1
|
Shamsad A, Gautam T, Singh R, Banerjee M. Genetic and epigenetic alterations associated with gestational diabetes mellitus and adverse neonatal outcomes. World J Clin Pediatr 2025; 14:99231. [DOI: 10.5409/wjcp.v14.i1.99231] [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: 07/17/2024] [Revised: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 12/20/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is a metabolic disorder, recognised during 24-28 weeks of pregnancy. GDM is linked with adverse newborn outcomes such as macrosomia, premature delivery, metabolic disorder, cardiovascular, and neurological disorders. Recent investigations have focused on the correlation of genetic factors such as β-cell function and insulin secretary genes (transcription factor 7 like 2, potassium voltage-gated channel subfamily q member 1, adiponectin etc.) on maternal metabolism during gestation leading to GDM. Epigenetic alterations like DNA methylation, histone modification, and miRNA expression can influence gene expression and play a dominant role in feto-maternal metabolic pathways. Interactions between genes and environment, resulting in differential gene expression patterns may lead to GDM. Researchers suggested that GDM women are more susceptible to insulin resistance, which alters intrauterine surroundings, resulting hyperglycemia and hyperinsulinemia. Epigenetic modifications in genes affecting neuroendocrine activities, and metabolism, increase the risk of obesity and type 2 diabetes in offspring. There is currently no treatment or effective preventive method for GDM, since the molecular processes of insulin resistance are not well understood. The present review was undertaken to understand the pathophysiology of GDM and its effects on adverse neonatal outcomes. In addition, the study of genetic and epigenetic alterations will provide lead to researchers in the search for predictive molecular biomarkers.
Collapse
Affiliation(s)
- Amreen Shamsad
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Tanu Gautam
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Renu Singh
- Department of Obstetrics and Gynecology, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Monisha Banerjee
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| |
Collapse
|
2
|
Lai F, Zhou K, Ma Y, Lv H, Wang W, Wang R, Xu T, Huang R. Single-cell RNA sequencing identifies endothelial-derived HBEGF as promoting pancreatic beta cell proliferation in mice via the EGFR-Kmt5a-H4K20me pathway. Diabetologia 2024:10.1007/s00125-024-06341-y. [PMID: 39694915 DOI: 10.1007/s00125-024-06341-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 12/20/2024]
Abstract
AIMS/HYPOTHESIS Pancreatic beta cell mass is dynamically regulated in response to increased physiological and pathological demands. Understanding the mechanisms that control physiological beta cell proliferation could provide valuable insights into novel therapeutic approaches to diabetes. Here, we aimed to analyse the intracellular and extracellular signalling pathways involved in regulating the physiological proliferation of beta cells using single-cell RNA-seq (scRNA-seq) and in vitro functional assays. METHODS Islets isolated from nulliparous mice, mice at different time points of gestation and mice at day 4 after delivery were analysed using scRNA-seq. Bioinformatics analyses of scRNA-seq data were performed to determine the heterogeneous transcriptomic characteristics of beta cells and to identify the proliferating subpopulation. CellChat was used to analyse cell-cell communication and identify the ligand-receptor pairs between beta cell subclusters as well as between non-beta cells and proliferating beta cells. In vitro functional assays were conducted in mouse and rat beta cell lines and isolated mouse primary islets to validate the role of Kmt5a- mono-methylation of histone H4 at lysine 20 (H4K20me) signalling and endothelial-derived heparin-binding EGF-like growth factor (HBEGF) in beta cell proliferation. RESULTS Of 43,724 endocrine and non-endocrine cells within islets analysed by scRNA-seq, 15,569 beta cells were clustered into eight distinct populations, each exhibiting unique heterogeneity. A proliferating beta cell subcluster was identified that highly expressed the histone methyltransferase Kmt5a. Activation of Kmt5a-H4K20me signalling upregulated the expression of Cdk1 and promoted beta cell proliferation. The crosstalk between endothelial cells and the proliferating beta cell subcluster, mediated by the HBEGF-EGF receptor (EGFR) ligand-receptor interaction, increased as beta cell mass expanded. HBEGF increased the expression levels of genes involved in the cell cycle and promoted beta cell proliferation by regulating the Kmt5a-H4K20me signalling pathway. CONCLUSIONS/INTERPRETATION Our study demonstrates that, under physiological conditions, endothelial-derived HBEGF regulates beta cell proliferation through the Kmt5a-H4K20me signalling pathway, which may serve as a potential target to promote beta cell expansion and treat diabetes. DATA AVAILABILITY The scRNA-seq and RNA-seq datasets are available from the Gene Expression Omnibus (GEO) using the accession numbers GSE278860 and GSE278861, respectively.
Collapse
Affiliation(s)
- Fengling Lai
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kaixin Zhou
- Guangzhou National Laboratory, Guangzhou, China
| | - Yingjie Ma
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Lv
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Weilin Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Rundong Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tao Xu
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Rong Huang
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
3
|
Seedat F, Kandzija N, Ellis M, Jiang S, Sarbalina A, Bancroft J, Drydale E, Hester S, Fischer R, Wade A, Stefana M, Todd J, Vatish M. Placental small extracellular vesicles from normal pregnancy and gestational diabetes increase insulin gene transcription and content in β cells. Clin Sci (Lond) 2024; 138:1481-1502. [PMID: 39432712 PMCID: PMC11579211 DOI: 10.1042/cs20241782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
Insulin secretion increases progressively during pregnancy to maintain normal maternal blood glucose levels. The placenta plays a crucial role in this process by releasing hormones and extracellular vesicles into the maternal circulation, which drive significant changes in pregnancy physiology. Placental extracellular vesicles, which are detectable in the plasma of pregnant women, have been shown to signal peripheral tissues and contribute to pregnancy-related conditions. While studies using murine models have demonstrated that extracellular vesicles can modulate insulin secretion in pancreatic islets, it remains unclear whether these effects translate to human biology. Understanding how placental signals enhance insulin synthesis and secretion from β cells could be pivotal in developing new therapies for diabetes. In our study, we isolated placental small extracellular vesicles from human placentae and utilised the human β cell line, EndoC-βH3, to investigate their effects on β-cell function in vitro. Our results indicate that human β cells internalise placental small extracellular vesicles, leading to enhanced insulin gene expression and increased insulin content within the β cells. Moreover, these vesicles up-regulated the expression of Annexin A1, a protein known to increase insulin content. This up-regulation of Annexin A1 holds promise as a potential mechanism by which placental small extracellular vesicles enhance insulin biosynthesis.
Collapse
Affiliation(s)
- Faheem Seedat
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, U.K
| | - Neva Kandzija
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, U.K
| | - Michael J. Ellis
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Shuhan Jiang
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, U.K
| | - Asselzhan Sarbalina
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, U.K
| | - James Bancroft
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Edward Drydale
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Svenja S. Hester
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
| | - Roman Fischer
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
| | - Alisha N. Wade
- Research in Metabolism and Endocrinology, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
- Division of Endocrinology, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, U.S.A
| | - M. Irina Stefana
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - John A. Todd
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Manu Vatish
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, U.K
| |
Collapse
|
4
|
Li Q, Yang RH, Hu Y, Tang BB, Jiang YJ, Zheng CB, Song TZ. Zika virus infection induces glycometabolic disorder in northern pig-tailed macaques. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2527-2529. [PMID: 39235558 DOI: 10.1007/s11427-024-2663-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/24/2024] [Indexed: 09/06/2024]
Affiliation(s)
- Qing Li
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ren-Hua Yang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Yan Hu
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bei-Bei Tang
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- KIZ-SU Joint Laboratory of Animal Model and Drug Development, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215021, China
| | - Ying-Jie Jiang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China.
| | - Tian-Zhang Song
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
5
|
Musa E, Salazar-Petres E, Vatish M, Levitt N, Sferruzzi-Perri AN, Matjila MJ. Kisspeptin signalling and its correlation with placental ultrastructure and clinical outcomes in pregnant South African women with obesity and gestational diabetes. Placenta 2024; 154:49-59. [PMID: 38878622 DOI: 10.1016/j.placenta.2024.05.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 09/03/2024]
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) is a major pregnancy metabolic disorder and is strongly linked with obesity. Kisspeptin is a hormone that increases several thousand-fold in the maternal circulation during human pregnancy, with placenta as its main source. Studies have suggested that kisspeptin regulates trophoblast invasion and promotes pancreatic insulin secretion and peripheral insulin sensitivity. METHODS In a well-characterized cohort of pregnant South African women and molecular and histological techniques, this study explored the impact and interaction of maternal obesity and GDM on kisspeptin (KISS1) signalling in relation to placental morphology and maternal and neonatal parameters. RESULTS We found that GDM had no effect on placental KISS1 and KISS1R (KISS1 receptor) mRNA and/or protein expression. However, obesity reduced placental KISS1R mRNA expression even though overall KISS1 protein abundance or localization was not different from the non-obese group. Maternal and cord circulating KISS1 concentrations did not vary with obesity or GDM, but maternal circulating KISS1 was positively correlated with placenta weight in non-GDM obese women, and negatively correlated with placental intervillous space volume in non-GDM non-obese women. Cord serum KISS1 was positively correlated with infant weight in GDM obese women, but negatively correlated with maternal BMI in the non-obese GDM group. Placental syncytiotrophoblast extracellular vesicles exhibited detectable KISS1 and its abundance was ∼50 % lower in those from obese GDM compared to non-GDM women. DISCUSSION This study shows maternal obesity and GDM can modulate placental kisspeptin signalling and placental morphological development with potential pathophysiological implications for clinically-relevant pregnancy and perinatal outcomes.
Collapse
Affiliation(s)
- Ezekiel Musa
- Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa; Department of Internal Medicine, Kaduna State University, Kaduna, Nigeria
| | - Esteban Salazar-Petres
- Carrera de Obstetricia, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Valdivia, Chile
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Naomi Levitt
- Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Mushi J Matjila
- Department of Obstetrics and Gynaecology, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
6
|
Ruiz-Otero N, Tessem JS, Banerjee RR. Pancreatic islet adaptation in pregnancy and postpartum. Trends Endocrinol Metab 2024; 35:834-847. [PMID: 38697900 DOI: 10.1016/j.tem.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024]
Abstract
Pancreatic islets, particularly insulin-producing β-cells, are central regulators of glucose homeostasis capable of responding to a variety of metabolic stressors. Pregnancy is a unique physiological stressor, necessitating the islets to adapt to the complex interplay of maternal and fetal-placental factors influencing the metabolic milieu. In this review we highlight studies defining gestational adaptation mechanisms within maternal islets and emerging studies revealing islet adaptations during the early postpartum and lactation periods. These include adaptations in both β and in 'non-β' islet cells. We also discuss insights into how gestational and postpartum adaptation may inform pregnancy-specific and general mechanisms of islet responses to metabolic stress and contribute to investigation of gestational diabetes.
Collapse
Affiliation(s)
- Nelmari Ruiz-Otero
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Jeffery S Tessem
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT 84601, USA
| | - Ronadip R Banerjee
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA.
| |
Collapse
|
7
|
Sousa D, Magalhães C, Matafome P, Pereira S. Adipose tissue-liver cross-talk: a route to hepatic dysfunction in pregnant women with obesity. Biosci Rep 2024; 44:BSR20231679. [PMID: 39083072 PMCID: PMC11327218 DOI: 10.1042/bsr20231679] [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: 02/20/2024] [Revised: 05/24/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Obesity during pregnancy has been escalating, becoming a huge problem that poses consequences not only for the health of the offspring but also for the maternal well-being. Women's adipose and hepatic tissue metabolism undergoes significant changes during the gestational period. During pregnancy, obesity is a primary instigator of steatosis, increasing the risk of non-alcholic fatty liver disease (NAFLD), now recognized under the updated nomenclature metabolic dysfunction-associated steatotic liver disease (MASLD). Pregnant women with obesity present higher levels of free fatty acids and glucose, reduction in insulin sensitivity, and adipose tissue endocrine dysregulation. Furthermore, obesity-induced modifications in clock genes and lipid-associated gene expression within adipose tissue disrupt crucial metabolic adaptations, potentially culminating in adipose tissue dysfunction. Thus, the liver experiences increased exposure to free fatty acids through the portal vein. Higher uptake of free fatty acids into the liver disrupts hepatic lipid oxidation while enhances lipogenesis, thereby predisposing to ectopic fat deposition within the liver. This review focuses on the obesity-induced changes during pregnancy in both liver and adipose tissue metabolism, elucidating how the metabolic crosstalk between these two organs can be dysregulated in pregnant women living with obesity.
Collapse
Affiliation(s)
- Diana Sousa
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Carina C. Magalhães
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Paulo Matafome
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Polytechnic University of Coimbra, Coimbra Health School, Rua 5 de Outubro—S. Martinho do Bispo, 3046-854 Coimbra, Portugal
| | - Susana P. Pereira
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra,3004-504 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra; 3004-517 Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory of for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
8
|
Chernysheva МB, Ruchko ЕS, Karimova МV, Vorotelyak ЕA, Vasiliev АV. Development, regeneration, and physiological expansion of functional β-cells: Cellular sources and regulators. Front Cell Dev Biol 2024; 12:1424278. [PMID: 39045459 PMCID: PMC11263198 DOI: 10.3389/fcell.2024.1424278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024] Open
Abstract
Pancreatic regeneration is a complex process observed in both normal and pathological conditions. The aim of this review is to provide a comprehensive understanding of the emergence of a functionally active population of insulin-secreting β-cells in the adult pancreas. The renewal of β-cells is governed by a multifaceted interaction between cellular sources of genetic and epigenetic factors. Understanding the development and heterogeneity of β-cell populations is crucial for functional β-cell regeneration. The functional mass of pancreatic β-cells increases in situations such as pregnancy and obesity. However, the specific markers of mature β-cell populations and postnatal pancreatic progenitors capable of increasing self-reproduction in these conditions remain to be elucidated. The capacity to regenerate the β-cell population through various pathways, including the proliferation of pre-existing β-cells, β-cell neogenesis, differentiation of β-cells from a population of progenitor cells, and transdifferentiation of non-β-cells into β-cells, reveals crucial molecular mechanisms for identifying cellular sources and inducers of functional cell renewal. This provides an opportunity to identify specific cellular sources and mechanisms of regeneration, which could have clinical applications in treating various pathologies, including in vitro cell-based technologies, and deepen our understanding of regeneration in different physiological conditions.
Collapse
Affiliation(s)
- М. B. Chernysheva
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| | - Е. S. Ruchko
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| | - М. V. Karimova
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
- Department of Biology and Biotechnologies Charles Darwin, The Sapienza University of Rome, Rome, Italy
| | - Е. A. Vorotelyak
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| | - А. V. Vasiliev
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| |
Collapse
|
9
|
Saucedo R, Ferreira-Hermosillo A, Robledo-Clemente M, Díaz-Velázquez MF, Valencia-Ortega J. Association of DNA Methylation with Infant Birth Weight in Women with Gestational Diabetes. Metabolites 2024; 14:361. [PMID: 39057684 PMCID: PMC11278577 DOI: 10.3390/metabo14070361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Offspring exposed to gestational diabetes mellitus (GDM) exhibit greater adiposity at birth. This early-life phenotype may increase offspring risk of developing obesity, metabolic syndrome, type 2 diabetes, and cardiovascular disease later in life. Infants born to women with GDM have a dysregulation of several hormones, cytokines, and growth factors related to fetal fat mass growth. One of the molecular mechanisms of GDM influencing these factors is epigenetic alterations, such as DNA methylation (DNAm). This review will examine the role of DNAm as a potential biomarker for monitoring fetal growth during pregnancy in women with GDM. This information is relevant since it may provide useful new biomarkers for the diagnosis, prognosis, and treatment of fetal growth and its later-life health consequences.
Collapse
Affiliation(s)
- Renata Saucedo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.S.); (A.F.-H.)
| | - Aldo Ferreira-Hermosillo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.S.); (A.F.-H.)
| | - Magalhi Robledo-Clemente
- Hospital de Gineco Obstetricia 3, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City 02990, Mexico; (M.R.-C.); (M.F.D.-V.)
| | - Mary Flor Díaz-Velázquez
- Hospital de Gineco Obstetricia 3, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City 02990, Mexico; (M.R.-C.); (M.F.D.-V.)
| | - Jorge Valencia-Ortega
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico
| |
Collapse
|
10
|
Bline AP, Ellis LB, Pelch KE, Lam J, Sen S, Zlatnik M, Varshavsky J. The effect of per and polyfluoroalkyl substance (PFAS) exposure on gestational diabetes mellitus and its subclinical risk factors: A systematic review and meta-analysis protocol. ENVIRONMENT INTERNATIONAL 2024; 188:108711. [PMID: 38754246 DOI: 10.1016/j.envint.2024.108711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/30/2024] [Accepted: 04/27/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Multiple lines of evidence suggest that exposure to per- and polyfluoroalkyl substances (PFAS) may alter glucose homeostasis, particularly during pregnancy, and may affect risk for developing gestational diabetes mellitus (GDM). While previous systematic reviews have been conducted on this topic, they did not assess internal validity of the included studies and their search strategies were narrowly focused. OBJECTIVE The objective of this study is to assess the effect of higher PFAS exposure (defined by individual compounds or mixtures measured before or during pregnancy) on GDM and subclinical measures of impaired glucose homeostasis (measured during pregnancy) compared to lower PFAS exposure in pregnant. METHODS We developed our systematic review protocol in accordance with the Navigation Guide. Peer-reviewed journal and grey literature searches were piloted in to identify relevant studies and refine our search terms and strategy. We also piloted the study screening criteria and data extraction form in DistillerSR, and refined our protocol accordingly. The risk of bias assessment protocol was adapted from Navigation Guide guidance and will be piloted and performed in DistillerSR. Pending the identification of comparable studies, quantitative meta-analyses will be performed where possible. Study results that cannot be quantitatively synthesized will be included in a narrative synthesis. The quality and strength of the body of evidence will be evaluated using Navigation Guide methodology, which is informed by guidance from the Cochrane Collaboration and Grading of Recommendations Assessment, Development and Evaluation (GRADE). We also made refinements to the quality of evidence considerations based on guidance from the National Institute of Environmental Health Sciences (NIEHS) Office of Health Assessment and Translation (OHAT). FUNDING This work was supported by the Systematizing Data on Per- and Polyfluoroalkyl Substances and Health Northeastern University TIER 1 Award.
Collapse
Affiliation(s)
- Abigail P Bline
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA, United States; Silent Spring Institute, Newton, MA, United States.
| | - Lauren B Ellis
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA, United States; Department of Health Sciences, Northeastern University, Boston, MA, United States.
| | - Katherine E Pelch
- Natural Resources Defense Council, San Francisco, CA, United States.
| | - Juleen Lam
- Department of Public Health, California State University, East Bay, Hayward, CA, United States.
| | - Saunak Sen
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.
| | - Marya Zlatnik
- Department of Obstetrics, Gynecology and Reproductive Sciences, Program on Reproductive Health and the Environment, University of California San Francisco, San Francisco, CA, United States.
| | - Julia Varshavsky
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA, United States; Department of Health Sciences, Northeastern University, Boston, MA, United States; Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, United States.
| |
Collapse
|
11
|
Zhang L, Wang H, Zu P, Li X, Ma S, Zhu Y, Xie T, Tao F, Zhu DM, Zhu P. Association between exposure to outdoor artificial light at night during pregnancy and glucose homeostasis: A prospective cohort study. ENVIRONMENTAL RESEARCH 2024; 247:118178. [PMID: 38220082 DOI: 10.1016/j.envres.2024.118178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Outdoor artificial light at night (ALAN) has been linked to an elevated risk of diabetes, but the available literature on the relationships between ALAN and glucose homeostasis in pregnancy is limited. METHODS A prospective cohort study of 6730 pregnant women was conducted in Hefei, China. Outdoor ALAN exposure was estimated using satellite data with individual addresses at a spatial resolution of approximately 1 km, and the average ALAN intensity was calculated. Gestational diabetes mellitus (GDM) was diagnosed based on a standard 75-g oral glucose tolerance test. Multivariable linear regression and logistic regression were used to estimate the relationships between ALAN and glucose homeostasis. RESULTS Outdoor ALAN was associated with elevated glucose homeostasis markers in the first trimester, but not GDM risk. An increase in the interquartile range of outdoor ALAN values was related to a 0.02 (95% confidence interval [CI]: 0.00, 0.03) mmol/L higher fasting plasma glucose, a 0.42 (95% CI: 0.30, 0.54) μU/mL increase in insulin and a 0.09 (95% CI: 0.07, 0.12) increase in homeostatic model assessment of insulin resistance (HOMA-IR) during the first trimester. Subgroup analyses showed that the associations between outdoor ALAN exposure and fasting plasma glucose, insulin, and HOMA-IR were more pronounced among pregnant women who conceived in summer and autumn. CONCLUSIONS The results provided evidence that brighter outdoor ALAN in the first trimester was related to elevated glucose intolerance in pregnancy, especially in pregnant women conceived in summer and autumn, and effective strategies are needed to prevent and manage light pollution.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, China
| | - Haixia Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, China
| | - Ping Zu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, China
| | - Xinyu Li
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei, China; Hefei Fourth People's Hospital, Hefei, China; Anhui Mental Health Center, Hefei, China
| | | | - Yuanyuan Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, China
| | - Tianqin Xie
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei, China; Hefei Fourth People's Hospital, Hefei, China; Anhui Mental Health Center, Hefei, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, China
| | - Dao-Min Zhu
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei, China; Hefei Fourth People's Hospital, Hefei, China; Anhui Mental Health Center, Hefei, China.
| | - Peng Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China; MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, China.
| |
Collapse
|
12
|
Calvo MJ, Parra H, Santeliz R, Bautista J, Luzardo E, Villasmil N, Martínez MS, Chacín M, Cano C, Checa-Ros A, D'Marco L, Bermúdez V, De Sanctis JB. The Placental Role in Gestational Diabetes Mellitus: A Molecular Perspective. TOUCHREVIEWS IN ENDOCRINOLOGY 2024; 20:10-18. [PMID: 38812661 PMCID: PMC11132656 DOI: 10.17925/ee.2024.20.1.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/01/2023] [Indexed: 05/31/2024]
Abstract
During pregnancy, women undergo several metabolic changes to guarantee an adequate supply of glucose to the foetus. These metabolic modifications develop what is known as physiological insulin resistance. When this process is altered, however, gestational diabetes mellitus (GDM) occurs. GDM is a multifactorial disease, and genetic and environmental factors play a crucial role in its aetiopathogenesis. GDM has been linked to both macroscopic and molecular alterations in placental tissues that affect placental physiology. This review summarizes the role of the placenta in the development of GDM from a molecular perspective, including hormonal and pro-inflammatory changes. Inflammation and hormonal imbalance, the characteristics dominating the GDM microenvironment, are responsible for placental changes in size and vascularity, leading to dysregulation in maternal and foetal circulations and to complications in the newborn. In conclusion, since the hormonal mechanisms operating in GDM have not been fully elucidated, more research should be done to improve the quality of life of patients with GDM and their future children.
Collapse
Affiliation(s)
- María José Calvo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Raquel Santeliz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Jordan Bautista
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Eliana Luzardo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Nelson Villasmil
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - María Sofía Martínez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Maricamen Chacín
- Facultad de Ciencias de la Salud, Barranquilla, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Ana Checa-Ros
- Research Group on Cardiorenal and Metabolic Diseases, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Luis D'Marco
- Research Group on Cardiorenal and Metabolic Diseases, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Barranquilla, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
13
|
Davis D, Kurz E, Hooper ME, Atchan M, Spiller S, Blackburn J, Bushell M, Lewis V, Leung M, Samarawickrema I, Knight-Agarwal C. The holistic maternity care needs of women with Gestational Diabetes Mellitus: A systematic review with thematic synthesis. Women Birth 2024; 37:166-176. [PMID: 37684120 DOI: 10.1016/j.wombi.2023.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023]
Abstract
PROBLEM Models of care for women with gestational diabetes mellitus (GDM) have evolved in an ad hoc way and do not meet women's needs. BACKGROUND GDM affects 50,000 Australian women per annum with prevalence quadrupling in the last ten years. Many health services are struggling to provide a quality service. People with diabetes are calling for care that focuses on their wellbeing more broadly. AIM To examine the holistic (emotional, social, economic, and spiritual) care needs of women with GDM. METHODS Qualitative and mixed-methods studies capturing the healthcare experiences of women with GDM were searched for in CINAHL, Medline, Web of Science and Scopus. English-language studies published between 2011 and 2023 were included. Quality of studies was assessed using Crowe Critical Appraisal Tool and NVIVO was used to identify key themes and synthesise data. FINDINGS Twenty-eight studies were included, representing the experiences of 958 women. Five themes reflect women's holistic needs through their journey from initial diagnosis to postpartum: psychological impact, information and education, making change for better health, support, and care transition. DISCUSSION The biomedical, fetal-centric model of care neglects the woman's holistic wellbeing resulting in high levels of unmet need. Discontinuity between tertiary and primary services results in a missed opportunity to assist women to make longer term changes that would benefit themselves (and their families) into the future. CONCLUSIONS The provision of holistic models of care for this cohort is pivotal to improving clinical outcomes and the experiences of women with GDM.
Collapse
Affiliation(s)
- Deborah Davis
- University of Canberra, Faculty of Health, Australia; ACT Government, Health Directorate, Australia.
| | - Ella Kurz
- University of Canberra, Faculty of Health, Australia
| | | | | | | | | | - Mary Bushell
- University of Canberra, Faculty of Health, Australia
| | | | - Myra Leung
- University of Canberra, Faculty of Health, Australia
| | | | | |
Collapse
|
14
|
Zhao C, Liu H, Deng Y, Wu H, Wang S, Lyu X, Lei J, Yang H, Hu M, Zhao Y, Ma X, Zou X, Yang Y. Maternal fasting serum C-peptide concentrations in the first and second trimesters and subsequent risk of gestational diabetes mellitus: A nested case-control study among Chinese women. Diabetes Res Clin Pract 2024; 208:111111. [PMID: 38266822 DOI: 10.1016/j.diabres.2024.111111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/11/2024] [Accepted: 01/21/2024] [Indexed: 01/26/2024]
Abstract
OBJECTIVE To examine the association of serum connecting peptide (C-peptide) concentrations with gestational diabetes mellitus (GDM) risk among Chinese women. METHODS A nested case-control study was conducted on 436 reproductive-aged women, involving 218 GDM cases and 218 controls matched at 1:1 by maternal age, in Beijing, China between January 2016 and December 2017. Fasting serum C-peptide were successively determined at 10-14 and 15-20 weeks of gestation. Restricted cubic spline and logistic regression analyses were utilized, and receiver operating characteristic (ROC) curves were generated to evaluate the predictive capacity of C-peptide for GDM. RESULTS Fasting serum C-peptide concentrations exhibited a significant decrease from the initial to the subsequent trimester in females with normal glucose tolerance (NGT). For each 1 log ng/mL increase of fasting serum C-peptide during the first and second trimesters, GDM risk increased by 2.38-fold [odds ratio (OR): 2.38, 95% confidence intervals (95%CI): 1.33-4.40] and 3.07-fold (OR: 3.07, 95%CI: 1.49-6.62), respectively. The areas under the ROC curves for the first- and second-trimester C-peptide were 80.4% and 82.4%. CONCLUSION Our findings revealed a positive correlation between fasting serum C-peptide during the first and second trimesters and the risk of GDM or its subtypes, underscoring the potential of C-peptide as a predictor for GDM development.
Collapse
Affiliation(s)
- Chuanyu Zhao
- Graduate School of Peking Union Medical College, Beijing Municipality, China; National Research Institute for Family Planning, Beijing Municipality, China; National Human Genetic Resources Center, Beijing Municipality, China
| | - Haiyan Liu
- Haidian District Maternal and Child Health Care Hospital, Beijing Municipality, China
| | - Yuzhi Deng
- National Research Institute for Family Planning, Beijing Municipality, China; National Human Genetic Resources Center, Beijing Municipality, China
| | - Hanbin Wu
- National Research Institute for Family Planning, Beijing Municipality, China; National Human Genetic Resources Center, Beijing Municipality, China
| | - Shuo Wang
- Haidian District Maternal and Child Health Care Hospital, Beijing Municipality, China
| | - Xinyi Lyu
- Graduate School of Peking Union Medical College, Beijing Municipality, China; National Research Institute for Family Planning, Beijing Municipality, China; National Human Genetic Resources Center, Beijing Municipality, China
| | - Jueming Lei
- National Research Institute for Family Planning, Beijing Municipality, China; National Human Genetic Resources Center, Beijing Municipality, China
| | - Haishan Yang
- Haidian District Maternal and Child Health Care Hospital, Beijing Municipality, China
| | - Meina Hu
- Haidian District Maternal and Child Health Care Hospital, Beijing Municipality, China
| | - Yinzhu Zhao
- Haidian District Maternal and Child Health Care Hospital, Beijing Municipality, China
| | - Xu Ma
- Graduate School of Peking Union Medical College, Beijing Municipality, China; National Research Institute for Family Planning, Beijing Municipality, China; National Human Genetic Resources Center, Beijing Municipality, China
| | - Xiaoxuan Zou
- Haidian District Maternal and Child Health Care Hospital, Beijing Municipality, China.
| | - Ying Yang
- Graduate School of Peking Union Medical College, Beijing Municipality, China; National Research Institute for Family Planning, Beijing Municipality, China; National Human Genetic Resources Center, Beijing Municipality, China.
| |
Collapse
|
15
|
Mittendorfer B, Patterson BW, Haire-Joshu D, Cahill AG, Cade WT, Stein RI, Klein S. Insulin Sensitivity and β-Cell Function During Early and Late Pregnancy in Women With and Without Gestational Diabetes Mellitus. Diabetes Care 2023; 46:2147-2154. [PMID: 37262059 PMCID: PMC10698210 DOI: 10.2337/dc22-1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 05/04/2023] [Indexed: 06/03/2023]
Abstract
OBJECTIVE To evaluate the metabolic alterations associated with gestational diabetes mellitus (GDM) in women with overweight or obesity. RESEARCH DESIGN AND METHODS We compared fasting and postprandial plasma glucose and free fatty acid (FFA) concentrations, insulin sensitivity (IS; Matsuda index), and β-cell function (i.e., β-cell responsiveness to glucose) by using a frequently sampled oral glucose tolerance test (OGTT) at 15 and 35 weeks' gestation in women with overweight or obesity who had GDM (n = 29) or did not have GDM (No-GDM; n = 164) at 35 weeks. RESULTS At 15 weeks, IS and β-cell function were lower, and fasting, 1-h, and total area-under-the-curve plasma glucose concentrations during the OGTT were higher (all P < 0.05) in the GDM than in the No-GDM group. At 35 weeks compared with 15 weeks, IS decreased, β-cell function increased, and postprandial suppression of plasma FFA was blunted in both the GDM and No-GDM groups, but the decrease in IS and the increase in postprandial FFA concentration were greater and the increase in β-cell function was less (all P ≤ 0.05) in the GDM than in the No-GDM group. A receiver operating characteristic curve analysis showed that both fasting plasma glucose and 1-h OGTT glucose concentration at 15 weeks are predictors of GDM, but the predictive power was <30%. CONCLUSIONS Women with overweight or obesity and GDM, compared with those without GDM, have worse IS and β-cell function early during pregnancy and a greater subsequent decline in IS and blunted increase in β-cell function. Increased fasting and 1-h OGTT plasma glucose concentration early during pregnancy are markers of increased GDM risk, albeit with weak predictive power.
Collapse
Affiliation(s)
| | | | | | - Alison G. Cahill
- Department of Obstetrics and Gynecology, Washington University, St. Louis, MO
- Department of Women’s Health, The University of Texas at Austin, Dell Medical School, Austin, TX
| | - W. Todd Cade
- Program in Physical Therapy, Washington University, St. Louis, MO
| | - Richard I. Stein
- Center for Human Nutrition, Washington University, St. Louis, MO
| | - Samuel Klein
- Center for Human Nutrition, Washington University, St. Louis, MO
- Sansum Diabetes Research Institute, Santa Barbara, CA
| |
Collapse
|
16
|
Zangeneh FZ, Hantoushzadeh S. The physiological basis with uterine myometrium contractions from electro-mechanical/hormonal myofibril function to the term and preterm labor. Heliyon 2023; 9:e22259. [PMID: 38034762 PMCID: PMC10687101 DOI: 10.1016/j.heliyon.2023.e22259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
Background Most labor-related problems can be attributed to the uterine myometrium muscle, as this irritable tissue must suppress its irritability potential during pregnancy. Unfortunately, fewer studies have investigated the causes of this lack of suppression in preterm labor. Methods We conducted a scoping narrative review using three online databases (PubMed, Scopus, and Science Direct). Results The review focused on ion channel functions in the myometrium, including sodium channels [Na K-ATPase, Na-activated K channels (Slo2), voltage-gated (SCN) Na+, Na+ leaky channels, nonselective (NALCN) channels], potassium channels [KATP (Kir6) channels, voltage-dependent K channels (Kv4, Kv7, and Kv11), twin-pore domain K channels (TASK, TREK), inward rectifier Kir7.1, Ca2+-activated K+ channels with large (KCNMA1, Slo1), small (KCNN1-3), intermediate (KCNN4) conductance], and calcium channels [L-Type and T-type Ca2+ channels, calcium-activated chloride channels (CaCC)], as well as hyperpolarization-activated cation channels. These channels' functions are associated with hormonal effects such as oxytocin, estrogen/progesterone, and local prostaglandins. Conclusion Electromechanical/hormonal activity and environmental autocrine factors can serve as the primary practical basis for premature uterine contractions in term/preterm labor. Our findings highlight the significance of.1.the amplitude rate of hyperpolarization and the frequency of contractions,2.changes in the estrogen/progesterone ratio,3.Prostaglandins E/F involvement in initiating potential spikes and the increase of intracytoplasmic Ca2+.This narrative study highlights the range of hyperpolarization and the frequency of myometrium contractions as crucial factors. The synchronized complex progress of estrogen to progesterone ratio and prostaglandins plays a significant role in initiating potential spikes and increasing intracytoplasmic Ca2+, which further influences the contraction process during labor. Insights into myometrium physiology gained from this study may pave the way for much-needed new treatments to reduce problems associated with normal and preterm labor.
Collapse
Affiliation(s)
- Farideh Zafari Zangeneh
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedighe Hantoushzadeh
- Department of Fetal-Maternal Medicine, Tehran University of Medical Sciences, Imam Khomeini Hospital, Tehran, Iran
| |
Collapse
|
17
|
Baumel-Alterzon S, Tse I, Heidery F, Garcia-Ocaña A, Scott DK. NRF2 Dysregulation in Mice Leads to Inadequate Beta-Cell Mass Expansion during Pregnancy and Gestational Diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555207. [PMID: 37693560 PMCID: PMC10491153 DOI: 10.1101/2023.08.28.555207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The late stages of the mammalian pregnancy are accompanied with increased insulin resistance due to the increased glucose demand of the growing fetus. Therefore, as a compensatory response to maintain the maternal normal blood glucose levels, maternal beta-cell mass expands leading to increased insulin release. Defects in beta-cell adaptive expansion during pregnancy can lead to gestational diabetes mellitus (GDM). Although the exact mechanisms that promote GDM are poorly understood, GDM strongly associates with impaired beta-cell proliferation and with increased levels of reactive oxygen species (ROS). Here, we show that NRF2 levels are upregulated in mouse beta-cells at gestation day 15 (GD15) concomitant with increased beta-cell proliferation. Importantly, mice with tamoxifen-induced beta-cell-specific NRF2 deletion display inhibition of beta-cell proliferation, increased beta-cell oxidative stress and elevated levels of beta-cell death at GD15. This results in attenuated beta-cell mass expansion and disturbed glucose homeostasis towards the end of pregnancy. Collectively, these results highlight the importance of NRF2-oxidative stress regulation in beta-cell mass adaptation to pregnancy and suggest NRF2 as a potential therapeutic target for treating GDM.
Collapse
Affiliation(s)
- Sharon Baumel-Alterzon
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Isabelle Tse
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Fatema Heidery
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adolfo Garcia-Ocaña
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute at City of Hope, Duarte, CA
| | - Donald K. Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
18
|
Su H, Jiang C, Zhang W, Zhu F, Jin Y, Cheng K, Lam T, Xu L. Parity and incident type 2 diabetes in older Chinese women: Guangzhou Biobank Cohort Study. Sci Rep 2023; 13:9504. [PMID: 37308533 DOI: 10.1038/s41598-023-36786-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023] Open
Abstract
This study examined the association between parity and incident type 2 diabetes in older Chinese women and estimated the mediation effect of adiposity indicators. A total of 11,473 women without diabetes at baseline from 2003 to 2008 were followed up until 2012. We used Cox proportional hazards regression to assess the association between parity and incident type 2 diabetes, and mediation analysis to estimate the mediation effect of adiposity indicators. Compared to women with one parity, the hazard ratio (HR) (95% confidence interval (CI)) for incident type 2 diabetes was 0.85 (0.44-1.63), 1.20 (1.11-1.30), 1.28 (1.16-1.41) and 1.27 (1.14-1.42) for women with parity of 0, 2, 3, and ≥ 4, respectively. The proportion of indirect effect (95% CI) mediated by body mass index, waist circumference, hip circumference, waist-to-hip ratio, waist-to-height ratio and body fat percentage was 26.5% (19.2-52.2%), 54.5% (39.4-108.7%), 25.1% (18.2-49.1%), 35.9% (25.6-74.1%), 50.3% (36.5-98.6%) and 15.1% (- 66.4 to 112.3%), respectively. Compared to women with one parity, women with multiparity (≥ 2) had a higher risk of incident type 2 diabetes and up to half of the association was mediated by abdominal obesity.
Collapse
Affiliation(s)
- Huimin Su
- School of Public Health, Sun Yat-Sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Chaoqiang Jiang
- Molecular Epidemiology Research Centre, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Weisen Zhang
- Molecular Epidemiology Research Centre, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China.
| | - Feng Zhu
- Molecular Epidemiology Research Centre, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Yali Jin
- Molecular Epidemiology Research Centre, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Karkeung Cheng
- Institute of Applied Health Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Taihing Lam
- Molecular Epidemiology Research Centre, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
- School of Public Health, The University of Hong Kong, Hong Kong, 999077, China
| | - Lin Xu
- School of Public Health, Sun Yat-Sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
- School of Public Health, The University of Hong Kong, Hong Kong, 999077, China.
| |
Collapse
|
19
|
Linares-Pineda TM, Gutiérrez-Repiso C, Peña-Montero N, Molina-Vega M, Rubio FL, Arana MS, Tinahones FJ, Picón-César MJ, Morcillo S. Higher β cell death in pregnant women, measured by DNA methylation patterns of cell-free DNA, compared to new-onset type 1 and type 2 diabetes subjects: a cross-sectional study. Diabetol Metab Syndr 2023; 15:115. [PMID: 37264478 DOI: 10.1186/s13098-023-01096-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/23/2023] [Indexed: 06/03/2023] Open
Abstract
Diabetes is a metabolic disorder of glucose homeostasis in which β cell destruction occurs silently and is detected mainly when symptoms appear. In the last few years, it has emerged a great interest in developing markers capable of detecting pancreatic β cell death focused on improving early diagnosis and getting a better treatment response, mainly in type 1 diabetes. But other types of diabetes would also benefit from early detection of β cell death. Differentially methylated circulating DNA is being studied as minimally invasive biomarker of cell death. We aimed to explore whether the unmethylated/methylated ratio of the insulin and amylin genes might be a good biomarker of β cell death in different types of diabetes. A lower index ∆Ct indicates a higher rate of β-cell death. Plasma samples from subjects without diabetes, pregnant women, pregnant with gestational diabetes (GDM), type 1 diabetes and type 2 diabetes were analyzed. A qPCR reaction with specific primers for both methylated and unmethylated fragments of insulin and amylin genes were carried out. Pregnant women, GDM and non- GDM, showed a higher β-cell death for both markers (∆INS = 3.8 ± 2.1 and ∆Amylin = 8.5 ± 3.6), whereas T1D presented lower rate (∆INS = 6.2 ± 2.1 and ∆Amylin = 10.7 ± 2.9) comparable to healthy subjects. The insulin methylation index was associated with the newborn birth weight (r = 0.46; p = 0.033) and with insulin resistance (r = -0.533; p = 0.027) in the GDM group. The higher rate of β-cell death was observed in pregnant women independently of their metabolic status. These indexes could be a good indicator of β cell death in processes caused by defects on insulin secretion, insulin action, or both.
Collapse
Affiliation(s)
- Teresa María Linares-Pineda
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Carolina Gutiérrez-Repiso
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Nerea Peña-Montero
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - María Molina-Vega
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Fuensanta Lima Rubio
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - María Suárez Arana
- Department of Obstetrics and Gynecology, Hospital Regional Universitario de Málaga, IBIMA, Málaga, Spain
| | - Francisco J Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina y Dermatología, Universidad de Málaga, Málaga, Spain
| | - María José Picón-César
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain.
| | - Sonsoles Morcillo
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
20
|
Martínez-Pinna J, Sempere-Navarro R, Medina-Gali RM, Fuentes E, Quesada I, Sargis RM, Trasande L, Nadal A. Endocrine disruptors in plastics alter β-cell physiology and increase the risk of diabetes mellitus. Am J Physiol Endocrinol Metab 2023; 324:E488-E505. [PMID: 37134142 PMCID: PMC10228669 DOI: 10.1152/ajpendo.00068.2023] [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: 03/01/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/05/2023]
Abstract
Plastic pollution breaks a planetary boundary threatening wildlife and humans through its physical and chemical effects. Of the latter, the release of endocrine disrupting chemicals (EDCs) has consequences on the prevalence of human diseases related to the endocrine system. Bisphenols (BPs) and phthalates are two groups of EDCs commonly found in plastics that migrate into the environment and make low-dose human exposure ubiquitous. Here we review epidemiological, animal, and cellular studies linking exposure to BPs and phthalates to altered glucose regulation, with emphasis on the role of pancreatic β-cells. Epidemiological studies indicate that exposure to BPs and phthalates is associated with diabetes mellitus. Studies in animal models indicate that treatment with doses within the range of human exposure decreases insulin sensitivity and glucose tolerance, induces dyslipidemia, and modifies functional β-cell mass and serum levels of insulin, leptin, and adiponectin. These studies reveal that disruption of β-cell physiology by EDCs plays a key role in impairing glucose homeostasis by altering the mechanisms used by β-cells to adapt to metabolic stress such as chronic nutrient excess. Studies at the cellular level demonstrate that BPs and phthalates modify the same biochemical pathways involved in adaptation to chronic excess fuel. These include changes in insulin biosynthesis and secretion, electrical activity, expression of key genes, and mitochondrial function. The data summarized here indicate that BPs and phthalates are important risk factors for diabetes mellitus and support a global effort to decrease plastic pollution and human exposure to EDCs.
Collapse
Affiliation(s)
- Juan Martínez-Pinna
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Alicante, Spain
| | - Roberto Sempere-Navarro
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Regla M Medina-Gali
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Fuentes
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ivan Quesada
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Robert M Sargis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Leonardo Trasande
- Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, United States
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, United States
- Wagner School of Public Service, New York University, New York, New York, United States
| | - Angel Nadal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
21
|
Musa E, Salazar-Petres E, Arowolo A, Levitt N, Matjila M, Sferruzzi-Perri AN. Obesity and gestational diabetes independently and collectively induce specific effects on placental structure, inflammation and endocrine function in a cohort of South African women. J Physiol 2023; 601:1287-1306. [PMID: 36849131 DOI: 10.1113/jp284139] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/19/2023] [Indexed: 03/01/2023] Open
Abstract
Maternal obesity and gestational diabetes mellitus (GDM) are associated with insulin resistance and health risks for mother and offspring. Obesity is also characterized by low-grade inflammation, which in turn, impacts insulin sensitivity. The placenta secretes inflammatory cytokines and hormones that influence maternal glucose and insulin handling. However, little is known about the effect of maternal obesity, GDM and their interaction, on placental morphology, hormones and inflammatory cytokines. In a South African cohort of non-obese and obese pregnant women with and without GDM, this study examined placental morphology using stereology, placental hormone and cytokine expression using real-time PCR, western blotting and immunohistochemistry, and circulating TNFα and IL-6 concentrations using ELISA. Placental expression of endocrine and growth factor genes was not altered by obesity or GDM. However, LEPTIN gene expression was diminished, syncytiotrophoblast TNFα immunostaining elevated and stromal and fetal vessel IL-6 staining reduced in the placenta of obese women in a manner that was partly influenced by GDM status. Placental TNFα protein abundance and maternal circulating TNFα concentrations were reduced in GDM. Both maternal obesity and, to a lesser extent, GDM were accompanied by specific changes in placental morphometry. Maternal blood pressure and weight gain and infant ponderal index were also modified by obesity and/or GDM. Thus, obesity and GDM have specific impacts on placental morphology and endocrine and inflammatory states that may relate to pregnancy outcomes. These findings may contribute to developing placenta-targeted treatments that improve mother and offspring outcomes, which is particularly relevant given increasing rates of obesity and GDM worldwide. KEY POINTS: Rates of maternal obesity and gestational diabetes (GDM) are increasing worldwide, including in low-middle income countries (LMIC). Despite this, much of the work in the field is conducted in higher-income countries. In a well-characterised cohort of South African women, this study shows that obesity and GDM have specific impacts on placental structure, hormone production and inflammatory profile. Moreover, such placental changes were associated with pregnancy and neonatal outcomes in women who were obese and/or with GDM. The identification of specific changes in the placenta may help in the design of diagnostic and therapeutic approaches to improve pregnancy and neonatal outcomes with particular significant benefit in LMICs.
Collapse
Affiliation(s)
- Ezekiel Musa
- Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- Department of Internal Medicine, Kaduna State University, Kaduna, Nigeria
| | - Esteban Salazar-Petres
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Afolake Arowolo
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Naomi Levitt
- Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Mushi Matjila
- Department of Obstetrics and Gynaecology, University of Cape Town, Cape Town, South Africa
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Lopez-Tello J, Sferruzzi-Perri AN. Characterization of placental endocrine function and fetal brain development in a mouse model of small for gestational age. Front Endocrinol (Lausanne) 2023; 14:1116770. [PMID: 36843585 PMCID: PMC9950515 DOI: 10.3389/fendo.2023.1116770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Conditions such as small for gestational age (SGA), which is defined as birthweight less than 10th percentile for gestational age can predispose to neurodevelopmental abnormalities compared to babies with normal birthweight. Fetal growth and birthweight depend on placental function, as this organ transports substrates to the developing fetus and it acts as a source of endocrine factors, including steroids and prolactins that are required for fetal development and pregnancy maintenance. To advance our knowledge on the aetiology of fetal growth disorders, the vast majority of the research has been focused on studying the transport function of the placenta, leaving practically unexplored the contribution of placental hormones in the regulation of fetal growth. Here, using mice and natural variability in fetal growth within the litter, we compared fetuses that fell on or below the 10th percentile (classified as SGA) with those that had adequate weight for their gestational age (AGA). In particular, we compared placental endocrine metabolism and hormone production, as well as fetal brain weight and expression of developmental, growth and metabolic genes between SGA and AGA fetuses. We found that compared to AGA fetuses, SGA fetuses had lower placental efficiency and reduced capacity for placental production of hormones (e.g. steroidogenic gene Cyp17a1, prolactin Prl3a1, and pregnancy-specific glycoproteins Psg21). Brain weight was reduced in SGA fetuses, although this was proportional to the reduction in overall fetal size. The expression of glucose transporter 3 (Slc2a3) was reduced despite the abundance of AKT, FOXO and ERK proteins were similar. Developmental (Sv2b and Gabrg1) and microglia genes (Ier3), as well as the pregnancy-specific glycoprotein receptor (Cd9) were lower in the brain of SGA versus AGA fetuses. In this mouse model of SGA, our results therefore demonstrate that placental endocrine dysfunction is associated with changes in fetal growth and fetal brain development.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Centre for Trophoblast Research – Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research – Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
23
|
Systematic Bayesian posterior analysis guided by Kullback-Leibler divergence facilitates hypothesis formation. J Theor Biol 2023; 558:111341. [PMID: 36335999 DOI: 10.1016/j.jtbi.2022.111341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/24/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Bayesian inference produces a posterior distribution for the parameters of a mathematical model that can be used to guide the formation of hypotheses; specifically, the posterior may be searched for evidence of alternative model hypotheses, which serves as a starting point for hypothesis formation and model refinement. Previous approaches to search for this evidence are largely qualitative and unsystematic; further, demonstrations of these approaches typically stop at hypothesis formation, leaving the questions they raise unanswered. Here, we introduce a Kullback-Leibler (KL) divergence-based ranking to expedite Bayesian hypothesis formation and investigate the hypotheses it generates, ultimately generating novel, biologically significant insights. Our approach uses KL divergence to rank parameters by how much information they gain from experimental data. Subsequently, rather than searching all model parameters at random, we use this ranking to prioritize examining the posteriors of the parameters that gained the most information from the data for evidence of alternative model hypotheses. We test our approach with two examples, which showcase the ability of our approach to systematically uncover different types of alternative hypothesis evidence. First, we test our KL divergence ranking on an established example of Bayesian hypothesis formation. Our top-ranked parameter matches the one previously identified to produce alternative hypotheses. In the second example, we apply our ranking in a novel study of a computational model of prolactin-induced JAK2-STAT5 signaling, a pathway that mediates beta cell proliferation. Within the top 3 ranked parameters (out of 33), we find a bimodal posterior revealing two possible ranges for the prolactin receptor degradation rate. We go on to refine the model, incorporating new data and determining which degradation rate is most plausible. Overall, while the effectiveness of our approach depends on having a properly formulated prior and on the form of the posterior distribution, we demonstrate that our approach offers a novel and generalizable quantitative framework for Bayesian hypothesis formation and use it to produce a novel, biologically-significant insight into beta cell signaling.
Collapse
|
24
|
Gurlo T, Prakash TP, Wang Z, Archang M, Pei L, Rosenberger M, Pirie E, Lee RG, Butler PC. Efficacy of IAPP suppression in mouse and human islets by GLP-1 analogue conjugated antisense oligonucleotide. Front Mol Biosci 2023; 10:1096286. [PMID: 36814640 PMCID: PMC9939749 DOI: 10.3389/fmolb.2023.1096286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023] Open
Abstract
Insulin resistance is the major risk factor for Type 2 diabetes (T2D). In vulnerable individuals, insulin resistance induces a progressive loss of insulin secretion with islet pathology revealing a partial deficit of beta cells and islet amyloid derived from islet amyloid polypeptide (IAPP). IAPP is co-expressed and secreted with insulin by beta cells, expression of both proteins being upregulated in response to insulin resistance. If IAPP expression exceeds the threshold for clearance of misfolded proteins, beta cell failure occurs exacerbated by the action of IAPP toxicity to compromise the autophagy lysosomal pathway. We postulated that suppression of IAPP expression by an IAPP antisense oligonucleotide delivered to beta cells by the GLP-1 agonist exenatide (eGLP1-IAPP-ASO) is a potential disease modifying therapy for T2D. While eGLP1-IAPP-ASO suppressed mouse IAPP and transgenic human IAPP expression in mouse islets, it had no discernable effects on IAPP expression in human islets under the conditions studied. Suppression of transgenic human IAPP expression in mouse islets attenuated disruption of the autophagy lysosomal pathway in beta cells, supporting the potential of this strategy.
Collapse
Affiliation(s)
- Tatyana Gurlo
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States,*Correspondence: Tatyana Gurlo, ; Peter C. Butler,
| | | | - Zhongying Wang
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maani Archang
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lina Pei
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Madeline Rosenberger
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elaine Pirie
- IONIS Pharmaceuticals, Carlsbad, CA, United States
| | | | - Peter C. Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States,*Correspondence: Tatyana Gurlo, ; Peter C. Butler,
| |
Collapse
|
25
|
Molina-Vega M, Picón-César MJ, Lima-Rubio F, Gutiérrez-Repiso C, Linares-Pineda TM, Suárez-Arana M, Fernández-Ramos AM, Tinahones FJ, Morcillo S. Insulin Requirement for Gestational Diabetes Control Is Related to Higher Vitamin D Levels up to 1 Year Postpartum: A Prospective Cohort Study. Antioxidants (Basel) 2022; 11:2230. [PMID: 36421415 PMCID: PMC9687061 DOI: 10.3390/antiox11112230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 01/04/2025] Open
Abstract
Vitamin D deficiency is highly prevalent in pregnant women and has been related to a higher risk of gestational diabetes mellitus (GDM). The aim of this study is to analyze vitamin D status evolution in a population of pregnant women with and without GDM. Two-hundred women were included from January 2019 to February 2022 as follows: Control group -CG-, Lifestyle group -LG- (GDM not requiring insulin), and Insulin group -IG- (GDM requiring insulin). Visits were carried out at baseline, antenatal, postpartum, and 1 year after birth. Vitamin D levels, weight, and insulin resistance were measured at every visit. Data about the season, vitamin D supplementation, Mediterranean diet adherence, and physical activity were included. In the three groups, 134 women were included in the CG, 43 in the LG, and 23 in the IG. Vitamin D levels were similar among the groups at baseline, but they were significantly higher in the LG and IG in comparison with the CG at the antenatal visit and significantly higher in the IG vs. CG and LG at the postpartum and 1 year after birth visits. Vitamin D levels were independently related to vitamin D supplementation and the season at baseline, to the season and belonging to the LG or IG at the antenatal visit, and were only independently associated with belonging to the IG at postpartum and 1 year after birth visits. In conclusion, in our population, women with GDM requiring insulin had higher levels of vitamin D in comparison with those not requiring insulin and healthy controls at postpartum and 1 year after pregnancy. Requiring insulin during pregnancy seems to be a factor that independently determines the levels of vitamin D until 1 year after birth. More studies are required to reproduce these data in other populations and to elucidate the mechanisms underlying these findings.
Collapse
Affiliation(s)
- María Molina-Vega
- Departamento de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (S.M.)
- Laboratorio de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.L.-R.); (C.G.-R.); (T.M.L.-P.)
| | - María José Picón-César
- Departamento de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (S.M.)
- Laboratorio de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.L.-R.); (C.G.-R.); (T.M.L.-P.)
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Fuensanta Lima-Rubio
- Laboratorio de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.L.-R.); (C.G.-R.); (T.M.L.-P.)
| | - Carolina Gutiérrez-Repiso
- Laboratorio de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.L.-R.); (C.G.-R.); (T.M.L.-P.)
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Teresa María Linares-Pineda
- Laboratorio de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.L.-R.); (C.G.-R.); (T.M.L.-P.)
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, 28029 Madrid, Spain
| | - María Suárez-Arana
- Departmento de Obstetricia y Ginecología, Hospital Regional Universitario de Málaga, IBIMA, 29009 Málaga, Spain;
| | - Ana María Fernández-Ramos
- Departamento de Análisis Clínicos, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain;
| | - Francisco J. Tinahones
- Departamento de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (S.M.)
- Laboratorio de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.L.-R.); (C.G.-R.); (T.M.L.-P.)
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Sonsoles Morcillo
- Departamento de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (S.M.)
- Laboratorio de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (F.L.-R.); (C.G.-R.); (T.M.L.-P.)
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
26
|
Cechinel LR, Batabyal RA, Freishtat RJ, Zohn IE. Parental obesity-induced changes in developmental programming. Front Cell Dev Biol 2022; 10:918080. [PMID: 36274855 PMCID: PMC9585252 DOI: 10.3389/fcell.2022.918080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Many studies support the link between parental obesity and the predisposition to develop adult-onset metabolic syndromes that include obesity, high blood pressure, dyslipidemia, insulin resistance, and diabetes in the offspring. As the prevalence of obesity increases in persons of childbearing age, so does metabolic syndrome in their descendants. Understanding how parental obesity alters metabolic programs in the progeny, predisposing them to adult-onset metabolic syndrome, is key to breaking this cycle. This review explores the basis for altered metabolism of offspring exposed to overnutrition by focusing on critical developmental processes influenced by parental obesity. We draw from human and animal model studies, highlighting the adaptations in metabolism that occur during normal pregnancy that become maladaptive with obesity. We describe essential phases of development impacted by parental obesity that contribute to long-term alterations in metabolism in the offspring. These encompass gamete formation, placentation, adipogenesis, pancreas development, and development of brain appetite control circuits. Parental obesity alters the developmental programming of these organs in part by inducing epigenetic changes with long-term consequences on metabolism. While exposure to parental obesity during any of these phases is sufficient to alter long-term metabolism, offspring often experience multiple exposures throughout their development. These insults accumulate to increase further the susceptibility of the offspring to the obesogenic environments of modern society.
Collapse
|
27
|
Lean SC, Candia AA, Gulacsi E, Lee GCL, Sferruzzi-Perri AN. Obesogenic diet in mice compromises maternal metabolic physiology and lactation ability leading to reductions in neonatal viability. Acta Physiol (Oxf) 2022; 236:e13861. [PMID: 35880402 PMCID: PMC9787084 DOI: 10.1111/apha.13861] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/12/2022] [Accepted: 07/20/2022] [Indexed: 01/29/2023]
Abstract
AIMS Diets containing high-fat and high sugar (HFHS) lead to overweight/obesity. Overweight/obesity increases the risk of infertility, and of the pregnant mother and her child for developing metabolic conditions. Overweight/obesity has been recreated in mice, but most studies focus on the effects of chronic, long-term HFHS diet exposure. Here, we exposed mice to HFHS from 3 weeks prior to pregnancy with the aim of determining impacts on fertility, and gestational and neonatal outcomes. METHODS Time-domain NMR scanning was used to assess adiposity, glucose, and insulin tolerance tests were employed to examine metabolic physiology, and morphological and proteomic analyses conducted to assess structure and nutrient levels of maternal organs and placenta. RESULTS Fertility measures of HFHS dams were largely the same as controls. HFHS dams had increased adiposity pre-pregnancy, however, exhibited exacerbated lipolysis/hyper-mobilization of adipose stores in late pregnancy. While there were no differences in glucose or insulin tolerance, HFHS dams were hyperglycemic and hyperinsulinemic in pregnancy. HFHS dams had fatty livers and altered pancreatic islet morphology. Although fetuses were hyperglycemic and hyperinsulinemic, there was no change in fetal growth in HFHS dams. There were also reductions in placenta formation. Moreover, there was increased offspring loss during lactation, which was related to aberrant mammary gland development and milk protein composition in HFHS dams. CONCLUSIONS These findings are relevant given current dietary habits and the development of maternal and offspring alterations in the absence of an increase in maternal weight and adiposity during pregnancy, which are the current clinical markers to determine risk across gestation.
Collapse
Affiliation(s)
- Samantha C Lean
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alejandro A Candia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Department for the Woman and Newborn Health Promotion, Universidad de Chile, Santiago, Chile
| | - Edina Gulacsi
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Giselle C L Lee
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Cade WT, Mittendorfer B, Patterson BW, Haire-Joshu D, Cahill AG, Stein RI, Schechtman KB, Tinius RA, Brown K, Klein S. Effect of excessive gestational weight gain on insulin sensitivity and insulin kinetics in women with overweight/obesity. Obesity (Silver Spring) 2022; 30:2014-2022. [PMID: 36150208 PMCID: PMC9512396 DOI: 10.1002/oby.23533] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Obesity increases the risk for pregnancy complications and maternal hyperglycemia. The Institute of Medicine developed guidelines for gestational weight gain (GWG) targets for women with overweight/obesity, but it is unclear whether exceeding these targets has adverse effects on maternal glucose metabolism. METHODS Insulin sensitivity (assessed using the Matsuda Insulin Sensitivity Index), β-cell function (assessed as insulin secretion rate in relation to plasma glucose), and plasma insulin clearance rate were evaluated using a frequently sampled oral glucose tolerance test at 15 and 35 weeks of gestation in 184 socioeconomically disadvantaged African American women with overweight/obesity. RESULTS Insulin sensitivity decreased, whereas β-cell function and insulin clearance increased from 15 to 35 weeks of gestation in the entire group. Compared with women who achieved the recommended GWG, excessive GWG was associated with a greater decrease in insulin sensitivity between 15 and 35 weeks. β-cell function and plasma insulin clearance were not affected by excessive GWG. CONCLUSIONS These data demonstrate that gaining more weight during pregnancy than recommended by the Institute of Medicine is associated with functional effects on glucose metabolism.
Collapse
Affiliation(s)
- W. Todd Cade
- Program in Physical Therapy, Washington University, St. Louis, Missouri, USA
| | | | - Bruce W. Patterson
- Center for Human Nutrition, Washington University, St. Louis, Missouri, USA
| | | | - Alison G. Cahill
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri, USA
- Department of Women’s Health, The University of Texas at Austin, Dell Medical School, Austin TX USA
| | - Richard I. Stein
- Center for Human Nutrition, Washington University, St. Louis, Missouri, USA
| | | | - Rachel A. Tinius
- Program in Physical Therapy, Washington University, St. Louis, Missouri, USA
| | - Katherine Brown
- Program in Physical Therapy, Washington University, St. Louis, Missouri, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
29
|
Song QX, Sun Y, Deng K, Mei JY, Chermansky CJ, Damaser MS. Potential role of oxidative stress in the pathogenesis of diabetic bladder dysfunction. Nat Rev Urol 2022; 19:581-596. [PMID: 35974244 DOI: 10.1038/s41585-022-00621-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
Diabetes mellitus is a chronic metabolic disease, posing a considerable threat to global public health. Treating systemic comorbidities has been one of the greatest clinical challenges in the management of diabetes. Diabetic bladder dysfunction, characterized by detrusor overactivity during the early stage of the disease and detrusor underactivity during the late stage, is a common urological complication of diabetes. Oxidative stress is thought to trigger hyperglycaemia-dependent tissue damage in multiple organs; thus, a growing body of literature has suggested a possible link between functional changes in urothelium, muscle and the corresponding innervations. Improved understanding of the mechanisms of oxidative stress could lead to the development of novel therapeutics to restore the redox equilibrium and scavenge excessive free radicals to normalize bladder function in patients with diabetes.
Collapse
Affiliation(s)
- Qi-Xiang Song
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Sun
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kangli Deng
- Department of Urology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Yi Mei
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | | | - Margot S Damaser
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA. .,Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA. .,Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
30
|
Lopez-Tello J, Salazar-Petres E, Webb L, Fowden AL, Sferruzzi-Perri AN. Ablation of PI3K-p110alpha Impairs Maternal Metabolic Adaptations to Pregnancy. Front Cell Dev Biol 2022; 10:928210. [PMID: 35846351 PMCID: PMC9283861 DOI: 10.3389/fcell.2022.928210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/10/2022] [Indexed: 01/03/2023] Open
Abstract
Pregnancy requires adaptations in maternal metabolism to support fetal growth. The phosphoinositol-3-kinase (PI3K) signalling pathway controls multiple biological processes and defects in this pathway are linked to metabolic disorders including insulin resistance and glucose intolerance in non-pregnant animals. However, relatively little is known about the contribution of PI3K signalling to the maternal metabolic adaptations during pregnancy. Using mice with partial inactivation of the PI3K isoform, p110α (due to a heterozygous dominant negative mutation; Pik3ca-D933A), the effects of impaired PI3K-p110α signalling on glucose and insulin handling were examined in the pregnant and non-pregnant states and related to the morphological, molecular, and mitochondrial changes in key metabolic organs. The results show that non-pregnant mice lacking PI3K-p110α are glucose intolerant but exhibit compensatory increases in pancreatic glucose-stimulated insulin release and adipose tissue mitochondrial respiratory capacity and fatty acid oxidation. However, in pregnancy, mutant mice failed to show the normal increment in glucose intolerance and pancreatic β-cell mass observed in wild-type pregnant dams and exhibited further enhanced adipose tissue mitochondrial respiratory capacity. These maladaptations in pregnant mutant mice were associated with fetal growth restriction. Hence, PI3K-p110α is a key regulator of metabolic adaptations that support fetal growth during normal pregnancy.
Collapse
Affiliation(s)
| | | | | | | | - Amanda N. Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
31
|
Napso T, Lean SC, Lu M, Mort EJ, Desforges M, Moghimi A, Bartels B, El‐Bacha T, Fowden AL, Camm EJ, Sferruzzi‐Perri AN. Diet-induced maternal obesity impacts feto-placental growth and induces sex-specific alterations in placental morphology, mitochondrial bioenergetics, dynamics, lipid metabolism and oxidative stress in mice. Acta Physiol (Oxf) 2022; 234:e13795. [PMID: 35114078 PMCID: PMC9286839 DOI: 10.1111/apha.13795] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023]
Abstract
AIM The current study investigated the impact of maternal obesity on placental phenotype in relation to fetal growth and sex. METHODS Female C57BL6/J mice were fed either a diet high in fat and sugar or a standard chow diet, for 6 weeks prior to, and during, pregnancy. At day 19 of gestation, placental morphology and mitochondrial respiration and dynamics were assessed using high-resolution respirometry, stereology, and molecular analyses. RESULTS Diet-induced maternal obesity increased the rate of small for gestational age fetuses in both sexes, and increased blood glucose concentrations in offspring. Placental weight, surface area, and maternal blood spaces were decreased in both sexes, with reductions in placental trophoblast volume, oxygen diffusing capacity, and an increased barrier to transfer in males only. Despite these morphological changes, placental mitochondrial respiration was unaffected by maternal obesity, although the influence of fetal sex on placental respiratory capacity varied between dietary groups. Moreover, in males, but not females, maternal obesity increased mitochondrial complexes (II and ATP synthase) and fission protein DRP1 abundance. It also reduced phosphorylated AMPK and capacity for lipid synthesis, while increasing indices of oxidative stress, specifically in males. In females only, placental mitochondrial biogenesis and capacity for lipid synthesis, were both enhanced. The abundance of uncoupling protein-2 was decreased by maternal obesity in both fetal sexes. CONCLUSION Maternal obesity exerts sex-dependent changes in placental phenotype in association with alterations in fetal growth and substrate supply. These findings may inform the design of personalized lifestyle interventions or therapies for obese pregnant women.
Collapse
Affiliation(s)
- Tina Napso
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Samantha C. Lean
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Minhui Lu
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Emily J. Mort
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Michelle Desforges
- Division of Developmental Biology and Medicine Maternal & Fetal Health Research Centre University of Manchester Manchester UK
| | - Ali Moghimi
- The Children’s Hospital at Westmead Westmead New South Wales Australia
- Department of Paediatrics Monash University Monash Victoria Australia
| | - Beverly Bartels
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Tatiana El‐Bacha
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Abigail L. Fowden
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Emily J. Camm
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Amanda N. Sferruzzi‐Perri
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| |
Collapse
|
32
|
Aldiss P, Lewis JE. The physiology of obesity; from mechanisms to medicine. Part two. J Physiol 2022; 600:999-1000. [PMID: 35229296 DOI: 10.1113/jp282466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Peter Aldiss
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jo Edward Lewis
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
33
|
Yeo E, Brubaker PL, Sloboda DM. The intestine and the microbiota in maternal glucose homeostasis during pregnancy. J Endocrinol 2022; 253:R1-R19. [PMID: 35099411 PMCID: PMC8942339 DOI: 10.1530/joe-21-0354] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/31/2022] [Indexed: 11/23/2022]
Abstract
It is now well established that, beyond its role in nutrient processing and absorption, the intestine and its accompanying gut microbiome constitute a major site of immunological and endocrine regulation that mediates whole-body metabolism. Despite the growing field of host-microbe research, few studies explore what mechanisms govern this relationship in the context of pregnancy. During pregnancy, significant maternal metabolic adaptations are made to accommodate the additional energy demands of the developing fetus and to prevent adverse pregnancy outcomes. Recent data suggest that the maternal gut microbiota may play a role in these adaptations, but changes to maternal gut physiology and the underlying intestinal mechanisms remain unclear. In this review, we discuss selective aspects of intestinal physiology including the role of the incretin hormone, glucagon-like peptide 1 (GLP-1), and the role of the maternal gut microbiome in the maternal metabolic adaptations to pregnancy. Specifically, we discuss how bacterial components and metabolites could mediate the effects of the microbiota on host physiology, including nutrient absorption and GLP-1 secretion and action, and whether these mechanisms may change maternal insulin sensitivity and secretion during pregnancy. Finally, we discuss how these pathways could be altered in disease states during pregnancy including maternal obesity and diabetes.
Collapse
Affiliation(s)
- Erica Yeo
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Obstetrics, Gynecology and Pediatrics, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
34
|
Mittendorfer B. Insulin, nobel laureates, and the journal of physiology. J Physiol 2022; 600:1269-1270. [DOI: 10.1113/jp282823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Bettina Mittendorfer
- Center for Human Nutrition Washington University School of Medicine St. Louis MO USA
| |
Collapse
|
35
|
Baumel-Alterzon S, Scott DK. Regulation of Pdx1 by oxidative stress and Nrf2 in pancreatic beta-cells. Front Endocrinol (Lausanne) 2022; 13:1011187. [PMID: 36187092 PMCID: PMC9521308 DOI: 10.3389/fendo.2022.1011187] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/26/2022] [Indexed: 01/05/2023] Open
Abstract
The beta-cell identity gene, pancreatic duodenal homeobox 1 (Pdx1), plays critical roles in many aspects of the life of beta-cells including differentiation, maturation, function, survival and proliferation. High levels of reactive oxygen species (ROS) are extremely toxic to cells and especially to beta-cells due to their relatively low expression of antioxidant enzymes. One of the major mechanisms for beta-cell dysfunction in type-2 diabetes results from oxidative stress-dependent inhibition of PDX1 levels and function. ROS inhibits Pdx1 by reducing Pdx1 mRNA and protein levels, inhibiting PDX1 nuclear localization, and suppressing PDX1 coactivator complexes. The nuclear factor erythroid 2-related factor (Nrf2) antioxidant pathway controls the redox balance and allows the maintenance of high Pdx1 levels. Therefore, pharmacological activation of the Nrf2 pathway may alleviate diabetes by preserving Pdx1 levels.
Collapse
Affiliation(s)
- Sharon Baumel-Alterzon
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Sharon Baumel-Alterzon,
| | - Donald K. Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
36
|
Vorobjova T, Tagoma A, Talja I, Janson H, Kirss A, Uibo R. FABP4 and I-FABP Levels in Pregnant Women Are Associated with Body Mass Index but Not Gestational Diabetes. J Diabetes Res 2022; 2022:1089434. [PMID: 35647197 PMCID: PMC9142318 DOI: 10.1155/2022/1089434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Gestational diabetes mellitus (GDM) is glucose intolerance detected initially during pregnancy. GDM poses an increased risk for the development of diabetes later in life. Fatty acid-binding protein 4 (FABP4) is a regulator of lipid metabolism and is associated with obesity, insulin resistance, and type 2 diabetes. Increased level of intestinal fatty acid-binding protein (I-FABP) may indicate impaired intestinal permeability, which may be an important contributor to the pathogenesis of type 1 diabetes and GDM. We aimed to compare FABP4 and I-FABP levels in pregnant women with GDM and in healthy pregnant controls, taking into consideration their prepregnancy body mass index (BMI), past exposures to enteroviruses (EV), and adipokine and cytokine levels, which have been shown to decrease insulin sensitivity. Material and Methods. Forty patients with GDM (median age 30.5) and 40 pregnant healthy controls (median age 31.1) were divided on the basis of their prepregnancy BMI into two groups: normal weight (BMI < 25, n = 20) and overweight (BMI ≥ 25, n = 20). FABP4 and I-FABP were measured from serum samples using commercial ELISA kits. RESULTS FABP4 and I-FABP levels did not differ between women with GDM and healthy pregnant controls (p > 0.05 for both comparisons). However, both levels were associated with BMI (p < 0.001 for both comparisons). Median I-FABP level was the highest in healthy controls with lower BMI (<25) (p = 0.0009). FABP4 levels correlated with BMI and C-peptide values in both groups (p < 0.001). Anti-EV antibody levels did not correlate with FABP4 or I-FABP levels. FABP4 and adiponectin levels were negatively correlated in controls (r = -0.61, p = 0.0009), while I-FABP correlated positively with adiponectin (r = 0.58, p = 0.04) and resistin (r = 0.67, p = 0.04) levels in the GDM group. CONCLUSION FABP4 and I-FABP levels were not dependent on the diagnosis of GDM, but rather on BMI. The correlation of I-FABP with adiponectin and resistin levels in women with GDM may suggests the importance of lipid metabolism in GDM-associated changes in intestinal permeability.
Collapse
Affiliation(s)
- Tamara Vorobjova
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Aili Tagoma
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Ija Talja
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Helis Janson
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Anne Kirss
- Women's Clinic, Tartu University Hospital, L. Puusepa 8, Tartu 51014, Estonia
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| |
Collapse
|
37
|
Aykroyd BRL, Tunster SJ, Sferruzzi-Perri AN. Loss of imprinting of the Igf2-H19 ICR1 enhances placental endocrine capacity via sex-specific alterations in signalling pathways in the mouse. Development 2022; 149:dev199811. [PMID: 34982814 PMCID: PMC8783045 DOI: 10.1242/dev.199811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022]
Abstract
Imprinting control region (ICR1) controls the expression of the Igf2 and H19 genes in a parent-of-origin specific manner. Appropriate expression of the Igf2-H19 locus is fundamental for normal fetal development, yet the importance of ICR1 in the placental production of hormones that promote maternal nutrient allocation to the fetus is unknown. To address this, we used a novel mouse model to selectively delete ICR1 in the endocrine junctional zone (Jz) of the mouse placenta (Jz-ΔICR1). The Jz-ΔICR1 mice exhibit increased Igf2 and decreased H19 expression specifically in the Jz. This was accompanied by an expansion of Jz endocrine cell types due to enhanced rates of proliferation and increased expression of pregnancy-specific glycoprotein 23 in the placenta of both fetal sexes. However, changes in the endocrine phenotype of the placenta were related to sexually-dimorphic alterations to the abundance of Igf2 receptors and downstream signalling pathways (Pi3k-Akt and Mapk). There was no effect of Jz-ΔICR1 on the expression of targets of the H19-embedded miR-675 or on fetal weight. Our results demonstrate that ICR1 controls placental endocrine capacity via sex-dependent changes in signalling.
Collapse
Affiliation(s)
| | | | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
38
|
Khant Aung Z, Kokay IC, Grattan DR, Ladyman SR. Prolactin-Induced Adaptation in Glucose Homeostasis in Mouse Pregnancy Is Mediated by the Pancreas and Not in the Forebrain. Front Endocrinol (Lausanne) 2021; 12:765976. [PMID: 34867810 PMCID: PMC8632874 DOI: 10.3389/fendo.2021.765976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/06/2021] [Indexed: 12/28/2022] Open
Abstract
Adaptive changes in glucose homeostasis during pregnancy require proliferation of insulin-secreting beta-cells in the pancreas, together with increased sensitivity for glucose-stimulated insulin secretion. Increased concentrations of maternal prolactin/placental lactogen contribute to these changes, but the site of action remains uncertain. Use of Cre-lox technology has generated pancreas-specific prolactin receptor (Prlr) knockouts that demonstrate the development of a gestational diabetic like state. However, many Cre-lines for the pancreas also express Cre in the hypothalamus and prolactin could act centrally to modulate glucose homeostasis. The aim of the current study was to examine the relative contribution of prolactin action in the pancreas and brain to these pregnancy-induced adaptations in glucose regulation. Deletion of prolactin receptor (Prlr) from the pancreas using Pdx-cre or Rip-cre led to impaired glucose tolerance and increased non-fasting blood glucose levels during pregnancy. Prlrlox/lox /Pdx-Cre mice also had impaired glucose-stimulated insulin secretion and attenuated pregnancy-induced increase in beta-cell fraction. Varying degrees of Prlr recombination in the hypothalamus with these Cre lines left open the possibility that central actions of prolactin could contribute to the pregnancy-induced changes in glucose homeostasis. Targeted deletion of Prlr specifically from the forebrain, including areas of expression induced by Pdx-Cre and Rip-cre, had no effect on pregnancy-induced adaptations in glucose homeostasis. These data emphasize the pancreas as the direct target of prolactin/placental lactogen action in driving adaptive changes in glucose homeostasis during pregnancy.
Collapse
Affiliation(s)
- Zin Khant Aung
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Ilona C. Kokay
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - David R. Grattan
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Sharon R. Ladyman
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- *Correspondence: Sharon R. Ladyman,
| |
Collapse
|