1
|
Yi Y, Wang T, Xu W, Zhang SH. Epigenetic modifications of placenta in women with gestational diabetes mellitus and their offspring. World J Diabetes 2024; 15:378-391. [PMID: 38591094 PMCID: PMC10999040 DOI: 10.4239/wjd.v15.i3.378] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/30/2023] [Accepted: 02/06/2024] [Indexed: 03/15/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is a pregnancy-related complication characterized by abnormal glucose metabolism in pregnant women and has an important impact on fetal development. As a bridge between the mother and the fetus, the placenta has nutrient transport functions, endocrine functions, etc., and can regulate placental nutrient transport and fetal growth and development according to maternal metabolic status. Only by means of placental transmission can changes in maternal hyperglycemia affect the fetus. There are many reports on the placental pathophysiological changes associated with GDM, the impacts of GDM on the growth and development of offspring, and the prevalence of GDM in offspring after birth. Placental epigenetic changes in GDM are involved in the programming of fetal development and are involved in the pathogenesis of later chronic diseases. This paper summarizes the effects of changes in placental nutrient transport function and hormone secretion levels due to maternal hyperglycemia and hyperinsulinemia on the development of offspring as well as the participation of changes in placental epigenetic modifications due to maternal hyperglycemia in intrauterine fetal programming to promote a comprehensive understanding of the impacts of placental epigenetic modifications on the development of offspring from patients with GDM.
Collapse
Affiliation(s)
- Yan Yi
- Department of Ultrasonography, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Tao Wang
- Clinical Molecular Immunology Center, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Wei Xu
- Department of Ultrasonography, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - San-Hong Zhang
- Department of Pediatric, Xiantao First People’s Hospital, Xiantao 433000, Hubei Province, China
| |
Collapse
|
2
|
Szrok-Jurga S, Czumaj A, Turyn J, Hebanowska A, Swierczynski J, Sledzinski T, Stelmanska E. The Physiological and Pathological Role of Acyl-CoA Oxidation. Int J Mol Sci 2023; 24:14857. [PMID: 37834305 PMCID: PMC10573383 DOI: 10.3390/ijms241914857] [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: 08/25/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Fatty acid metabolism, including β-oxidation (βOX), plays an important role in human physiology and pathology. βOX is an essential process in the energy metabolism of most human cells. Moreover, βOX is also the source of acetyl-CoA, the substrate for (a) ketone bodies synthesis, (b) cholesterol synthesis, (c) phase II detoxication, (d) protein acetylation, and (d) the synthesis of many other compounds, including N-acetylglutamate-an important regulator of urea synthesis. This review describes the current knowledge on the importance of the mitochondrial and peroxisomal βOX in various organs, including the liver, heart, kidney, lung, gastrointestinal tract, peripheral white blood cells, and other cells. In addition, the diseases associated with a disturbance of fatty acid oxidation (FAO) in the liver, heart, kidney, lung, alimentary tract, and other organs or cells are presented. Special attention was paid to abnormalities of FAO in cancer cells and the diseases caused by mutations in gene-encoding enzymes involved in FAO. Finally, issues related to α- and ω- fatty acid oxidation are discussed.
Collapse
Affiliation(s)
- Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Jacek Turyn
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Areta Hebanowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Julian Swierczynski
- Institue of Nursing and Medical Rescue, State University of Applied Sciences in Koszalin, 75-582 Koszalin, Poland;
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Ewa Stelmanska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| |
Collapse
|
3
|
Pinto GDA, Murgia A, Lai C, Ferreira CS, Goes VA, Guimarães DDAB, Ranquine LG, Reis DL, Struchiner CJ, Griffin JL, Burton GJ, Torres AG, El-Bacha T. Sphingolipids and acylcarnitines are altered in placentas from women with gestational diabetes mellitus. Br J Nutr 2023; 130:921-932. [PMID: 36539977 DOI: 10.1017/s000711452200397x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gestational diabetes mellitus (GDM) is the most common medical complication of pregnancy and a severe threat to pregnant people and offspring health. The molecular origins of GDM, and in particular the placental responses, are not fully known. The present study aimed to perform a comprehensive characterisation of the lipid species in placentas from pregnancies complicated with GDM using high-resolution MS lipidomics, with a particular focus on sphingolipids and acylcarnitines in a semi-targeted approach. The results indicated that despite no major disruption in lipid metabolism, placentas from GDM pregnancies showed significant alterations in sphingolipids, mostly lower abundance of total ceramides. Additionally, very long-chain ceramides and sphingomyelins with twenty-four carbons were lower, and glucosylceramides with sixteen carbons were higher in placentas from GDM pregnancies. Semi-targeted lipidomics revealed the strong impact of GDM on the placental acylcarnitine profile, particularly lower contents of medium and long-chain fatty-acyl carnitine species. The lower contents of sphingolipids may affect the secretory function of the placenta, and lower contents of long-chain fatty acylcarnitines is suggestive of mitochondrial dysfunction. These alterations in placental lipid metabolism may have consequences for fetal growth and development.
Collapse
Affiliation(s)
- Gabriela D A Pinto
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | | | - Carla Lai
- University of Cagliari, Department of Life and Environmental Science, Cagliari Via Ospedale, Cagliari, Italy
| | - Carolina S Ferreira
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Vanessa A Goes
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Deborah de A B Guimarães
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Layla G Ranquine
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Desirée L Reis
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Claudio J Struchiner
- School of Applied Mathematics, Fundação Getúlio Vargas, Rio de Janeiro, Brazil
- Institute of Social Medicine, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julian L Griffin
- Department of Biochemistry, Cambridge, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alexandre G Torres
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Watkins OC, Pillai RA, Selvam P, Yong HE, Cracknell‐Hazra VK, Sharma N, Cazenave‐Gassiot A, Bendt AK, Godfrey KM, Lewis RM, Wenk MR, Chan S. Myo-inositol alters the effects of glucose, leptin and insulin on placental palmitic acid and oleic acid metabolism. J Physiol 2023; 601:4151-4169. [PMID: 37602663 PMCID: PMC10952252 DOI: 10.1113/jp285036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Well-regulated placental palmitic acid (PA) and oleic acid (OA) metabolism is vital for optimal placental function and fetal development, but dysregulation occurs with gestational diabetes (GDM). We hypothesized that such dysregulation might arise from increased maternofetal glucose, leptin or insulin concentrations present in GDM, and that dysregulated PA and OA lipid metabolism could be moderated by myo-inositol, a natural polyol and potential GDM intervention. Placental explants from 21 women were incubated with stable isotope-labelled 13 C-PA or 13 C-OA for 48 h. Explants were treated with glucose (5, 10 mm) or leptin (13 nm) or insulin (150 nm) in combination with myo-inositol (0.3, 30, 60 μm). Forty-seven 13 C-PA lipids and 37 13 C-OA lipids were measured by liquid chromatography-mass spectrometry (LCMS). Compared with controls (5 mm glucose), glucose (10 mm) increased 19 13 C-OA lipids and nine 13 C-PA lipids, but decreased 13 C-OA phosphatidylethanolamine 38:5 and 13 C-PA phosphatidylethanolamine 36:4. The effects of leptin and insulin were less prominent than glucose, with leptin increasing 13 C-OA acylcarnitine 18:1, and insulin increasing four 13 C-PA triacylglycerides. Most glucose, leptin and insulin-induced alterations in lipids were attenuated by co-incubation with myo-inositol (30 or 60 μm), with attenuation also occurring in all subgroups stratified by GDM status and fetal sex. However, glucose-induced increases in acylcarnitine were not attenuated by myo-inositol and were even exaggerated in some instances. Myo-inositol therefore appears to generally act as a moderator, suppressing the perturbation of lipid metabolic processes by glucose, leptin and insulin in placenta in vitro. Whether myo-inositol protects the fetus and pregnancy from unfavourable outcomes requires further research. KEY POINTS: Incubation of placental explants with additional glucose, or to a lesser extent insulin or leptin, alters the placental production of 13 C-lipids from 13 C-palmitic acid (PA) and 13 C-oleic acid (OA) in vitro compared with untreated controls from the same placenta. Co-incubation with myo-inositol attenuated most alterations induced by glucose, insulin or leptin in 13 C-lipids, but did not affect alterations in 13 C-acylcarnitines. Alterations induced by glucose and leptin in 13 C-PA triacylglycerides and 13 C-PA phospholipids were influenced by fetal sex and gestational diabetes status, but were all still attenuated by myo-inositol co-incubation. Insulin differently affected 13 C-PA triacylglycerides and 13 C-PA phospholipids depending on fetal sex, with alterations also attenuated by myo-inositol co-incubation.
Collapse
Affiliation(s)
- Oliver C. Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Reshma Appukuttan Pillai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Preben Selvam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Hannah E.J. Yong
- Singapore Institute for Clinical SciencesAgency for Science, Technology and ResearchSingapore
| | - Victoria K.B. Cracknell‐Hazra
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Singapore Institute for Clinical SciencesAgency for Science, Technology and ResearchSingapore
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustUK
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Amaury Cazenave‐Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine and Precision Medicine TRPNational University of SingaporeSingapore
- Singapore Lipidomics Incubator, Life Sciences InstituteNational University of SingaporeSingapore
| | - Anne K. Bendt
- Singapore Lipidomics Incubator, Life Sciences InstituteNational University of SingaporeSingapore
| | - Keith M. Godfrey
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustUK
| | - Rohan M. Lewis
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustUK
| | - Markus R. Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine and Precision Medicine TRPNational University of SingaporeSingapore
- Singapore Lipidomics Incubator, Life Sciences InstituteNational University of SingaporeSingapore
| | - Shiao‐Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Singapore Institute for Clinical SciencesAgency for Science, Technology and ResearchSingapore
| |
Collapse
|
5
|
Easton ZJW, Sarr O, Zhao L, Buzatto AZ, Luo X, Zhao S, Li L, Regnault TRH. An Integrated Multi-OMICS Approach Highlights Elevated Non-Esterified Fatty Acids Impact BeWo Trophoblast Metabolism and Lipid Processing. Metabolites 2023; 13:883. [PMID: 37623828 PMCID: PMC10456680 DOI: 10.3390/metabo13080883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Maternal obesity and gestational diabetes mellitus (GDM) are linked with impaired placental function and early onset of non-communicable cardiometabolic diseases in offspring. Previous studies have highlighted that the dietary non-esterified fatty acids (NEFAs) palmitate (PA) and oleate (OA), key dietary metabolites associated with maternal obesity and GDM, are potential modulators of placental lipid processing. Using the BeWo cell line model, the current study integrated transcriptomic (mRNA microarray), metabolomic, and lipidomic readouts to characterize the underlying impacts of exogenous PA and OA on placental villous trophoblast cell metabolism. Targeted gas chromatography and thin-layer chromatography highlighted that saturated and monounsaturated NEFAs differentially impact BeWo cell lipid profiles. Furthermore, cellular lipid profiles differed when exposed to single and multiple NEFA species. Additional multi-omic analyses suggested that PA exposure is associated with enrichment in β-oxidation pathways, while OA exposure is associated with enrichment in anti-inflammatory and antioxidant pathways. Overall, this study further demonstrated that dietary PA and OA are important regulators of placental lipid metabolism. Encouraging appropriate dietary advice and implementing dietary interventions to maintain appropriate placental function by limiting excessive exposure to saturated NEFAs remain crucial in managing at-risk obese and GDM pregnancies.
Collapse
Affiliation(s)
- Zachary J. W. Easton
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
| | - Ousseynou Sarr
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
| | - Lin Zhao
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
| | - Adriana Zardini Buzatto
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
| | - Xian Luo
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
| | - Shuang Zhao
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
| | - Liang Li
- The Metabolomics Innovation Centre (TMIC), University of Alberta, Edmonton, AB T6G 2G2, Canada; (A.Z.B.); (X.L.); (S.Z.); (L.L.)
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Timothy R. H. Regnault
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada; (Z.J.W.E.); (O.S.); (L.Z.)
- Department of Obstetrics and Gynaecology, Western University, B2-401 London Health Science Centre-Victoria Hospital, 800 Commissioners Rd E, London, ON N6H 5W9, Canada
- Children’s Health Research Institute, 800 Commissioners Rd E, London, ON N6C 2V5, Canada
- Lawson Health Research Institute, 750 Base Line Rd E, London, ON N6C 2R5, Canada
| |
Collapse
|
6
|
Visiedo F, Vázquez-Fonseca L, Ábalos-Martínez J, Broullón-Molanes JR, Quintero-Prado R, Mateos RM, Bugatto F. Maternal elevated inflammation impairs placental fatty acids β-oxidation in women with gestational diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1146574. [PMID: 37214247 PMCID: PMC10196201 DOI: 10.3389/fendo.2023.1146574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/03/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction An adverse proinflammatory milieu contributes to abnormal cellular energy metabolism response. Gestational diabetes mellitus (GDM) is closely related to an altered maternal inflammatory status. However, its role on lipid metabolism regulation in human placenta has not yet been assessed. The aim of this study was to examine the impact of maternal circulating inflammatory mediators ([TNF]-α, [IL]-6, and Leptin) on placental fatty acid metabolism in GDM pregnancies. Methods Fasting maternal blood and placental tissues were collected at term deliveries from 37 pregnant women (17 control and 20 GDM). Molecular approach techniques as radiolabeled lipid tracers, ELISAs, immunohistochemistry and multianalyte immunoassay quantitative analysis, were used to quantify serum inflammatory factors' levels, to measure lipid metabolic parameters in placental villous samples (mitochondrial fatty acid oxidation [FAO] rate and lipid content [Triglycerides]), and to analyze their possible relationships. The effect of potential candidate cytokines on fatty acid metabolism in ex vivo placental explants culture following C-section a term was also examined. Results Maternal serum IL-6, TNF-α and leptin levels were significantly increased in GDM patients compared with control pregnant women (9,9±4,5 vs. 3,00±1,7; 4,5±2,8 vs. 2,1±1,3; and 10026,7±5628,8 vs. 5360,2±2499,9 pg/ml, respectively). Placental FAO capacity was significantly diminished (~30%; p<0.01), whereas triglyceride levels were three-fold higher (p<0.01) in full-term GDM placentas. Uniquely the maternal IL-6 levels showed an inverse and positive correlation with the ability to oxidize fatty acids and triglyceride amount in placenta, respectively (r= -0,602, p=0.005; r= 0,707, p=0.001). Additionally, an inverse correlation between placental FAO and triglycerides was also found (r=-0.683; p=0.001). Interestingly, we ex vivo demonstrated by using placental explant cultures that a prolonged exposure with IL-6 (10 ng/mL) resulted in a decline in the fatty acid oxidation rate (~25%; p=0.001), along to acute increase (2-fold times) in triglycerides accumulation (p=0.001), and in lipid neutral and lipid droplets deposits. Conclusions Enhanced maternal proinflammatory cytokines levels (essentially IL-6) is closely associated with an altered placental fatty acid metabolism in pregnancies with GDM, which may interfere with adequate delivery of maternal fat across the placenta to the fetus.
Collapse
Affiliation(s)
- Francisco Visiedo
- Inflammation and Metabolic Syndrome in Pregnancy Group (CO25), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Cádiz, Spain
| | - Luis Vázquez-Fonseca
- Inflammation and Metabolic Syndrome in Pregnancy Group (CO25), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Cádiz, Spain
| | - Jessica Ábalos-Martínez
- Inflammation and Metabolic Syndrome in Pregnancy Group (CO25), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Cádiz, Spain
| | - J. Román Broullón-Molanes
- Inflammation and Metabolic Syndrome in Pregnancy Group (CO25), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Cádiz, Spain
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology “Puerta del Mar” University Hospital, University of Cádiz, Cádiz, Spain
- Area of Obstetrics and Gynaecology, Department of Child and Mother Health and Radiology, School of Medicine, University of Cádiz, Cádiz, Spain
| | - Rocío Quintero-Prado
- Department of Obstetrics and Gynecology, Puerto Real University Hospital, Cadiz, Spain
| | - Rosa María Mateos
- Area of Biochemistry and Molecular Biology, Department of Biomedicine, Biotechnology and Public Health, University of Cádiz, Cádiz, Spain
| | - Fernando Bugatto
- Inflammation and Metabolic Syndrome in Pregnancy Group (CO25), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Cádiz, Spain
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology “Puerta del Mar” University Hospital, University of Cádiz, Cádiz, Spain
- Area of Obstetrics and Gynaecology, Department of Child and Mother Health and Radiology, School of Medicine, University of Cádiz, Cádiz, Spain
| |
Collapse
|
7
|
Ferreira CS, Pinto GDA, Reis DL, Vigor C, Goes VA, Guimarães DDAB, Mucci DB, Belcastro L, Saraiva MA, Oger C, Galano JM, Sardinha FLC, Torres AG, Durand T, Burton GJ, El-Bacha T. Placental F 4-Neuroprostanes and F 2-Isoprostanes are altered in gestational diabetes mellitus and maternal obesity. Prostaglandins Leukot Essent Fatty Acids 2023; 189:102529. [PMID: 36608621 DOI: 10.1016/j.plefa.2022.102529] [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: 08/28/2022] [Revised: 11/30/2022] [Accepted: 12/23/2022] [Indexed: 12/26/2022]
Abstract
We investigated whether gestational diabetes mellitus (GDM) associated with maternal obesity modifies the placental profile of F4-Neuroprostanes and F2-Isoprostanes, metabolites of non-enzymatic oxidation of docosahexaenoic acid (DHA) and arachidonic acid (AA), respectively. Twenty-five placental samples were divided into lean (n=11), obesity (n=7) and overweight/obesity+GDM (n=7) groups. F4-Neuroprostanes and F2-Isoprostanes were higher in obesity compared to lean controls, but reduced to levels similar to lean women when obesity is further complicated with GDM. Lower content of F2-Isoprostanes suggests adaptive placental responses in GDM attenuating oxidative stress. However, low levels of placental F4-Neuroprostanes may indicate impaired DHA metabolism in GDM, affecting fetal development and offspring health. These results were not related to differences in placental content of DHA, AA and polyunsaturated fatty acids status nor to maternal diet or gestational weight gain. Placental DHA and AA metabolism differs in obesity and GDM, highlighting the importance of investigating the signalling roles of F4-Neuroprostanes and F2-Isoprostanes in the human term placenta.
Collapse
Affiliation(s)
- Carolina S Ferreira
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Gabriela D A Pinto
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Desirée L Reis
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 route de Mende, CEDEX 5, 34293 Montpellier, France
| | - Vanessa A Goes
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Deborah de A B Guimarães
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Daniela B Mucci
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Livia Belcastro
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Marcelle A Saraiva
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 route de Mende, CEDEX 5, 34293 Montpellier, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 route de Mende, CEDEX 5, 34293 Montpellier, France
| | - Fátima L C Sardinha
- Laboratory of Nutritional Biochemistry, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Alexandre G Torres
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; Laboratory of Biochemistry and Chemistry of Lipids, Department of Chemistry, Universidade Federal do Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, Bâtiment Balard, 1919 route de Mende, CEDEX 5, 34293 Montpellier, France
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EG, United Kingdom
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EG, United Kingdom.
| |
Collapse
|
8
|
Easton ZJW, Luo X, Li L, Regnault TRH. The impact of hyperglycemia upon BeWo trophoblast cell metabolic function: A multi-OMICS and functional metabolic analysis. PLoS One 2023; 18:e0283118. [PMID: 36930661 PMCID: PMC10022812 DOI: 10.1371/journal.pone.0283118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
Pre-existing and gestationally-developed diabetes mellitus have been linked with impairments in placental villous trophoblast cell metabolic function, that are thought to underlie the development of metabolic diseases early in the lives of the exposed offspring. Previous research using placental cell lines and ex vivo trophoblast preparations have highlighted hyperglycemia is an important independent regulator of placental function. However, it is poorly understood if hyperglycemia directly influences aspects of placental metabolic function, including nutrient storage and mitochondrial respiration, that are altered in term diabetic placentae. The current study examined metabolic and mitochondrial function as well as nutrient storage in both undifferentiated cytotrophoblast and differentiated syncytiotrophoblast BeWo cells cultured under hyperglycemia conditions (25 mM glucose) for 72 hours to further characterize the direct impacts of placental hyperglycemic exposure. Hyperglycemic-exposed BeWo trophoblasts displayed increased glycogen and triglyceride nutrient stores, but real-time functional readouts of metabolic enzyme activity and mitochondrial respiratory activity were not altered. However, specific investigation into mitochondrial dynamics highlighted increased expression of markers associated with mitochondrial fission that could indicate high glucose-exposed trophoblasts are transitioning towards mitochondrial dysfunction. To further characterize the impacts of independent hyperglycemia, the current study subsequently utilized a multi-omics approach and evaluated the transcriptomic and metabolomic signatures of BeWo cytotrophoblasts. BeWo cytotrophoblasts exposed to hyperglycemia displayed increased mRNA expression of ACSL1, HSD11B2, RPS6KA5, and LAP3 and reduced mRNA expression of CYP2F1, and HK2, concomitant with increased levels of: lactate, malonate, and riboflavin metabolites. These changes highlighted important underlying alterations to glucose, glutathione, fatty acid, and glucocorticoid metabolism in BeWo trophoblasts exposed to hyperglycemia. Overall, these results demonstrate that hyperglycemia is an important independent regulator of key areas of placental metabolism, nutrient storage, and mitochondrial function, and these data continue to expand our knowledge on mechanisms governing the development of placental dysfunction.
Collapse
Affiliation(s)
- Zachary J W Easton
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Xian Luo
- The Metabolomics Innovation Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Liang Li
- The Metabolomics Innovation Centre, University of Alberta, Edmonton, Alberta, Canada
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Timothy R H Regnault
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Department of Obstetrics and Gynaecology, London Health Science Centre-Victoria Hospital, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
9
|
Fan X, Dai J, He J, Tian R, Xu J, Song J, Bai J, Liu Y, Zou Z, Chen X. Optimal gestational weight gain in Chinese pregnant women with gestational diabetes mellitus: A large retrospective cohort study. J Obstet Gynaecol Res 2023; 49:182-193. [PMID: 36184564 DOI: 10.1111/jog.15448] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/11/2022] [Accepted: 09/16/2022] [Indexed: 01/19/2023]
Abstract
AIM To examine the optimal gestational weight gain (GWG) for Chinese pregnant women with gestational diabetes mellitus (GDM) based on the Chinese-specific body mass index (BMI) classification. METHODS A retrospective cohort study was conducted using the 2017-2020 data from pregnant women with GDM in a tertiary hospital. A quadratic function model and the total predicted probability of adverse pregnancy outcomes were developed to obtain the optimal GWG. Differences in the incidence of adverse pregnancy outcomes between our optimal GWG recommendations and the Institute of Medicine (IOM) 2009 GWG guidelines were also analyzed. RESULTS A total of 8103 pregnant women with GDM were analyzed. Based on the Chinese-specific BMI classification, the optimal GWG range was 11.0-17.5 kg for underweight women, 3.7-9.7 kg for normal-weight women, -0.6 to 4.8 kg for overweight women, and - 9.8 to 4.2 kg for obese women. Excessive GWG had a higher risk of large for gestational age (LGA) (OR: 2.99, 95% CI: 2.42-3.70), macrosomia (OR: 2.35, 95% CI: 1.77-3.12), pre-eclampsia (OR: 1.91, 95% CI: 1.37-2.65), gestational hypertension (OR: 1.65, 95% CI: 1.24-2.19), cesarean section (OR: 1.29, 95% CI: 1.15-1.44), postpartum hemorrhage (OR: 1.29, 95% CI: 1.02-1.64); insufficient GWG had a higher risk of small for gestational age (OR: 1.82, 95% CI: 1.20-2.75). Compared to the IOM 2009 GWG guidelines, the prevalence of macrosomia, LGA, and postpartum hemorrhage were significantly lower in pregnant women following the implementation of our recommended GWG range (p < 0.05). CONCLUSIONS Compared to the IOM 2009 GWG recommendations, our optimal GWG recommendations for Chinese pregnant women were more sensitive.
Collapse
Affiliation(s)
- Xiaoxiao Fan
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Jiamiao Dai
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Jing He
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Ruixue Tian
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Jingqi Xu
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Jiayang Song
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Jinbing Bai
- Emory University Nell Hodgson Woodruff School of Nursing, Atlanta, Georgia, USA
| | - Yanqun Liu
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Zhijie Zou
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| | - Xiaoli Chen
- Wuhan University School of Nursing, Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Chemical inhibition of mitochondrial fission improves insulin signaling and subdues hyperglycemia induced stress in placental trophoblast cells. Mol Biol Rep 2023; 50:493-506. [PMID: 36352179 DOI: 10.1007/s11033-022-07959-0] [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: 05/31/2022] [Accepted: 09/17/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is a metabolic complication that affects millions of pregnant women in the world. Placental tissue function is endangered by hyperglycemia during GDM, which is correlated to increased incidences of pregnancy complications. Recently we showed that due to a significant decrease in mitochondrial fusion, mitochondrial dynamics equilibrium is altered in placental tissues from GDM patients. Evidence for the role of reduced mitochondrial fusion in the disruption of mitochondrial function in placental cells is limited. METHODS AND RESULTS Here we show that chemical inhibition of mitochondrial fission in cultured placental trophoblast cells leads to an increase in mitochondrial fusion and improves the physiological state of these cells and hence, their capacity to cope in a hyperglycemic environment. Specifically, mitochondrial fission inhibition led to a reduction in reactive oxygen species (ROS) generation, mitochondrial unfolded protein marker expressions, and mitochondrial depolarization. It supported the increase in mitochondrial antioxidant enzyme expressions as well. Mitochondrial fission inhibition also increases the placental cell insulin sensitivity during hyperglycemia. CONCLUSION Our results suggest that mitochondrial fusion/fission equilibrium is critical for placental cell function and signify the therapeutic potential of small molecule inhibitors of fission during GDM.
Collapse
|
11
|
Kuentzel KB, Bradić I, Mihalič ZN, Korbelius M, Rainer S, Pirchheim A, Kargl J, Kratky D. Dysregulation of Placental Lipid Hydrolysis by High-Fat/High-Cholesterol Feeding and Gestational Diabetes Mellitus in Mice. Int J Mol Sci 2022; 23:12286. [PMID: 36293139 PMCID: PMC9603336 DOI: 10.3390/ijms232012286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Advanced maternal age and obesity are the main risk factors to develop gestational diabetes mellitus (GDM). Obesity is a consequence of the increased storage of triacylglycerol (TG). Cytosolic and lysosomal lipid hydrolases break down TG and cholesteryl esters (CE) to release fatty acids (FA), free cholesterol, and glycerol. We have recently shown that intracellular lipases are present and active in the mouse placenta and that deficiency of lysosomal acid lipase alters placental and fetal lipid homeostasis. To date, intracellular lipid hydrolysis in GDM has been poorly studied despite the important role of FA in this condition. Therefore, we hypothesized that intracellular lipases are dysregulated in pregnancies complicated by maternal high-fat/high-cholesterol (HF/HCD) feeding with and without GDM. Placentae of HF/HCD-fed mice with and without GDM were more efficient, indicating increased nutrient transfer to the fetus. The increased activity of placental CE but not TG hydrolases in placentae of dams fed HF/HCD with or without GDM resulted in upregulated cholesterol export to the fetus and placental TG accumulation. Our results indicate that HF/HCD-induced dysregulation of placental lipid hydrolysis contributes to fetal hepatic lipid accumulation and possibly to fetal overgrowth, at least in mice.
Collapse
Affiliation(s)
- Katharina B. Kuentzel
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Ivan Bradić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Zala N. Mihalič
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Melanie Korbelius
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Silvia Rainer
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Julia Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
12
|
Garcia-Santillan JA, Lazo-de-la-Vega-Monroy ML, Rodriguez-Saldaña GC, Solis-Barbosa MA, Corona-Figueroa MA, Gonzalez-Dominguez MI, Gomez-Zapata HM, Malacara JM, Barbosa-Sabanero G. Placental Nutrient Transporters and Maternal Fatty Acids in SGA, AGA, and LGA Newborns From Mothers With and Without Obesity. Front Cell Dev Biol 2022; 10:822527. [PMID: 35399516 PMCID: PMC8990844 DOI: 10.3389/fcell.2022.822527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/25/2022] [Indexed: 12/01/2022] Open
Abstract
Adverse environmental factors in early life result in fetal metabolic programming and increased risk of adult diseases. Birth weight is an indirect marker of the intrauterine environment, modulated by nutrient availability and placental transport capacity. However, studies of placental transporters in idiopathic birth weight alterations and in maternal obesity in relation to neonatal metabolic outcomes are scarce. We aimed to analyze the placental nutrient transporter protein expression in small (SGA, n = 14), adequate (AGA, n = 18), and large (LGA n = 10) gestational age term for newborns from healthy or obese mothers (LGA-OB, n = 9) and their association with maternal fatty acids, metabolic status, placental triglycerides, and neonatal outcomes. The transporter expression was determined by Western blot. The fatty acid profile was evaluated by gas chromatography, and placental triglycerides were quantified by an enzymatic colorimetric method. GLUT1 was higher in LGA and lower in SGA and positively correlated with maternal HbA1c and placental weight (PW). SNAT2 was lower in SGA, while SNAT4 was lower in LGA-OB. FATP1 was lower in SGA and higher in LGA. SNAT4 correlated negatively and FATP1 correlated positively with the PW and birth anthropometry (BA). Placental triglycerides were higher in LGA and LGA-OB and correlated with pregestational BMI, maternal insulin, and BA. Maternal docosahexaenoic acid (DHA) was higher in SGA, specifically in male placentas, correlating negatively with maternal triglycerides, PW, cord glucose, and abdominal perimeter. Palmitic acid (PA) correlated positively with FATP4 and cord insulin, linoleic acid correlated negatively with PA and maternal cholesterol, and arachidonic acid correlated inversely with maternal TG and directly with FATP4. Our study highlights the importance of placental programming in birth weight both in healthy and obese pregnancies.
Collapse
Affiliation(s)
| | | | | | - Miguel-Angel Solis-Barbosa
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Campus Leon, Guanajuato, Mexico
| | | | | | | | - Juan-Manuel Malacara
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Campus Leon, Guanajuato, Mexico
| | - Gloria Barbosa-Sabanero
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Campus Leon, Guanajuato, Mexico
- *Correspondence: Gloria Barbosa-Sabanero,
| |
Collapse
|
13
|
Ortega MA, Sáez MA, Fraile-Martínez O, Álvarez-Mon MA, García-Montero C, Guijarro LG, Asúnsolo Á, Álvarez-Mon M, Bujan J, García-Honduvilla N, De León-Luis JA, Bravo C. Overexpression of glycolysis markers in placental tissue of pregnant women with chronic venous disease: a histological study. Int J Med Sci 2022; 19:186-194. [PMID: 34975312 PMCID: PMC8692115 DOI: 10.7150/ijms.65419] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic Venous Disease (CVD) refers to a wide variety of venous disorders being the varicose veins its most common manifestation. It is well-established the link between pregnancy and the risk of suffering CVD, due to hormonal or haematological factors, especially during the third trimester. In the same manner, previous studies have demonstrated the detrimental effect of this condition in the placental tissue of pregnant women, including in the normal physiology and the metabolomic profile of this organ. In this context, the aim of this study was to evaluate the glucose homeostasis in the placental tissue of women presenting CVD. Through immunohistochemistry, we studied the protein expression of the glucose transporter 1 (GLUT-1), Phosphoglycerate kinase 1 (PGK1), aldolase (ALD), Glyceraldehyde-3-phosphate dehydrogenase (GA3PDH) and lactate dehydrogenase (LDH). Our results have reported a significative increase in the expression of GLUT-1, PGK1, ALD, GA3PDH and the isoenzyme LDHA in placentas of women with CVD. This work has proven for the first-time an altered glucose metabolism in the placental tissue of women affected by CVD, what may aid to understand the pathophysiological mechanisms of this condition in more distant organs such as placenta. Furthermore, our research also supports the basis for further studies in the metabolic phenotyping of the human placenta due to CVD, which may be considered during the late pregnancy in these women.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Spain
| | - Miguel A Sáez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Miguel A Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Luis G Guijarro
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital Príncipe de Asturias, CIBEREHD, Alcalá de Henares, Madrid, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Juan A De León-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid 28009, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid 28009, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| |
Collapse
|
14
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
15
|
Zhang C, Zhao D. MicroRNA-362-5p promotes the proliferation and inhibits apoptosis of trophoblast cells via targeting glutathione-disulfide reductase. Bioengineered 2021; 12:2410-2419. [PMID: 34107852 PMCID: PMC8806602 DOI: 10.1080/21655979.2021.1933678] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gestational diabetes mellitus (GDM), a common complication of pregnancy, harms the health of pregnant women and fetuses. MicroRNAs (miRNAs) dysregulation in placenta is involved in GDM. Herein, we explored the roles of miR-362-5p in GDM. After high glucose (HG) treated HTR-8/SVneo cells, CCK-8 and flow cytometry were conducted to assess the capability of the proliferation and apoptosis, respectively. The data demonstrated that HG inhibited proliferation and induced apoptosis of HTR-8/SVneo cells. MiR-362-5p level was reduced in HG-treated cells and placenta tissues of GDM patients, measured by qPCR. Overexpressed miR-362-5p accelerated the proliferation and restrained apoptosis of HG-treated cells. Furthermore, glutathione-disulfide reductase (GSR) was verified as a target of miR-362-5p, through TargetScan database and dual-luciferase reporter assay. GSR was upregulated in GDM placenta tissues and was negatively regulated by miR-362-5p. Enforced GSR level abolished the effects of miR-362-5p overexpression on the proliferation and apoptosis of HTR-8/SVneo cells. Furthermore, miR-362-5p increased p-PI3K, p-AKT and bcl-2, while reduced bax and cleaved caspase3, which were abolished by GSR. In conclusion, miR-362-5p promoted cell proliferation and inhibited apoptosis via targeting GSR and activating PI3K/AKT pathway. The findings mentioned above suggested that miR-362-5p might be a therapy target of GDM.
Collapse
Affiliation(s)
- Cuihua Zhang
- First Department of Obstetrics, Chongqing Maternal and Child Health Hospital, Chongqing, China
| | - Dan Zhao
- First Department of Obstetrics, Chongqing Maternal and Child Health Hospital, Chongqing, China
| |
Collapse
|
16
|
Li Y, Yuan X, Shi Z, Wang H, Ren D, Zhang Y, Fan Y, Liu Y, Cui Z. LncRNA XIST serves as a diagnostic biomarker in gestational diabetes mellitus and its regulatory effect on trophoblast cell via miR-497-5p/FOXO1 axis. Cardiovasc Diagn Ther 2021; 11:716-725. [PMID: 34295698 DOI: 10.21037/cdt-21-110] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/05/2021] [Indexed: 12/22/2022]
Abstract
Background Gestational diabetes mellitus (GDM) is increasingly common in pregnancy. This study's purpose was to identify the expression of XIST and manifest the potential mechanism of XIST in GDM. Methods Ninety-three patients with GDM and 93 normal pregnant women were included in this investigation. qRT-PCR was conducted to evaluate the expression of miR-497-5p and XIST and the relationship between XIST and fasting blood glucose (FBG) was explored by Pearson assay. The clinical diagnosis of XIST on GDM patients was validated by the receiver operator characteristic (ROC) curve. Cell counting kit-8 (CCK-8) was applied to elucidate cell viability. Luciferase reporter assay was performed to document the relationship among XIST, miR-497-5p, and FOXO1. Results The expression of XIST was increased in GDM patients and HTR-8/SVneo cell models caused by high glucose (HG). The expression of XIST was associated with the FBG levels and appeared to be a feasible indicator in discriminating GDM patients. The expression of miR-497-5p was prominently reduced in GDM patients and cell models. Inhibition of XIST might alleviate the adverse function of HG on cell viability via sponging miR-497-5p. FOXO1 was proved to be a downstream target gene of miR-497-5p. Conclusions Overexpression of XIST and downregulation of miR-497-5p were indicated in this publication. XIST might serve as a promising diagnostic marker for GDM patients. XIST/miR-497-5p/FOXO1 axis played a critical role in the regulation of trophoblast cells.
Collapse
Affiliation(s)
- Yanchuan Li
- Department of Obstetrics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiaohua Yuan
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Ziyun Shi
- Department of Obstetrics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Haili Wang
- Department of Obstetrics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Duomei Ren
- Department of Obstetrics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Ya Zhang
- Department of Obstetrics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yangyang Fan
- Department of Obstetrics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yanfeng Liu
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, China
| | - Zhangxia Cui
- Department of Obstetrics, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine (Xi Xian Central Hospital), Xianyang, China
| |
Collapse
|
17
|
Wang N, Song L, Sun B, Peng Y, Fei S, Cui J, Mi Y, Cui W. Contribution of gestational diabetes mellitus heterogeneity and prepregnancy body mass index to large-for-gestational-age infants-A retrospective case-control study. J Diabetes 2021; 13:307-317. [PMID: 32935481 DOI: 10.1111/1753-0407.13113] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/20/2020] [Accepted: 09/10/2020] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE To study the associations between heterogeneity of gestational diabetes mellitus (GDM) subtype/prepregnancy body mass index (pre-BMI) and large-for-gestational-age (LGA) infants of Chinese women. METHODS We performed a retrospective case-control study of 299 women with GDM and 204 women with normal glucose tolerance (NGT), using oral glucose tolerance test-based indices performed at 24-25 weeks of gestation. Women with GDM were classified into the following three physiologic subtypes: GDM with a predominant insulin-secretion defect (GDM-dysfunction), GDM with a predominant insulin-sensitivity defect (GDM-resistance), or GDM with both defects (GDM-mixed). We then used a binary logistic regression model to evaluate the potential associations of GDM subtypes and pre-BMI with newborn macrosomia or LGA. RESULTS Women with GDM-resistance had a higher pre-BMI (P < 0.001), whereas women in the GDM-dysfunction and GDM-mixed groups had pre-BMIs comparable to the NGT group. In the logistic regression model, women in the GDM-mixed group exhibited an increased risk of bearing newborns with macrosomia and LGA, and women in the GDM-dysfunction group tended to have newborns with LGA after adjusting for pre-BMI and other potential confounders. Women who were overweight or obese prepregnancy manifested an increased risk of having newborns with macrosomia and LGA relative to normal-weight women, regardless of whether values were unadjusted or adjusted for all potential confounders. There was no significant interaction between GDM subtype and pre-BMI for any of the studied outcomes. CONCLUSIONS Heterogeneity of GDM (GDM-dysfunction and GDM-mixed) and prepregnancy overweight/obesity were independently associated with LGA in Chinese women. There was no significant interaction between GDM subtypes and pre-BMI for LGA.
Collapse
Affiliation(s)
- Ning Wang
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yanqi Peng
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sijia Fei
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaqi Cui
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Mi
- The Second Department of Obstetrics, Northwest Women and Children's Hospital, Xi'an, China
| | - Wei Cui
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
18
|
VALENT AM, CHOI H, KOLAHI KS, THORNBURG KL. Hyperglycemia and gestational diabetes suppress placental glycolysis and mitochondrial function and alter lipid processing. FASEB J 2021; 35:e21423. [PMID: 33605480 PMCID: PMC8906558 DOI: 10.1096/fj.202000326rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/15/2022]
Abstract
The degree that maternal glycemia affects placental metabolism of trophoblast cell types [cytotrophoblast (CTB) and syncytiotrophoblast (SCT)] in pregnant persons with gestational diabetes mellitus (GDM) is unknown. We tested the hypotheses that (a) hyperglycemia suppresses the metabolic rates of CTB and SCT; and (b) low placental metabolic activity from GDM placentas is due to decreased oxygen consumption of CTB. Trophoblast cells isolated from GDM and non-GDM term placentas were cultured for 8-hour (CTB) and following syncytialization at 72-hour (SCT) in 5 mM of glucose or 25 mM of glucose. Oxygen consumption rates, glycolysis, ATP levels, and lipid droplet morphometries were determined in CTB and SCT. In CTB from GDM placentas compared to control CTB: (a) oxidative phosphorylation was decreased by 44% (41.8 vs 74.2 pmol O2 /min/100 ng DNA, P = .002); (b) ATP content was 39% lower (1.1 × 10-7 vs 1.8 × 10-7 nM/ng DNA, P = .046); and (c) lipid droplets were two times larger (31.0 vs 14.4 µm2 /cell, P < .001) and 1.7 times more numerous (13.5 vs 7.9 lipid droplets/cell, P < .001). Hyperglycemia suppressed CTB glycolysis by 55%-60% (mean difference 20.4 [GDM, P = .008] and 15.4 [non-GDM, P = .029] mpH/min/100 ng DNA). GDM SCT was not metabolically different from non-GDM SCT. However, GDM SCT had significantly decreased expression of genes associated with differentiation including hCG, GCM1, and syncytin-1. We conclude that suppressed metabolic activity by the GDM placenta is attributable to metabolic dysfunction of CTB, not SCT. Critical placental hormone expression and secretion are decreased in GDM trophoblasts.
Collapse
Affiliation(s)
- Amy M. VALENT
- Oregon Health and Science University, Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Portland, OR 97239 USA
| | - Haeri CHOI
- Oregon Health and Science University, Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Portland, OR 97239 USA,Oregon Health and Science University, Knight Cardiovascular Institute, Center for Developmental Health, Portland, OR 97239 USA
| | - Kevin S. KOLAHI
- Oregon Health and Science University, School of Medicine, Portland, OR 97239 USA
| | - Kent L. THORNBURG
- Oregon Health and Science University, Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Portland, OR 97239 USA,Oregon Health and Science University, School of Medicine, Portland, OR 97239 USA,Oregon Health and Science University, Knight Cardiovascular Institute, Center for Developmental Health, Portland, OR 97239 USA
| |
Collapse
|
19
|
Abascal-Saiz A, Fuente-Luelmo E, Haro M, de la Calle M, Ramos-Álvarez MP, Perdomo G, Bartha JL. Placental Compartmentalization of Lipid Metabolism: Implications for Singleton and Twin Pregnancies. Reprod Sci 2020; 28:1150-1160. [PMID: 33171514 DOI: 10.1007/s43032-020-00385-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/01/2020] [Indexed: 11/27/2022]
Abstract
The study of placental lipid metabolism in uncomplicated pregnancies has not been developed in the literature to date. Its importance lies in expanding the knowledge of placental function to enable comparison with pathological pregnancies in future research. The aim of the present study was to compare the lipid metabolic activity and storage of the maternal and fetal sides of the placenta in healthy pregnancies. Moreover, we compare singleton vs. twin pregnancies to determine if placental metabolic needs differ. We analyzed placental explants from uncomplicated pregnancies, 20 from singleton and 8 from bichorial-biamniotic twin pregnancies (n = 28). Six cotyledon fragments were collected from each placenta at different distances from the umbilical cord, three close to the chorionic plate (hereinafter, we will refer to them as "fetal side") and another three close to the anchoring villi into the decidua basalis (referred to as "maternal side"). The samples were analyzed for quantitative assay placental fatty acid oxidation (FAO) and esterification (FAE) activities and triglyceride levels. The location of lipid storage in the chorionic villi was assessed by Oil red-O staining. Placental fatty acid oxidation did not show differences when comparing the maternal and fetal sides of the placenta or between single and twin pregnancies. When comparing placental sides, FAE was increased twofold in the maternal side compared to the fetal side of the placenta (P = 0.013). The tendency for lipogenesis in the placenta was exemplified by the FAE/FAO ratio, which was a 37.1% higher on the maternal side (P = 0.019). Despite this, triglyceride levels were five times higher in the fetal side than in the maternal one (P = 0.024). When analyzing singleton vs. twins, FAE was superior in the fetal side in multiple pregnancies (× 2.6, P = 0.007) and the FAE/FAO ratio was significantly higher in twins than in singleton pregnancies, on both sides of the placenta. Despite this finding, triglyceride levels were similar in twin and singleton pregnancies. Comparing the placentas of twins in the same pregnancy, there were no differences in lipid metabolism (FAO or FAE) or placental triglyceride levels between the two co-twins. Using Oil red-O staining, lipid storage in chorionic villi was found to be located on the syncytiotrophoblast cells and not in the connecting axis. The maternal side of the placenta is more active in the esterification of fatty acids, while the storage of neutral lipids concentrates on the fetal side. Moreover, multiple gestations have increased esterification without changes in the concentration of placental triglycerides, probably due to a higher transfer to the fetal circulation in response to the greater energy demand from twin fetuses.
Collapse
Affiliation(s)
- Alejandra Abascal-Saiz
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Eva Fuente-Luelmo
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María Haro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María de la Calle
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - María P Ramos-Álvarez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - Germán Perdomo
- Department of Health Sciences, University of Burgos, Burgos, Spain.,Institute of Molecular Biology and Genetic (IMBG), CSIC - University of Valladolid, Valladolid, Spain
| | - José L Bartha
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
20
|
Steinhauser CB, Askelson K, Lambo CA, Hobbs KC, Bazer FW, Satterfield MC. Lipid metabolism is altered in maternal, placental, and fetal tissues of ewes with small for gestational age fetuses†. Biol Reprod 2020; 104:170-180. [PMID: 33001151 DOI: 10.1093/biolre/ioaa180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/23/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Nutrient restriction (NR) has the potential to negatively impact birthweight, an indicator of neonatal survival and lifelong health. Those fetuses are termed as small for gestational age (SGA). Interestingly, there is a spectral phenotype of fetal growth rates in response to NR associated with changes in placental development, nutrient and waste transport, and lipid metabolism. A sheep model with a maternal diet, starting at Day 35, of 100% National Research Council (NRC) nutrient requirements (n = 8) or 50% NRC (n = 28) was used to assess alterations in fetuses designated NR SGA (n = 7) or NR NonSGA (n = 7) based on fetal weight at Day 135 of pregnancy. Allantoic fluid concentrations of triglycerides were greater in NR SGA fetuses than 100% NRC and NR NonSGA fetuses at Day 70 (P < 0.05). There was a negative correlation between allantoic fluid concentrations of triglycerides (R2 = 0.207) and bile acids (R2 = 0.179) on Day 70 and fetal weight at Day 135 for NR ewes (P < 0.05). Bile acids were more abundant in maternal and fetal blood for NR SGA compared to 100% NRC and NR NonSGA ewes (P < 0.05). Maternal blood concentrations of NEFAs increased in late pregnancy in NR NonSGA compared to NR SGA ewes (P < 0.05). Protein expression of fatty acid transporter SLC27A6 localized to placentomal maternal and fetal epithelia and decreased in Day 70 NR SGA compared to 100% NRC and NR NonSGA placentomes (P < 0.05). These results identify novel factors associated with an ability of placentae and fetuses in NR NonSGA ewes to adapt to, and overcome, nutritional hardship during pregnancy.
Collapse
Affiliation(s)
| | - Katharine Askelson
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Colleen A Lambo
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Kenneth C Hobbs
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - M Carey Satterfield
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
21
|
Mitochondrial dysfunction in the fetoplacental unit in gestational diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165948. [PMID: 32866635 DOI: 10.1016/j.bbadis.2020.165948] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Gestational diabetes mellitus (GDM) is a disease of pregnancy that is associated with d-glucose intolerance and foeto-placental vascular dysfunction. GMD causes mitochondrial dysfunction in the placental endothelium and trophoblast. Additionally, GDM is associated with reduced placental oxidative phosphorylation due to diminished activity of the mitochondrial F0F1-ATP synthase (complex V). This phenomenon may result from a higher generation of reactive superoxide anion and nitric oxide. Placental mitochondrial biogenesis and mitophagy work in concert to maintain cell homeostasis and are vital mechanisms securing the efficient generation of ATP, whose demand is higher in pregnancy, ensuring foetal growth and development. Additional factors disturbing placental ATP synthase activity in GDM include pre-gestational maternal obesity or overweight, intracellular pH, miRNAs, fatty acid oxidation, and foetal (and 'placental') sex. GDM is also associated with maternal and foetal hyperinsulinaemia, altered circulating levels of adiponectin and leptin, and the accumulation of extracellular adenosine. Here, we reviewed the potential interplay between these molecules or metabolic conditions on the mechanisms of mitochondrial dysfunction in the foeto-placental unit in GDM pregnancies.
Collapse
|
22
|
Placental function in maternal obesity. Clin Sci (Lond) 2020; 134:961-984. [PMID: 32313958 DOI: 10.1042/cs20190266] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Maternal obesity is associated with pregnancy complications and increases the risk for the infant to develop obesity, diabetes and cardiovascular disease later in life. However, the mechanisms linking the maternal obesogenic environment to adverse short- and long-term outcomes remain poorly understood. As compared with pregnant women with normal BMI, women entering pregnancy obese have more pronounced insulin resistance, higher circulating plasma insulin, leptin, IGF-1, lipids and possibly proinflammatory cytokines and lower plasma adiponectin. Importantly, the changes in maternal levels of nutrients, growth factors and hormones in maternal obesity modulate placental function. For example, high insulin, leptin, IGF-1 and low adiponectin in obese pregnant women activate mTOR signaling in the placenta, promoting protein synthesis, mitochondrial function and nutrient transport. These changes are believed to increase fetal nutrient supply and contribute to fetal overgrowth and/or adiposity in offspring, which increases the risk to develop disease later in life. However, the majority of obese women give birth to normal weight infants and these pregnancies are also associated with activation of inflammatory signaling pathways, oxidative stress, decreased oxidative phosphorylation and lipid accumulation in the placenta. Recent bioinformatics approaches have expanded our understanding of how maternal obesity affects the placenta; however, the link between changes in placental function and adverse outcomes in obese women giving birth to normal sized infants is unclear. Interventions that specifically target placental function, such as activation of placental adiponectin receptors, may prevent the transmission of metabolic disease from obese women to the next generation.
Collapse
|
23
|
Physical Activity During Pregnancy Is Associated with Increased Placental FATP4 Protein Expression. Reprod Sci 2020; 27:1909-1919. [PMID: 32519158 DOI: 10.1007/s43032-020-00210-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022]
Abstract
Placental function is of utmost importance to ensure proper fetal development in utero. Among the placenta's many roles includes the passage of sufficient macronutrients, such as glucose, amino acids, and fatty acids, to the fetus. Macronutrients are carried from maternal circulation to the fetus across transporters within the placenta. The objective of this study was to examine the impact of (i) an acute bout of exercise and (ii) chronic exercise participation on placenta nutrient transporter expression and localization. To investigate the effect of acute exercise, pre- and post-exercise serum was collected from pregnant (n = 5) and non-pregnant (n = 5) women who underwent a moderate-intensity exercise session and used to treat BeWo cells. To assess chronic physical activity, we analyzed term placenta from women categorized as active (n = 10) versus non-active (n = 10). Protein expression and localization for the transporters GLUT1, SNAT1, and FATP4 were examined for both groups. GLUT1 expression in BeWo cells treated with serum from pregnant women was higher compared with that from non-pregnant, independent of exercise. FATP4 protein expression was elevated in the term placenta of active women. Immunohistochemistry analysis of term placenta illustrated increased staining of FATP4 in placental tissue from active women and differential staining pattern of GLUT1 depending on physical activity status. Chronic exercise during pregnancy increases the expression of placental FATP4 in vivo, suggesting greater metabolism and usage of fatty acids. Additionally, serum from pregnant women could contain factors that increase GLUT1 protein expression in vitro. BeWo cells treated with pre- and post-exercise serum from pregnant women resulted in greater GLUT1 expression compared with those treated with pre- and post-exercise serum from non-pregnant women. Physical activity appears to differentially impact key placental transporters involved in the transfer and availability of nutrients from mother to fetus. Future research ought to examine the mechanisms involved in regulating these changes and their impact on fetal growth and health.
Collapse
|
24
|
Jadhav A, Khaire A, Joshi S. Exploring the role of oxidative stress, fatty acids and neurotrophins in gestational diabetes mellitus. Growth Factors 2020; 38:226-234. [PMID: 33703982 DOI: 10.1080/08977194.2021.1895143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Gestational diabetes mellitus (GDM) constitutes an unfavorable intrauterine environment for embryonic and feto-placental development. Women with GDM are at higher risk for materno-fetal complications and placental abnormalities. The placenta acts as an interface between the maternal and fetal circulations and also plays an important role in protecting the fetus from adverse effects of maternal metabolic conditions. One of the earliest abnormalities observed in GDM pregnancies is increased oxidative stress in the placenta which affects fetal development. Imbalances in maternal nutrition particularly long-chain polyunsaturated fatty acid (LCPUFA) intake and/or metabolism lead to increased oxidative stress. Reports indicate that oxidative stress and LCPUFA such as docosahexaenoic acid affect the levels of neurotrophins. The present review aims to provide insights into a mechanistic link between oxidative stress, LCPUFA and neurotrophin in the placenta in women with GDM and its implications for neurodevelopmental outcomes in children.
Collapse
Affiliation(s)
- Anjali Jadhav
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Amrita Khaire
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| |
Collapse
|