects metformin treatment will induce on their bodies. A systematic profiling of the molecular landscape of metformin treatment is expected to provide crucial implications for this issue. Here, we delineated the first transcriptomic landscape induced by metformin in 10 tissues (aorta, brown adipose, brain, eye, heart, liver, kidney, skeletal muscle, stomach and testis) of healthy mice by using RNA-sequencing technique. A comprehensive computational analysis was performed. The overrepresentation of cardiovascular disease-related gene sets, positive correlation with hypertension-related transcriptomic signatures and the associations of drugs with hypertensive side effect together indicate that although metformin does exert various beneficial effects, it would also increase the risk of hypertension in healthy mice. This prediction was experimentally validated by an independent animal experiments. Together, this study provided important resource necessary for investigating metformin's beneficial/deleterious effects on various healthy tissues, when it is potentially prescribed to healthy individual for prophylactic use.
Collapse
Affiliation(s)
- Yuhong Meng
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| | - Rui Xiang
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| | - Han Yan
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| | - Yiran Zhou
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| | - Yuntao Hu
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| | - Jichun Yang
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| | - Yuan Zhou
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| | - Qinghua Cui
- Department of Physiology and PathophysiologyDepartment of Biomedical InformaticsCenter for Non‐coding RNA MedicineMOE Key Lab of Cardiovascular SciencesSchool of Basic Medical SciencesPeking UniversityBeijingChina
| |
Collapse
|
39
|
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
|
40
|
Kilcoyne KR, Mitchell RT. Effect of environmental and pharmaceutical exposures on fetal testis development and function: a systematic review of human experimental data. Hum Reprod Update 2020; 25:397-421. [PMID: 30869130 PMCID: PMC6601394 DOI: 10.1093/humupd/dmz004] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/06/2018] [Accepted: 01/23/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Overall, the incidence of male reproductive disorders has increased in recent decades. Testicular development during fetal life is crucial for subsequent male reproductive function. Non-genomic factors such as environmental chemicals, pharmaceuticals and lifestyle have been proposed to impact on human fetal testicular development resulting in subsequent effects on male reproductive health. Whilst experimental studies using animal models have provided support for this hypothesis, more recently a number of experimental studies using human tissues and cells have begun to translate these findings to determine direct human relevance. OBJECTIVE AND RATIONALE The objective of this systematic review was to provide a comprehensive description of the evidence for effects of prenatal exposure(s) on human fetal testis development and function. We present the effects of environmental, pharmaceutical and lifestyle factors in experimental systems involving exposure of human fetal testis tissues and cells. Comparison is made with existing epidemiological data primarily derived from a recent meta-analysis. SEARCH METHODS For identification of experimental studies, PubMed and EMBASE were searched for articles published in English between 01/01/1966 and 13/07/2018 using search terms including ‘endocrine disruptor’, ‘human’, ‘fetal’, ‘testis’, ‘germ cells’, ‘testosterone’ and related search terms. Abstracts were screened for selection of full-text articles for further interrogation. Epidemiological studies involving exposure to the same agents were extracted from a recent systematic review and meta-analysis. Additional studies were identified through screening of bibliographies of full-texts of articles identified through the initial searches. OUTCOMES A total of 25 experimental studies and 44 epidemiological studies were included. Consistent effects of analgesic and phthalate exposure on human fetal germ cell development are demonstrated in experimental models, correlating with evidence from epidemiological studies and animal models. Furthermore, analgesic-induced reduction in fetal testosterone production, which predisposes to the development of male reproductive disorders, has been reported in studies involving human tissues, which also supports data from animal and epidemiological studies. However, whilst reduced testosterone production has been demonstrated in animal studies following exposure(s) to a variety of environmental chemicals including phthalates and bisphenol A, these effects are not reproduced in experimental approaches using human fetal testis tissues. WIDER IMPLICATIONS Direct experimental evidence for effects of prenatal exposure(s) on human fetal testis development and function exists. However, for many exposures the data is limited. The increasing use of human-relevant models systems in which to determine the effects of environmental exposure(s) (including mixed exposures) on development and function of human tissues should form an important part of the process for assessment of such exposures by regulatory bodies to take account of animal–human differences in susceptibility.
Collapse
Affiliation(s)
- Karen R Kilcoyne
- MRC Centre for Reproductive Health, The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, UK.,Royal Hospital for Sick Children, Edinburgh, UK
| |
Collapse
|
41
|
Yang Y, Chen H, Weng S, Pan T, Chen W, Wang F, Luo T, Tang Y. In vitro exposure to metformin activates human spermatozoa at therapeutically relevant concentrations. Andrology 2020; 8:663-670. [PMID: 31944615 DOI: 10.1111/andr.12755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/18/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Metformin, a drug used to treat type 2 diabetes, has gained attention for its multiple therapeutic applications. However, little is known about its effects on human sperm function at therapeutically relevant concentration. OBJECTIVES The aim of this study was to elucidate the in vitro actions of metformin on human sperm function and explore the underlying mechanism of any effects. MATERIALS AND METHODS Human ejaculated spermatozoa were treated with therapeutically relevant concentrations (0.25, 5, 10, 20, 40, and 80 µM) of metformin in vitro. Fertilization-essential functions of spermatozoa were examined, including viability, motility, capacitation, acrosome reaction, hyperactivation, and penetration ability. The signaling pathways mediated by 5'-AMP-activated protein kinase (AMPK), intracellular calcium concentration ([Ca2+ ]i ), and tyrosine phosphorylation of spermatozoa were also measured. RESULTS Although metformin did not affect sperm viability, motility, and [Ca2+ ]i , it significantly increased the percentages of capacitated spermatozoa, acrosomal-reacted spermatozoa, and hyperactivated spermatozoa as well as penetration ability of human spermatozoa at the concentrations of 40 and 80 µM (P < .05). These concentrations of metformin also elevated the levels of phosphorylated AMPK and tyrosine phosphorylation in human spermatozoa. In addition, activation of AMPK by A769662 (an AMPK activator) had similar effects to metformin on human spermatozoa, while inhibition of AMPK by Compound C (an AMPK inhibitor) suppressed the enhancement of metformin on human spermatozoa. CONCLUSION Our findings indicate that metformin activates human sperm function through an AMPK-related mechanism which increases tyrosine phosphorylation at therapeutically relevant concentrations, thereby suggesting its improvement on human sperm function when treating subfertile males of type 2 diabetes.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Houyang Chen
- Reproductive Medical Center, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Shiqi Weng
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, China
| | - Tingting Pan
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, China
| | - Wenqiong Chen
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, China
| | - Fang Wang
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, China
| | - Tao Luo
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, China
| | - Yuxin Tang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, China.,Reproductive Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
42
|
Calle-Guisado V, Gonzalez-Fernandez L, Martin-Hidalgo D, Garcia-Marin LJ, Bragado MJ. Metformin inhibits human spermatozoa motility and signalling pathways mediated by protein kinase A and tyrosine phosphorylation without affecting mitochondrial function. Reprod Fertil Dev 2019; 31:787-795. [PMID: 30562475 DOI: 10.1071/rd18256] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/12/2018] [Indexed: 12/18/2022] Open
Abstract
Metformin is a leading antidiabetic drug that is used worldwide in the treatment of diabetes mellitus. This biguanide exerts metabolic and pleiotropic effects in somatic cells, although its invitro actions on human spermatozoa remain unknown. The present study investigated the effects of metformin on human sperm function. Human spermatozoa were incubated in the presence or absence of 10mM metformin for 8 or 20h, and motility was measured by computer-aided sperm analysis (CASA); other parameters were evaluated by flow cytometry. Metformin significantly reduced the percentage of motile, progressive and rapid spermatozoa and significantly decreased sperm velocity. Metformin did not affect viability, mitochondrial membrane potential (MMP) or mitochondrial superoxide anion generation of human spermatozoa at any time studied. However, metformin clearly inhibited the protein kinase (PK) A pathway and protein tyrosine phosphorylation at 8 and 20h, key regulatory pathways for correct sperm function. In summary, metformin treatment of human spermatozoa had a detrimental effect on motility and inhibited essential sperm signalling pathways, namely PKA and protein tyrosine phosphorylation, without affecting physiological parameters (viability, MMP, mitochondrial superoxide anion generation). Given the growing clinical use of metformin in different pathologies in addition to diabetes, this study highlights an adverse effect of metformin on spermatozoa and its relevance in terms of human fertility in patients who potentially could be treated with metformin in the future.
Collapse
Affiliation(s)
- V Calle-Guisado
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), Avda Universidad, University of Extremadura, 10003 Caceres, Spain
| | - L Gonzalez-Fernandez
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), Avda Universidad, University of Extremadura, 10003 Caceres, Spain
| | - D Martin-Hidalgo
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), Avda Universidad, University of Extremadura, 10003 Caceres, Spain
| | - L J Garcia-Marin
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), Avda Universidad, University of Extremadura, 10003 Caceres, Spain
| | - M J Bragado
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), Avda Universidad, University of Extremadura, 10003 Caceres, Spain
| |
Collapse
|
43
|
Tavares RS, Escada-Rebelo S, Sousa MI, Silva A, Ramalho-Santos J, Amaral S. Can Antidiabetic Drugs Improve Male Reproductive (Dys)Function Associated with Diabetes? Curr Med Chem 2019; 26:4191-4222. [PMID: 30381064 DOI: 10.2174/0929867325666181101111404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 07/25/2018] [Accepted: 10/05/2018] [Indexed: 12/19/2022]
Abstract
The alarming increase in the number of diabetic patients worldwide raises concerns regarding the impact of the disease on global health, not to mention on social and economic aspects. Furthermore, the association of this complex metabolic disorder with male reproductive impairment is worrying, mainly due to the increasing chances that young individuals, at the apex of their reproductive window, could be affected by the disease, further contributing to the disturbing decline in male fertility worldwide. The cornerstone of diabetes management is glycemic control, proven to be effective in avoiding, minimizing or preventing the appearance or development of disease-related complications. Nonetheless, the possible impact of these therapeutic interventions on male reproductive function is essentially unexplored. To address this issue, we have made a critical assessment of the literature on the effects of several antidiabetic drugs on male reproductive function. While the crucial role of insulin is clear, as shown by the recovery of reproductive impairments in insulin-deficient individuals after treatment, the same clearly does not apply to other antidiabetic strategies. In fact, there is an abundance of controversial reports, possibly related to the various study designs, experimental models and compounds used, which include biguanides, sulfonylureas, meglitinides, thiazolidinediones/glitazones, bile acid sequestrants, amylin mimetics, as well as sodiumglucose co-transporter 2 (SGLT2) inhibitors, glucagon-like peptide 1 (GLP1), α-glucosidase inhibitors and dipeptidyl peptidase 4 (DPP4) inhibitors. These aspects constitute the focus of the current review.
Collapse
Affiliation(s)
- R S Tavares
- Biology of Reproduction and Stem Cell Group, CNC- Center for Neuroscience and Cell Biology, University of Coimbra 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - S Escada-Rebelo
- Biology of Reproduction and Stem Cell Group, CNC- Center for Neuroscience and Cell Biology, University of Coimbra 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - M I Sousa
- Biology of Reproduction and Stem Cell Group, CNC- Center for Neuroscience and Cell Biology, University of Coimbra 3004-504 Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - A Silva
- Biology of Reproduction and Stem Cell Group, CNC- Center for Neuroscience and Cell Biology, University of Coimbra 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - J Ramalho-Santos
- Biology of Reproduction and Stem Cell Group, CNC- Center for Neuroscience and Cell Biology, University of Coimbra 3004-504 Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - S Amaral
- Biology of Reproduction and Stem Cell Group, CNC- Center for Neuroscience and Cell Biology, University of Coimbra 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
44
|
Cannarella R, Arato I, Condorelli RA, Mongioì LM, Lilli C, Bellucci C, La Vignera S, Luca G, Mancuso F, Calogero AE. Effects of Insulin on Porcine Neonatal Sertoli Cell Responsiveness to FSH In Vitro. J Clin Med 2019; 8:jcm8060809. [PMID: 31174276 PMCID: PMC6617126 DOI: 10.3390/jcm8060809] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022] Open
Abstract
There is ongoing debate as to whether the decline of sperm production in recent times may be related to a parallel increase in the rate of obesity and diabetes. Lower anti-Müllerian hormone (AMH) and inhibin B secretion have been observed in young hyperinsulinemic patients compared to healthy controls, suggesting a Sertoli cell (SC) dysfunction. The pathophysiological mechanisms underlying SC dysfunction in these patients are poorly understood. To the best of our knowledge, no evidence is available on the effects of insulin on SC function. Therefore, this study was undertaken to assess the effects of insulin on basal and follicle-stimulating hormone (FSH)-stimulated SC function in vitro. To accomplish this, we evaluated the expression of AMH, inhibin B and FSHR genes, the secretion of AMH and inhibin B and the phosphorylation of AKT473 and SC proliferation on neonatal porcine SC after incubation with FSH and/or insulin. We found that similar to FSH, the expression and secretion of AMH is suppressed by insulin. Co-incubation with FSH and insulin decreased AMH secretion significantly more than with FSH alone. Insulin had no effect on the expression and secretion of the inhibin B gene, but co-incubation with FSH and insulin had a lower effect on inhibin B secretion than that found with FSH alone. FSH and/or insulin increased AKT473 phosphorylation and SC proliferation. In conclusion, the results of this study showed that insulin modulates SC function. We hypothesize that hyperinsulinemia may therefore influence testicular function even before puberty begins. Therefore, particular care should be taken to avoid the onset of hyperinsulinemia in children to prevent a future deleterious effect on fertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Iva Arato
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Laura M Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Cinzia Lilli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Catia Bellucci
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Francesca Mancuso
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
45
|
Sun X, Tavenier A, Deng W, Leishman E, Bradshaw HB, Dey SK. Metformin attenuates susceptibility to inflammation-induced preterm birth in mice with higher endocannabinoid levels. Biol Reprod 2019; 98:208-217. [PMID: 29228105 DOI: 10.1093/biolre/iox164] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/29/2017] [Indexed: 12/11/2022] Open
Abstract
Premature decidual senescence is a contributing factor to preterm birth. Fatty acid amide hydrolase mutant females (Faah-/-) with higher endocannabinoid levels are also more susceptible to preterm birth upon lipopolysaccharide (LPS) challenge due to enhanced decidual senescence; this is associated with mitogen-activated protein kinase p38 activation. Previous studies have shown that mechanistic target of rapamycin complex 1 (mTORC1) contributes to decidual senescence and promotes the incidence of preterm birth. In this study, we sought to attenuate premature decidual aging in Faah-/- females by targeting mTORC1 and p38 signaling pathways. Because metformin is known to inhibit mTOR and p38 signaling pathways, Faah-/- females were treated with metformin. These mice had a significantly lower preterm birth incidence with a higher rate of live birth after an LPS challenge on day 16 of pregnancy; metformin treatment did not affect placentation or neonatal birth weight. These results were associated with decreased levels of p38, as well as pS6, a downstream mediator of mTORC1 activity, in day 16 Faah-/-decidual tissues. Since metformin treatment attenuates premature decidual senescence with limited side effects during pregnancy, careful use of this drug may be effective in ameliorating specific adverse pregnancy events.
Collapse
Affiliation(s)
- Xiaofei Sun
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alexandra Tavenier
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Wenbo Deng
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Emma Leishman
- Department of Psychological and Brain Sciences, Kinsey Institute for Research in Sex, Gender, and Reproduction, Indiana University, Bloomington, Indiana, USA
| | - Heather B Bradshaw
- Department of Psychological and Brain Sciences, Kinsey Institute for Research in Sex, Gender, and Reproduction, Indiana University, Bloomington, Indiana, USA
| | - Sudhansu K Dey
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
46
|
Forcato S, Montagnini BG, de Góes MLM, da Silva Novi DRB, Inhasz Kiss AC, Ceravolo GS, Ceccatto Gerardin DC. Reproductive evaluations in female rat offspring exposed to metformin during intrauterine and intrauterine/lactational periods. Reprod Toxicol 2019; 87:1-7. [PMID: 31055052 DOI: 10.1016/j.reprotox.2019.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 02/08/2023]
Abstract
Metformin (MET) is a widely-used drug for the treatment of type 2 diabetes mellitus and gestational diabetes. It is known that metformin crosses the placenta and can to be transferred through milk. In vitro studies show that MET decreases gonadotropin-releasing hormone and gonadotropins release in rat neurons, and decreases progesterone and estradiol in rat granulosa cells and androstenedione synthesis in human theca cells. This study evaluated whether MET maternal exposure might interfere with reproductive parameters of female offspring. Wistar female rats were treated with MET 293 mg/kg/day, by gavage, from gestational day (GD) 0 to GD 21 (METG) or GD 0 until lactation day (LD) 21 (METGL). Controls groups received water. An increase in plasmatic estradiol levels was observed during the estrus stage in the METGL group. This result suggests that exposure to MET during gestational and lactational periods might be related to programming in theca and/or granulosa cells during development.
Collapse
Affiliation(s)
- Simone Forcato
- Department of Physiological Sciences, Center of Biological Sciences, Londrina State University, Londrina, PR, Brazil
| | - Bruno Garcia Montagnini
- Department of Physiological Sciences, Center of Biological Sciences, Londrina State University, Londrina, PR, Brazil
| | - Maria Luiza Marino de Góes
- Department of Physiological Sciences, Center of Biological Sciences, Londrina State University, Londrina, PR, Brazil
| | | | | | - Graziela Scalianti Ceravolo
- Department of Physiological Sciences, Center of Biological Sciences, Londrina State University, Londrina, PR, Brazil
| | | |
Collapse
|
47
|
Xu Q, Xie Q. Long-term effects of prenatal exposure to metformin on the health of children based on follow-up studies of randomized controlled trials: a systematic review and meta-analysis. Arch Gynecol Obstet 2019; 299:1295-1303. [PMID: 30953188 DOI: 10.1007/s00404-019-05124-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/16/2019] [Indexed: 01/11/2023]
Abstract
PURPOSE Oral antidiabetic medication of metformin is increasingly used in pregnant women with gestational diabetes mellitus (GDM), polycystic ovary syndrome (PCOS) and obesity. The drug passes through the placenta and can potentially influence the fetus. The aim of the study is to investigate the possible long-term effects of prenatal exposure to metformin on growth and development of the offspring. METHODS A systematic review and meta-analysis was conducted to examine the longer term outcomes by the follow-up studies of the already published RCTs focusing on the body composition, metabolic parameters and neurophysiological development of the children prenatally exposed to metformin. The primary sources of the reviewed studies through August 2018, with restriction on the language of English, were Pubmed and Embase. RESULTS 11 follow-up studies were included, with a maximal age of children being 13 years, comprising 823 children of mothers with GDM or PCOS who were randomized to either metformin or insulin/placebo during pregnancy. From the pooled meta-analysis we found that children prenatal exposure to metformin were associated with a significantly heavier weight (MD = 0.48 kg, 95% CI 0.24 kg, 0.73 kg; P = 0.0001, I2 = 0). As for other parameters of body composition, metabolic parameters and neurophysiological development, the results were similar between metformin and placebo/insulin use. CONCLUSION Increased offspring weight was more observed in children prenatal exposure to metformin. Heathcare providers and patients should be aware that metformin is increasingly prescribed in pregnancy based on the relatively limited evidences but nonetheless encouraging long-term offspring data are available.
Collapse
Affiliation(s)
- Qing Xu
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Putian College, No. 181 East Meiyuan Road, Lichen District, Putian City, 351100, Fujian, China.
| | - Qinglian Xie
- Health Evaluation Center, Affiliated Hospital of Putian College, Putian City, 351100, Fujian, China
| |
Collapse
|
48
|
Hart R. Generational Health Impact of PCOS on Women and their Children. Med Sci (Basel) 2019; 7:medsci7030049. [PMID: 30889922 PMCID: PMC6473601 DOI: 10.3390/medsci7030049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a metabolic disorder with reproductive consequences. Hence, the synergy of the dual maternal challenges of difficulties with conception, set on a background of metabolic disorder and inflammation, understandably leads to increased obstetric risk for the woman. Furthermore, she is more likely than her peers to require assistance with conception, either through induction of ovulation with the attendant risk of a multiple gestation, or in vitro fertilization (IVF) with its recognized increased obstetric risk for woman and her child. The increased obstetric risk for a woman with PCOS is manifested with an increased rate of miscarriage, gestational diabetes, hypertensive disorder and premature delivery. These obstetric complications are due to impairment of placental function, systemic inflammation and metabolic disorder and are markers for the woman herself of her predisposition to cardiometabolic disorder in later life. Consequently, it is inevitable that this environment may induce changes in the fetus during pregnancy, leading to an intergenerational risk from maternal PCOS.
Collapse
Affiliation(s)
- Roger Hart
- Division of Obstetrics and Gynaecology, Medical School, University of Western Australia, Perth, WA 6008, Australia.
- Fertility Specialists of Western Australia, Bethesda Hospital, 25 Queenslea Drive, Claremont, WA 6010, Australia.
- Division of Obstetrics & Gynaecology, King Edward Memorial Hospital, Subiaco, Perth, WA 6008, Australia.
| |
Collapse
|
49
|
Meroni SB, Galardo MN, Rindone G, Gorga A, Riera MF, Cigorraga SB. Molecular Mechanisms and Signaling Pathways Involved in Sertoli Cell Proliferation. Front Endocrinol (Lausanne) 2019; 10:224. [PMID: 31040821 PMCID: PMC6476933 DOI: 10.3389/fendo.2019.00224] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/21/2019] [Indexed: 12/16/2022] Open
Abstract
Sertoli cells are somatic cells present in seminiferous tubules which have essential roles in regulating spermatogenesis. Considering that each Sertoli cell is able to support a limited number of germ cells, the final number of Sertoli cells reached during the proliferative period determines sperm production capacity. Only immature Sertoli cells, which have not established the blood-testis barrier, proliferate. A number of hormonal cues regulate Sertoli cell proliferation. Among them, FSH, the insulin family of growth factors, activin, and cytokines action must be highlighted. It has been demonstrated that cAMP/PKA, ERK1/2, PI3K/Akt, and mTORC1/p70SK6 pathways are the main signal transduction pathways involved in Sertoli cell proliferation. Additionally, c-Myc and hypoxia inducible factor are transcription factors which participate in the induction by FSH of various genes of relevance in cell cycle progression. Cessation of proliferation is a pre-requisite to Sertoli cell maturation accompanied by the establishment of the blood-testis barrier. With respect to this barrier, the participation of androgens, estrogens, thyroid hormones, retinoic acid and opioids has been reported. Additionally, two central enzymes that are involved in sensing cell energy status have been associated with the suppression of Sertoli cell proliferation, namely AMPK and Sirtuin 1 (SIRT1). Among the molecular mechanisms involved in the cessation of proliferation and in the maturation of Sertoli cells, it is worth mentioning the up-regulation of the cell cycle inhibitors p21Cip1, p27Kip, and p19INK4, and of the gap junction protein connexin 43. A decrease in Sertoli cell proliferation due to administration of certain therapeutic drugs and exposure to xenobiotic agents before puberty has been experimentally demonstrated. This review focuses on the hormones, locally produced factors, signal transduction pathways, and molecular mechanisms controlling Sertoli cell proliferation and maturation. The comprehension of how the final number of Sertoli cells in adulthood is established constitutes a pre-requisite to understand the underlying causes responsible for the progressive decrease in sperm production that has been observed during the last 50 years in humans.
Collapse
|
50
|
Nna VU, Bakar ABA, Ahmad A, Mohamed M. Down-regulation of steroidogenesis-related genes and its accompanying fertility decline in streptozotocin-induced diabetic male rats: ameliorative effect of metformin. Andrology 2018; 7:110-123. [PMID: 30515996 DOI: 10.1111/andr.12567] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/19/2018] [Accepted: 10/29/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Metformin has long been used for glycemic control in diabetic state. Recently, other benefits of metformin beyond blood glucose regulation have emerged. OBJECTIVES To investigate the effect of metformin on the expression of testicular steroidogenesis-related genes, spermatogenesis, and fertility of male diabetic rats. MATERIALS AND METHODS Eighteen adult male Sprague Dawley rats were divided into three groups, namely normal control (NC), diabetic control (DC), and metformin-treated (300 mg/kg body weight/day) diabetic rats (D+Met). Diabetes was induced using a single intraperitoneal injection of streptozotocin (60 mg/kg b.w.), followed by oral treatment with metformin for four weeks. RESULTS Diabetes decreased serum and intratesticular testosterone levels and increased serum but not intratesticular levels of luteinizing hormone. Sperm count, motility, viability, and normal morphology were decreased, while sperm nuclear DNA fragmentation was increased in DC group, relative to NC group. Testicular mRNA levels of androgen receptor, luteinizing hormone receptor, cytochrome P450 enzyme (CYP11A1), steroidogenic acute regulatory (StAR) protein, 3β-hydroxysteroid dehydrogenase (HSD), and 17β-HSD, as well as the level of StAR protein and activities of CYP11A1, 3β-HSD, and 17β-HSD, were decreased in DC group. Similarly, decreased activities of epididymal antioxidant enzymes and increased lipid peroxidation were observed in DC group. Consequently, decreased litter size, fetal weight, mating and fertility indices, and increased pre- and post-implantation losses were recorded in DC group. Following intervention with metformin, we observed increases in serum and intratesticular testosterone levels, Leydig cell count, improved sperm parameters, and decreased sperm nuclear DNA fragmentation. Furthermore, mRNA levels and activities of steroidogenesis-related enzymes were increased, with improved fertility outcome. DISCUSSION AND CONCLUSION Diabetes mellitus is associated with dysregulation of steroidogenesis, abnormal spermatogenesis, and fertility decline. Controlling hyperglycemia is therefore crucial in preserving male reproductive function. Metformin not only regulates blood glucose level, but also preserves male fertility in diabetic state.
Collapse
Affiliation(s)
- V U Nna
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - A B A Bakar
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - A Ahmad
- Basic Science and Oral Biology Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - M Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Unit of Integrative Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|