1
|
Amarsi R, Furse S, Cleaton MAM, Maurel S, Mitchell AL, Ferguson-Smith AC, Cenac N, Williamson C, Koulman A, Charalambous M. A co-ordinated transcriptional programme in the maternal liver supplies long chain polyunsaturated fatty acids to the conceptus using phospholipids. Nat Commun 2024; 15:6767. [PMID: 39117683 PMCID: PMC11310303 DOI: 10.1038/s41467-024-51089-z] [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: 07/11/2023] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
The long and very long chain polyunsaturated fatty acids (LC-PUFAs) are preferentially transported by the mother to the fetus. Failure to supply LC-PUFAs is strongly linked with stillbirth, fetal growth restriction, and impaired neurodevelopmental outcomes. However, dietary supplementation during pregnancy is unable to simply reverse these outcomes, suggesting imperfectly understood interactions between dietary fatty acid intake and the molecular mechanisms of maternal supply. Here we employ a comprehensive approach combining untargeted and targeted lipidomics with transcriptional profiling of maternal and fetal tissues in mouse pregnancy. Comparison of wild-type mice with genetic models of impaired lipid metabolism allows us to describe maternal hepatic adaptations required to provide LC-PUFAs to the developing fetus. A late pregnancy-specific, selective activation of the Liver X Receptor signalling pathway dramatically increases maternal supply of LC-PUFAs within circulating phospholipids. Crucially, genetic ablation of this pathway in the mother reduces LC-PUFA accumulation by the fetus, specifically of docosahexaenoic acid (DHA), a critical nutrient for brain development.
Collapse
Affiliation(s)
- Risha Amarsi
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, SE19RT, UK
- Pregnancy Physiology Laboratory, Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK
| | - Samuel Furse
- Biological chemistry group, Jodrell laboratory, Royal Botanic Gardens Kew, Kew Road, Richmond, Surrey, TW9 3DS, UK
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Mary A M Cleaton
- Department of Genetics, Downing Street, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Sarah Maurel
- IRSD, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Alice L Mitchell
- Department of Women and Children's Health, King's College London, Guy's Campus, London, UK
| | - Anne C Ferguson-Smith
- Department of Genetics, Downing Street, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Nicolas Cenac
- IRSD, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Catherine Williamson
- Department of Women and Children's Health, King's College London, Guy's Campus, London, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Marika Charalambous
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, SE19RT, UK.
- Pregnancy Physiology Laboratory, Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK.
| |
Collapse
|
2
|
Arcot A, Walker RE, Gallagher K, Klatt KC, Gernand AD. Maternal Vitamin B12 in Pregnancy and Placental Development. J Nutr Metab 2024; 2024:3439995. [PMID: 39148633 PMCID: PMC11326881 DOI: 10.1155/2024/3439995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Accepted: 06/28/2024] [Indexed: 08/17/2024] Open
Abstract
Vitamin B12, or cobalamin, is an essential nutrient required for diverse physiological functions secondary to its role as a critical cofactor for two mammalian enzymes, methionine synthase and methylmalonyl-CoA mutase. While essential throughout all life stages, several pathways that require vitamin B12, including hematopoiesis, myelination, and DNA/histone methylation, are particularly critical during pregnancy and fetal development. This narrative review aims to describe vitamin B12 in pregnancy, with emphasis on the placenta's role in ensuring adequate nutrition of the fetus and impacts of vitamin B12 deficiency on placental development and function. Our literature search included preclinical model systems and human cohorts and interventions. Our review identified evidence of B12 deficiency resulting in impaired placental development, greater placental inflammation, and modulation of placental docosahexaenoic acid concentration, collectively suggestive of vitamin B12 deficiency as a determinant of both maternal and fetal health outcomes. Heterogeneity in study design complicated generalization of findings. Future studies should consider selecting a B12 marker that is relatively stable across pregnancy, such as holotranscobalamin, while accounting for important confounders such as maternal folate.
Collapse
Affiliation(s)
- Amrita Arcot
- Department of Nutritional Sciences The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Rachel E Walker
- Department of Nutritional Sciences The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Kelly Gallagher
- Ross and Carol Nese College of Nursing The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Kevin C Klatt
- Department of Nutritional Sciences and Toxicology University of California, Berkeley, California, USA
| | - Alison D Gernand
- Department of Nutritional Sciences The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
3
|
Barzen C, Vogel M, Kiess W, Poulain T. Associations between gestational weight gain and weight development of the offspring: Differences depending on maternal pre-pregnancy BMI. Arch Gynecol Obstet 2024; 310:395-403. [PMID: 38609672 PMCID: PMC11169002 DOI: 10.1007/s00404-024-07487-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/17/2024] [Indexed: 04/14/2024]
Abstract
PURPOSE Obesity rates are rising, and the gestational weight gain (GWG) of most women does not comply with current guidelines. This study assesses the association of pre-pregnancy BMI (ppBMI) and GWG with the child's weight development and investigates whether associations with GWG differ depending on ppBMI. METHODS Data were obtained from the cohort study LIFE Child (Germany), comprising 691 mother-child pairs. Children's weight was followed until age five. Associations between maternal ppBMI, GWG, and children's weight were evaluated using regression analyses. RESULTS The association between GWG and birth weight (BW) was significantly positive in normal and underweight (n/u) women (βGWG = 0.05, p < 0.01, 95% confidence interval (CI) 0.03-0.07), but not in women with overweight or obesity (o/o) (βGWG = 0.0002, p = 0.99, 95% CI -0.03 to 0.03). The risk of giving birth to an infant who was large for gestational age (LGA) increased with rising GWG in n/u women (OR = 1.6, p < 0.01, 95% CI 1.23-2.25). Women with o/o were at increased risk for a LGA baby regardless of GWG (OR = 3, p < 0.01, 95% CI 1.34-6.97). This trend persisted in the child's weight development during the first 5 years of life. CONCLUSION Women with o/o might increase their offspring's risk for higher weight at birth and in early childhood. In n/u women, GWG might be the more influential factor. Women should strive for normal weight before conception and should be more attentive to GWG.
Collapse
Affiliation(s)
- Charlotte Barzen
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Philipp-Rosenthal-Strasse 27, 04103, Leipzig, Germany.
| | - Mandy Vogel
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Philipp-Rosenthal-Strasse 27, 04103, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents and Center for Paediatric Research (CPL), Leipzig University, Liebigstraße 20a, Haus 6, 04103, Leipzig, Germany
| | - Wieland Kiess
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Philipp-Rosenthal-Strasse 27, 04103, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents and Center for Paediatric Research (CPL), Leipzig University, Liebigstraße 20a, Haus 6, 04103, Leipzig, Germany
| | - Tanja Poulain
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Philipp-Rosenthal-Strasse 27, 04103, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents and Center for Paediatric Research (CPL), Leipzig University, Liebigstraße 20a, Haus 6, 04103, Leipzig, Germany
| |
Collapse
|
4
|
Du H, Li D, Molive LM, Wu N. Advances in free fatty acid profiles in gestational diabetes mellitus. J Transl Med 2024; 22:180. [PMID: 38374136 PMCID: PMC10875910 DOI: 10.1186/s12967-024-04922-4] [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: 11/26/2023] [Accepted: 01/21/2024] [Indexed: 02/21/2024] Open
Abstract
The morbidity of gestational diabetes mellitus (GDM) is increasing and is associated with adverse perinatal outcomes and long-term maternal and infant health. The exact mechanism underlying changes in plasma free fatty acid (FFA) profiles in patients with GDM is unknown. However, it is believed that changes in diet and lipid metabolism may play a role. Fatty acids contain many specific FFAs, and the type of FFA has different impacts on physiological processes; hence, determining changes in FFAs in individual plasma is essential. Alterations in FFA concentration or profile may facilitate insulin resistance. Additionally, some FFAs show potential to predict GDM in early pregnancy and are strongly associated with the growth and development of the fetus and occurrence of macrosomia. Here, we aimed to review changes in FFAs in women with GDM and discuss the relationship of FFAs with GDM incidence and adverse outcomes.
Collapse
Affiliation(s)
- Haoyi Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Danyang Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Laura Monjowa Molive
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Na Wu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
- Medical Department, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, People's Republic of China.
| |
Collapse
|
5
|
Romero-Córdoba S, Chirinos M, Noyola-Martínez N, Torres-Ramírez N, García-Olivares M, Aragón-Hernández JP, Ramírez-Camacho I, Zúñiga R, Larrea F, Halhali A, Barrera D. Transcriptional landscape of human trophoblast cells treated with calcitriol and TGF-β1. Mol Cell Endocrinol 2024; 579:112088. [PMID: 37832930 DOI: 10.1016/j.mce.2023.112088] [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: 05/17/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Calcitriol and transforming growth factor beta 1 (TGF-β1) are unrelated molecules that regulate biological processes according to the genetic target, cell type, and context. Several studies have shown independent effects of calcitriol and TGF-βs on the placenta, but there is no information regarding the impact of their combination on these cells. Therefore, this study analyzed the effects of calcitriol, TGF-β1, and their combination in primary cultures of human trophoblast cells using a whole genome expression microarray. Data analysis revealed a set of differentially expressed genes induced by each treatment. Enrichment pathway analysis identified modulatory effects of calcitriol on genes related to metabolic processes such as vitamin D, steroid, and fat-soluble vitamins as well as antimicrobial and immune responses. In relation to TGF-β1, the analysis showed a few differentially expressed genes that were mainly associated with the neutrophil immune response. Lastly, the analysis revealed that the combination of calcitriol and TGF-β1 up-regulated genes involving both immunologic processes and the biosynthesis of unsaturated fatty acids, eicosanoids, and lipoxins, among others. In contrast, pathways down-regulated by the combination were mostly associated with the catabolic process of acylglycerols and peptides, PPAR signaling pathway, cellular response to low-density lipoprotein stimulus, renin angiotensin system and digestion, mobilization and transport of lipids. Consistent with these results, the combined treatment on human trophoblast cells induced the accumulation of intracellular neutral lipid droplets and stimulated both gene and protein expression of 15-hydroxyprostaglandin dehydrogenase. In conclusion, the results revealed that differentially expressed genes induced by the combination modified the transcriptional landscape compared to each treatment alone, mainly altering the storage, activity and metabolism of lipids, which might have an impact on placental development.
Collapse
Affiliation(s)
- Sandra Romero-Córdoba
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico; Departamento de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Mayel Chirinos
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico
| | - Nancy Noyola-Martínez
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico
| | - Nayeli Torres-Ramírez
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Avenida Universidad 3000, México, 04510, Mexico
| | - Mitzi García-Olivares
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico
| | - Juan Pablo Aragón-Hernández
- Departamento de la Unidad Tocoquirúrgica, Hospital General "Dr. Manuel Gea González", Ciudad de México, 14080, Mexico
| | - Ixchel Ramírez-Camacho
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico
| | - Rosa Zúñiga
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico
| | - Fernando Larrea
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico
| | - Ali Halhali
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico
| | - David Barrera
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Ciudad de México, 14080, Mexico.
| |
Collapse
|
6
|
Song Y, Lu R, Yu G, Rahman ML, Chen L, Zhu Y, Tsai MY, Fiehn O, Chen Z, Zhang C. Longitudinal lipidomic profiles during pregnancy and associations with neonatal anthropometry: findings from a multiracial cohort. EBioMedicine 2023; 98:104881. [PMID: 38006745 PMCID: PMC10709105 DOI: 10.1016/j.ebiom.2023.104881] [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: 06/04/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Maternal lipidomic profiling offers promise for characterizing lipid metabolites during pregnancy, but longitudinal data are limited. This study aimed to examine associations of longitudinal lipidomic profiles during pregnancy with multiple neonatal anthropometry using data from a multiracial cohort. METHODS We measured untargeted plasma lipidome profiles among 321 pregnant women from the NICHD Fetal Growth Study-Singletons using plasma samples collected longitudinally during four study visits at gestational weeks (GW) 10-14, 15-26, 23-31, and 33-39, respectively. We evaluated individual lipidomic metabolites at each study visit in association with neonatal anthropometry. We also evaluated the associations longitudinally by constructing lipid networks using weighted correlation network analysis and common networks using consensus network analysis across four visits using linear mixed-effects models with the adjustment of false discover rate. FINDINGS Multiple triglycerides (TG) were positively associated with birth weight (BW), BW Z-score, length and head circumference, while some cholesteryl ester (CE), phosphatidylcholine (PC), sphingomyelines (SM), phosphatidylethanolamines (PE), and lysophosphatidylcholines (LPC 20:3) families were inversely associated with BW, length, abdominal and head circumference at different GWs. Longitudinal trajectories of TG, PC, and glucosylcermides (GlcCer) were associated with BW, and CE (18:2) with BW z-score, length, and sum of skinfolds (SS), while some PC and PE were significantly associated with abdominal and head circumference. Modules of TG at GW 10-14 and 15-26 mainly were associated with BW. At GW 33-39, two networks of LPC (20:3) and of PC, TG, and CE, showed inverse associations with abdominal circumference. Distinct trajectories within two consensus modules with changes in TG, CE, PC, and LPC showed significant differences in BW and length. INTERPRETATION The results demonstrated that longitudinal changes of TGs during early- and mid-pregnancy and changes of PC, LPC, and CE during late-pregnancy were significantly associated with neonatal anthropometry. FUNDING National Institute of Child Health and Human Development intramural funding.
Collapse
Affiliation(s)
- Yiqing Song
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Ruijin Lu
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Guoqi Yu
- Global Center for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mohammad L Rahman
- National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Liwei Chen
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Yeiyi Zhu
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, 451 Health Sciences Drive, Davis, CA, USA
| | - Zhen Chen
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Cuilin Zhang
- Global Center for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
7
|
Guo T, Yang Y, Jia J, Deng Y, Wang Y, Zhang Y, Zhang H, He Y, Zhao J, Peng Z, Wang Q, Shen H, Zhang Y, Yan D, Ma X. Preconception paternal/maternal BMI and risk of small/large for gestational age infant in over 4·7 million Chinese women aged 20-49 years: a population-based cohort study in China. Br J Nutr 2023; 129:1645-1655. [PMID: 35184774 DOI: 10.1017/s000711452200054x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Evidence of couples' BMI and its influence on birth weight is limited and contradictory. Therefore, this study aims to assess the association between couple's preconception BMI and the risk of small for gestational age (SGA)/large for gestational age (LGA) infant, among over 4·7 million couples in a retrospective cohort study based on the National Free Pre-pregnancy Checkups Project between 1 December 2013 and 30 November 2016 in China. Among the live births, 256 718 (5·44 %) SGA events and 506 495 (10·73 %) LGA events were documented, respectively. After adjusting for confounders, underweight men had significantly higher risk (OR 1·17 (95 % CI 1·15, 1·19)) of SGA infants compared with men with normal BMI, while a significant and increased risk of LGA infants was obtained for overweight and obese men (OR 1·08 (95 % CI 1·06, 1·09); OR 1·19 (95 % CI 1·17, 1·20)), respectively. The restricted cubic spline result revealed a non-linear decreasing dose-response relationship of paternal BMI (less than 22·64) with SGA. Meanwhile, a non-linear increasing dose-response relationship of paternal BMI (more than 22·92) with LGA infants was observed. Moreover, similar results about the association between maternal preconception BMI and SGA/LGA infants were obtained. Abnormal preconception BMI in either women or men were associated with increased risk of SGA/LGA infants, respectively. Overall, couple's abnormal weight before pregnancy may be an important preventable risk factor for SGA/LGA infants.
Collapse
Affiliation(s)
- Tonglei Guo
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
| | - Ying Yang
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- Graduate School of Peking Union Medical College, Building 18, No. 9, Dongdan Santiao, Dongcheng District, 100730Beijing, People's Republic of China
| | - Jiajing Jia
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- Graduate School of Peking Union Medical College, Building 18, No. 9, Dongdan Santiao, Dongcheng District, 100730Beijing, People's Republic of China
| | - Yuzhi Deng
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- Graduate School of Peking Union Medical College, Building 18, No. 9, Dongdan Santiao, Dongcheng District, 100730Beijing, People's Republic of China
| | - Yuanyuan Wang
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
| | - Ya Zhang
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
| | - Hongguang Zhang
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
| | - Yuan He
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- Graduate School of Peking Union Medical College, Building 18, No. 9, Dongdan Santiao, Dongcheng District, 100730Beijing, People's Republic of China
| | - Jun Zhao
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
| | - Zuoqi Peng
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
| | - Qiaomei Wang
- Department of Maternal and Child Health, National Health Commission of the PRC, No. 1, Xizhimenwai South Road, Xicheng District, 100044Beijing, People's Republic of China
| | - Haiping Shen
- Department of Maternal and Child Health, National Health Commission of the PRC, No. 1, Xizhimenwai South Road, Xicheng District, 100044Beijing, People's Republic of China
| | - Yiping Zhang
- Department of Maternal and Child Health, National Health Commission of the PRC, No. 1, Xizhimenwai South Road, Xicheng District, 100044Beijing, People's Republic of China
| | - Donghai Yan
- Department of Maternal and Child Health, National Health Commission of the PRC, No. 1, Xizhimenwai South Road, Xicheng District, 100044Beijing, People's Republic of China
| | - Xu Ma
- National Research Institute for Family Planning, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- National Human Genetic Resource Center, No. 12, Dahuisi Road, Haidian District, 100081Beijing, People's Republic of China
- Graduate School of Peking Union Medical College, Building 18, No. 9, Dongdan Santiao, Dongcheng District, 100730Beijing, People's Republic of China
| |
Collapse
|
8
|
Berger N, van der Wel T, Hirschmugl B, Baernthaler T, Gindlhuber J, Fawzy N, Eichmann T, Birner-Gruenberger R, Zimmermann R, van der Stelt M, Wadsack C. Inhibition of diacylglycerol lipase β modulates lipid and endocannabinoid levels in the ex vivo human placenta. Front Endocrinol (Lausanne) 2023; 14:1092024. [PMID: 36864832 PMCID: PMC9971001 DOI: 10.3389/fendo.2023.1092024] [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: 11/07/2022] [Accepted: 01/27/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Lipids and fatty acids are key components in metabolic processes of the human placenta, thereby contributing to the development of the fetus. Placental dyslipidemia and aberrant activity of lipases have been linked to diverse pregnancy associated complications, such as preeclampsia and preterm birth. The serine hydrolases, diacylglycerol lipase α and β (DAGLα, DAGLβ) catalyze the degradation of diacylglycerols, leading to the formation of monoacylglycerols (MAG), including one main endocannabinoid 2-arachidonoylglycerol (2-AG). The major role of DAGL in the biosynthesis of 2-AG is evident from various studies in mice but has not been investigated in the human placenta. Here, we report the use of the small molecule inhibitor DH376, in combination with the ex vivo placental perfusion system, activity-based protein profiling (ABPP) and lipidomics, to determine the impact of acute DAGL inhibition on placental lipid networks. Methods DAGLα and DAGLβ mRNA expression was detected by RT-qPCR and in situ hybridization in term placentas. Immunohistochemistry staining for CK7, CD163 and VWF was applied to localize DAGLβ transcripts to different cell types of the placenta. DAGLβ activity was determined by in- gel and MS-based activity-based protein profiling (ABPP) and validated by addition of the enzyme inhibitors LEI-105 and DH376. Enzyme kinetics were measured by EnzChek™ lipase substrate assay. Ex vivo placental perfusion experiments were performed +/- DH376 [1 µM] and changes in tissue lipid and fatty acid profiles were measured by LC-MS. Additionally, free fatty acid levels of the maternal and fetal circulations were determined. Results We demonstrate that mRNA expression of DAGLβ prevails in placental tissue, compared to DAGLα (p ≤ 0.0001) and that DAGLβ is mainly located to CK7 positive trophoblasts (p ≤ 0.0001). Although few DAGLα transcripts were identified, no active enzyme was detected applying in-gel or MS-based ABPP, which underlined that DAGLβ is the principal DAGL in the placenta. DAGLβ dependent substrate hydrolysis in placental membrane lysates was determined by the application of LEI-105 and DH376. Ex vivo pharmacological inhibition of DAGLβ by DH376 led to reduced MAG tissue levels (p ≤ 0.01), including 2-AG (p≤0.0001). We further provide an activity landscape of serine hydrolases, showing a broad spectrum of metabolically active enzymes in the human placenta. Discussion Our results emphasize the role of DAGLβ activity in the human placenta by determining the biosynthesis of 2-AG. Thus, this study highlights the special importance of intra-cellular lipases in lipid network regulation. Together, the activity of these specific enzymes may contribute to the lipid signaling at the maternal-fetal interface, with implications for function of the placenta in normal and compromised pregnancies.
Collapse
Affiliation(s)
- Natascha Berger
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Leiden, Netherlands
| | - Birgit Hirschmugl
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Thomas Baernthaler
- Otto Loewi Research Center, Division of Pharmacology, University of Graz, Graz, Austria
| | - Juergen Gindlhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Diagnostic and Research Center of Molecular Medicine, Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Nermeen Fawzy
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Thomas Eichmann
- BioTechMed-Graz, Graz, Austria
- Core Facility Mass Spectrometry, Center for Medical Research (ZMF), Medical University of Graz, Graz, Austria
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Center of Molecular Medicine, Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
| | - Robert Zimmermann
- BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Leiden, Netherlands
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
9
|
The ANGPTL3-4-8 Axis in Normal Gestation and in Gestational Diabetes, and Its Potential Involvement in Fetal Growth. Int J Mol Sci 2023; 24:ijms24032486. [PMID: 36768809 PMCID: PMC9917010 DOI: 10.3390/ijms24032486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Dyslipidemia in gestational diabetes has been associated with worse perinatal outcomes. The ANGPTL3-4-8 axis regulates lipid metabolism, especially in the transition from fasting to feeding. In this study, we evaluated the response of ANGPTL3, 4, and 8 after the intake of a mixed meal in women with normal glucose tolerance and gestational diabetes, and we assessed their gene expressions in different placental locations. Regarding the circulating levels of ANGPTL3, 4, and 8, we observed an absence of ANGPTL4 response after the intake of the meal in the GDM group compared to its presence in the control group. At the placental level, we observed a glucose tolerance-dependent expression pattern of ANGPTL3 between the two placental sides. When we compared the GDM pregnancies with the control pregnancies, a downregulation of the maternal side ANGPTL3 expression was observed. This suggests a dysregulation of the ANGPTL3-4-8 axis in GDM, both at the circulating level after ingestion and at the level of placental expression. Furthermore, we discerned that the expressions of ANGPTL3, 4, and 8 were related to birth weight and placental weight in the GDM group, but not in the control group, which suggests that they may play a role in regulating the transplacental passage of nutrients.
Collapse
|
10
|
Berger N, Allerkamp H, Wadsack C. Serine Hydrolases in Lipid Homeostasis of the Placenta-Targets for Placental Function? Int J Mol Sci 2022; 23:6851. [PMID: 35743292 PMCID: PMC9223866 DOI: 10.3390/ijms23126851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/01/2023] Open
Abstract
The metabolic state of pregnant women and their unborn children changes throughout pregnancy and adapts to the specific needs of each gestational week. These adaptions are accomplished by the actions of enzymes, which regulate the occurrence of their endogenous substrates and products in all three compartments: mother, placenta and the unborn. These enzymes determine bioactive lipid signaling, supply, and storage through the generation or degradation of lipids and fatty acids, respectively. This review focuses on the role of lipid-metabolizing serine hydrolases during normal pregnancy and in pregnancy-associated pathologies, such as preeclampsia, gestational diabetes mellitus, or preterm birth. The biochemical properties of each class of lipid hydrolases are presented, with special emphasis on their role in placental function or dysfunction. While, during a normal pregnancy, an appropriate tonus of bioactive lipids prevails, dysregulation and aberrant signaling occur in diseased states. A better understanding of the dynamics of serine hydrolases across gestation and their involvement in placental lipid homeostasis under physiological and pathophysiological conditions will help to identify new targets for placental function in the future.
Collapse
Affiliation(s)
- Natascha Berger
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (N.B.); (H.A.)
| | - Hanna Allerkamp
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (N.B.); (H.A.)
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (N.B.); (H.A.)
- BioTechMed-Graz, 8036 Graz, Austria
| |
Collapse
|
11
|
Maternal lipid profile in pregnancy and embryonic size: a population-based prospective cohort study. BMC Pregnancy Childbirth 2022; 22:333. [PMID: 35436866 PMCID: PMC9016996 DOI: 10.1186/s12884-022-04647-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background Lipids are crucial for fetal growth and development. Maternal lipid concentrations are associated with fetal growth in the second and third trimester of pregnancy and with birth outcomes. However, it is unknown if this association starts early in pregnancy or arises later during fetal development. The aim of this study was to investigate the association between the maternal lipid profile in early pregnancy and embryonic size. Methods We included 1474 women from the Generation R Study, a population based prospective birth cohort. Both embryonic size and the maternal lipid profile were measured between 10 weeks + 1 day and 13 weeks + 6 days gestational age. The maternal lipid profile was defined as total cholesterol, triglycerides (TG), high-density lipoprotein cholesterol (HDL-c), low-density lipoprotein cholesterol (LDL-c), remnant cholesterol, non-high-density (non-HDL-c) lipoprotein cholesterol concentrations and the triglycerides/high-density lipoprotein (TG/HDL-c) ratio. Additionally, maternal glucose concentrations were assessed. Embryonic size was assessed using crown-rump length (CRL) measurements. Associations were studied with linear regression models, adjusted for confounding factors: maternal age, pre-pregnancy body mass index (BMI), parity, educational level, ethnicity, smoking and folic acid supplement use. Results Triglycerides and remnant cholesterol concentrations are positively associated with embryonic size (fully adjusted models, 0.17 SDS CRL: 95% CI 0.03; 0.30, and 0.17 SDS: 95% CI 0.04; 0.31 per 1 MoM increase, respectively). These associations were not present in women with normal weight (triglycerides and remnant cholesterol: fully adjusted model, 0.44 SDS: 95% CI 0.15; 0.72). Associations between maternal lipid concentrations and embryonic size were not attenuated after adjustment for glucose concentrations. Total cholesterol, HDL-c, LDL-c, non-HDL-c concentrations and the TG/HDL-c ratio were not associated with embryonic size. Conclusions Higher triglycerides and remnant cholesterol concentrations in early pregnancy are associated with increased embryonic size, most notably in overweight women. Trial registration The study protocol has been approved by the Medical Ethics Committee of the Erasmus University Medical Centre (Erasmus MC), Rotterdam (MEC-2007-413). Written informed consent was obtained from all participants. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04647-6.
Collapse
|
12
|
Gago-Dominguez M, Redondo CM, Calaza M, Matabuena M, Bermudez MA, Perez-Fernandez R, Torres-Español M, Carracedo Á, Castelao JE. LIPG endothelial lipase and breast cancer risk by subtypes. Sci Rep 2021; 11:10436. [PMID: 34001944 PMCID: PMC8129130 DOI: 10.1038/s41598-021-89669-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 04/21/2021] [Indexed: 11/09/2022] Open
Abstract
Experimental data showed that endothelial lipase (LIPG) is a crucial player in breast cancer. However, very limited data exists on the role of LIPG on the risk of breast cancer in humans. We examined the LIPG-breast cancer association within our population-based case-control study from Galicia, Spain, BREOGAN (BREast Oncology GAlicia Network). Plasma LIPG and/or OxLDL were measured on 114 breast cancer cases and 82 controls from our case-control study, and were included in the present study. The risk of breast cancer increased with increasing levels of LIPG (multivariable OR for the highest category (95% CI) 2.52 (1.11-5.81), P-trend = 0.037). The LIPG-breast cancer association was restricted to Pre-menopausal breast cancer (Multivariable OR for the highest LIPG category (95% CI) 4.76 (0.94-28.77), P-trend = 0.06, and 1.79 (0.61-5.29), P-trend = 0.372, for Pre-menopausal and Post-menopausal breast cancer, respectively). The LIPG-breast cancer association was restricted to Luminal A breast cancers (Multivariable OR for the highest LIPG category (95% CI) 3.70 (1.42-10.16), P-trend = 0.015, and 2.05 (0.63-7.22), P-trend = 0.311, for Luminal A and non-Luminal A breast cancers, respectively). Subset analysis only based on HER2 receptor indicated that the LIPG-breast cancer relationship was restricted to HER2-negative breast cancers (Multivariable OR for the highest LIPG category (95% CI) 4.39 (1.70-12.03), P-trend = 0.012, and 1.10 (0.28-4.32), P-trend = 0.745, for HER2-negative and HER2-positive tumors, respectively). The LIPG-breast cancer association was restricted to women with high total cholesterol levels (Multivariable OR for the highest LIPG category (95% CI) 6.30 (2.13-20.05), P-trend = 0.018, and 0.65 (0.11-3.28), P-trend = 0.786, among women with high and low cholesterol levels, respectively). The LIPG-breast cancer association was also restricted to non-postpartum breast cancer (Multivariable OR for the highest LIPG category (95% CI) 3.83 (1.37-11.39), P-trend = 0.003, and 2.35 (0.16-63.65), P-trend = 0.396, for non-postpartum and postpartum breast cancer, respectively), although we lacked precision. The LIPG-breast cancer association was more pronounced among grades II and III than grade I breast cancers (Multivariable ORs for the highest category of LIPG (95% CI) 2.73 (1.02-7.69), P-trend = 0.057, and 1.90 (0.61-6.21), P-trend = 0.170, for grades II and III, and grade I breast cancers, respectively). No association was detected for OxLDL levels and breast cancer (Multivariable OR for the highest versus the lowest category (95% CI) 1.56 (0.56-4.32), P-trend = 0.457).
Collapse
Affiliation(s)
- Manuela Gago-Dominguez
- Galician Public Foundation of Genomic Medicine (FPGMX), Servicio Galego de Saúde (SERGAS), Santiago de Compostela, Spain.
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Centro en Red de Enfermedades Raras (CIBERER), University of Santiago de Compostela, Santiago de Compostela, Spain.
- Galician Public Foundation of Genomic Medicine (FPGMX), Genomic Medicine Group, International Cancer Genetics and Epidemiology Group, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.
| | - Carmen M Redondo
- Oncology and Genetics Unit, Instituto de Investigación Sanitaria Galicia Sur, Vigo, Spain
| | - Manuel Calaza
- Conselleria de Educación, Xunta de Galicia, Santiago de Compostela, Spain
| | - Marcos Matabuena
- Centro de Investigación en Tecnoloxías da Información (CiTIUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria A Bermudez
- Department of Biology, Faculty of Science, University of A Coruña, A Coruña, Spain
| | - Roman Perez-Fernandez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Torres-Español
- Galician Public Foundation of Genomic Medicine (FPGMX), Servicio Galego de Saúde (SERGAS), Santiago de Compostela, Spain
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Centro en Red de Enfermedades Raras (CIBERER), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ángel Carracedo
- Galician Public Foundation of Genomic Medicine (FPGMX), Servicio Galego de Saúde (SERGAS), Santiago de Compostela, Spain
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Centro en Red de Enfermedades Raras (CIBERER), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - J Esteban Castelao
- Oncology and Genetics Unit, Instituto de Investigación Sanitaria Galicia Sur, Vigo, Spain
| |
Collapse
|
13
|
da Costa Faria NR, Chaves-Filho AB, Alcantara LCJ, de Siqueira IC, Calcagno JI, Miyamoto S, de Filippis AMB, Yoshinaga MY. Plasma lipidome profiling of newborns with antenatal exposure to Zika virus. PLoS Negl Trop Dis 2021; 15:e0009388. [PMID: 33930014 PMCID: PMC8115770 DOI: 10.1371/journal.pntd.0009388] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/12/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023] Open
Abstract
The 2015–2016 Zika virus (ZIKV) outbreak in Brazil was remarkably linked to the incidence of microcephaly and other deleterious clinical manifestations, including eye abnormalities, in newborns. It is known that ZIKV targets the placenta, triggering an inflammatory profile that may cause placental insufficiency. Transplacental lipid transport is delicately regulated during pregnancy and deficiency on the delivery of lipids such as arachidonic and docosahexaenoic acids may lead to deficits in both brain and retina during fetal development. Here, plasma lipidome profiles of ZIKV exposed microcephalic and normocephalic newborns were compared to non-infected controls. Our results reveal major alterations in circulating lipids from both ZIKV exposed newborns with and without microcephaly relative to controls. In newborns with microcephaly, the plasma concentrations of hydroxyoctadecadienoic acid (HODE), primarily as 13-HODE isomer, derived from linoleic acid were higher as compared to normocephalic ZIKV exposed newborns and controls. Total HODE concentrations were also positively associated with levels of other oxidized lipids and several circulating free fatty acids in newborns, indicating a possible plasma lipidome signature of microcephaly. Moreover, higher concentrations of lysophosphatidylcholine in ZIKV exposed normocephalic newborns relative to controls suggest a potential disruption of polyunsaturated fatty acids transport across the blood-brain barrier of fetuses. The latter data is particularly important given the neurocognitive and neurodevelopmental abnormalities observed in follow-up studies involving children with antenatal ZIKV exposure, but normocephalic at birth. Taken together, our data reveal that plasma lipidome alterations associated with antenatal exposure to ZIKV could contribute to identification and monitoring of the wide spectrum of clinical phenotypes at birth and further, during childhood. Antenatal exposure to Zika virus (ZIKV) is linked to a wide range of clinical presentations at birth, from asymptomatic cases to microcephaly, and other neurocognitive and neurodevelopmental abnormalities manifested in the early childhood. Stratification of these clinical phenotypes in newborns with suspected antenatal ZIKV exposure is challenging, but critical to improve early assessment of rehabilitative interventions. In this study, plasma lipidome profiling of 274 lipid species was performed in both normocephalic and microcephalic newborns with antenatal ZIKV exposure and compared to non-infected controls. Multiple lipid species were independent predictors of antenatal ZIKV exposure. More specifically, microcephaly was strongly associated with an oxidized free fatty acid and ZIKV exposed normocephalic newborns exhibited higher plasma concentrations of lysophosphatidylcholine relative to controls. These findings emphasize the need for studies focused on the role of individual lipids in neuropathogenesis of ZIKV and raise the potential of plasma lipidome profiling for early diagnosis of newborns with suspected antenatal ZIKV exposure. To validate the predictive ability of this approach, prospective studies with a larger cohort of newborns are now required.
Collapse
Affiliation(s)
| | | | | | | | - Juan Ignacio Calcagno
- Maternidade Prof. José Maria de Magalhães Netto, State Health Secretary (Salvador), Bahia, Brazil
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Marcos Yukio Yoshinaga
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
14
|
Maternal proteomic profiling reveals alterations in lipid metabolism in late-onset fetal growth restriction. Sci Rep 2020; 10:21033. [PMID: 33273667 PMCID: PMC7713381 DOI: 10.1038/s41598-020-78207-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/20/2020] [Indexed: 12/18/2022] Open
Abstract
Fetal growth restriction defined as the failure to achieve the fetal genetic growth potential is a major cause of perinatal morbidity and mortality. The role of maternal adaptations to placental insufficiency in this disorder is still not fully understood. We aimed to investigate the biological processes and protein–protein interactions involved in late-onset fetal growth restriction in particular. We applied 2D nano LC–MS/MS proteomics analysis on maternal blood samples collected at the time of delivery from 5 singleton pregnancies with late-onset fetal growth restriction and 5 uncomplicated pregnancies. Data were analyzed using R package “limma” and Ingenuity Pathway Analysis. 25 proteins showed significant changes in their relative abundance in late-onset fetal growth restriction (p value < 0.05). Direct protein–protein interactions network demonstrated that Neurogenic locus notch homolog protein 1 (NOTCH1) was the most significant putative upstream regulator of the observed profile. Gene ontology analysis of these proteins revealed the involvement of 14 canonical pathways. The most significant biological processes were efflux of cholesterol, efflux of phospholipids, adhesion of blood cells, fatty acid metabolism and dyslipidemia. Future studies are warranted to validate the potential role of the detected altered proteins as potential therapeutic targets in the late-onset form of fetal growth restriction.
Collapse
|
15
|
Godhamgaonkar AA, Wadhwani NS, Joshi SR. Exploring the role of LC-PUFA metabolism in pregnancy complications. Prostaglandins Leukot Essent Fatty Acids 2020; 163:102203. [PMID: 33227645 DOI: 10.1016/j.plefa.2020.102203] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/09/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022]
Abstract
Maternal nutrition during pregnancy plays a significant role in growth and development of the placenta and influencing pregnancy outcome. Suboptimal nutritional status during early gestational period compromises the normal course of pregnancy leading to adverse maternal and fetal outcomes. Omega-3 and omega-6 long chain polyunsaturated fatty acids (LC-PUFA) are important for the growth and development of the placenta. Maternal fatty acids and their metabolites influence the normal course of pregnancy by regulating cell growth and development, cell signaling, regulate angiogenesis, modulate inflammatory responses and influence various structural and functional processes. Alterations in LC-PUFA and their metabolites may result in inadequate spiral artery remodeling or placental angiogenesis leading to structural and functional deficiency of the placenta which contributes to several pregnancy complications like preeclampsia, gestational diabetes mellitus, intrauterine growth restriction, and results in adverse birth outcomes. In this review, we summarize studies examining the role of fatty acids and their metabolites in pregnancy. We also discuss the possible molecular mechanisms through which LC-PUFA influences placental growth and development. Studies have demonstrated that omega-3 fatty acid supplementation lowers the incidence of preterm births, but its effect on reducing pregnancy complications are inconclusive.
Collapse
Affiliation(s)
- Aditi A Godhamgaonkar
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India
| | - Nisha S Wadhwani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India.
| |
Collapse
|
16
|
Rabinowicz S, Levkovitz O, Leibovitch L, Schushan-Eisen I, Morag I, Rosen C, Maayan-Metzger A, Strauss T. Increased risk for early hypertriglyceridemia in small for gestational age preterm infants. Eur J Pediatr 2020; 179:1873-1879. [PMID: 32770487 DOI: 10.1007/s00431-020-03764-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 11/29/2022]
Abstract
Small for gestational age preterm are at increased risk for future metabolic syndrome. Early indication for the disrupted metabolism may be found in the perinatal period. We aimed to evaluate whether small for gestational age preterm infants are at increased risk for hypertriglyceridemia when treated with lipid emulsions, and to investigate the association between triglyceride levels and morbidity. Small for gestational age infants ≤ 34 weeks' gestation age born during 2013-2016 were matched and compared with appropriate for gestational age counterparts. Triglyceride concentration > 250 mg/dL during treatment with parenteral nutrition was considered high. The study included 71 pairs of preterm infants. Hypertriglyceridemia was documented among 22.5% of the small for gestational age infants vs. 5.6% of the appropriate for gestational age infants (p = 0.007). Mean triglyceride levels were 194.4 ± 192.3 mg/dL and 99.9 ± 82.8 mg/dL, respectively (p < 0.001). Small for gestational age was predictive of hypertriglyceridemia (OR = 6.41; 95% CI 1.8-22.9). No significant association was found between triglyceride levels and morbidities in multivariate analysis.Conclusion: Small for gestational age preterm infants receiving lipid emulsions might be at a higher risk for hypertriglyceridemia. Routine monitoring of triglyceride levels will enable identification of the necessity for a slower increase in lipid emulsion therapy. What is Known: • Moderate and very preterm infants are routinely treated with lipid emulsions. • Small for gestational age (SGA) infants may have different metabolism, as they demonstrate higher risk for metabolic syndrome. What is New: • • SGA infants had a higher mean triglyceride level and more commonly had early hypertriglyceridemia (triglycerides > 250 mg/dL) compared with appropriate for gestational age infants treated with the same intravenous lipid dose. Small for gestational age was predictive of hypertriglyceridemia. • No significant association was found between triglyceride levels and morbidities in multivariate analysis.
Collapse
Affiliation(s)
- Shira Rabinowicz
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orly Levkovitz
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262000, Tel Hashomer, Israel
| | - Leah Leibovitch
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262000, Tel Hashomer, Israel
| | - Irit Schushan-Eisen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262000, Tel Hashomer, Israel
| | - Iris Morag
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262000, Tel Hashomer, Israel
| | - Chava Rosen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262000, Tel Hashomer, Israel
| | - Ayala Maayan-Metzger
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262000, Tel Hashomer, Israel
| | - Tzipora Strauss
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262000, Tel Hashomer, Israel.
| |
Collapse
|
17
|
Huhtala MS, Tertti K, Rönnemaa T. Serum lipids and their association with birth weight in metformin and insulin treated patients with gestational diabetes. Diabetes Res Clin Pract 2020; 170:108456. [PMID: 32979417 DOI: 10.1016/j.diabres.2020.108456] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/14/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022]
Abstract
AIMS To compare the effects of metformin and insulin treatment on maternal serum lipids in patients with gestational diabetes (GDM), and to analyse the associations between individual lipids and birth weight (BW). METHODS This is a secondary analysis of a randomized trial comparing metformin (n = 110) and insulin (n = 107) treatment of GDM. Fasting serum lipidome was measured at baseline (the time of diagnosis, mean 30 gestational weeks, gw) and at 36 gw using nuclear magnetic resonance spectroscopy. RESULTS Total and VLDL triglycerides, and VLDL cholesterol increased from baseline to 36 gw in both treatment groups. The rise in triglycerides was greater in the metformin treated patients (p < 0.01). Baseline total and VLDL triglycerides, VLDL cholesterol, and apolipoprotein B to A-1 ratio (apoB/apoA-1) associated positively with BW, more strongly in the metformin group. Among patients in the highest baseline VLDL cholesterol or apoB/apoA-1 quartile, those treated with insulin had lower BWs than those treated with metformin (p < 0.03). CONCLUSION Compared to insulin, metformin treatment of GDM led to higher maternal serum concentrations of triglyceride-rich lipoproteins. Especially triglycerides and cholesterol in VLDL were positively associated with BW. Women with high VLDL cholesterol or high apoB/apoA-1 may benefit from insulin treatment over metformin with respect to offspring BW.
Collapse
Affiliation(s)
- Mikael S Huhtala
- Department of Obstetrics and Gynecology, University of Turku, 20014 Turku, Finland; Department of Obstetrics and Gynecology, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland.
| | - Kristiina Tertti
- Department of Obstetrics and Gynecology, University of Turku, 20014 Turku, Finland; Department of Obstetrics and Gynecology, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Tapani Rönnemaa
- Department of Medicine, University of Turku, 20014 Turku, Finland; Department of Medicine, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| |
Collapse
|
18
|
Lindberg R, Lindqvist M, Trupp M, Vinnars MT, Nording ML. Polyunsaturated Fatty Acids and Their Metabolites in Hyperemesis Gravidarum. Nutrients 2020; 12:nu12113384. [PMID: 33158081 PMCID: PMC7694173 DOI: 10.3390/nu12113384] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/26/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) have been studied in relation to pregnancy. However, there is limited knowledge on PUFAs and their metabolites in relation to hyperemesis gravidarum (HG), a pregnancy complication associated with nutritional deficiencies and excessive vomiting. In order to survey the field, a systematic review of the literature was performed, which also included nausea and vomiting of pregnancy (NVP) due to its close relationship with HG. In the very few published studies found, the main subjects of the research concerned free fatty acids (four records), lipid profiles (three records), and bioactive lipids (one article about prostaglandin E2 and one about endocannabinoids). The authors of these studies concluded that, although no cause-and-effect relationship can be established, HG is linked to increased sympathetic responsiveness, thermogenic activity and metabolic rate. In addition, NVP is linked to a metabolic perturbance (which lasts throughout pregnancy). The low number of retrieved records underlines the need for more research in the area of PUFAs and HG, especially with regard to the underlying mechanism for the detected effects, potentially involving growth differentiation factor 15 (GDF15) since evidence for GDF15 regulation of lipid metabolism and the role for GDF15 and its receptor in nausea and vomiting is emerging.
Collapse
Affiliation(s)
| | - Maria Lindqvist
- Department of Nursing, Umeå University, 901 87 Umeå, Sweden;
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, 901 87 Umeå, Sweden;
| | - Miles Trupp
- Department of Clinical Sciences, Neurosciences, Umeå University, 901 87 Umeå, Sweden;
| | - Marie-Therese Vinnars
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, 901 87 Umeå, Sweden;
- Division of Obstetrics and Gynecology, Örnsköldsvik Hospital, 891 89 Örnsköldsvik, Sweden
| | - Malin L. Nording
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden;
- Correspondence:
| |
Collapse
|
19
|
Jesuino BG, Foratori-Junior GA, Missio ALT, Mascoli LS, Sales-Peres SHDC. Periodontal status of women with excessive gestational weight gain and the association with their newborns' health. Int Dent J 2020; 70:396-404. [PMID: 32501575 PMCID: PMC9379182 DOI: 10.1111/idj.12580] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE The aim of this study was to assess the association between high gestational weight gain and systemic and periodontal status of women between the 32nd and 36th gestational weeks of pregnancy (T1) and after delivery (T2), and the association of these factors with newborns' health. METHODS The sample was divided into excessive gestational weight gain (GE = 25) and normal gestational weight gain (GN = 25) and was evaluated regarding: (i) socio-economic status; (ii) systemic and periodontal status; and (iii) newborns' health. The results were analysed using the Mann-Whitney U-test, the t-test, the Friedman test, analysis of variance (ANOVA) and Cochran's Q test (P < 0.05). RESULTS Women with GE had lower household income (P = 0.010) and higher body mass index (BMI) at both T1 and T2. The prevalence of hypertension at T1 was higher in women with GE, but the condition resolved post-delivery (P = 0.001). Worsening in oral hygiene was observed at T2 in both groups (P < 0.001). Sixty-eight percent of women with GE and 16% of women with GN had periodontitis at T1, and 52% and 12%, respectively, had periodontitis at T2. In women with GE, the BMI of newborns was higher (P = 0.031). CONCLUSIONS Women with high gestational weight gain also showed a high prevalence of hypertension and periodontitis during pregnancy, and persistent periodontitis after delivery. High gestational weight gain was related to high BMI of newborns.
Collapse
Affiliation(s)
- Bruno Gualtieri Jesuino
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Bauru, São Paulo, Brazil
| | - Gerson Aparecido Foratori-Junior
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Bauru, São Paulo, Brazil
| | - Alana Luiza Trenhago Missio
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Bauru, São Paulo, Brazil
| | - Leonardo Silva Mascoli
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Bauru, São Paulo, Brazil
| | | |
Collapse
|
20
|
Role of adipose tissue in regulating fetal growth in gestational diabetes mellitus. Placenta 2020; 102:39-48. [PMID: 33218577 DOI: 10.1016/j.placenta.2020.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
Gestational diabetes mellitus (GDM) is a global health issue with significant short and long-term complications for both mother and baby. There is a strong need to identify an effective therapeutic that can prevent the development of GDM. A better understanding of the pathophysiology of GDM and the relationship between the adipose tissue, the placenta and fetal growth is required. The placenta regulates fetal growth by modulating nutrient transfer of glucose, amino acids and fatty acids. Various factors secreted by the adipose tissue, such as adipokines, adipocytokines and more recently identified extracellular vesicles, can influence inflammation and interact with placental nutrient transport. In this review, the role of the placental nutrient transporters and the adipose-derived factors that can influence their function will be discussed. A better understanding of these factors and their relationship may make a potential target for therapeutic interventions to prevent the development of GDM and its consequences.
Collapse
|
21
|
Guo J, Fang M, Zhuang S, Qiao Y, Huang W, Gong Q, Xu D, Zhang Y, Wang H. Prenatal dexamethasone exposure exerts sex-specific effect on placental oxygen and nutrient transport ascribed to the differential expression of IGF2. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:233. [PMID: 32309380 PMCID: PMC7154419 DOI: 10.21037/atm.2019.12.156] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Clinical studies have showed that dexamethasone exposure during pregnancy could cause fetal growth retardation, but the mechanism by which prenatal dexamethasone exposure influences placental nutrient transport is still unclear. This study investigated the impacts of prenatal dexamethasone on the placental oxygen and nutrient transport. Methods Pregnant Wistar rats were subcutaneously administered with dexamethasone from day 9 to day 20 of gestation at 0.2 or 0.8 mg/kg·d. Pregnant rats were sacrificed on gestational day 20. The placental tissue was collected for analysis. Results Prenatal dexamethasone exposure (PDE) declined the fetal weight and increased the intrauterine growth retardation (IUGR) rate in a dose-dependent manner. The total placental volume and the length, density and surface area of fetal capillaries in the labyrinth zone reduced in a dose-dependent manner. In addition, the thickness of syncytial membrane dose-dependently increased, resulting in a dose-dependent decrease in oxygen diffusion capacity. Furthermore, after PDE, the nutrient transport area and oxygen diffusion capacity of male placenta were lower than that of female placenta. The mRNA and protein expression of placental nutrient transporters including glucose transporter 1 (GLUT1), glucose transporter 3 (GLUT3), L-type amino acid transporter 1 (LAT1), lipoprotein lipase (LPL) and scavenger receptor class B type 1 (SRB1) increased in female placenta. However, in male placenta, the expression of LAT1, LPL and SRB1 was significantly decreased and GLUT1 and GLUT3 have a decrease trend. We further investigated the expression of insulin-like growth factor 1 (IGF1) and insulin-like growth factor 2 (IGF2) related to placental and fetal growth and development. Our study showed that the expression of IGF1 was significantly decreased both in male and female placentas after PDE. But the expression of IGF2 was significantly increased in female placentas while significantly decreased in male placentas. Conclusions Our study shows prenatal dexamethasone exposure exerts sex-specific influence on the placental oxygen and nutrient transport. This might be ascribed to the differential expression of IGF2 after PDE. These findings provide evidence on the dexamethasone-induced toxicity to the placenta and fetal development.
Collapse
Affiliation(s)
- Juanjuan Guo
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Man Fang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Siying Zhuang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuan Qiao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Wen Huang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qing Gong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Dan Xu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Yuanzhen Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.,Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| |
Collapse
|
22
|
Chassen S, Jansson T. Complex, coordinated and highly regulated changes in placental signaling and nutrient transport capacity in IUGR. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165373. [PMID: 30684642 PMCID: PMC6650384 DOI: 10.1016/j.bbadis.2018.12.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023]
Abstract
The most common cause of intrauterine growth restriction (IUGR) in the developed world is placental insufficiency, a concept often used synonymously with reduced utero-placental and umbilical blood flows. However, placental insufficiency and IUGR are associated with complex, coordinated and highly regulated changes in placental signaling and nutrient transport including inhibition of insulin and mTOR signaling and down-regulation of specific amino acid transporters, Na+/K+-ATPase, the Na+/H+-exchanger, folate and lactate transporters. In contrast, placental glucose transport capacity is unaltered and Ca2+-ATPase activity and the expression of proteins involved in placental lipid transport are increased in IUGR. These findings are not entirely consistent with the traditional view that the placenta is dysfunctional in IUGR, but rather suggest that the placenta adapts to reduce fetal growth in response to an inability of the mother to allocate resources to the fetus. This new model has implications for the understanding of the mechanisms underpinning IUGR and for the development of intervention strategies.
Collapse
Affiliation(s)
- Stephanie Chassen
- Department of Pediatrics, Division of Neonatology, University of Colorado, Anschutz Medical Campus, Aurora, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, USA.
| |
Collapse
|
23
|
Balachandiran M, Bobby Z, Dorairajan G, Jacob SE, Gladwin V, Vinayagam V, Packirisamy RM. Placental Accumulation of Triacylglycerols in Gestational Diabetes Mellitus and Its Association with Altered Fetal Growth are Related to the Differential Expressions of Proteins of Lipid Metabolism. Exp Clin Endocrinol Diabetes 2020; 129:803-812. [PMID: 31968385 DOI: 10.1055/a-1017-3182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) exhibit altered placental lipid metabolism. The molecular basis of this altered metabolism is not clear. Altered placental expression of proteins of lipogenesis and fatty acid oxidation may be involved in the placental accumulation of triacylglycerols (TG). The present study was aimed at investigating the differential expressions of placental proteins related to lipid metabolism among GDM women in comparison with control pregnant women (CPW) and to correlate them with maternal and fetal lipid parameters as well as altered fetal growth. MATERIALS AND METHODS Maternal blood, cord blood, and placental samples were collected from GDM and CPW. The biochemical parameters, glucose, lipid profile and free fatty acids (FFA) were measured. The placental TG content was measured. Differential placental expressions of proteins; phosphatidylinositol-3-kinase (PI3K) p85α, PI3K p110α,liver X receptor alpha (LXRα), sterol regulatory element binding protein1(SREBP1), fatty acid synthase (FAS), stearyl CoA desaturase1 (SCD1), lipoprotein lipase (LPL),Peroxisome proliferator-activated receptor (PPAR)α and PPARγ were analysed by western blotting and immunohistochemistry. RESULTS Placental protein expressions of PI3K p110α, LXRα, FAS, SCD1, and LPL were found to be significantly higher, whereas PPARα and PPARγ were lower in GDM women compared with CPW. The placental TG content and cord plasma FFA were increased in GDM women compared with CPW. The placental TG content positively correlated with Ponderal index of GDM new-borns. CONCLUSION Differential expressions of placental proteins related to lipid metabolism in GDM might have led to placental TG accumulation. This might have contributed to the fetal overgrowth in GDM.
Collapse
Affiliation(s)
- Manoharan Balachandiran
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Zachariah Bobby
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Gowri Dorairajan
- Department of Obstetrics & Gynaecology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Sajini Elizabeth Jacob
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Victorraj Gladwin
- Department of Anatomy, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Vickneshwaran Vinayagam
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Rajaa Muthu Packirisamy
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
24
|
The effect of high glucose on lipid metabolism in the human placenta. Sci Rep 2019; 9:14114. [PMID: 31575970 PMCID: PMC6773712 DOI: 10.1038/s41598-019-50626-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) during pregnancy can result in fetal overgrowth, likely due to placental dysfunction, which has health consequences for the infant. Here we test our prediction from previous work using a placental cell line that high glucose concentrations affect placental lipid metabolism. Placentas from women with type 1 (n = 13), type 2 (n = 6) or gestational (n = 12) DM, BMI-matched to mothers without DM (n = 18), were analysed for lipase and fatty acid transport proteins and fatty acid and triglyceride content. Explants from uncomplicated pregnancies (n = 6) cultured in physiological or high glucose were similarly analysed. High glucose levels did not alter placental lipase or transporter expression or the profile and abundance of fatty acids, but triglyceride levels were higher (p < 0.05), suggesting reduced β- oxidation. DM did not affect placental protein expression or fatty acid profile. Triglyceride levels of placentas from mothers with pre-existing DM were similar to controls, but higher in obese women with gestational DM. Maternal hyperglycemia may not affect placental fatty acid uptake and transport. However, placental β-oxidation is affected by high glucose and reduced in a subset of women with DM. Abnormal placental lipid metabolism could contribute to increased maternal-fetal lipid transfer and excess fetal growth in some DM pregnancies.
Collapse
|
25
|
Pre-pregnancy BMI, gestational weight gain and birth outcomes in Lebanon and Qatar: Results of the MINA cohort. PLoS One 2019; 14:e0219248. [PMID: 31265481 PMCID: PMC6605672 DOI: 10.1371/journal.pone.0219248] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/20/2019] [Indexed: 11/19/2022] Open
Abstract
Accumulating evidence has highlighted the role of maternal nutritional status on fetal development, birth outcomes and child health. The Mother and Infant Nutritional Assessment (MINA) cohort is a 3-year follow-up study of pregnant women and their children in Qatar and Lebanon. This study reports on the characteristics and determinants of pre-pregnancy BMI and Gestational Weight Gain (GWG) of MINA particiants, as well as birth outcomes. A total of 272 pregnant women were recruited during their first trimester from primary healthcare centers as well as private clinics in Beirut (n = 194) and Doha (n = 147). During the first visit, data collection included pre-pregnancy weight, sociodemographic and lifestyle characteristics. The weight before delivery and neonatal outcomes were extracted from the medical records. GWG was calculated as the difference between weight before delivery and pre-pregnancy weight and was classified into insufficient, adequate, and excessive, as per the IOM criteria. Overall, 42.1% of women had a pre-pregnancy BMI≥25 Kg/m2 (58% in Qatar vs 30.8% in Lebanon, p<0.001). Only 30.2% of women had adequate GWG, while 25.7% and 44.1% of women had insufficient and excessive GWG, respectively. In the cohort 68.7% of infants had a weight adequate-for-gestational age (AGA), 6.7% were SGA and 24.6% were LGA. The proportions of LGA were higher with greater GWG (p<0.05). After adjustment, Qatari women were 3 times more likely to be overweight or obese before pregnancy while a higher education level was associated with significantly lower odds of pre-pregnancy BMI≥25 Kg/m2. Pre-pregnancy BMI≥25 Kg/m2 and regular breakfast consumption were predictors of excessive GWG (OR: 3.20, CI: 1.48–6.91; OR: 2.84, CI: 1.15–7.02, respectively). The high prevalence of pre-pregnancy overweight and excessive GWG among MINA participants underscores the need for culture-specific intervention programs to promote healthy body weight in women of childbearing age, and prevent excessive weight gain during pregnancy.
Collapse
|
26
|
Ferchaud-Roucher V, Kramer A, Silva E, Pantham P, Weintraub ST, Jansson T, Powell TL. A potential role for lysophosphatidylcholine in the delivery of long chain polyunsaturated fatty acids to the fetal circulation. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:394-402. [PMID: 30572119 DOI: 10.1016/j.bbalip.2018.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/10/2018] [Accepted: 12/15/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Véronique Ferchaud-Roucher
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Anita Kramer
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elena Silva
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Priyadarshini Pantham
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, IL, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center San Antonio, TX, USA
| | - Thomas Jansson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa L Powell
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
27
|
Akerele OA, Cheema SK. A diet enriched in longer chain omega-3 fatty acids reduced placental inflammatory cytokines and improved fetal sustainability of C57BL/6 mice. Prostaglandins Leukot Essent Fatty Acids 2018; 137:43-51. [PMID: 30293596 DOI: 10.1016/j.plefa.2018.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 10/28/2022]
Abstract
Omega (n)-3 polyunsaturated fatty acids (PUFA) are important regulators of inflammatory response that may impact pregnancy outcome. The effects of breeding chow diets containing n-3 PUFA from either fish oil (FO) or soybean oil (SO) were investigated on tissue fatty acid composition, inflammatory cytokines and pregnancy outcome. Female C57BL/6 mice (7 weeks old) were fed FO or SO diets for 2 weeks before mating and throughout pregnancy. Animals were sacrificed before and during pregnancy at day 6.5, 12.5 and 18.5. The FO diet increased the incorporation of n-3 PUFA in placenta, with a concomitant decrease in the concentration of pro-inflammatory cytokines. The FO diet increased the mRNA expression of placental specific PUFA transporter, which coincided with accretion of n-3 PUFA in fetal brain. Sites of fetal resorption were noticeable in the SO group but not in the FO group. N-3 PUFA may improve fetal sustainability via altering cytokine levels.
Collapse
Affiliation(s)
- O A Akerele
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada
| | - S K Cheema
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
28
|
Miranda J, Simões RV, Paules C, Cañueto D, Pardo-Cea MA, García-Martín ML, Crovetto F, Fuertes-Martin R, Domenech M, Gómez-Roig MD, Eixarch E, Estruch R, Hansson SR, Amigó N, Cañellas N, Crispi F, Gratacós E. Metabolic profiling and targeted lipidomics reveals a disturbed lipid profile in mothers and fetuses with intrauterine growth restriction. Sci Rep 2018; 8:13614. [PMID: 30206284 PMCID: PMC6134091 DOI: 10.1038/s41598-018-31832-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022] Open
Abstract
Fetal growth may be impaired by poor placental function or maternal conditions, each of which can influence the transfer of nutrients and oxygen from the mother to the developing fetus. Large-scale studies of metabolites (metabolomics) are key to understand cellular metabolism and pathophysiology of human conditions. Herein, maternal and cord blood plasma samples were used for NMR-based metabolic fingerprinting and profiling, including analysis of the enrichment of circulating lipid classes and subclasses, as well as the number of sub-fraction particles and their size. Changes in phosphatidylcholines and glycoproteins were prominent in growth-restricted fetuses indicating significant alterations in their abundance and biophysical properties. Lipoprotein profiles showed significantly lower plasma concentrations of cholesterol-intermediate density lipoprotein (IDL), triglycerides-IDL and high-density lipoprotein (HDL) in mothers of growth-restricted fetuses compared to controls (p < 0.05). In contrast, growth-restricted fetuses had significantly higher plasma concentrations of cholesterol and triglycerides transporting lipoproteins [LDL, IDL, and VLDL, (p < 0.005; all)], as well as increased VLDL particle types (large, medium and small). Significant changes in plasma concentrations of formate, histidine, isoleucine and citrate in growth-restricted fetuses were also observed. Comprehensive metabolic profiling reveals that both, mother and fetuses of pregnancies complicated with fetal growth restriction have a substantial disruption in lipid metabolism.
Collapse
Affiliation(s)
- Jezid Miranda
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rui V Simões
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Cristina Paules
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Daniel Cañueto
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
| | | | - María L García-Martín
- BIONAND, Andalusian Centre for Nanomedicine and Biotechnology, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | - Francesca Crovetto
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rocio Fuertes-Martin
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
- Biosfer Teslab, Reus, Spain
| | - Monica Domenech
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - María D Gómez-Roig
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Elisenda Eixarch
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Ramon Estruch
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Lund University, Sweden; Skåne University Hospital, Lund, Sweden
| | | | - Nicolau Cañellas
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
- CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Fatima Crispi
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain.
| | - Eduard Gratacós
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
29
|
Wadhwani N, Patil V, Joshi S. Maternal long chain polyunsaturated fatty acid status and pregnancy complications. Prostaglandins Leukot Essent Fatty Acids 2018; 136:143-152. [PMID: 28888333 DOI: 10.1016/j.plefa.2017.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 07/06/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022]
Abstract
Maternal nutrition plays a crucial role in influencing fetal growth and birth outcome. Any nutritional insult starting several weeks before pregnancy and during critical periods of gestation is known to influence fetal development and increase the risk for diseases during later life. Literature suggests that chronic adult diseases may have their origin during early life - a concept referred to as Developmental Origins of Health and Disease (DOHaD) which states that adverse exposures early in life "program" risks for later chronic disorders. Long chain polyunsaturated fatty acids (LCPUFA), mainly omega-6 and omega-3 fatty acids are known to have an effect on fetal programming. The placental supply of optimal levels of LCPUFA to the fetus during early life is extremely important for the normal growth and development of both placenta and fetus. Any alteration in placental development will result in adverse pregnancy outcome such as gestational diabetes mellitus (GDM), preeclampsia, and intrauterine growth restriction (IUGR). A disturbed materno-fetal LCPUFA supply is known to be linked with each of these pathologies. Further, a disturbed LCPUFA metabolism is reported to be associated with a number of metabolic disorders. It is likely that LCPUFA supplementation during early pregnancy may be beneficial in improving the health of the mother, improving birth outcome and thereby reducing the risk of diseases in later life.
Collapse
Affiliation(s)
- Nisha Wadhwani
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Vidya Patil
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Sadhana Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India.
| |
Collapse
|
30
|
Mohan R, Baumann D, Alejandro EU. Fetal undernutrition, placental insufficiency, and pancreatic β-cell development programming in utero. Am J Physiol Regul Integr Comp Physiol 2018; 315:R867-R878. [PMID: 30110175 DOI: 10.1152/ajpregu.00072.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The prevalence of obesity and type 2 (T2D) diabetes is a major health concern in the United States and around the world. T2D is a complex disease characterized by pancreatic β-cell failure in association with obesity and insulin resistance in peripheral tissues. Although several genes associated with T2D have been identified, it is speculated that genetic variants account for only <10% of the risk for this disease. A strong body of data from both human epidemiological and animal studies shows that fetal nutrient factors in utero confer significant susceptibility to T2D. Numerous studies done in animals have shown that suboptimal maternal environment or placental insufficiency causes intrauterine growth restriction (IUGR) in the fetus, a critical factor known to predispose offspring to obesity and T2D, in part by causing permanent consequences in total functional β-cell mass. This review will focus on the potential contribution of the placenta in fetal programming of obesity and TD and its likely impact on pancreatic β-cell development and growth.
Collapse
Affiliation(s)
- Ramkumar Mohan
- Department of Integrative Biology and Physiology, University of Minnesota , Minneapolis, Minnesota
| | - Daniel Baumann
- Department of Integrative Biology and Physiology, University of Minnesota , Minneapolis, Minnesota
| | - Emilyn Uy Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
31
|
Afolabi OA, Alagbonsi AI, Oyinloye RT, Salahdeen HM, Salman TM, Olatunji LA. High-fat diet reduces weight gain but increases other cardio-metabolic indices in offspring of normotensive and hypertensive rats. Arch Physiol Biochem 2018; 124:218-225. [PMID: 28952789 DOI: 10.1080/13813455.2017.1383441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study investigated the effect of post-natal consumption of high-fat diet (HFD) on cardio-metabolic indices in male offspring of hypertensive female rats. There were neither significant differences in body weight gain either in pups from normotensive or hypertensive dams that received normal diet during the post-weaning periods (except at 7th and 9th weeks), nor in both pup groups that received HFD. However, both pup groups that received HFD had reduced body weight gain when compared to their counterparts that received normal diet. Post-weaning administration of HFD to pups of hypertensive and normotensive dams significantly increased their blood glucose, pressure and lipid profiles when compared to those weaned to normal diet. It was concluded that male offspring consumption of HFD diet elicits cardio-metabolic disturbance that slightly depended of maternal cardiovascular status but majorly depended on post-weaning weight gain, while that elicited by maternal hypertension is not related to post-weaning weight gain.
Collapse
Affiliation(s)
- Oladele Ayobami Afolabi
- a Department of Physiology, College of Health Sciences , Ladoke Akintola University of Technology , Ogbosomo , Oyo , Nigeria
| | - Abdullateef Isiaka Alagbonsi
- b Department of Physiology, Faculty of Medicine and Health Sciences , University of Gitwe , Gitwe , Republic of Rwanda
| | - Roseline Toyosi Oyinloye
- a Department of Physiology, College of Health Sciences , Ladoke Akintola University of Technology , Ogbosomo , Oyo , Nigeria
| | | | - Toyin Mohammed Salman
- d Department of Physiology, College of Health Sciences , University of Ilorin , Ilorin , Kwara , Nigeria
| | - Lawrence Aderemi Olatunji
- d Department of Physiology, College of Health Sciences , University of Ilorin , Ilorin , Kwara , Nigeria
| |
Collapse
|
32
|
Heerwagen MJR, Gumina DL, Hernandez TL, Van Pelt RE, Kramer AW, Janssen RC, Jensen DR, Powell TL, Friedman JE, Winn VD, Barbour LA. Placental lipoprotein lipase activity is positively associated with newborn adiposity. Placenta 2018; 64:53-60. [PMID: 29626981 DOI: 10.1016/j.placenta.2018.03.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/16/2018] [Accepted: 03/04/2018] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Recent data suggest that in addition to glucose, fetal growth is related to maternal triglycerides (TG). To reach the fetus, TG must be hydrolyzed to free fatty acids (FFA) and transported across the placenta, but regulation is uncertain. Placental lipoprotein lipase (pLPL) hydrolyzes TG, both dietary chylomicron TG (CM-TG) and very-low density lipoprotein TG (VLDL-TG), to FFA. This may promote fetal fat accretion by increasing the available FFA pool for placental uptake. We tested the novel hypothesis that pLPL activity, but not maternal adipose tissue LPL activity, is associated with newborn adiposity and higher maternal TG. METHODS Twenty mothers (n = 13 normal-weight; n = 7 obese) were prospectively recruited. Maternal glucose, insulin, TG (total, CM-TG, VLDL-TG), and FFA were measured at 14-16, 26-28, and 36-37 weeks, and adipose tissue LPL was measured at 26-28 weeks. At term delivery, placental villous biopsies were immediately analyzed for pLPL enzymatic activity. Newborn percent body fat (newborn %fat) was assessed by skinfolds. RESULTS Placental LPL activity was positively correlated with birthweight (r = 0.48;P = 0.03) and newborn %fat (r = 0.59;P = 0.006), further strengthened by correcting for gestational age at delivery (r = 0.75;P = 0.0001), but adipose tissue LPL was not. Maternal TG and BMI were not correlated with pLPL activity. Additionally, pLPL gene expression, while modestly correlated with enzymatic activity (r = 0.53;P < 0.05), was not correlated with newborn adiposity. DISCUSSION This is the first study to show a positive correlation between pLPL activity and newborn %fat. Placental lipase regulation and the role of pLPL in pregnancies characterized by nutrient excess and fetal overgrowth warrant further investigation.
Collapse
Affiliation(s)
- Margaret J R Heerwagen
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Diane L Gumina
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Teri L Hernandez
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; College of Nursing, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rachael E Van Pelt
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anita W Kramer
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rachel C Janssen
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dalan R Jensen
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa L Powell
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob E Friedman
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Linda A Barbour
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
33
|
Zhao R, Xu L, Wu M, Huang S, Cao X. Maternal pre-pregnancy body mass index, gestational weight gain influence birth weight. Women Birth 2018; 31:e20-e25. [DOI: 10.1016/j.wombi.2017.06.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/04/2017] [Accepted: 06/06/2017] [Indexed: 02/06/2023]
|
34
|
Chavan-Gautam P, Rani A, Freeman DJ. Distribution of Fatty Acids and Lipids During Pregnancy. Adv Clin Chem 2018; 84:209-239. [PMID: 29478515 DOI: 10.1016/bs.acc.2017.12.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal fatty acid and lipid metabolism undergoes changes during pregnancy to facilitate fetal growth and development. Different types of fatty acids have different roles in maintaining a successful pregnancy and they are incorporated into different forms of lipids for the purpose of storage and transport. This chapter aims to provide an understanding of the distribution and metabolism of fatty acids and lipids in the maternal, placental, and fetal compartments. We further describe how this distribution is altered in maternal obesity, preterm birth, and pregnancy complications such as gestational diabetes mellitus, preeclampsia, and intrauterine growth restriction.
Collapse
Affiliation(s)
- Preeti Chavan-Gautam
- Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India.
| | - Alka Rani
- Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India
| | - Dilys J Freeman
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
35
|
Chalil A, Kitson AP, Aristizabal Henao JJ, Marks KA, Elzinga JL, Lamontagne-Kam DME, Chalil D, Badoud F, Mutch DM, Stark KD. PEMT, Δ6 desaturase, and palmitoyldocosahexaenoyl phosphatidylcholine are increased in rats during pregnancy. J Lipid Res 2017; 59:123-136. [PMID: 29167412 DOI: 10.1194/jlr.m080309] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/07/2017] [Indexed: 01/14/2023] Open
Abstract
DHA is important for fetal neurodevelopment. During pregnancy, maternal plasma DHA increases, but the mechanism is not fully understood. Using rats fed a fixed-formula diet (DHA as 0.07% total energy), plasma and liver were collected for fatty acid profiling before pregnancy, at 15 and 20 days of pregnancy, and 7 days postpartum. Phosphatidylethanolamine methyltransferase (PEMT) and enzymes involved in PUFA synthesis were examined in liver. Ad hoc transcriptomic and lipidomic analyses were also performed. With pregnancy, DHA increased in liver and plasma lipids, with a large increase in plasma DHA between day 15 and day 20 that was mainly attributed to an increase in 16:0/DHA phosphatidylcholine (PC) in liver (2.6-fold) and plasma (3.9-fold). Increased protein levels of Δ6 desaturase (FADS2) and PEMT at day 20 and increased Pemt expression and PEMT activity at day 15 suggest that during pregnancy, both DHA synthesis and 16:0/DHA PC synthesis are upregulated. Transcriptomic analysis revealed minor changes in the expression of genes related to phospholipid synthesis, but little insight on DHA metabolism. Hepatic PEMT appears to be the mechanism for increased plasma 16:0/DHA PC, which is supported by increased DHA biosynthesis based on increased FADS2 protein levels.
Collapse
Affiliation(s)
- Alan Chalil
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Alex P Kitson
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | | | - Kristin A Marks
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Jason L Elzinga
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | | | - Daniel Chalil
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Flavia Badoud
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David M Mutch
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Ken D Stark
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
36
|
Gallo L, Barrett H, Dekker Nitert M. Review: Placental transport and metabolism of energy substrates in maternal obesity and diabetes. Placenta 2017; 54:59-67. [DOI: 10.1016/j.placenta.2016.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 11/29/2022]
|
37
|
Ferchaud-Roucher V, Rudolph MC, Jansson T, Powell TL. Fatty acid and lipid profiles in primary human trophoblast over 90h in culture. Prostaglandins Leukot Essent Fatty Acids 2017; 121:14-20. [PMID: 28651693 DOI: 10.1016/j.plefa.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 05/18/2017] [Accepted: 06/01/2017] [Indexed: 01/15/2023]
Abstract
Little is known about the mechanisms underlying the preferential transport of long chain polyunsaturated fatty acids (LCPUFA) to the fetus by the syncytiotrophoblast and the role of cytotrophoblasts in placental lipid metabolism and transport. We studied primary human trophoblast (PHT) cells cultured for 90h to determine the fatty acid and lipid composition of cytotrophoblast (18h culture) and syncytiotrophoblast (90h culture) cells. In cultured PHT total lipid fatty acids were significantly (P < 0.05) reduced at 90h compared to 18h in culture including lower levels of palmitic acid (PA, 16:0, -37%), palmitoleic acid (POA, 16:1n-7, -30%), oleic acid (OA, 18:1n-9, -31%), LCPUFA arachidonic acid (AA, 20:4n-6, -28%) and α-linolenic acid (ALA, 18:3n-3, -55%). In major lipid classes, OA and most of the n-3 and n-6 LCPUFA were markedly lower at 90h in TG (-57 to -76%; p < 0.05). In the cellular NEFA, n-6 LCPUFA, dihomo-γ-linolenic acid (DGLA, 20:3n-6) and AA were both reduced by -51% and DHA was -55% lower (p < 0.05) at 90h. In contrast, phospholipid FA content did not change between cytotrophoblasts and syncytiotrophoblast except for OA, which decreased by -62% (p < 0.05). Decreasing PHT TG and NEFA lipid content at 90h in culture is likely due to processes related to differentiation such as alterations in lipase activity that occur as cytotrophoblast cells differentiate. We speculate that syncytiotrophoblast prioritizes PL containing AA and DHA for transfer to the fetus by mobilizing FA from storage lipids.
Collapse
Affiliation(s)
- Véronique Ferchaud-Roucher
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Michael C Rudolph
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa L Powell
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
38
|
田 鎏, 廖 晖, 杨 慧, 马 妮, 张 昌, 刁 红. [Expression of FABP7 in mouse placenta tissue and human trophoblast HTR-8/Svneo cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:594-599. [PMID: 28539280 PMCID: PMC6780475 DOI: 10.3969/j.issn.1673-4254.2017.05.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To detect the expression of FABP7 in the placenta of pregnant mice and in HTR-8/Svneo cells. METHODS Real-time PCR and immunofluorescence were used to detect FABP7 mRNA and protein expressions in the uterine and placental tissue of pregnant mice at different days of gestation. FABP7 expression was also detected in cultured HTR-8/Svneo cells using immunofluorescence assay. The mice were treated with E2, P4 or their combination for 6 and 24 h and Fabp7 mRNA level in the uterus was detected with real-time PCR. RESULTS At 7.5-10.5 days of gestation, the pregnant mice showed positive expressions of Fabp7 mRNA in the uterus and placenta, and FABP7 protein was detected in the decidualized cells and trophoblast giant cells. The expressions of FABP7 were detected at both the mRNA and protein levels in cultured HTR-8/Svneo cells. In mice treated with P4 alone or with E2+P4 for 6 and 24 h, the expression level of Fabp7 mRNA was upregulated in the uterus. Fabp7 upregulation was observed in mice at 24 h following E2 treatment but not at 6 h. CONCLUSION FABP7 is expressed in trophoblast giant cells and decidual cells in the placental tissue of mice and in cultured HTR-8/Svneo cells, suggesting the involvement of FABP7 in placental development and in maintenance of pregnancy. E2 and P4 can regulate the expression of FABP7 in mouse uterus.
Collapse
Affiliation(s)
- 鎏 田
- 湖北医药学院附属人民医院生殖医学中心,湖北 十堰 442000Reproductive Medicine Center, People's Hospital Affiliated to Hubei University of Medicine, Shiyan 442000, China
- 湖北医药学院生物工程学院,湖北 十堰 442000College of Bioengineering, Shiyan 442000, China
| | - 晖淇 廖
- 湖北医药学院附属人民医院生殖医学中心,湖北 十堰 442000Reproductive Medicine Center, People's Hospital Affiliated to Hubei University of Medicine, Shiyan 442000, China
- 湖北医药学院生物工程学院,湖北 十堰 442000College of Bioengineering, Shiyan 442000, China
| | - 慧 杨
- 湖北医药学院附属人民医院生殖医学中心,湖北 十堰 442000Reproductive Medicine Center, People's Hospital Affiliated to Hubei University of Medicine, Shiyan 442000, China
| | - 妮 马
- 湖北医药学院附属人民医院生殖医学中心,湖北 十堰 442000Reproductive Medicine Center, People's Hospital Affiliated to Hubei University of Medicine, Shiyan 442000, China
- 湖北医药学院生物工程学院,湖北 十堰 442000College of Bioengineering, Shiyan 442000, China
| | - 昌军 张
- 湖北医药学院附属人民医院生殖医学中心,湖北 十堰 442000Reproductive Medicine Center, People's Hospital Affiliated to Hubei University of Medicine, Shiyan 442000, China
- 湖北医药学院生物工程学院,湖北 十堰 442000College of Bioengineering, Shiyan 442000, China
- 湖北医药学院胚胎干细胞湖北省重点实验室,湖北 十堰 442000Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan 442000, China
| | - 红录 刁
- 湖北医药学院附属人民医院生殖医学中心,湖北 十堰 442000Reproductive Medicine Center, People's Hospital Affiliated to Hubei University of Medicine, Shiyan 442000, China
- 湖北医药学院生物工程学院,湖北 十堰 442000College of Bioengineering, Shiyan 442000, China
- 湖北医药学院胚胎干细胞湖北省重点实验室,湖北 十堰 442000Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan 442000, China
| |
Collapse
|
39
|
Gagné-Ouellet V, Houde AA, Guay SP, Perron P, Gaudet D, Guérin R, Jean-Patrice B, Hivert MF, Brisson D, Bouchard L. Placental lipoprotein lipase DNA methylation alterations are associated with gestational diabetes and body composition at 5 years of age. Epigenetics 2017; 12:616-625. [PMID: 28486003 DOI: 10.1080/15592294.2017.1322254] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is associated with obesity in childhood. This suggests that consequences of in utero exposure to maternal hyperglycemia extend beyond the fetal development, possibly through epigenetic programming. The aims of this study were to assess whether placental DNA methylation (DNAm) marks were associated with maternal GDM status and to offspring body composition at 5 years old in a prospective birth cohort. DNAm levels were measured in the fetal side of the placenta in 66 samples (24 from GDM mothers) using bisDNA-pyrosequencing. Anthropometric and body composition (bioimpedance) were measured in children at 5 years of age. Mann-Whitney and Spearman tests were used to assess associations between GDM, placental DNAm levels at the lipoprotein lipase (LPL) locus and children's weight, height, body mass index (BMI), body fat, and lean masses at 5 years of age. Weight, height, and BMI z-scores were computed according to the World Health Organization growth chart. Analyses were adjusted for gestational age at birth, child sex, maternal age, and pre-pregnancy BMI. LPL DNAm levels were positively correlated with birth weight z-scores (r = 0.252, P = 0.04), and with mid-childhood weight z-scores (r = 0.314, P = 0.01) and fat mass (r = 0.275, P = 0.04), and negatively correlated with lean mass (r = -0.306, P = 0.02). We found a negative correlation between LPL DNAm and mRNA levels in placenta (r = -0.459; P < 0.001), which highlights the regulation of transcriptional activity by these epivariations. We demonstrated that alterations in fetal placental DNAm levels at the LPL gene locus are associated with the anthropometric profile in children at 5 years of age. These findings support the concept of fetal metabolic programming through epigenetic changes.
Collapse
Affiliation(s)
- Valérie Gagné-Ouellet
- a Department of Biochemistry , Université de Sherbrooke , Sherbrooke , QC , Canada.,b ECOGENE-21 Biocluster , Chicoutimi , Quebec , Canada , QC , Canada
| | - Andrée-Anne Houde
- c Department of Medicine , Université de Montréal , Montréal , QC , Canada
| | - Simon-Pierre Guay
- a Department of Biochemistry , Université de Sherbrooke , Sherbrooke , QC , Canada.,b ECOGENE-21 Biocluster , Chicoutimi , Quebec , Canada , QC , Canada.,e Department of Medicine , Université de Sherbrooke , Sherbrooke , QC , Canada
| | - Patrice Perron
- b ECOGENE-21 Biocluster , Chicoutimi , Quebec , Canada , QC , Canada.,e Department of Medicine , Université de Sherbrooke , Sherbrooke , QC , Canada
| | - Daniel Gaudet
- b ECOGENE-21 Biocluster , Chicoutimi , Quebec , Canada , QC , Canada.,f Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine , Université de Montréal Community Gene Medicine Center, Chicoutimi Department of Medicine, Université de Montréal , Montréal , QC , Canada
| | - Renée Guérin
- d Department of Medical Biology , CIUSSS Saguenay-Lac-Saint-Jean - Chicoutimi Hospital , Saguenay , QC , Canada
| | | | - Marie-France Hivert
- e Department of Medicine , Université de Sherbrooke , Sherbrooke , QC , Canada.,g Department of Population Medicine , Harvard Pilgrim Health Care Institute, Harvard Medical School , Boston , MA , USA
| | - Diane Brisson
- b ECOGENE-21 Biocluster , Chicoutimi , Quebec , Canada , QC , Canada.,f Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine , Université de Montréal Community Gene Medicine Center, Chicoutimi Department of Medicine, Université de Montréal , Montréal , QC , Canada
| | - Luigi Bouchard
- a Department of Biochemistry , Université de Sherbrooke , Sherbrooke , QC , Canada.,b ECOGENE-21 Biocluster , Chicoutimi , Quebec , Canada , QC , Canada.,d Department of Medical Biology , CIUSSS Saguenay-Lac-Saint-Jean - Chicoutimi Hospital , Saguenay , QC , Canada
| |
Collapse
|
40
|
Abstract
Epidemiological evidence links an individual's susceptibility to chronic disease in adult life to events during their intrauterine phase of development. Biologically this should not be unexpected, for organ systems are at their most plastic when progenitor cells are proliferating and differentiating. Influences operating at this time can permanently affect their structure and functional capacity, and the activity of enzyme systems and endocrine axes. It is now appreciated that such effects lay the foundations for a diverse array of diseases that become manifest many years later, often in response to secondary environmental stressors. Fetal development is underpinned by the placenta, the organ that forms the interface between the fetus and its mother. All nutrients and oxygen reaching the fetus must pass through this organ. The placenta also has major endocrine functions, orchestrating maternal adaptations to pregnancy and mobilizing resources for fetal use. In addition, it acts as a selective barrier, creating a protective milieu by minimizing exposure of the fetus to maternal hormones, such as glucocorticoids, xenobiotics, pathogens, and parasites. The placenta shows a remarkable capacity to adapt to adverse environmental cues and lessen their impact on the fetus. However, if placental function is impaired, or its capacity to adapt is exceeded, then fetal development may be compromised. Here, we explore the complex relationships between the placental phenotype and developmental programming of chronic disease in the offspring. Ensuring optimal placentation offers a new approach to the prevention of disorders such as cardiovascular disease, diabetes, and obesity, which are reaching epidemic proportions.
Collapse
Affiliation(s)
- Graham J Burton
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Abigail L Fowden
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Kent L Thornburg
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
41
|
Papacleovoulou G, Nikolova V, Oduwole O, Chambers J, Vazquez-Lopez M, Jansen E, Nicolaides K, Parker M, Williamson C. Gestational disruptions in metabolic rhythmicity of the liver, muscle, and placenta affect fetal size. FASEB J 2017; 31:1698-1708. [PMID: 28082353 PMCID: PMC5566176 DOI: 10.1096/fj.201601032r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/03/2017] [Indexed: 11/12/2022]
Abstract
Maternal metabolic adaptations are essential for successful pregnancy outcomes. We investigated how metabolic gestational processes are coordinated, whether there is a functional link with internal clocks, and whether disruptions are related to metabolic abnormalities in pregnancy, by studying day/night metabolic pathways in murine models and samples from pregnant women with normally grown and large-for-gestational age infants. In early mouse pregnancy, expression of hepatic lipogenic genes was up-regulated and uncoupled from the hepatic clock. In late mouse pregnancy, rhythmicity of energy metabolism-related genes in the muscle followed the patterns of internal clock genes in this tissue, and coincided with enhanced lipid transporter expression in the fetoplacental unit. Diurnal triglyceride patterns were disrupted in human placentas from pregnancies with large-for-gestational age infants and this overlapped with an increase in BMAL1 expression. Metabolic adaptations in early pregnancy are uncoupled from the circadian clock, whereas in late pregnancy, energy availability is mediated by coordinated muscle-placenta metabolic adjustments linked to internal clocks. Placental triglyceride oscillations in the third trimester of human pregnancy are lost in large-for-gestational age infants and may be regulated by BMAL1. In summary, disruptions in metabolic and circadian rhythmicity are associated with increased fetal size, with implications for the pathogenesis of macrosomia.-Papacleovoulou, G., Nikolova, V., Oduwole, O., Chambers, J., Vazquez-Lopez, M., Jansen, E., Nicolaides, K., Parker, M., Williamson, C. Gestational disruptions in metabolic rhythmicity of the liver, muscle, and placenta affect fetal size.
Collapse
Affiliation(s)
| | - Vanya Nikolova
- Division of Women's Health, Guy's Campus, King's College London, London, United Kingdom
| | - Olayiwola Oduwole
- Institute of Reproductive and Developmental Biology, Surgery and Cancer, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Jenny Chambers
- Women's Health Research Centre, Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Marta Vazquez-Lopez
- Women's Health Research Centre, Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Eugene Jansen
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands; and
| | - Kypros Nicolaides
- Harris Birthright Centre for Fetal Medicine, King's College London, London, United Kingdom
| | - Malcolm Parker
- Institute of Reproductive and Developmental Biology, Surgery and Cancer, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Catherine Williamson
- Division of Women's Health, Guy's Campus, King's College London, London, United Kingdom;
- Institute of Reproductive and Developmental Biology, Surgery and Cancer, Hammersmith Hospital, Imperial College London, London, United Kingdom
| |
Collapse
|
42
|
Zeng Z, Liu F, Li S. Metabolic Adaptations in Pregnancy: A Review. ANNALS OF NUTRITION AND METABOLISM 2017; 70:59-65. [PMID: 28297696 DOI: 10.1159/000459633] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/02/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pregnancy is a dynamic state involving multiple adaptations that are necessary in order to ensure a continuous supply of essential metabolites to support the growth and the development of the fetus. OBJECTIVES This review article is aimed to discuss important adaptations in metabolism that take place during non-complicated pregnancy. MATERIALS AND METHODS We searched the electronic database PubMed for pre-clinical as well as clinical controlled trials reporting the importance of metabolic adaptations during a non-complicated pregnancy. The preferred language was English and the most recent reports were selected to get an updated review. RESULTS It was observed clearly in the searched literature that metabolic adaptations are a crucial part of pregnancy, as they provide the mother with sufficient energy stores to meet the demands of pregnancy. These adaptions also help in preparing the mother for lactation and also help in providing proper environment for the proper growth of fetus in the womb. Moreover, multiple biomolecules including glucose, fatty acids, ketone bodies, hormones collectively contribute toward these metabolic adaptations. CONCLUSIONS This review article concludes that metabolic adaptations are crucial for proper fetus development.
Collapse
Affiliation(s)
- Zhandong Zeng
- Department of Pediatric Surgery, Xuzhou Children's Hospital, Xuzhou, PR China
| | | | | |
Collapse
|
43
|
Herrera E, Desoye G. Maternal and fetal lipid metabolism under normal and gestational diabetic conditions. Horm Mol Biol Clin Investig 2017; 26:109-27. [PMID: 26351960 DOI: 10.1515/hmbci-2015-0025] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/08/2015] [Indexed: 12/18/2022]
Abstract
Maternal lipids are strong determinants of fetal fat mass. Here we review the overall lipid metabolism in normal and gestational diabetes mellitus (GDM) pregnancies. During early pregnancy, the increase in maternal fat depots is facilitated by insulin, followed by increased adipose tissue breakdown and subsequent hypertriglyceridemia, mainly as a result of insulin resistance (IR) and estrogen effects. The response to diabetes is variable as a result of greater IR but decreased estrogen levels. The vast majority of fatty acids (FAs) in the maternal circulation are esterified and associated with lipoproteins. These are taken up by the placenta and hydrolyzed by lipases. The released FAs enter various metabolic routes and are released into fetal circulation. Although these determinants are modified in maternal GDM, the fetus does not seem to receive more FAs than in non-GDM pregnancies. Long-chain polyunsaturated FAs are essential for fetal development and are obtained from the mother. Mitochondrial FA oxidation occurs in fetal tissue and in placenta and contributes to energy production. Fetal fat accretion during the last weeks of gestation occurs very rapidly and is sustained not only by FAs crossing the placenta, but also by fetal lipogenesis. Fetal hyperinsulinemia in GDM mothers promotes excess accretion of adipose tissue, which gives rise to altered adipocytokine profiles. Fetal lipoproteins are low at birth, but the GDM effects are unclear. The increase in body fat in neonates of GDM women is a risk factor for obesity in early childhood and later life.
Collapse
|
44
|
Abstract
Fatty acids are critical nutrient regulators of intracellular signaling and influence key pathways including inflammatory responses, hemostasis as well as central nervous system development and function. Preterm birth interrupts the maternal-fetal transfer of essential fatty acids including docosahexaenoic and arachidonic acids, which occurs during the third trimester. Postnatal deficits of these nutrients accrue in preterm infants during the first week and they remain throughout the first months. Due to the regulatory roles of these fatty acids, such deficits contribute an increased risk of developing prematurity-related morbidities including impaired growth and neurodevelopment. The fatty acid contents of parenteral and enteral nutrition are insufficient to meet current recommendations. This chapter summarizes the regulatory roles of fatty acids, current recommendations and limitations of parenteral and enteral nutrition in meeting these recommendations in preterm infants. Suggested areas for research on the roles of fatty acids in preterm infant health are also provided.
Collapse
|
45
|
Hay SM, McArdle HJ, Hayes HE, Stevens VJ, Rees WD. The effect of iron deficiency on the temporal changes in the expression of genes associated with fat metabolism in the pregnant rat. Physiol Rep 2016; 4:4/21/e12908. [PMID: 27905292 PMCID: PMC5112487 DOI: 10.14814/phy2.12908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 12/22/2022] Open
Abstract
Iron is essential for the oxidative metabolism of lipids. Lipid metabolism changes during gestation to meet the requirements of the growing fetus and to prepare for lactation. The temporal effects of iron deficiency during gestation were studied in female rats fed complete or iron‐deficient diets. Plasma triglycerides were elevated in the iron‐deficient group throughout gestation. There were time‐dependent changes in the triglyceride content of the maternal liver, falling at the midpoint of gestation and then increasing on d21.5. Compared to the control, triglycerides in the maternal liver were not different in the iron‐deficient group prior to pregnancy and on d12.5, but were markedly reduced by d21.5. The abundance of mRNAs in the maternal liver suggests that lipogenesis is unchanged and beta‐oxidation is reduced on d21.5 by iron deficiency. On d21.5 of gestation, the expression of placental lipase was unchanged by iron deficiency, however, the abundance of mRNAs for SREBP‐1c, FABP4 were reduced, suggesting that there were changes in fatty acid handling. In the fetal liver, iron deficiency produced a marked decrease in the abundance of the L‐CPT‐1 mRNA, suggesting that beta‐oxidation is reduced. This study shows that the major effect of iron deficiency on maternal lipid metabolism occurs late in gestation and that perturbed lipid metabolism may be a common feature of models of fetal programming.
Collapse
Affiliation(s)
- Susan M Hay
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, U.K
| | - Harry J McArdle
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, U.K
| | - Helen E Hayes
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, U.K
| | - Valerie J Stevens
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, U.K
| | - William D Rees
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, U.K.
| |
Collapse
|
46
|
Maternal obesity modulates intracellular lipid turnover in the human term placenta. Int J Obes (Lond) 2016; 41:317-323. [PMID: 27780978 PMCID: PMC5309341 DOI: 10.1038/ijo.2016.188] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/22/2016] [Accepted: 09/25/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Obesity before pregnancy is associated with impaired metabolic status of the mother and the offspring later in life. These adverse effects have been attributed to epigenetic changes in utero, but little is known about the role of placental metabolism and its contribution to fetal development. OBJECTIVES We examined the impact of maternal pre-pregnancy obesity on the expression of genes involved in placental lipid metabolism in lean and obese women. SUBJECTS/METHODS Seventy-three lean and obese women with healthy pregnancy were recruited at term elective cesarean delivery. Metabolic parameters were measured on maternal venous blood samples. Expression of 88 genes involved in lipid metabolism was measured in whole placenta tissue. Proteins of genes differently expressed in response to maternal obesity were quantified, correlated with maternal parameters and immunolocalized in placenta sections. Isolated primary trophoblasts were used for in vitro assays. RESULTS Triglyceride (TG) content was increased in placental tissue of obese (1.10, CI 1.04-1.24 mg g-1, P<0.05) vs lean (0.84, CI 0.72-1.02 mg g-1) women. Among target genes examined, six showed positive correlation (P<0.05) with maternal pre-pregnancy BMI, namely ATGL (PNPLA2), FATP1 (SLC27A1), FATP3 (SLC27A3), PLIN2, PPARG and CGI-58 (ABHD5). CGI-58 protein abundance was twofold higher (P<0.001) in placentas of obese vs lean women. CGI-58 protein levels correlated positively with maternal insulin levels and pre-pregnancy body mass index (R=0.63, P<0.001 and R=0.64, P<0.001, respectively). CGI-58 and PLIN2 were primarily located in the syncytiotrophoblast and, were upregulated (1.38- and 500-fold, respectively) upon oleic acid and insulin treatment of cultured trophoblast cells. CONCLUSION Pre-gravid obesity significantly modifies the expression of placental genes related to transport and storage of neutral lipids. We propose that the upregulation of CGI-58, a master regulator of TG hydrolysis, contributes to the turnover of intracellular lipids in placenta of obese women, and is tightly regulated by metabolic factors of the mother.
Collapse
|
47
|
Berti C, Cetin I, Agostoni C, Desoye G, Devlieger R, Emmett PM, Ensenauer R, Hauner H, Herrera E, Hoesli I, Krauss-Etschmann S, Olsen SF, Schaefer-Graf U, Schiessl B, Symonds ME, Koletzko B. Pregnancy and Infants' Outcome: Nutritional and Metabolic Implications. Crit Rev Food Sci Nutr 2016; 56:82-91. [PMID: 24628089 DOI: 10.1080/10408398.2012.745477] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pregnancy is a complex period of human growth, development, and imprinting. Nutrition and metabolism play a crucial role for the health and well-being of both mother and fetus, as well as for the long-term health of the offspring. Nevertheless, several biological and physiological mechanisms related to nutritive requirements together with their transfer and utilization across the placenta are still poorly understood. In February 2009, the Child Health Foundation invited leading experts of this field to a workshop to critically review and discuss current knowledge, with the aim to highlight priorities for future research. This paper summarizes our main conclusions with regards to maternal preconceptional body mass index, gestational weight gain, placental and fetal requirements in relation to adverse pregnancy and long-term outcomes of the fetus (nutritional programming). We conclude that there is an urgent need to develop further human investigations aimed at better understanding of the basis of biochemical mechanisms and pathophysiological events related to maternal-fetal nutrition and offspring health. An improved knowledge would help to optimize nutritional recommendations for pregnancy.
Collapse
Affiliation(s)
- C Berti
- a Unit of Obstetrics & Gynecology, Department of Biomedical and Clinical Sciences, Hospital 'L. Sacco', and Center for Fetal Research Giorgio Pardi , University of Milan , Milan , Italy
| | - I Cetin
- a Unit of Obstetrics & Gynecology, Department of Biomedical and Clinical Sciences, Hospital 'L. Sacco', and Center for Fetal Research Giorgio Pardi , University of Milan , Milan , Italy.,b Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - C Agostoni
- b Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - G Desoye
- c Department of Obstetrics & Gynaecology , Medical University of Graz , Graz , Austria
| | - R Devlieger
- d Department of Obstetrics & Gynaecology , University Hospitals K.U. Leuven , Leuven , Belgium
| | - P M Emmett
- e Centre for Child & Adolescent Health, School of Social & Community Medicine , University of Bristol , Bristol , United Kingdom
| | - R Ensenauer
- f Research Center, Dr. von Hauner Children's Hospital, Klinikum der Ludwig-Maximilians-Universität München , Munich , Germany
| | - H Hauner
- g Else Kroener-Fresenius-Centre for Nutritional Medicine, Klinikum rechts der Isar , Technical University of Munich , Munich , Germany
| | - E Herrera
- h Unit of Biochemistry & Molecular Biology, Universidad San Pablo CEU , Madrid , Spain
| | - I Hoesli
- i Department of Obstetrics and Gynaecology , University Hospital of Basel , Basel , Switzerland
| | - S Krauss-Etschmann
- j Comprehensive Pneumology Center, Ludwig Maximilians University Hospital , Munich , Germany.,k Helmholtz Zentrum München , Munich , Germany
| | - S F Olsen
- l Centre for Fetal Programming, Statens Serum Institut , Copenhagen , Denmark
| | - U Schaefer-Graf
- m Department of Obstetrics & Gynecology , Berlin Center for Diabetes & Pregnancy, St. Joseph Hospital , Berlin , Germany
| | - B Schiessl
- n Fetal Maternal Unit, Department of Obstetrics & Gynecology , University of Munich , Munich , Germany
| | - M E Symonds
- o The Early Life Nutrition Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital of Nottingham , Nottingham , United Kingdom
| | - B Koletzko
- p Dr. von Hauner Children's Hospital , University of Munich Medical Centre , Munich , Germany
| |
Collapse
|
48
|
Lipoprotein lipase and lipid profiles in plasma and placenta from normal pregnancies compared with patients with intrahepatic cholestasis of pregnancy. Eur J Obstet Gynecol Reprod Biol 2016; 203:279-85. [PMID: 27400425 DOI: 10.1016/j.ejogrb.2016.06.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 05/05/2016] [Accepted: 06/21/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To analyse lipoprotein lipase (LPL) expression and lipid levels in placenta and plasma of patients with intrahepatic cholestasis of pregnancy (ICP) and normal pregnancies. METHODS This prospective study included 30 patients with ICP and 30 gestational-age-matched pregnancies without any complications. Enzyme-linked immunosorbent assays were used to investigate plasma LPL levels from 28 weeks of gestation, at 4-weekly intervals, to 38 weeks of gestation, and data were assessed longitudinally. Immunohistochemistry, Western blotting and real-time polymerase chain reaction were used to detect placental LPL expression and activity. Placental triglyceride and total cholesterol levels were also analysed. The clinical data related to ICP and lipid profiles were collected retrospectively. RESULTS Plasma LPL concentration increased with gestational age in both groups, but the increase was limited in the ICP group. Immunohistochemistry revealed LPL staining mainly in syncytiotrophoblasts, and 3,3'-diamino-benzidine tetrahydrochloride wt% was lower in ICP placenta compared with normal placenta (p<0.01). LPL protein and mRNA expression in ICP placenta were significantly lower than in normal placenta (p<0.01). LPL activity was not significantly different in both groups. Correlation analysis indicated that the plasma LPL level was negatively associated with the corresponding concentration of total bile acid (r=-0.57) in the ICP group. CONCLUSION Reduced LPL expression in placenta, limited increase in LPL level in maternal plasma, and abnormal lipid profiles were found in patients with ICP. LPL was possibly related to ICP by participating abnormal lipid metabolism.
Collapse
|
49
|
Kolahi K, Louey S, Varlamov O, Thornburg K. Real-Time Tracking of BODIPY-C12 Long-Chain Fatty Acid in Human Term Placenta Reveals Unique Lipid Dynamics in Cytotrophoblast Cells. PLoS One 2016; 11:e0153522. [PMID: 27124483 PMCID: PMC4849650 DOI: 10.1371/journal.pone.0153522] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/30/2016] [Indexed: 01/10/2023] Open
Abstract
While the human placenta must provide selected long-chain fatty acids to support the developing fetal brain, little is known about the mechanisms underlying the transport process. We tracked the movement of the fluorescently labeled long-chain fatty acid analogue, BODIPY-C12, across the cell layers of living explants of human term placenta. Although all layers took up the fatty acid, rapid esterification of long-chain fatty acids and incorporation into lipid droplets was exclusive to the inner layer cytotrophoblast cells rather than the expected outer syncytiotrophoblast layer. Cytotrophoblast is a progenitor cell layer previously relegated to a repair role. As isolated cytotrophoblasts differentiated into syncytialized cells in culture, they weakened their lipid processing capacity. Syncytializing cells suppress previously active genes that regulate fatty-acid uptake (SLC27A2/FATP2, FABP4, ACSL5) and lipid metabolism (GPAT3, LPCAT3). We speculate that cytotrophoblast performs a previously unrecognized role in regulating placental fatty acid uptake and metabolism.
Collapse
Affiliation(s)
- Kevin Kolahi
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, United States of America
- Center for Developmental Health, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Samantha Louey
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Oleg Varlamov
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Kent Thornburg
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, United States of America
- Center for Developmental Health, Oregon Health and Science University, Portland, Oregon, United States of America
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
50
|
Kwon SG, Hwang JH, Park DH, Kim TW, Kang DG, Kang KH, Kim IS, Park HC, Na CS, Ha J, Kim CW. Identification of Differentially Expressed Genes Associated with Litter Size in Berkshire Pig Placenta. PLoS One 2016; 11:e0153311. [PMID: 27078025 PMCID: PMC4831801 DOI: 10.1371/journal.pone.0153311] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 03/28/2016] [Indexed: 01/24/2023] Open
Abstract
Improvement in litter size has become of great interest in the pig industry because fecundity is directly related to sow reproductive life. Improved reproduction has thus been achieved by elucidating the molecular functions of genes associated with fecundity. In the present study, we identified differentially expressed genes (DEGs) via transcriptomic analysis using RNA-sequencing (RNA-Seq) in Berkshire pig placentas from larger (LLG, mean litter size >12) and smaller (SLG, mean litter size < 6.5) litter size groups. In total 588 DEGs were identified (p < 0.05, > 1.5-fold change), of which 98 were upregulated, while 490 were downregulated in the LLG compared with the SLG. Gene Ontology (GO) enrichment was also performed. We concluded that 129 of the 588 DEGs were closely related to litter size according to reproduction related genes selected based on previous reports, as 110 genes were downregulated and 19 upregulated in the LLG compared with the SLG. RT-qPCR utilizing specific primers targeting the early growth response 2 (EGR2), pheromaxein c subunit (PHEROC) and endothelial lipase (LIPG) genes showed high accordance with RNA-Seq results. Furthermore, we investigated the upstream regulators of these three genes in the placenta. We found that WNT9B, a Wnt signaling pathway molecule, and IL-6, known inducers of EGR2 and LIPG, respectively, were significantly increased in LLG compared with SLG. We believe that the induction of IL-6 and LIPG may play an important role in increasing nutrition supply through the placenta from the sow to the piglet during gestation. These results provide novel molecular insights into pig reproduction.
Collapse
Affiliation(s)
- Seul Gi Kwon
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Jung Hye Hwang
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Da Hye Park
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Tae Wan Kim
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Deok Gyeong Kang
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Kyung Hee Kang
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Il-Suk Kim
- Department of Animal Resource Technology, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | | | - Chong-Sam Na
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, South Korea
| | - Jeongim Ha
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
- * E-mail: (JH); (CWK)
| | - Chul Wook Kim
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
- * E-mail: (JH); (CWK)
| |
Collapse
|