1
|
Pastor FM, de Melo Ocarino N, Silva JF, Reis AMS, Serakides R. Bone development in fetuses with intrauterine growth restriction caused by maternal endocrine-metabolic dysfunctions. Bone 2024; 186:117169. [PMID: 38880170 DOI: 10.1016/j.bone.2024.117169] [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: 03/28/2024] [Revised: 05/21/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Intrauterine growth restriction (IUGR) affects a large proportion of infants, particularly in underdeveloped countries. Among the main causes of IUGR, maternal endocrine-metabolic dysfunction is highlighted, either due to its high incidence or due to the severity of the immediate and mediated changes that these dysfunctions cause in the fetus and the mother. Although the effects of endocrine and metabolic disorders have been widely researched, there are still no reviews that bring together and summarize the effects of these conditions on bone development in cases of IUGR. Therefore, the present literature review was conducted with the aim of discussing bone changes observed in fetuses with IUGR caused by maternal endocrine-metabolic dysfunction. The main endocrine dysfunctions that occur with IUGR include maternal hyperthyroidism, hypothyroidism, and hypoparathyroidism. Diabetes mellitus, hypertensive disorders, and obesity are the most important maternal metabolic dysfunctions that compromise fetal growth. The bone changes reported in the fetus are, for the most part, due to damage to cell proliferation and differentiation, as well as failures in the synthesis and mineralization of the extracellular matrix, which results in shortening and fragility of the bones. Some maternal dysfunctions, such as hyperthyroidism, have been widely studied, whereas conditions such as hypoparathyroidism and gestational hypertensive disorders require further study regarding the mechanisms underlying the development of bone changes. Similarly, there is a gap in the literature regarding changes related to intramembranous ossification, as most published articles only describe changes in endochondral bone formation associated with IUGR. Furthermore, there is a need for more research aimed at elucidating the late postnatal changes that occur in the skeletons of individuals affected by IUGR and their possible relationships with adult diseases, such as osteoarthritis and osteoporosis.
Collapse
Affiliation(s)
- Felipe Martins Pastor
- Departamento de Cínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Natália de Melo Ocarino
- Departamento de Cínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, Bahia, Brazil
| | - Amanda Maria Sena Reis
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Rogéria Serakides
- Departamento de Cínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
2
|
Zhu M, Sun R, Jin L, Yu D, Huang X, Zhu T, Gong Y, Chen Y, Shi J, Wang Q, Lu C, Wang D. Metabolomics profiling of maternal and umbilical cord blood in normoglycemia macrosomia. J Matern Fetal Neonatal Med 2023; 36:2270761. [PMID: 37848386 DOI: 10.1080/14767058.2023.2270761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023]
Abstract
Background: Macrosomia is a common disorder that occurs during pregnancy. We investigated the comprehensive metabolite profiles of pregnant maternal and fetal sera in normoglycemic macrosomia in a Chinese population. Methods: Forty pregnant women and their fetuses were included in the study (twenty macrosomia patients and twenty normal-weight controls). Maternal and umbilical cord serum metabolites were identified using ultra-performance liquid chromatography coupled with tandem mass spectrometry. Results: In total, 203 metabolites were identified. Lipids and lipid-like molecules were the predominant metabolites. Fifty-three metabolites with significant differences were obtained in the maternal samples. In the macrosomia group, the levels of docosahexaenoic acid, eicosapentaenoic acid, and arachidonic acid were significantly higher than those in the control group. Umbilical cord serum samples were obtained for 24 different metabolites. The maternal-fetal gradient of polyunsaturated fatty acids was decreased in the macrosomia group. Aconitic acid, citric acid, isocitric acid, 2-methylhexanoic acid, and 12-hydroxystearic acid were the common differential metabolites in the maternal and umbilical cord serum samples. Conclusion: There were obvious metabolic abnormalities in the sera of pregnant women and fetuses with macrosomia. Lipids and lipid-like molecules were the predominant differential metabolites but had different classifications in the maternal and umbilical cord serum. These results may provide new insights into the long-term metabolic disorders associated with macrosomia.
Collapse
Affiliation(s)
- Mianmian Zhu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Rongyue Sun
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Lixu Jin
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Dandan Yu
- Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Xiaoxia Huang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Ting Zhu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Yujing Gong
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Yuan Chen
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Jiamin Shi
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Qiu Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Chaosheng Lu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Dan Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| |
Collapse
|
3
|
Sun B, Reynolds KS, Garland MA, McMahon M, Saha SK, Zhou CJ. Epigenetic implications in maternal diabetes and metabolic syndrome-associated risk of orofacial clefts. Birth Defects Res 2023; 115:1835-1850. [PMID: 37497595 PMCID: PMC11526419 DOI: 10.1002/bdr2.2226] [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: 04/03/2023] [Revised: 06/12/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023]
Abstract
Orofacial clefts (OFCs) are one of the most common types of structural birth defects. The etiologies are complicated, involving with genetic, epigenetic, and environmental factors. Studies have found that maternal diabetes and metabolic syndrome are associated with a higher risk of OFCs in offspring. Metabolic syndrome is a clustering of several disease risk factors, including hyperglycemia, dyslipidemia, obesity, and hypertension. Metabolic disease during pregnancy can increase risk of adverse outcomes and significantly influence fetal development, including orofacial formation and fusion. An altered metabolic state may contribute to developmental disorders or congenital defects including OFCs, potentially through epigenetic modulations, such as histone modification, DNA methylation, and noncoding RNA expression to alter activities of critical morphogenetic signaling or related developmental genes. This review summarizes the currently available evidence and underlying mechanisms of how the maternal metabolic syndrome is associated with OFCs in mostly human and some animal studies. It may provide a better understanding of the interactions between intrauterine metabolic status and fetal orofacial development which might be applied toward prevention and treatments of OFCs.
Collapse
Affiliation(s)
- Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Kurt S. Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Michael A. Garland
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Moira McMahon
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Subbroto K. Saha
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Chengji J. Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| |
Collapse
|
4
|
Ng DZW, Lee SXY, Ooi DSQ, Ta LDH, Yap GC, Tay CJX, Huang CH, Tham EH, Loo EXL, Shek LPC, Goh A, Bever HPSV, Teoh OH, Lee YS, Yap F, Tan KH, Chong YS, Chan SY, Eriksson JG, Godfrey KM, Lee BW, Chan ECY. Sensitive LC-MS/MS method for the temporal profiling of bile acids, fatty acids and branched-chain alpha-keto acids in maternal plasma during pregnancy and cord blood plasma at delivery. Clin Chim Acta 2023:117449. [PMID: 37331549 DOI: 10.1016/j.cca.2023.117449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/22/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND AND AIMS There are significant changes to the maternal inflammatory profile across pregnancy. Recent studies suggest that perturbations in maternal gut microbial and dietary-derived plasma metabolites over the course of pregnancy mediate inflammation through a complex interplay of immunomodulatory effects. Despite this body of evidence, there is currently no analytical method that is suitable for the simultaneous profiling of these metabolites within human plasma. MATERIALS AND METHODS We developed a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the high-throughput analysis of these metabolites in human plasma without derivatization. Plasma samples were processed using liquid-liquid extraction method with varying proportions of methyl tert-butyl ether, methanol, and water in a 3:10:2.5 ratio to reduce matrix effects. RESULTS LC-MS/MS detection was sufficiently sensitive to quantify these gut microbial and dietary-derived metabolites at physiological concentrations and linear calibration curves with r2>0.99 were obtained. Recovery was consistent across concentration levels. Stability experiments confirmed that up to 160 samples could be analyzed within a single batch. The method was validated and applied to analyse maternal plasma during the first and third trimester and cord blood plasma of 5 mothers. CONCLUSION This study validated a straightforward and sensitive LC/MS-MS method for the simultaneous quantitation of gut microbial and dietary-derived metabolites in human plasma within 9 minutes without prior sample derivatization.
Collapse
Affiliation(s)
- Daniel Zhi Wei Ng
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | - Sean Xian Yu Lee
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | - Delicia Shu Qin Ooi
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228
| | - Le Duc Huy Ta
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228
| | - Gaik Chin Yap
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228
| | - Carina Jing Xuan Tay
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228
| | - Chiung-Hui Huang
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228
| | - Elizabeth Huiwen Tham
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228; Division of Allergy & Immunology, Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Evelyn Xiu Ling Loo
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Lynette P C Shek
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anne Goh
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Hugo P S Van Bever
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Oon Hoe Teoh
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yung Seng Lee
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Division of Paediatric Endocrinology and Diabetes, Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Fabian Yap
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Kok Hian Tan
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Obstetrics & Gynaecology, National University of Singapore, Singapore
| | - Shiao Yng Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Obstetrics & Gynaecology, National University of Singapore, Singapore
| | - Johan Gunnar Eriksson
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Obstetrics & Gynaecology, National University of Singapore, Singapore; Folkhälsan Research Center, Helsinki, Finland and Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Bee Wah Lee
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228.
| | - Eric Chun Yong Chan
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543.
| |
Collapse
|
5
|
Stadler JT, Scharnagl H, Wadsack C, Marsche G. Preeclampsia Affects Lipid Metabolism and HDL Function in Mothers and Their Offspring. Antioxidants (Basel) 2023; 12:antiox12040795. [PMID: 37107170 PMCID: PMC10135112 DOI: 10.3390/antiox12040795] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
Preeclampsia (PE) is linked to an overall increased cardiovascular risk for both the mother and child. Functional impairment of high-density lipoproteins (HDL) may contribute to the excess cardiovascular risk associated with PE. In this study, we investigated the effects of PE on maternal and neonatal lipid metabolism, and the parameters of HDL composition and function. The study cohort included 32 normotensive pregnant women, 18 women diagnosed with early-onset PE, and 14 women with late-onset PE. In mothers, early- and late-onset PE was associated with atherogenic dyslipidemia, characterized by high plasma triglycerides and low HDL-cholesterol levels. We observed a shift from large HDL to smaller HDL subclasses in early-onset PE, which was associated with an increased plasma antioxidant capacity in mothers. PE was further associated with markedly increased levels of HDL-associated apolipoprotein (apo) C-II in mothers, and linked to the triglyceride content of HDL. In neonates of early-onset PE, total cholesterol levels were increased, whereas HDL cholesterol efflux capacity was markedly reduced in neonates from late-onset PE. In conclusion, early- and late-onset PE profoundly affect maternal lipid metabolism, potentially contributing to disease manifestation and increased cardiovascular risk later in life. PE is also associated with changes in neonatal HDL composition and function, demonstrating that complications of pregnancy affect neonatal lipoprotein metabolism.
Collapse
Affiliation(s)
- Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
6
|
Grupe K, Scherneck S. Mouse Models of Gestational Diabetes Mellitus and Its Subtypes: Recent Insights and Pitfalls. Int J Mol Sci 2023; 24:ijms24065982. [PMID: 36983056 PMCID: PMC10058162 DOI: 10.3390/ijms24065982] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is currently the most common complication of pregnancy and is defined as a glucose intolerance disorder with recognition during pregnancy. GDM is considered a uniform group of patients in conventional guidelines. In recent years, evidence of the disease's heterogeneity has led to a growing understanding of the value of dividing patients into different subpopulations. Furthermore, in view of the increasing incidence of hyperglycemia outside pregnancy, it is likely that many cases diagnosed as GDM are in fact patients with undiagnosed pre-pregnancy impaired glucose tolerance (IGT). Experimental models contribute significantly to the understanding of the pathogenesis of GDM and numerous animal models have been described in the literature. The aim of this review is to provide an overview of the existing mouse models of GDM, in particular those that have been obtained by genetic manipulation. However, these commonly used models have certain limitations in the study of the pathogenesis of GDM and cannot fully describe the heterogeneous spectrum of this polygenic disease. The polygenic New Zealand obese (NZO) mouse is introduced as a recently emerged model of a subpopulation of GDM. Although this strain lacks conventional GDM, it exhibits prediabetes and an IGT both preconceptionally and during gestation. In addition, it should be emphasized that the choice of an appropriate control strain is of great importance in metabolic studies. The commonly used control strain C57BL/6N, which exhibits IGT during gestation, is discussed in this review as a potential model of GDM.
Collapse
Affiliation(s)
- Katharina Grupe
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, Mendelssohnstraße 1, D-38106 Braunschweig, Germany
| | - Stephan Scherneck
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, Mendelssohnstraße 1, D-38106 Braunschweig, Germany
| |
Collapse
|
7
|
Zhang M, Brady TM, Buckley JP, Appel LJ, Hong X, Wang G, Liang L, Wang X, Mueller NT. Metabolome-Wide Association Study of Cord Blood Metabolites With Blood Pressure in Childhood and Adolescence. Hypertension 2022; 79:2806-2820. [PMID: 36111548 PMCID: PMC9649875 DOI: 10.1161/hypertensionaha.122.20139] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/28/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND No studies have examined whether the cord blood metabolome-a reflection of in utero metabolism-influences blood pressure (BP) in children. OBJECTIVES To examine prospective associations of cord blood metabolites with systolic BP (SBP), diastolic BP (DBP), and risk of elevated BP in childhood and adolescence. METHODS In the Boston Birth Cohort, we measured metabolites in cord blood plasma, and SBP and DBP at clinic visits between 3 and 18 years. We examined associations of cord metabolites with SBP and DBP percentiles using linear mixed models and with elevated BP using mixed-effects Poisson regression. RESULTS Our study included 902 mother-child dyads (60% Black, 23% Hispanic, 45% female). Children were followed for a median of 9.2 (interquartile range, 6.7-11.7) years, and the median number of BP observations per child was 7 (interquartile range, 4-11). After false discovery rate correction, 3 metabolites were associated with SBP, 96 with DBP, and 24 with elevated BP; 2 metabolites (1-methylnicotinamide, dimethylguanidino valeric acid) were associated with all 3 outcomes, and 21 metabolites were associated with both DBP and elevated BP. After multivariable adjustment, 48 metabolites remained significantly associated with DBP. Metabolites that showed the strongest associations with SBP, DBP, and elevated BP included nucleotides (eg, xanthosine, hypoxanthine, xanthine) and acylcarnitines (eg, C6 and C7 carnitines), which represent fatty acid oxidation and purine metabolism pathways. CONCLUSIONS In our urban and predominantly racial/ethnic minority cohort, we provide evidence that metabolomic alterations in utero, in particular, acylcarnitine- and purine-metabolism metabolites, may be involved in the early life origins of hypertension.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
| | - Tammy M Brady
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jessie P Buckley
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Lawrence J Appel
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
- Division of General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Johns Hopkins University, Baltimore, MD
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Johns Hopkins University, Baltimore, MD
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Xiaobin Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
- Center on the Early Life Origins of Disease, Johns Hopkins University, Baltimore, MD
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
8
|
Mirza FG, Tahlak MA, Rjeili RB, Hazari K, Ennab F, Hodgman C, Khamis AH, Atiomo W. Polycystic Ovarian Syndrome (PCOS): Does the Challenge End at Conception? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph192214914. [PMID: 36429632 PMCID: PMC9690374 DOI: 10.3390/ijerph192214914] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 05/14/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent condition that not only has the potential to impede conception but also represents the most common endocrine dysfunction in fertile women. It is considered a heterogeneous and multifaceted disorder, with multiple reproductive and metabolic phenotypes which differently affect the early- and long-term syndrome's risks. Undoubtedly, the impact of PCOS on infertility has attracted most of the attention of healthcare providers and investigators. However, there is growing evidence that even after conception is achieved, PCOS predisposes the parturient to several adverse pregnancy outcomes including a high risk of pregnancy-induced hypertension, spontaneous abortion, gestational diabetes, preeclampsia, and preterm birth, which increase the risks of stillbirth and neonatal death. Fetal growth abnormalities may also be more common, but the relationship is less well defined. This narrative review aims to summarize current knowledge regarding these conditions as they interplay with PCOS and concludes that although there appears to be an increase in these complications during the pregnancy of women with PCOS, there is a need for further research to clarify the possible confounding impact of obesity. Implications for clinical practice and future research are outlined.
Collapse
Affiliation(s)
- Fadi G. Mirza
- Latifa Women and Children Hospital, Dubai P.O. Box 9115, United Arab Emirates
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
- Department of Obstetrics and Gynaecology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Muna A. Tahlak
- Latifa Women and Children Hospital, Dubai P.O. Box 9115, United Arab Emirates
| | - Rachelle Bou Rjeili
- Faculty of Medicine, American University of Beirut, Beruit P.O. Box 11-0236, Lebanon
| | - Komal Hazari
- Latifa Women and Children Hospital, Dubai P.O. Box 9115, United Arab Emirates
| | - Farah Ennab
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
| | - Charlie Hodgman
- School of Biosciences, University of Nottingham, Loughborough LE12 5RD, UK
| | - Amar Hassan Khamis
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
| | - William Atiomo
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
- Correspondence:
| |
Collapse
|
9
|
Östling H, Lodefalk M, Backman H, Kruse R. Global microRNA and protein expression in human term placenta. Front Med (Lausanne) 2022; 9:952827. [PMID: 36330066 PMCID: PMC9622934 DOI: 10.3389/fmed.2022.952827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Description of the global expression of microRNAs (miRNAs) and proteins in healthy human term placentas may increase our knowledge of molecular biological pathways that are important for normal fetal growth and development in term pregnancy. The aim of this study was to explore the global expression of miRNAs and proteins, and to point out functions of importance in healthy term placentas. Materials and methods Placental samples (n = 19) were identified in a local biobank. All samples were from uncomplicated term pregnancies with vaginal births and healthy, normal weight newborns. Next-generation sequencing and nano-scale liquid chromatographic tandem mass spectrometry were used to analyse miRNA and protein expression, respectively. Results A total of 895 mature miRNAs and 6,523 proteins were detected in the placentas, of which 123 miRNAs and 346 proteins were highly abundant. The miRNAs were in high degree mapped to chromosomes 19, 14, and X. Analysis of the highly abundant miRNAs and proteins showed several significantly predicted functions in common, including immune and inflammatory response, lipid metabolism and development of the nervous system. Discussion The predicted function inflammatory response may reflect normal vaginal delivery, while lipid metabolism and neurodevelopment may be important processes for the term fetus. The data presented in this study, with complete miRNA and protein findings, will enhance the knowledge base for future research in the field of placental function and pathology.
Collapse
Affiliation(s)
- Hanna Östling
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- *Correspondence: Hanna Östling,
| | - Maria Lodefalk
- Department of Paediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- University Health Care Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Helena Backman
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Robert Kruse
- iRiSC - Inflammatory Response and Infection Susceptibility Centre, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
10
|
Ponsuksili S, Murani E, Hadlich F, Iqbal MA, Fuchs B, Galuska CE, Perdomo-Sabogal A, Sarais F, Trakooljul N, Reyer H, Oster M, Wimmers K. Prenatal transcript levels and metabolomics analyses reveal metabolic changes associated with intrauterine growth restriction and sex. Open Biol 2022; 12:220151. [PMID: 36102059 PMCID: PMC9471991 DOI: 10.1098/rsob.220151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The metabolic changes associated with intrauterine growth restriction (IUGR) particularly affect the liver, which is a central metabolic organ and contributes significantly to the provision of energy and specific nutrients and metabolites. Therefore, our aim was to decipher and elucidate the molecular pathways of developmental processes mediated by miRNAs and mRNAs, as well as the metabolome in fetal liver tissue in IUGR compared to appropriate for gestational age groups (AGA). Discordant siblings representing the extremes in fetal weight at day 63 post conception (dpc) were selected from F2 fetuses of a cross of German Landrace and Pietrain. Most of the changes in the liver of IUGR at midgestation involved various lipid metabolic pathways, both on transcript and metabolite levels, especially in the category of sphingolipids and phospholipids. Differentially expressed miRNAs, such as miR-34a, and their differentially expressed mRNA targets were identified. Sex-specific phenomena were observed at both the transcript and metabolite levels, particularly in male. This suggests that sex-specific adaptations in the metabolic system occur in the liver during midgestation (63 dpc). Our multi-omics network analysis reveals interactions and changes in the metabolic system associated with IUGR and identified an important biosignature that differs between IUGR and AGA piglets.
Collapse
Affiliation(s)
- Siriluck Ponsuksili
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Eduard Murani
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Frieder Hadlich
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Muhammad Arsalan Iqbal
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Beate Fuchs
- Research Institute for Farm Animal Biology (FBN), Core Facility Metabolomics, 18196 Dummerstorf, Germany
| | - Christina E Galuska
- Research Institute for Farm Animal Biology (FBN), Core Facility Metabolomics, 18196 Dummerstorf, Germany
| | - Alvaro Perdomo-Sabogal
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Fabio Sarais
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Nares Trakooljul
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Henry Reyer
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Michael Oster
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.,Faculty of Agricultural and Environmental Sciences, University Rostock, 18059 Rostock, Germany
| |
Collapse
|
11
|
Heras-Molina A, Escudero R, Pesántez-Pacheco JL, García-Contreras C, Vázquez-Gómez M, Astiz S, Óvilo C, González-Bulnes A, Isabel B. Maternal Supplementation with Polyphenols and Omega-3 Fatty Acids during Pregnancy: Prenatal Effects on Fetal Fatty Acid Composition in the Iberian Pig. Animals (Basel) 2022; 12:ani12162140. [PMID: 36009731 PMCID: PMC9405247 DOI: 10.3390/ani12162140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary The present study aimed to determine the effects of maternal dietary supplementation combining hydroxytyrosol and n3 polyunsaturated fatty acids (n3-PUFA) from day 35 to day 100 of gestation on the fatty acid (FA) composition of the offspring tissues of the Iberian pig. No effects were found in the plasma FA composition of the dams but higher levels of n3-PUFA were found in the plasma and different tissues (muscle, liver, and brain) of the supplemented fetuses. These findings may have important implications for piglets’ health and may offer guidance for achieving human dietary n3-PUFA recommendations. Abstract Intrauterine Growth Restriction (IUGR) is a major problem in pig production and different strategies, mainly maternal supplementation with different agents, are currently being studied. The combination of hydroxytyrosol and n3-PUFA seems to be a promising treatment to counteract IUGR, since the combination may help improve n3-PUFA composition and lower the inflammatory status of IUGR piglets. The aim of the present study is to determine the effects of a maternal supplementation, from day 35 to day 100 of pregnancy, with linseed oil and hydroxytyrosol on the fetal FA composition. The results showed higher n3 levels, including eicosapentaenoic and docosahexaenoic FA in the offspring from treated gilts, which showed lower n6-PUFA/n3-PUFA (n6/n3) ratios. Saturated and monounsaturated fatty acids were also affected by treatment, especially in the muscle and brain. Thus, a maternal supplementation with linseed oil and hydroxytyrosol affected the fetal FA tissue composition, which could have implications in pig production due to the improvement of the piglets’ health status.
Collapse
Affiliation(s)
- Ana Heras-Molina
- CSIC-INIA, Ctra. De La Coruña Km. 7.5, 28040 Madrid, Spain
- Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Rosa Escudero
- Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - José L. Pesántez-Pacheco
- CSIC-INIA, Ctra. De La Coruña Km. 7.5, 28040 Madrid, Spain
- School of Veterinary Medicine and Zootechnics, Faculty of Agricultural Sciences, University of Cuenca, Avda. Doce de Octubre, Cuenca 010220, Ecuador
| | | | - Marta Vázquez-Gómez
- Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Nutrition and Obesities: Systemic Approaches Research Unit (NutriOmics), INSERM, Sorbonne Université, 75006 Paris, France
| | - Susana Astiz
- CSIC-INIA, Ctra. De La Coruña Km. 7.5, 28040 Madrid, Spain
| | - Cristina Óvilo
- CSIC-INIA, Ctra. De La Coruña Km. 7.5, 28040 Madrid, Spain
| | - Antonio González-Bulnes
- CSIC-INIA, Ctra. De La Coruña Km. 7.5, 28040 Madrid, Spain
- Departamento de Producción y Sanidad Animal, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, C/Tirant lo Blanc, 7. Alfara del Patriarca, 46115 Valencia, Spain
- Correspondence: (A.G.-B.); (B.I.)
| | - Beatriz Isabel
- Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Correspondence: (A.G.-B.); (B.I.)
| |
Collapse
|
12
|
Burdge GC. α-linolenic acid interconversion is sufficient as a source of longer chain ω-3 polyunsaturated fatty acids in humans: An opinion. Lipids 2022; 57:267-287. [PMID: 35908848 DOI: 10.1002/lipd.12355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 01/20/2023]
Abstract
α-linolenic acid (αLNA) conversion into the functionally important ω-3 polyunsaturated fatty acids (PUFA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), has been regarded as inadequate for meeting nutritional requirements for these PUFA. This view is based on findings of small αLNA supplementation trials and stable isotope tracer studies that have been interpreted as indicating human capacity for EPA and, in particular, DHA synthesis is limited. The purpose of this review is to re-evaluate this interpretation. Markedly differing study designs, inconsistent findings and lack of trial replication preclude robust consensus regarding the nutritional adequacy of αLNA as a source of EPC and DHA. The conclusion that αLNA conversion in humans is constrained is inaccurate because it presupposes the existence of an unspecified, higher level of metabolic activity. Since capacity for EPA and DHA synthesis is the product of evolution it may be argued that the levels of EPA and DHA it maintains are nutritionally appropriate. Dietary and supra-dietary EPA plus DHA intakes confer health benefits. Paradoxically, such health benefits are also found amongst vegetarians who do not consume EPA and DHA, and for whom αLNA conversion is the primary source of ω-3 PUFA. Since there are no reported adverse effects on health or cognitive development of diets that exclude EPA and DHA, their synthesis from αLNA appears to be nutritionally adequate. This is consistent with the dietary essentiality of αLNA and has implications for developing sustainable nutritional recommendations for ω-3 PUFA.
Collapse
Affiliation(s)
- Graham C Burdge
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
13
|
Satokar VV, Vickers MH, Reynolds CM, Ponnampalam AP, Firth EC, Garg ML, Bridge-Comer PE, Cutfield WS, Albert BB. Toxicity of oxidized fish oil in pregnancy - a dose response study in rats. Am J Physiol Regul Integr Comp Physiol 2022; 323:R244-R254. [PMID: 35726870 DOI: 10.1152/ajpregu.00042.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Fish oil (FO) supplements are consumed during pregnancy to increase dietary omega-3. However, FO is often oxidized past recommended limits. In rats, a large dose of highly oxidized FO substantially increased newborn mortality, but the effects of human-relevant doses of less oxidized oil are unknown. A dose-response study in rats was conducted to estimate the safe level of oxidation during pregnancy. METHODOLOGY Sprague-Dawley rat dams were mated, then individually housed and provided with a gel treatment on each day of pregnancy. Treatment groups differed only in the FO content of the gel; control (no oil), PV5, PV10, and PV40 (0.05ml of FO oxidized to a peroxide value (PV) of 5, 10, or 40meq/kg), or PV40(1ml) (1ml of PV40). A subset of dams was culled on gestational day 20 to enable sampling, and the remainder were allowed to give birth. Newborn mortality was recorded. Offspring were sampled at postnatal days 2 and 21, and dams at day 21. RESULTS There were no signs of unwellness during pregnancy. However, there was markedly increased neonatal mortality affecting the PV40(1ml) (12.8%) and PV40 (6.3%) groups, but not the control, PV5, or PV10 groups (1-1.4%). Dietary oxidized FO altered the expression of placental genes involved in antioxidant pathways and the production of free radicals. Conclusions Highly oxidized FO was toxic in rat pregnancy leading to a marked increase in mortality even at a human-relevant dose. We observed no toxic effects of FOs with PV≤ 10meq/kg, suggesting that this is an appropriate maximum limit.
Collapse
Affiliation(s)
- Vidit V Satokar
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Clare M Reynolds
- Liggins Institute, University of Auckland, Auckland, New Zealand.,University College Dublin Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Anna P Ponnampalam
- Department of Physiology and Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | | | - Manohar L Garg
- Nutraceuticals Research Program, School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia
| | | | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand.,A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand
| | - Benjamin B Albert
- Liggins Institute, University of Auckland, Auckland, New Zealand.,A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Liebmann M, Asuaje Pfeifer M, Grupe K, Scherneck S. Estradiol (E2) Improves Glucose-Stimulated Insulin Secretion and Stabilizes GDM Progression in a Prediabetic Mouse Model. Int J Mol Sci 2022; 23:ijms23126693. [PMID: 35743136 PMCID: PMC9223537 DOI: 10.3390/ijms23126693] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 01/27/2023] Open
Abstract
Female New Zealand obese (NZO) mice are an established model of preconceptional (pc.) prediabetes that progresses as gestational diabetes mellitus (GDM) during gestation. It is known that NZO mice show improvement in insulin sensitivity and glucose-stimulated insulin secretion (GSIS) during gestation in vivo. The latter is no longer detectable in ex vivo perifusion experiments in isolated islets of Langerhans, suggesting a modulation by extrapancreatic factors. Here, we demonstrated that plasma 17β-estradiol (E2) levels increased markedly in NZO mice during gestation. The aim of this work was to determine whether these increased E2 levels are responsible for the improvement in metabolism during gestation. To achieve this goal, we examined its effects in isolated islets and primary hepatocytes of both NZO and metabolically healthy NMRI mice. E2 increased GSIS in the islets of both strains significantly. Hepatic glucose production (HGP) failed to be decreased by insulin in NZO hepatocytes but was reduced by E2 in both strains. Hepatocytes of pregnant NZO mice showed significantly lower glucose uptake (HGU) compared with NMRI controls, whereby E2 stimulation diminished this difference. Hepatocytes of pregnant NZO showed reduced glycogen content, increased cyclic adenosine monophosphate (cAMP) levels, and reduced AKT activation. These differences were abolished after E2 stimulation. In conclusion, our data indicate that E2 stabilizes and prevents deterioration of the metabolic state of the prediabetic NZO mice. E2 particularly increases GSIS and improves hepatic glucose utilization to a lower extent.
Collapse
|
15
|
Effects of Low Protein-High Carbohydrate Diet during Early and Late Pregnancy on Respiratory Quotient and Visceral Adiposity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3878581. [PMID: 35432727 PMCID: PMC9010209 DOI: 10.1155/2022/3878581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 12/05/2022]
Abstract
Background Low Protein-High Carbohydrate (LPHC) diet during pregnancy is considered a nutritional and health problem related to the development of maternal metabolic alterations, such as fatty liver and obesity in the perinatal and postnatal period. It is known that increase in visceral adiposity tissue (VAT) modulates maternal metabolic rate, with the respiratory quotient (RQ) being a parameter related to that variable; however, it is unknown whether LPHC intake during pregnancy affects the VAT and the RQ. In this study, we examine if consumption of LPHC during pregnancy modifies the VAT and RQ in early and late periods of pregnancy. Methods This is a longitudinal and cross-sectional study with Wistar rats during gestation (G) (3, 8, 15, and 20) and nonpregnant rats. Rats were fed with a control diet with 63/18% carbohydrate/protein and an experimental diet with 79/6% carbohydrate/protein. We studied water and food consumption and metabolic parameters such as RQ and energy expenditure (EE), calculated by indirect calorimetry. In the cross-sectional study, we determined visceral fat, as well as the concentration of free fatty acids, insulin, glucose, and lipid profile in serum. Results Nonpregnant rats with LPHC intake decreased significantly in VAT (86%) and the RQ (18%); in pregnant rats in early (8G) and late pregnancy (15G) in LPHC diet, both parameters (VAT and RQ) (25%-92%) increased during light time. When comparing time points during pregnancy in the control and LPHC groups, the RQ increased in 15G during daytime compared to 8G during the night period (17 and 5%, respectively). In late pregnancy, LPHC intake and triacylglyceride levels increased and cholesterol and glucose decreased (45 and 26%, respectively), in comparison to nonpregnant rats. Conclusions LPHC intake in nonpregnant rats decreases the RQ and VAT. Interestingly, the opposite occurs in early pregnancy: the RQ and VAT increased, and this correlates with free fatty acid (FFA) levels. The increase in RQ and VAT during light time in early pregnancy increased mobilization of carbohydrate and protein metabolism. These results suggest that LPHC intake during pregnancy increases the glucose metabolism as a compensatory mechanism for energy needs in the fetus and the mother in early pregnancy.
Collapse
|
16
|
Human Placental Intracellular Cholesterol Transport: A Focus on Lysosomal and Mitochondrial Dysfunction and Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11030500. [PMID: 35326150 PMCID: PMC8944475 DOI: 10.3390/antiox11030500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
The placenta participates in cholesterol biosynthesis and metabolism and regulates exchange between the maternal and fetal compartments. The fetus has high cholesterol requirements, and it is taken up and synthesized at elevated rates during pregnancy. In placental cells, the major source of cholesterol is the internalization of lipoprotein particles from maternal circulation by mechanisms that are not fully understood. As in hepatocytes, syncytiotrophoblast uptake of lipoprotein cholesterol involves lipoprotein receptors such as low-density lipoprotein receptor (LDLR) and scavenger receptor class B type I (SR-BI). Efflux outside the cells requires proteins such as the ATP-binding cassette (ABC) transporters ABCA1 and ABCG1. However, mechanisms associated with intracellular traffic of cholesterol in syncytiotrophoblasts are mostly unknown. In hepatocytes, uptaken cholesterol is transported to acidic late endosomes (LE) and lysosomes (LY). Proteins such as Niemann–Pick type C 1 (NPC1), NPC2, and StAR related lipid transfer domain containing 3 (STARD3) are required for cholesterol exit from the LE/LY. These proteins transfer cholesterol from the lumen of the LE/LY into the LE/LY-limiting membrane and then export it to the endoplasmic reticulum, mitochondria, or plasma membrane. Although the production, metabolism, and transport of cholesterol in placental cells are well explored, there is little information on the role of proteins related to intracellular cholesterol traffic in placental cells during physiological or pathological pregnancies. Such studies would be relevant for understanding fetal and placental cholesterol management. Oxidative stress, induced by generating excess reactive oxygen species (ROS), plays a critical role in regulating various cellular and biological functions and has emerged as a critical common mechanism after lysosomal and mitochondrial dysfunction. This review discusses the role of cholesterol, lysosomal and mitochondrial dysfunction, and ROS in the development and progression of hypercholesterolemic pregnancies.
Collapse
|
17
|
Duttaroy AK, Basak S. Maternal Fatty Acid Metabolism in Pregnancy and Its Consequences in the Feto-Placental Development. Front Physiol 2022; 12:787848. [PMID: 35126178 PMCID: PMC8811195 DOI: 10.3389/fphys.2021.787848] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/30/2021] [Indexed: 12/16/2022] Open
Abstract
During pregnancy, maternal plasma fatty acids are critically required for cell growth and development, cell signaling, and the development of critical structural and functional aspects of the feto-placental unit. In addition, the fatty acids modulate the early stages of placental development by regulating angiogenesis in the first-trimester human placenta. Preferential transport of maternal plasma long-chain polyunsaturated fatty acids during the third trimester is critical for optimal fetal brain development. Maternal status such as obesity, diabetes, and dietary intakes may affect the functional changes in lipid metabolic processes in maternal-fetal lipid transport and metabolism. Fatty acids traverse the placental membranes via several plasma membrane fatty acid transport/binding proteins (FAT, FATP, p-FABPpm, and FFARs) and cytoplasmic fatty acid-binding proteins (FABPs). This review discusses the maternal metabolism of fatty acids and their effects on early placentation, placental fatty acid transport and metabolism, and their roles in feto-placental growth and development. The review also highlights how maternal fat metabolism modulates lipid processing, including transportation, esterification, and oxidation of fatty acids.
Collapse
Affiliation(s)
- Asim K. Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- *Correspondence: Asim K. Duttaroy,
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| |
Collapse
|
18
|
Dawid M, Mlyczyńska E, Jurek M, Respekta N, Pich K, Kurowska P, Gieras W, Milewicz T, Kotula-Balak M, Rak A. Apelin, APJ, and ELABELA: Role in Placental Function, Pregnancy, and Foetal Development-An Overview. Cells 2021; 11:cells11010099. [PMID: 35011661 PMCID: PMC8750556 DOI: 10.3390/cells11010099] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022] Open
Abstract
The apelinergic system, which includes the apelin receptor (APJ) as well as its two specific ligands, namely apelin and ELABELA (ELA/APELA/Toddler), have been the subject of many recent studies due to their pleiotropic effects in humans and other animals. Expression of these factors has been investigated in numerous tissues and organs—for example, the lungs, heart, uterus, and ovary. Moreover, a number of studies have been devoted to understanding the role of apelin and the entire apelinergic system in the most important processes in the body, starting from early stages of human life with regulation of placental function and the proper course of pregnancy. Disturbances in the balance of placental processes such as proliferation, apoptosis, angiogenesis, or hormone secretion may lead to specific pregnancy pathologies; therefore, there is a great need to search for substances that would help in their early diagnosis or treatment. A number of studies have indicated that compounds of the apelinergic system could serve this purpose. Hence, in this review, we summarized the most important reports about the role of apelin and the entire apelinergic system in the regulation of placental physiology and pregnancy.
Collapse
Affiliation(s)
- Monika Dawid
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
| | - Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
| | - Małgorzata Jurek
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
| | - Natalia Respekta
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
| | - Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
| | - Patrycja Kurowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
| | - Wiktoria Gieras
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
| | - Tomasz Milewicz
- Department of Gynecological Endocrinology, Jagiellonian University Medical College, 31-501 Krakow, Poland;
| | - Małgorzata Kotula-Balak
- University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, 30-059 Krakow, Poland;
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (M.D.); (E.M.); (M.J.); (N.R.); (K.P.); (P.K.); (W.G.)
- Correspondence: ; Tel.: +48-1-2664-5003
| |
Collapse
|
19
|
Omaña-Guzmán LI, Ortiz-Hernández L, Ancira-Moreno M, Morales-Hernández V, O’Neill MS, Vadillo-Ortega F. Association of pre-pregnancy body mass index and rate of weight gain during pregnancy with maternal indicators of cardiometabolic risk. Nutr Diabetes 2021; 11:36. [PMID: 34824195 PMCID: PMC8616911 DOI: 10.1038/s41387-021-00178-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/17/2021] [Accepted: 10/29/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND/OBJECTIVE Changes in metabolism and extensive hemodynamic adjustments occur during normal pregnancy. The presence of maternal obesity imposes an overload to these physiological adaptations that may result in increased risk for the development of cardiometabolic complications during and after pregnancy. The aim of this study is to describe total cholesterol (TC), triglycerides (TG), glucose, and arterial blood pressure (BP) trajectories and to analyze the association of these cardiometabolic risk indicators during pregnancy with pre-pregnancy body mass index (pBMI) and monthly gestational weight gain (MGWG). SUBJECTS/METHODS A prospective cohort study of pregnant women was conducted in Mexico City. Monthly samples of blood were taken during clinical follow-up and biochemical and blood pressure were measured during each visit. Adjusted linear mixed-effect regression models were fit to describe the trajectories of these biomarkers during pregnancy and to analyze the association with pBMI and MGWG. RESULTS Seven hundred and twenty women were included of which 16.6% had pre-gestational obesity, 33.2% had pre-gestational overweight, 45.8% had normal pBMI and 4.4% had pre-gestational underweight. Women with pre-gestational obesity had higher lipids concentrations in the beginning of pregnancy (TC: [Formula: see text] = 33.08, p = 0.010; TG: [Formula: see text] = 31.29, p = <0.001) but the concentrations increased less than in women with normal pBMI (TC: [Formula: see text] = -14.18, p = 0.001; TG: [Formula: see text] = -5.42, p < 0.001). By the end of pregnancy, women with pre-gestational obesity had lower concentrations of lipids than women with normal pBMI. By contrast, women with pre-gestational obesity had higher glucose concentrations and higher BP levels than women with normal pBMI over pregnancy. CONCLUSIONS pBMI is differentially associated with longitudinal trajectories of maternal biochemical markers of cardiometabolic risk. MGWG did not significantly affect the biochemical indicators or BP trajectories. Our results suggest that pBMI is more relevant to predicting adverse cardiometabolic markers trajectories during pregnancy than MGWG.
Collapse
Affiliation(s)
- Luz Isabel Omaña-Guzmán
- grid.7220.70000 0001 2157 0393Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico ,grid.452651.10000 0004 0627 7633Unidad de Vinculación Científica de la Facultad de Medicina UNAM, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Luis Ortiz-Hernández
- grid.7220.70000 0001 2157 0393Departamento de Atención a la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Mónica Ancira-Moreno
- grid.441047.20000 0001 2156 4794Departamento de Salud, Universidad Iberoamericana, Mexico City, Mexico
| | - Vanesa Morales-Hernández
- grid.419218.70000 0004 1773 5302Biología de la Reproducción, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Marie S. O’Neill
- grid.214458.e0000000086837370Epidemiology and Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI USA
| | - Felipe Vadillo-Ortega
- grid.452651.10000 0004 0627 7633Unidad de Vinculación Científica de la Facultad de Medicina UNAM, Instituto Nacional de Medicina Genómica, Mexico City, Mexico ,grid.214458.e0000000086837370Epidemiology and Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
20
|
Bandres-Meriz J, Majali-Martinez A, Hoch D, Morante M, Glasner A, van Poppel MNM, Desoye G, Herrera E. Maternal C-Peptide and Insulin Sensitivity, but Not BMI, Associate with Fatty Acids in the First Trimester of Pregnancy. Int J Mol Sci 2021; 22:10422. [PMID: 34638763 PMCID: PMC8508886 DOI: 10.3390/ijms221910422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/19/2021] [Accepted: 09/24/2021] [Indexed: 12/19/2022] Open
Abstract
Maternal obesity in pregnancy is a pro-inflammatory condition exposing the fetus to an adverse environment. Here, we tested associations of maternal obesity (primary exposures: BMI, leptin) and metabolic parameters (secondary exposures: glucose, C-peptide, and insulin sensitivity) with total serum concentrations of fatty acids in the first trimester of human pregnancy. This cross-sectional study included 123 non-smoking women with singleton pregnancy. In maternal serum, cotinine, leptin, and C-peptide (ELISA), glucose (hexokinase-based test) and fatty acids (gas chromatography) were quantified, and the insulin sensitivity index (ISHOMA) was calculated. Concentrations of fatty acid classes and total fatty acids did not differ between BMI or leptin categories. However, n-3 polyunsaturated fatty acids (PUFA) were decreased in the category with the highest C-peptide concentration (n-3 PUFA: CI -35.82--6.28, p < 0.006) and in the lowest ISHOMA category (n-3 PUFA: CI -36.48--5.61, p < 0.008). In a subcohort, in which fetal sex was determined (RT-qPCR of placental tissue), C-peptide was significantly associated with docosahexaenoic acid (DHA) in mothers bearing a female (n = 46), but not male (n = 37) fetus. In conclusion, pregnant women with high fasting C-peptide and low ISHOMA had decreased n-3 PUFA, and DHA was lower with higher C-peptide only in mothers bearing a female fetus.
Collapse
Affiliation(s)
- Julia Bandres-Meriz
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036 Graz, Austria; (A.M.-M.); (D.H.)
| | - Alejandro Majali-Martinez
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036 Graz, Austria; (A.M.-M.); (D.H.)
| | - Denise Hoch
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036 Graz, Austria; (A.M.-M.); (D.H.)
| | - Milagros Morante
- Faculty of Pharmacy, Universidad San Pablo CEU, 28668 Madrid, Spain; (M.M.); (E.H.)
| | | | | | - Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036 Graz, Austria; (A.M.-M.); (D.H.)
| | - Emilio Herrera
- Faculty of Pharmacy, Universidad San Pablo CEU, 28668 Madrid, Spain; (M.M.); (E.H.)
| |
Collapse
|
21
|
McConnell C, Thoene M, Van Ormer M, Furtado JD, Korade Z, Genaro-Mattos TC, Hanson C, Anderson-Berry A. Plasma Concentrations and Maternal-Umbilical Cord Plasma Ratios of the Six Most Prevalent Carotenoids across Five Groups of Birth Gestational Age. Antioxidants (Basel) 2021; 10:antiox10091409. [PMID: 34573041 PMCID: PMC8467757 DOI: 10.3390/antiox10091409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 11/26/2022] Open
Abstract
Carotenoids are antioxidant nutrients with the potential to provide protection against oxidative stress. Plasma carotenoid concentrations are lower in newborn infants compared to their mothers; however, limited information is available regarding how concentrations differ by gestational age. The objective of this research is to assess maternal and umbilical cord plasma carotenoid concentrations and maternal-umbilical cord plasma ratios across five groups of birth gestational age. Mother-infant dyads were enrolled at delivery for collection of maternal and umbilical cord blood. Plasma carotenoids were analyzed by HPLC and LC-MS/MS. Birth gestational age was categorized into five groups, and the Kruskal–Wallis test compared carotenoid concentrations and maternal-umbilical cord plasma ratios between these groups. A p-value of < 0.05 was considered statistically significant. 370 mother-infant dyads were included, with most infants delivered at early term (20.3%) or term (64.6%). Though maternal plasma concentrations increased with birth gestational age, we observed less variability in umbilical cord plasma concentrations, thus the maternal-umbilical cord plasma ratio also increased with birth CGA groups for lutein + zeaxanthin (p = 0.008), β-cryptoxanthin (p = 0.027), α-carotene (p = 0.030); β-carotene approached significance (p = 0.056). Additional research is needed to determine if carotenoid concentrations were physiologic to varying gestational ages or if they were impacted by factors associated with preterm birth.
Collapse
Affiliation(s)
- Chelsey McConnell
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.M.); (M.V.O.); (Z.K.); (A.A.-B.)
| | - Melissa Thoene
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.M.); (M.V.O.); (Z.K.); (A.A.-B.)
- Correspondence:
| | - Matthew Van Ormer
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.M.); (M.V.O.); (Z.K.); (A.A.-B.)
| | - Jeremy D. Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA;
| | - Zeljka Korade
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.M.); (M.V.O.); (Z.K.); (A.A.-B.)
| | | | - Corrine Hanson
- College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Ann Anderson-Berry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.M.); (M.V.O.); (Z.K.); (A.A.-B.)
| |
Collapse
|
22
|
Paquette AG, MacDonald J, Lapehn S, Bammler T, Kruger L, Day DB, Price ND, Loftus C, Kannan K, Marsit C, Mason WA, Bush NR, LeWinn KZ, Enquobahrie DA, Prasad B, Karr CJ, Sathyanarayana S. A Comprehensive Assessment of Associations between Prenatal Phthalate Exposure and the Placental Transcriptomic Landscape. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:97003. [PMID: 34478338 PMCID: PMC8415559 DOI: 10.1289/ehp8973] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
BACKGROUND Phthalates are commonly used endocrine-disrupting chemicals that are ubiquitous in the general population. Prenatal phthalate exposure may alter placental physiology and fetal development, leading to adverse perinatal and childhood health outcomes. OBJECTIVE We examined associations between prenatal phthalate exposure in the second and third trimesters and the placental transcriptome at birth, including genes and long noncoding RNAs (lncRNAs), to gain insight into potential mechanisms of action during fetal development. METHODS The ECHO PATHWAYs consortium quantified 21 urinary phthalate metabolites from 760 women enrolled in the CANDLE study (Shelby County, TN) using high-performance liquid chromatography-tandem mass spectrometry. Placental transcriptomic data were obtained using paired-end RNA sequencing. Linear models were fitted to estimate separate associations between maternal urinary phthalate metabolite concentration during the second and third trimester and placental gene expression at birth, adjusted for confounding variables. Genes were considered differentially expressed at a Benjamini-Hochberg false discovery rate (FDR) p<0.05. Associations between phthalate metabolites and biological pathways were identified using self-contained gene set testing and considered significantly altered with an FDR-adjusted p<0.2. RESULTS We observed significant associations between second-trimester phthalate metabolites mono (carboxyisooctyl) phthalate (MCIOP), mono-2-ethyl-5-carboxypentyl phthalate, and mono-2-ethyl-5-oxohexyl phthalate and 18 genes in total, including four lncRNAs. Specifically, placental expression of NEAT1 was associated with multiple phthalate metabolites. Third-trimester MCIOP and mono-isobutyl phthalate concentrations were significantly associated with placental expression of 18 genes and two genes, respectively. Expression of genes within 27 biological pathways was associated with mono-methyl phthalate, MCIOP, and monoethyl phthalate concentrations. DISCUSSION To our knowledge, this is the first genome-wide assessment of the relationship between the placental transcriptome at birth and prenatal phthalate exposure in a large and diverse birth cohort. We identified numerous genes and lncRNAs associated with prenatal phthalate exposure. These associations mirror findings from other epidemiological and in vitro analyses and may provide insight into biological pathways affected in utero by phthalate exposure. https://doi.org/10.1289/EHP8973.
Collapse
Affiliation(s)
- Alison G. Paquette
- Seattle Children’s Research Institute, Seattle, Washington, USA
- University of Washington, Seattle, Washington, USA
| | | | - Samantha Lapehn
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Theo Bammler
- University of Washington, Seattle, Washington, USA
| | - Laken Kruger
- Washington State University, Spokane, Washington, USA
| | - Drew B. Day
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Nathan D. Price
- Institute For Systems Biology, Seattle, Washington, USA
- Onegevity Health, New York City, New York, USA
| | | | | | | | - W. Alex Mason
- University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Nicole R. Bush
- University of California San Francisco, San Francisco California, USA
| | - Kaja Z. LeWinn
- University of California San Francisco, San Francisco California, USA
| | | | | | | | - Sheela Sathyanarayana
- Seattle Children’s Research Institute, Seattle, Washington, USA
- University of Washington, Seattle, Washington, USA
| |
Collapse
|
23
|
Andescavage NN, Limperopoulos C. Placental abnormalities in congenital heart disease. Transl Pediatr 2021; 10:2148-2156. [PMID: 34584887 PMCID: PMC8429875 DOI: 10.21037/tp-20-347] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/05/2020] [Indexed: 01/26/2023] Open
Abstract
Congenital heart disease (CHD) remains the most common birth defect in infants, and critical CHD is associated with significant rates of morbidity and mortality. With the advent of powerful yet noninvasive advanced fetal imaging, it is becoming increasingly evident that the presence of CHD in utero disrupts typical development and contributes to the lifelong morbidity in this population. Across healthy and high-risk populations, intrauterine influences can permanently alter fetal development that may manifest in complex morbidities later in life, the so-called fetal-onset-of-adult-disease (FOAD) phenomenon. The placenta plays a critical role in not only supporting fetal development, but also by adapting to specific intrauterine conditions. The role of placental health, adaptation and dysfunction, however, in CHD is not well understood. In this article, we will review current evidence relating placental health in CHD, appraise existing knowledge-gaps in the field and highlight promising new avenues to better understand the impact of placental function on fetal well-being. We will review evidence of ex vivo human placental studies that describe abnormal placental findings in pregnancies complicated by CHD, as well evidence for in vivo assessments of the human placenta. While overall clinical in vivo assessments of placental development are rather limited, we will also review emerging evidence from advanced quantitative and functional magnetic resonance imaging that are bringing new insights into placental structure and function throughout gestation. By providing novel information about placental development, we can now explore the maternal-fetal-placental connection in greater detail, and better understand the multi-factorial mechanisms that may contribute to adverse outcomes seen in survivors of CHD.
Collapse
Affiliation(s)
- Nickie N Andescavage
- Division of Neonatology, Children's National Health System, Washington, DC, USA.,Department of Pediatrics, George Washington University School of Medicine, Washington, DC, USA
| | - Catherine Limperopoulos
- Department of Pediatrics, George Washington University School of Medicine, Washington, DC, USA.,Division of Diagnostic Imaging & Radiology, Children's National Health System, Washington, DC, USA.,Department of Radiology, George Washington University School of Medicine, Washington, DC, USA
| |
Collapse
|
24
|
Ramirez-Hincapie S, Giri V, Keller J, Kamp H, Haake V, Richling E, van Ravenzwaay B. Influence of pregnancy and non-fasting conditions on the plasma metabolome in a rat prenatal toxicity study. Arch Toxicol 2021; 95:2941-2959. [PMID: 34327559 DOI: 10.1007/s00204-021-03105-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/17/2021] [Indexed: 11/25/2022]
Abstract
The current parameters for determining maternal toxicity (e.g. clinical signs, food consumption, body weight development) lack specificity and may underestimate the extent of effects of test compounds on the dams. Previous reports have highlighted the use of plasma metabolomics for an improved and mechanism-based identification of maternal toxicity. To establish metabolite profiles of healthy pregnancies and evaluate the influence of food consumption as a confounding factor, metabolite profiling of rat plasma was performed by gas- and liquid-chromatography-tandem mass spectrometry techniques. Metabolite changes in response to pregnancy, food consumption prior to blood sampling (non-fasting) as well as the interaction of both conditions were studied. In dams, both conditions, non-fasting and pregnancy, had a marked influence on the plasma metabolome and resulted in distinct individual patterns of changed metabolites. Non-fasting was characterized by increased plasma concentrations of amino acids and diet related compounds and lower levels of ketone bodies. The metabolic profile of pregnant rats was characterized by lower amino acids and glucose levels and higher concentrations of plasma fatty acids, triglycerides and hormones, capturing the normal biochemical changes undergone during pregnancy. The establishment of metabolic profiles of pregnant non-fasted rats serves as a baseline to create metabolic fingerprints for prenatal and maternal toxicity studies.
Collapse
Affiliation(s)
- S Ramirez-Hincapie
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - V Giri
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - J Keller
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - H Kamp
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - V Haake
- BASF Metabolome Solution GmbH, Berlin, Germany
| | - E Richling
- Food Chemistry and Toxicology, Department of Chemistry, University of Kaiserslautern, Kaiserslautern, Germany
| | - B van Ravenzwaay
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany.
| |
Collapse
|
25
|
Abstract
Almost 2 billion adults in the world are overweight, and more than half of them are classified as obese, while nearly one-third of children globally experience poor growth and development. Given the vast amount of knowledge that has been gleaned from decades of research on growth and development, a number of questions remain as to why the world is now in the midst of a global epidemic of obesity accompanied by the "double burden of malnutrition," where overweight coexists with underweight and micronutrient deficiencies. This challenge to the human condition can be attributed to nutritional and environmental exposures during pregnancy that may program a fetus to have a higher risk of chronic diseases in adulthood. To explore this concept, frequently called the developmental origins of health and disease (DOHaD), this review considers a host of factors and physiological mechanisms that drive a fetus or child toward a higher risk of obesity, fatty liver disease, hypertension, and/or type 2 diabetes (T2D). To that end, this review explores the epidemiology of DOHaD with discussions focused on adaptations to human energetics, placental development, dysmetabolism, and key environmental exposures that act to promote chronic diseases in adulthood. These areas are complementary and additive in understanding how providing the best conditions for optimal growth can create the best possible conditions for lifelong health. Moreover, understanding both physiological as well as epigenetic and molecular mechanisms for DOHaD is vital to most fully address the global issues of obesity and other chronic diseases.
Collapse
Affiliation(s)
- Daniel J Hoffman
- Department of Nutritional Sciences, Program in International Nutrition, and Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Theresa L Powell
- Department of Pediatrics and Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Daniel B Hardy
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
26
|
Reis LG, Silva TH, Ravagnani GM, Martinez CHG, Salles MSV, Andrade AFC, Cônsolo NRB, Martins SMMK, de Oliveira Bussiman F, Oliveira MXS, Lanna DPD, Saran Netto A. Maternal Supplementation with Cow's Milk Naturally Enriched with PUFA Alters the Metabolism of Sows and the Fatty Acid Profile of the Offspring. Nutrients 2021; 13:1942. [PMID: 34198804 PMCID: PMC8228345 DOI: 10.3390/nu13061942] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 01/21/2023] Open
Abstract
The study aimed to evaluate the supplementation of gilts with cow's milk naturally enriched with n-3 and n-6 polyunsaturated fatty acids (PUFA) on reproductive outcomes, and the serum biochemical and FA profile of swine females and their offspring. During 316 days, 30 gilts were distributed into three groups: (1) Control, fed a basal diet + milk from cows without oil; (2) n-3, fed a basal diet + milk from cows fed a diet enriched with linseed oil; (3) n-6, fed a basal diet + milk from cows fed a diet enriched with soybean oil. The gilts receiving the diets containing PUFA had higher serum urea and very-low-density lipoprotein levels and lower serum total protein and low-density lipoprotein levels compared to the Control group. Females supplemented with n-3 presented higher serum palmitic acid and γ-linolenic acid levels than those fed n-6. Piglets from the Control group were heavier at birth than those from females supplemented with enriched milk. The piglets from females receiving enriched milk had 140 g higher body weight from 1 to 21 days old compared to the Control group, and greater average daily weight gain from 7 to 14 days old. The serum eicosapentaenoic acid level of piglets fed n-3 was 69% higher than those fed n-6, which reduced the AA/EPA ratio. Gilts supplemented with PUFA-enriched cow's milk showed changes in their serum palmitic and γ-linolenic acid levels, in addition to improved performance, EPA concentration and consequently reduced AA/EPA ratio in their piglets, demonstrating beneficial results for their progeny.
Collapse
Affiliation(s)
- Leriana Garcia Reis
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (L.G.R.); (T.H.S.); (N.R.B.C.); (S.M.M.K.M.)
| | - Thiago Henrique Silva
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (L.G.R.); (T.H.S.); (N.R.B.C.); (S.M.M.K.M.)
| | - Gisele Mouro Ravagnani
- Department of Animal Reproduction, School of Veterinary and Animal Science, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (G.M.R.); (A.F.C.A.)
| | - Cristian Hernando Garcia Martinez
- Department of Animal Nutrition and Production, School of Veterinary and Animal Science, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (C.H.G.M.); (F.d.O.B.)
| | | | - André Furugen Cesar Andrade
- Department of Animal Reproduction, School of Veterinary and Animal Science, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (G.M.R.); (A.F.C.A.)
| | - Nara Regina Brandão Cônsolo
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (L.G.R.); (T.H.S.); (N.R.B.C.); (S.M.M.K.M.)
| | - Simone Maria Massami Kitamura Martins
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (L.G.R.); (T.H.S.); (N.R.B.C.); (S.M.M.K.M.)
| | - Fernando de Oliveira Bussiman
- Department of Animal Nutrition and Production, School of Veterinary and Animal Science, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (C.H.G.M.); (F.d.O.B.)
| | - Mauricio Xavier Silva Oliveira
- Department of Animal Science, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA;
| | - Dante Pazzanese Duarte Lanna
- Department of Animal Science, Luiz de Queiroz College Agriculture, University of São Paulo, Avenida Pádua Dias, 11, Piracicaba 13418-900, Brazil;
| | - Arlindo Saran Netto
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, Avenida Duque de Caxias Norte, 225, Pirassununga 13635-900, Brazil; (L.G.R.); (T.H.S.); (N.R.B.C.); (S.M.M.K.M.)
| |
Collapse
|
27
|
Huhtala MS, Rönnemaa T, Pellonperä O, Tertti K. Cord serum metabolome and birth weight in patients with gestational diabetes treated with metformin, insulin, or diet alone. BMJ Open Diabetes Res Care 2021; 9:e002022. [PMID: 34059525 PMCID: PMC8169462 DOI: 10.1136/bmjdrc-2020-002022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/09/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Recent research has demonstrated the benefits of metformin treatment in gestational diabetes (GDM) on short-term pregnancy outcomes (including excessive fetal growth and pre-eclampsia), but its effects on fetal metabolism remain mostly unknown. Our aim was to study the effects of metformin treatment compared with insulin or diet on the cord serum metabolome and also to assess how these metabolites are related to birth weight (BW) in pregnancies complicated by GDM. RESEARCH DESIGN AND METHODS Cord serum samples were available from 113, 97, and 98 patients with GDM treated with diet, insulin, and metformin, respectively. A targeted metabolome was measured using nuclear magnetic resonance spectroscopy. The patients in the metformin and insulin groups had participated in a previous randomized trial (NCT01240785). RESULTS Cord serum alanine was elevated in the metformin group (0.53 mmol/L) compared with the insulin (0.45 mmol/L, p<0.001) and the diet groups (0.46 mmol/L, p<0.0001). All other measured metabolites were similar between the groups. The triglyceride (TG)-to-phosphoglyceride ratio, average very low-density lipoprotein particle diameter, docosahexaenoic acid, omega-3 fatty acids (FAs), and ratios of omega-3 and monounsaturated FA to total FA were inversely related to BW. The omega-6-to-total-FA and omega-6-to-omega-3-FA ratios were positively related to BW. Cholesterol in very large and large high-density lipoprotein (HDL) was positively (p<0.01) associated with BW when adjusted for maternal prepregnancy body mass index, gestational weight gain, glycated hemoglobin, and mode of delivery. CONCLUSIONS Metformin treatment in GDM leads to an increase in cord serum alanine. The possible long-term implications of elevated neonatal alanine in this context need to be evaluated in future studies. Although previous studies have shown that metformin increased maternal TG levels, the cord serum TG levels were not affected. Cord serum HDL cholesterol and several FA variables are related to the regulation of fetal growth in GDM. Moreover, these associations seem to be independent of maternal confounding factors. TRIAL REGISTRATION NUMBER NCT01240785.
Collapse
Affiliation(s)
- Mikael S Huhtala
- Obstetrics and Gynecology, University of Turku, Turku, Finland
- Obstetrics and Gynecology, TYKS Turku University Hospital, Turku, Finland
| | - Tapani Rönnemaa
- Medicine, University of Turku, Turku, Finland
- Medicine, TYKS Turku University Hospital, Turku, Finland
| | - Outi Pellonperä
- Obstetrics and Gynecology, University of Turku, Turku, Finland
- Obstetrics and Gynecology, TYKS Turku University Hospital, Turku, Finland
| | - Kristiina Tertti
- Obstetrics and Gynecology, University of Turku, Turku, Finland
- Obstetrics and Gynecology, TYKS Turku University Hospital, Turku, Finland
| |
Collapse
|
28
|
Natarajan SK, Bruett T, Muthuraj PG, Sahoo PK, Power J, Mott JL, Hanson C, Anderson-Berry A. Saturated free fatty acids induce placental trophoblast lipoapoptosis. PLoS One 2021; 16:e0249907. [PMID: 33886600 PMCID: PMC8062006 DOI: 10.1371/journal.pone.0249907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Obesity during pregnancy increases the risk for maternal complications like gestational diabetes, preeclampsia, and maternal inflammation. Maternal obesity also increases the risk of childhood obesity, intrauterine growth restriction (IUGR) and diabetes to the offspring. Increased circulating free fatty acids (FFAs) in obesity due to adipose tissue lipolysis induces lipoapoptosis to hepatocytes, cholangiocytes, and pancreatic-β-cells. During the third trimester of human pregnancy, there is an increase in maternal lipolysis and release of FFAs into the circulation. It is currently unknown if increased FFAs during gestation as a result of maternal obesity cause placental cell lipoapoptosis. Increased exposure of FFAs during maternal obesity has been shown to result in placental lipotoxicity. The objective of the present study is to determine saturated FFA-induced trophoblast lipoapoptosis and also to test the protective role of monounsaturated fatty acids against FFA-induced trophoblast lipoapoptosis using in vitro cell culture model. Here, we hypothesize that saturated FFAs induce placental trophoblast lipoapoptosis, which was prevented by monounsaturated fatty acids. METHODS Biochemical and structural markers of apoptosis by characteristic nuclear morphological changes with DAPI staining, and caspase 3/7 activity was assessed. Cleaved PARP and cleaved caspase 3 were examined by western blot analysis. RESULTS Treatment of trophoblast cell lines, JEG-3 and JAR cells with palmitate (PA) or stearate (SA) induces trophoblast lipoapoptosis as evidenced by a significant increase in apoptotic nuclear morphological changes and caspase 3/7 activity. We observed that saturated FFAs caused a concentration-dependent increase in placental trophoblast lipoapoptosis. We also observed that monounsaturated fatty acids like palmitoleate and oleate mitigates placental trophoblast lipoapoptosis caused due to PA exposure. CONCLUSION We show that saturated FFAs induce trophoblast lipoapoptosis. Co-treatment of monounsaturated fatty acids like palmitoleate and oleate protects against FFA-induced trophoblast lipoapoptosis.
Collapse
Affiliation(s)
- Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- * E-mail:
| | - Taylor Bruett
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Philma Glora Muthuraj
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Prakash K. Sahoo
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Jillian Power
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Justin L. Mott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Corrine Hanson
- College of Allied Health Professions Medical Nutrition Education, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Ann Anderson-Berry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
29
|
The anabolic role of the Warburg, Cori-cycle and Crabtree effects in health and disease. Clin Nutr 2021; 40:2988-2998. [PMID: 33674148 DOI: 10.1016/j.clnu.2021.02.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
In evolution, genes survived that could code for metabolic pathways, promoting long term survival during famines or fasting when suffering from trauma, disease or during physiological growth. This requires utilization of substrates, already present in some form in the body. Carbohydrate stores are limited and to survive long, their utilization is restricted to survival pathways, by inhibiting glucose oxidation and glycogen synthesis. This leads to insulin resistance and spares muscle protein, because being the main supplier of carbon for new glucose production. In these survival pathways, part of the glucose is degraded in glycolysis in peripheral (muscle) tissues to pyruvate and lactate (Warburg effect), which are partly reutilized for glucose formation in liver and kidney, completing the Cori-cycle. Another part of the glucose taken up by muscle contributes, together with muscle derived amino acids, to the production of substrates consisting of a complete amino acid mix but extra non-essential amino acids like glutamine, alanine, glycine and proline. These support cell proliferation, matrix deposition and redox regulation in tissues, specifically active in host response and during growth. In these tissues, also glucose is taken up delivering glycolytic intermediates, that branch off and act as building blocks and produce reducing equivalents. Lactate is also produced and released in the circulation, adding to the lactate released by muscle in the Cori-cycle and completing secondary glucose cycles. Increased fluxes through these cycles lead to modest hyperglycemia and hyperlactatemia in states of healthy growth and disease and are often misinterpreted as induced by hypoxia.
Collapse
|
30
|
Álvarez D, Muñoz Y, Ortiz M, Maliqueo M, Chouinard-Watkins R, Valenzuela R. Impact of Maternal Obesity on the Metabolism and Bioavailability of Polyunsaturated Fatty Acids during Pregnancy and Breastfeeding. Nutrients 2020; 13:nu13010019. [PMID: 33374585 PMCID: PMC7822469 DOI: 10.3390/nu13010019] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Prenatal and postnatal development are closely related to healthy maternal conditions that allow for the provision of all nutritional requirements to the offspring. In this regard, an appropriate supply of fatty acids (FA), mainly n-3 and n-6 long-chain polyunsaturated fatty acids (LCPUFA), is crucial to ensure a normal development, because they are an integral part of cell membranes and participate in the synthesis of bioactive molecules that regulate multiple signaling pathways. On the other hand, maternal obesity and excessive gestational weight gain affect FA supply to the fetus and neonate, altering placental nutrient transfer, as well as the production and composition of breast milk during lactation. In this regard, maternal obesity modifies FA profile, resulting in low n-3 and elevated n-6 PUFA levels in maternal and fetal circulation during pregnancy, as well as in breast milk during lactation. These modifications are associated with a pro-inflammatory state and oxidative stress with short and long-term consequences in different organs of the fetus and neonate, including in the liver, brain, skeletal muscle, and adipose tissue. Altogether, these changes confer to the offspring a higher risk of developing obesity and its complications, as well as neuropsychiatric disorders, asthma, and cancer. Considering the consequences of an abnormal FA supply to offspring induced by maternal obesity, we aimed to review the effects of obesity on the metabolism and bioavailability of FA during pregnancy and breastfeeding, with an emphasis on LCPUFA homeostasis.
Collapse
Affiliation(s)
- Daniela Álvarez
- Endocrinology and Metabolism Laboratory, West Division, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (D.Á.); (Y.M.); (M.O.); (M.M.)
| | - Yasna Muñoz
- Endocrinology and Metabolism Laboratory, West Division, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (D.Á.); (Y.M.); (M.O.); (M.M.)
| | - Macarena Ortiz
- Endocrinology and Metabolism Laboratory, West Division, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (D.Á.); (Y.M.); (M.O.); (M.M.)
| | - Manuel Maliqueo
- Endocrinology and Metabolism Laboratory, West Division, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (D.Á.); (Y.M.); (M.O.); (M.M.)
| | - Raphaël Chouinard-Watkins
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada;
| | - Rodrigo Valenzuela
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada;
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
- Correspondence: or ; Tel.: +56-2-9786746
| |
Collapse
|
31
|
Parrettini S, Caroli A, Torlone E. Nutrition and Metabolic Adaptations in Physiological and Complicated Pregnancy: Focus on Obesity and Gestational Diabetes. Front Endocrinol (Lausanne) 2020; 11:611929. [PMID: 33424775 PMCID: PMC7793966 DOI: 10.3389/fendo.2020.611929] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
Pregnancy offers a window of opportunity to program the future health of both mothers and offspring. During gestation, women experience a series of physical and metabolic modifications and adaptations, which aim to protect the fetus development and are closely related to both pre-gestational nutritional status and gestational weight gain. Moreover, pre-gestational obesity represents a challenge of treatment, and nowadays there are new evidence as regard its management, especially the adequate weight gain. Recent evidence has highlighted the determinant role of nutritional status and maternal diet on both pregnancy outcomes and long-term risk of chronic diseases, through a transgenerational flow, conceptualized by the Development Origin of Health and Diseases (Dohad) theory. In this review we will analyse the physiological and endocrine adaptation in pregnancy, and the metabolic complications, thus the focal points for nutritional and therapeutic strategies that we must early implement, virtually before conception, to safeguard the health of both mother and progeny. We will summarize the current nutritional recommendations and the use of nutraceuticals in pregnancy, with a focus on the management of pregnancy complicated by obesity and hyperglycemia, assessing the most recent evidence about the effects of ante-natal nutrition on the long-term, on either maternal health or metabolic risk of the offspring.
Collapse
Affiliation(s)
- Sara Parrettini
- S. Maria della Misericordia Hospital, Division of Endocrinology and Metabolism, Perugia, Italy
- Department of Medicine, University of Perugia, Perugia, Italy
| | - Antonella Caroli
- S. Maria della Misericordia Hospital, Division of Endocrinology and Metabolism, Perugia, Italy
- Department of Medicine, University of Perugia, Perugia, Italy
| | - Elisabetta Torlone
- S. Maria della Misericordia Hospital, Division of Endocrinology and Metabolism, Perugia, Italy
| |
Collapse
|
32
|
Effects of dietary n-3-PUFA supplementation, post-insemination plane of nutrition and pregnancy status on the endometrial transcriptome of beef heifers. Sci Rep 2020; 10:20798. [PMID: 33247230 PMCID: PMC7695717 DOI: 10.1038/s41598-020-77604-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Supplementation of cattle diets with n-3-polyunsaturated fatty acids (PUFA) can improve reproductive efficiency. Conversely, short-term fluctuations in feed supply can impact pregnancy establishment. The objectives of this study were to examine the effects of (1) dietary supplementation with n-3-PUFA and (2) post-insemination plane of nutrition on the endometrial transcriptome. Beef crossbred heifers were offered concentrate based diets fortified with n-3-PUFA (PUFA; n = 32) or not (CONT; n = 28) for 30 days prior to breeding at a synchronised oestrous. Following artificial insemination, heifers were allocated within treatment to either a high or low plane of nutrition. Heifers were maintained on these diets for 16 days following which endometrial tissue was harvested at slaughter for subsequent RNAseq analysis. The influence of pregnancy status on the endomentrial transcriptome, within each dietary treatment group, was also examined. Post-insemination diet affected (P < 0.05) the endometrial transcriptome. Specifically, within n-3-PUFA-supplemented heifers, genes involved in embryonic development and mTOR signalling pathways, important in pregnancy establishment, were identified as differentially expressed. Results indicate that dietary supplementation of cattle diets with n-3-PUFA may have a positive effect on the expression of key fertility-related genes and pathways, during the critical window of maternal recognition of pregnancy, particularly where animals are underfed.
Collapse
|
33
|
Abascal-Saiz A, Fuente-Luelmo E, Haro M, de la Calle M, Ramos-Álvarez MP, Perdomo G, Bartha JL. Placental Compartmentalization of Lipid Metabolism: Implications for Singleton and Twin Pregnancies. Reprod Sci 2020; 28:1150-1160. [PMID: 33171514 DOI: 10.1007/s43032-020-00385-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/01/2020] [Indexed: 11/27/2022]
Abstract
The study of placental lipid metabolism in uncomplicated pregnancies has not been developed in the literature to date. Its importance lies in expanding the knowledge of placental function to enable comparison with pathological pregnancies in future research. The aim of the present study was to compare the lipid metabolic activity and storage of the maternal and fetal sides of the placenta in healthy pregnancies. Moreover, we compare singleton vs. twin pregnancies to determine if placental metabolic needs differ. We analyzed placental explants from uncomplicated pregnancies, 20 from singleton and 8 from bichorial-biamniotic twin pregnancies (n = 28). Six cotyledon fragments were collected from each placenta at different distances from the umbilical cord, three close to the chorionic plate (hereinafter, we will refer to them as "fetal side") and another three close to the anchoring villi into the decidua basalis (referred to as "maternal side"). The samples were analyzed for quantitative assay placental fatty acid oxidation (FAO) and esterification (FAE) activities and triglyceride levels. The location of lipid storage in the chorionic villi was assessed by Oil red-O staining. Placental fatty acid oxidation did not show differences when comparing the maternal and fetal sides of the placenta or between single and twin pregnancies. When comparing placental sides, FAE was increased twofold in the maternal side compared to the fetal side of the placenta (P = 0.013). The tendency for lipogenesis in the placenta was exemplified by the FAE/FAO ratio, which was a 37.1% higher on the maternal side (P = 0.019). Despite this, triglyceride levels were five times higher in the fetal side than in the maternal one (P = 0.024). When analyzing singleton vs. twins, FAE was superior in the fetal side in multiple pregnancies (× 2.6, P = 0.007) and the FAE/FAO ratio was significantly higher in twins than in singleton pregnancies, on both sides of the placenta. Despite this finding, triglyceride levels were similar in twin and singleton pregnancies. Comparing the placentas of twins in the same pregnancy, there were no differences in lipid metabolism (FAO or FAE) or placental triglyceride levels between the two co-twins. Using Oil red-O staining, lipid storage in chorionic villi was found to be located on the syncytiotrophoblast cells and not in the connecting axis. The maternal side of the placenta is more active in the esterification of fatty acids, while the storage of neutral lipids concentrates on the fetal side. Moreover, multiple gestations have increased esterification without changes in the concentration of placental triglycerides, probably due to a higher transfer to the fetal circulation in response to the greater energy demand from twin fetuses.
Collapse
Affiliation(s)
- Alejandra Abascal-Saiz
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Eva Fuente-Luelmo
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María Haro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María de la Calle
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - María P Ramos-Álvarez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - Germán Perdomo
- Department of Health Sciences, University of Burgos, Burgos, Spain.,Institute of Molecular Biology and Genetic (IMBG), CSIC - University of Valladolid, Valladolid, Spain
| | - José L Bartha
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
34
|
Bowman CE, Arany Z, Wolfgang MJ. Regulation of maternal-fetal metabolic communication. Cell Mol Life Sci 2020; 78:1455-1486. [PMID: 33084944 DOI: 10.1007/s00018-020-03674-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
Pregnancy may be the most nutritionally sensitive stage in the life cycle, and improved metabolic health during gestation and early postnatal life can reduce the risk of chronic disease in adulthood. Successful pregnancy requires coordinated metabolic, hormonal, and immunological communication. In this review, maternal-fetal metabolic communication is defined as the bidirectional communication of nutritional status and metabolic demand by various modes including circulating metabolites, endocrine molecules, and other secreted factors. Emphasis is placed on metabolites as a means of maternal-fetal communication by synthesizing findings from studies in humans, non-human primates, domestic animals, rabbits, and rodents. In this review, fetal, placental, and maternal metabolic adaptations are discussed in turn. (1) Fetal macronutrient needs are summarized in terms of the physiological adaptations in place to ensure their proper allocation. (2) Placental metabolite transport and maternal physiological adaptations during gestation, including changes in energy budget, are also discussed. (3) Maternal nutrient limitation and metabolic disorders of pregnancy serve as case studies of the dynamic nature of maternal-fetal metabolic communication. The review concludes with a summary of recent research efforts to identify metabolites, endocrine molecules, and other secreted factors that mediate this communication, with particular emphasis on serum/plasma metabolomics in humans, non-human primates, and rodents. A better understanding of maternal-fetal metabolic communication in health and disease may reveal novel biomarkers and therapeutic targets for metabolic disorders of pregnancy.
Collapse
Affiliation(s)
- Caitlyn E Bowman
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zoltan Arany
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
35
|
Steinhauser CB, Askelson K, Lambo CA, Hobbs KC, Bazer FW, Satterfield MC. Lipid metabolism is altered in maternal, placental, and fetal tissues of ewes with small for gestational age fetuses†. Biol Reprod 2020; 104:170-180. [PMID: 33001151 DOI: 10.1093/biolre/ioaa180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/23/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Nutrient restriction (NR) has the potential to negatively impact birthweight, an indicator of neonatal survival and lifelong health. Those fetuses are termed as small for gestational age (SGA). Interestingly, there is a spectral phenotype of fetal growth rates in response to NR associated with changes in placental development, nutrient and waste transport, and lipid metabolism. A sheep model with a maternal diet, starting at Day 35, of 100% National Research Council (NRC) nutrient requirements (n = 8) or 50% NRC (n = 28) was used to assess alterations in fetuses designated NR SGA (n = 7) or NR NonSGA (n = 7) based on fetal weight at Day 135 of pregnancy. Allantoic fluid concentrations of triglycerides were greater in NR SGA fetuses than 100% NRC and NR NonSGA fetuses at Day 70 (P < 0.05). There was a negative correlation between allantoic fluid concentrations of triglycerides (R2 = 0.207) and bile acids (R2 = 0.179) on Day 70 and fetal weight at Day 135 for NR ewes (P < 0.05). Bile acids were more abundant in maternal and fetal blood for NR SGA compared to 100% NRC and NR NonSGA ewes (P < 0.05). Maternal blood concentrations of NEFAs increased in late pregnancy in NR NonSGA compared to NR SGA ewes (P < 0.05). Protein expression of fatty acid transporter SLC27A6 localized to placentomal maternal and fetal epithelia and decreased in Day 70 NR SGA compared to 100% NRC and NR NonSGA placentomes (P < 0.05). These results identify novel factors associated with an ability of placentae and fetuses in NR NonSGA ewes to adapt to, and overcome, nutritional hardship during pregnancy.
Collapse
Affiliation(s)
| | - Katharine Askelson
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Colleen A Lambo
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Kenneth C Hobbs
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - M Carey Satterfield
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
36
|
Hernandez-Trejo M, Sámano R, Chico-Barba G, Pizano-Zarate ML, Herrera-González NE. Neonatal adiposity may increase plasmatic cytokines. PLoS One 2020; 15:e0238370. [PMID: 32886687 PMCID: PMC7473588 DOI: 10.1371/journal.pone.0238370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/15/2020] [Indexed: 11/18/2022] Open
Abstract
Maternal health and nutritional status before and during gestation may affect neonates' immune system and energy balance as they develop. The objective of this study was to associate certain clinical markers of maternal adiposity (body mass index and gestational weight gain) and neonatal adiposity (birth weight, abdominal circumference, and waist/height index) with the levels of pro- and anti-inflammatory cytokines in umbilical cord blood at birth: IL-1β, IL-1Rα, IL-4, IL-6, IL-10, IFN-γ, and TNF-α. An exploratory cross-sectional study was conducted with a convenience sample of women from one hospital recruited shortly before giving birth through scheduled cesarean section. Of 31 the pregnant women who agreed to participate and met the inclusion criteria, twenty-nine newborns from these women were analyzed. Three cases of tobacco smoking during pregnancy were identified as an unexpected maternal risk factor and were included in the analysis. Typical of the population treated at this hospital, ten of our participants had diabetes during pregnancy, and nine of them had a pre-pregnancy BMI> 25. Non-parametric statistical analyses and a generalized linear model with gamma scale response with a log link were performed. Results: Correlation analyses, differences in medians, and a prediction model all showed positive and significant results between cytokine levels in cord blood and neonatal abdominal circumference, birth weight, and waist-height index. For maternal variables, smoking during pregnancy showed significant associations with cytokine levels in cord blood. Conclusion: This study found a variety of associations suggesting that increased neonatal adiposity increases pro-inflammatory cytokine levels at birth.
Collapse
Affiliation(s)
- Maria Hernandez-Trejo
- Neurobiología del Desarrollo, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
- * E-mail:
| | - Reyna Sámano
- Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
| | - Gabriela Chico-Barba
- Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
| | - Maria Luisa Pizano-Zarate
- Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
| | - Norma Estela Herrera-González
- Sección de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México, México
| |
Collapse
|
37
|
Walejko JM, Chelliah A, Keller-Wood M, Wasserfall C, Atkinson M, Gregg A, Edison AS. Diabetes Leads to Alterations in Normal Metabolic Transitions of Pregnancy as Revealed by Time-Course Metabolomics. Metabolites 2020; 10:E350. [PMID: 32867274 PMCID: PMC7570364 DOI: 10.3390/metabo10090350] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/15/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Women with diabetes during pregnancy are at increased risk of poor maternal and neonatal outcomes. Despite this, the effects of pre-gestational (PGDM) or gestational diabetes (GDM) on metabolism during pregnancy are not well understood. In this study, we utilized metabolomics to identify serum metabolic changes in women with and without diabetes during pregnancy and the cord blood at birth. We observed elevations in tricarboxylic acid (TCA) cycle intermediates, carbohydrates, ketones, and lipids, and a decrease in amino acids across gestation in all individuals. In early gestation, PGDM had elevations in branched-chain amino acids and sugars compared to controls, whereas GDM had increased lipids and decreased amino acids during pregnancy. In both GDM and PGDM, carbohydrate and amino acid pathways were altered, but in PGDM, hemoglobin A1c and isoleucine were significantly increased compared to GDM. Cord blood from GDM and PGDM newborns had similar increases in carbohydrates and choline metabolism compared to controls, and these alterations were not maternal in origin. Our results revealed that PGDM and GDM have distinct metabolic changes during pregnancy. A better understanding of diabetic metabolism during pregnancy can assist in improved management and development of therapeutics and help mitigate poor outcomes in both the mother and newborn.
Collapse
Affiliation(s)
- Jacquelyn M. Walejko
- Department of Biochemistry & Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Anushka Chelliah
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Texas Health Science Center at Houston, UT Health, Houston, TX 77030, USA;
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA;
| | - Clive Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL 32610, USA; (C.W.); (M.A.)
| | - Mark Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL 32610, USA; (C.W.); (M.A.)
| | - Anthony Gregg
- Department of Obstetrics and Gynecology, Baylor University, Dallas, TX 75246, USA;
| | - Arthur S. Edison
- Departments of Genetics and Biochemistry & Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
38
|
Zhang T, Xia Y, Han TL, Zhang H, Baker PN. Five serum fatty acids are associated with subclinical hypothyroidism in a Chinese pregnant population. Sci Rep 2020; 10:6743. [PMID: 32317737 PMCID: PMC7174292 DOI: 10.1038/s41598-020-63513-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 03/23/2020] [Indexed: 12/11/2022] Open
Abstract
Subclinical hypothyroidism (SCH) is a common endocrine disorder affecting women of reproductive age. Although SCH and abnormal fatty acid composition are often associated with adverse pregnancy outcomes and metabolic syndrome later in maternal and fetal life, the longitudinal relationship between SCH and serum fatty acids during pregnancy has rarely been studied. Therefore, the aim of this study was to investigate the association between SCH and maternal serum fatty acids throughout gestation. A total of 240 women enrolled in the Complex Lipids in Mothers and Babies (CLIMB) study in Chongqing, China were included in our study. Clinical information and maternal serum samples were collected at three time points during pregnancy: 11-14th, 22-28th, and 32-34th weeks of gestation. Twenty serum fatty acids were quantified using gas chromatography-mass spectrometry (GC-MS) analysis. A majority of the 20 serum fatty acids increased as gestation progressed in women with a normal pregnancy and women experiencing SCH. Levels of arachidic acid, docosahexaenoic acid, and eicosenoic acid were significantly higher in the serum of women with SCH when compared to women with a normal pregnancy, in the second trimester. On the other hand, the levels of eicosadienoic acid and octadecanoic acid were significantly higher in SCH in the third trimester. Our findings demonstrate that serum fatty acid composition during the second and third trimesters was significantly associated with SCH in pregnant Chinese women.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing, 400016, China
| | - Yinyin Xia
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, China
| | - Ting-Li Han
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China.
- Liggins Institute, The University of Auckland, Auckland, 1023, New Zealand.
| | - Hua Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing, 400016, China.
| | - Philip N Baker
- College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK
| |
Collapse
|
39
|
Contreras-Duarte S, Carvajal L, Garchitorena MJ, Subiabre M, Fuenzalida B, Cantin C, Farías M, Leiva A. Gestational Diabetes Mellitus Treatment Schemes Modify Maternal Plasma Cholesterol Levels Dependent to Women´s Weight: Possible Impact on Feto-Placental Vascular Function. Nutrients 2020; 12:E506. [PMID: 32079298 PMCID: PMC7071311 DOI: 10.3390/nu12020506] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/11/2020] [Indexed: 12/19/2022] Open
Abstract
: Gestational diabetes mellitus (GDM) associates with fetal endothelial dysfunction (ED), which occurs independently of adequate glycemic control. Scarce information exists about the impact of different GDM therapeutic schemes on maternal dyslipidemia and obesity and their contribution to the development of fetal-ED. The aim of this study was to evaluate the effect of GDM-treatments on lipid levels in nonobese (N) and obese (O) pregnant women and the effect of maternal cholesterol levels in GDM-associated ED in the umbilical vein (UV). O-GDM women treated with diet showed decreased total cholesterol (TC) and low-density lipoproteins (LDL) levels with respect to N-GDM ones. Moreover, O-GDM women treated with diet in addition to insulin showed higher TC and LDL levels than N-GDM women. The maximum relaxation to calcitonin gene-related peptide of the UV rings was lower in the N-GDM group compared to the N one, and increased maternal levels of TC were associated with even lower dilation in the N-GDM group. We conclude that GDM-treatments modulate the TC and LDL levels depending on maternal weight. Additionally, increased TC levels worsen the GDM-associated ED of UV rings. This study suggests that it could be relevant to consider a specific GDM-treatment according to weight in order to prevent fetal-ED, as well as to consider the possible effects of maternal lipids during pregnancy.
Collapse
Affiliation(s)
- Susana Contreras-Duarte
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
| | - Lorena Carvajal
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
| | - María Jesús Garchitorena
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
| | - Mario Subiabre
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
| | - Bárbara Fuenzalida
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
| | - Claudette Cantin
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
| | - Marcelo Farías
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
| | - Andrea Leiva
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (L.C.); (M.J.G.); (M.S.); (B.F.); (C.C.); (M.F.)
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago 8330024, Chile
| |
Collapse
|
40
|
Alahakoon TI, Medbury HJ, Williams H, Lee VW. Lipid profiling in maternal and fetal circulations in preeclampsia and fetal growth restriction-a prospective case control observational study. BMC Pregnancy Childbirth 2020; 20:61. [PMID: 32000699 PMCID: PMC6993402 DOI: 10.1186/s12884-020-2753-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 01/20/2020] [Indexed: 12/18/2022] Open
Abstract
Background While many risk factors for preeclampsia, such as increased body mass index, advanced maternal age, chronic hypertension, diabetes, are now established in clinical practice, maternal lipid profile has not been included in the risk assessment for preeclampsia. We aim to characterize the serum levels of Total Cholesterol (TC), High density lipoprotein (HDL), Low density lipoprotein (LDL), Triglycerides (TG), Apolipoprotein A1, Apolipoprotein B and their ratios TC/HDL and ApoB/ApoA1 in the maternal and fetal circulations of normal pregnancy, preeclampsia (PE), fetal growth restriction (FGR) and PE + FGR. Methods A prospective cross-sectional case control study was conducted measuring maternal and fetal lipid levels by enzymatic analysis and immune-turbidimetric enzymatic assays. FGR was defined by elevated umbilical artery Doppler resistance in association with estimated fetal weight < 10%. Kruskal Wallis non-parametric analysis of variance was used to test for homogeneity across the clinical groups for each of the variables, Mann-Whitney tests for pairwise comparisons and Spearman rank correlation were used to quantify gestational age-related changes. Results (1) TG levels were elevated in maternal PE and cord blood PE + FGR groups compared to normal pregnancies. (2) A statistically significant elevation of fetal ApoB levels was observed in PE, FGR and PE + FGR compared to normal pregnancies. Apolipoprotein levels A1 and B were not different between maternal groups. (3) TC, HDL, LDL and TC/HDL levels did not show any significant gestational variation or between clinical groups in the maternal or fetal circulation. Conclusions Elevation in maternal TG levels may have a role in the pathogenesis of PE. The implications of elevated maternal and fetal TG levels and elevated fetal Apolipoprotein B levels deserves further exploration of their role in long term cardiovascular risk in the mother as well as the offspring.
Collapse
Affiliation(s)
- Thushari I Alahakoon
- University of Sydney, Sydney Medical School, Sydney, NSW, Australia. .,Westmead Institute for Maternal and Fetal Medicine, Westmead Hospital, Westmead, NSW, Australia.
| | - Heather J Medbury
- University of Sydney, Sydney Medical School, Sydney, NSW, Australia.,Department of Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Helen Williams
- University of Sydney, Sydney Medical School, Sydney, NSW, Australia.,Department of Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Vincent W Lee
- University of Sydney, Sydney Medical School, Sydney, NSW, Australia.,Department of Renal Medicine, Westmead Hospital and University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
41
|
Dumolt JH, Ma M, Mathew J, Patel MS, Rideout TC. Gestational hypercholesterolemia alters fetal hepatic lipid metabolism and microRNA expression in Apo-E-deficient mice. Am J Physiol Endocrinol Metab 2019; 317:E831-E838. [PMID: 31453710 PMCID: PMC6879864 DOI: 10.1152/ajpendo.00138.2019] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Maternal hypercholesterolemia (MHC) is a pathological condition characterized by an exaggerated rise in maternal serum cholesterol during gestation, which can alter offspring hepatic lipid metabolism. However, the extent that these maladaptations occur during gestation and the molecular mechanisms involved remain unknown. MicoRNAs (miRNA) are small, noncoding RNAs that contribute to the development and progression of nonalcoholic fatty liver disease. Therefore, we sought to determine the degree to which in utero exposure to excessive cholesterol affects fetal hepatic lipid metabolism and miRNA expression. Twelve female apoE-/- mice were randomly assigned to two different chow-based diets throughout gestation: control (CON) or the CON diet with cholesterol (0.15%). MHC reduced maternal fecundity and reduced litter size and weight. On gestational day 18, fetuses from MHC dams possessed increased placental cholesterol and hepatic triglycerides (TG), which were accompanied by a downregulation in the expression of hepatic lipogenic and TG synthesis and transport genes. Furthermore, fetal livers from MHC mothers showed increased miRNA-27a and reduced miRNA-200c expression. In summary, in utero exposure to MHC alters fetal lipid metabolism and lends mechanistic insight that implicates early changes in miRNA expression that may link to later-life programming of disease risk.
Collapse
Affiliation(s)
- Jerad H Dumolt
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Min Ma
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Joyce Mathew
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Mulchand S Patel
- Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| |
Collapse
|
42
|
Kadakia R, Talbot O, Kuang A, Bain JR, Muehlbauer MJ, Stevens RD, Ilkayeva OR, Lowe LP, Metzger BE, Newgard CB, Scholtens DM, Lowe WL. Cord Blood Metabolomics: Association With Newborn Anthropometrics and C-Peptide Across Ancestries. J Clin Endocrinol Metab 2019; 104:4459-4472. [PMID: 31498869 PMCID: PMC6735762 DOI: 10.1210/jc.2019-00238] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/28/2019] [Indexed: 12/18/2022]
Abstract
CONTEXT Newborn adiposity is associated with childhood obesity. Cord blood metabolomics is one approach that can be used to understand early-life contributors to adiposity and insulin resistance. OBJECTIVE To determine the association of cord blood metabolites with newborn adiposity and hyperinsulinemia in a multiethnic cohort of newborns. DESIGN Cross-sectional, observational study. SETTING Hyperglycemia and Adverse Pregnancy Outcome study. PARTICIPANTS One thousand six hundred multiethnic mother-newborn pairs. MAIN OUTCOME MEASURE Cord blood C-peptide, birthweight, and newborn sum of skinfolds. RESULTS Meta-analyses across four ancestry groups (Afro-Caribbean, Northern European, Thai, and Mexican American) demonstrated significant associations of cord blood metabolites with cord blood C-peptide, birthweight, and newborn sum of skinfolds. Several metabolites, including branched-chain amino acids (BCAAs), medium- and long-chain acylcarnitines, nonesterified fatty acids, and triglycerides were negatively associated with cord C-peptide but positively associated with birthweight and/or sum of skinfolds. 1,5-Anhydroglucitol, an inverse marker of recent maternal glycemia, was significantly inversely associated with birthweight and sum of skinfolds. Network analyses revealed groups of interrelated amino acid, acylcarnitine, and fatty acid metabolites associated with all three newborn outcomes. CONCLUSIONS Cord blood metabolites are associated with newborn size and cord blood C-peptide levels after adjustment for maternal body mass index and glucose during pregnancy. Negative associations of metabolites with C-peptide at birth were observed. 1,5-Anhydroglucitol appears to be a marker of adiposity in newborns. BCAAs were individually associated with birthweight and demonstrated possible associations with newborn adiposity in network analyses.
Collapse
Affiliation(s)
- Rachel Kadakia
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
- Correspondence and Reprint Requests: William L. Lowe, Jr., MD, Feinberg School of Medicine, Northwestern University, Rubloff Building, 12th Floor, 420 East Superior Street, Chicago, Ilinois 60611.
| | - Octavious Talbot
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Alan Kuang
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Robert D Stevens
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | - Lynn P Lowe
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Boyd E Metzger
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Durham, North Carolina
- Duke University School of Medicine, Durham, North Carolina
| | | | - William L Lowe
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | |
Collapse
|
43
|
Sureshchandra S, Marshall NE, Messaoudi I. Impact of pregravid obesity on maternal and fetal immunity: Fertile grounds for reprogramming. J Leukoc Biol 2019; 106:1035-1050. [PMID: 31483523 DOI: 10.1002/jlb.3ri0619-181r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022] Open
Abstract
Maternal pregravid obesity results in several adverse health outcomes during pregnancy, including increased risk of gestational diabetes, preeclampsia, placental abruption, and complications at delivery. Additionally, pregravid obesity and in utero exposure to high fat diet have been shown to have detrimental effects on fetal programming, predisposing the offspring to adverse cardiometabolic, endocrine, and neurodevelopmental outcomes. More recently, a deeper appreciation for the modulation of offspring immunity and infectious disease-related outcomes by maternal pregravid obesity has emerged. This review will describe currently available animal models for studying the impact of maternal pregravid obesity on fetal immunity and review the data from clinical and animal model studies. We also examine the burden of pregravid obesity on the maternal-fetal interface and the link between placental and systemic inflammation. Finally, we discuss future studies needed to identify key mechanistic underpinnings that link maternal inflammatory changes and fetal cellular reprogramming events.
Collapse
Affiliation(s)
- Suhas Sureshchandra
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Nicole E Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
44
|
Grunewald M, Hellmuth C, Kirchberg FF, Mearin ML, Auricchio R, Castillejo G, Korponay-Szabo IR, Polanco I, Roca M, Vriezinga SL, Werkstetter K, Koletzko B, Demmelmair H. Variation and Interdependencies of Human Milk Macronutrients, Fatty Acids, Adiponectin, Insulin, and IGF-II in the European PreventCD Cohort. Nutrients 2019; 11:E2034. [PMID: 31480373 PMCID: PMC6770528 DOI: 10.3390/nu11092034] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/17/2019] [Accepted: 08/23/2019] [Indexed: 01/16/2023] Open
Abstract
Human milk composition is variable. The identification of influencing factors and interdependencies of components may help to understand the physiology of lactation. In this study, we analyzed linear trends in human milk composition over time, the variation across different European countries and the influence of maternal celiac disease. Within a multicenter European study exploring potential prevention of celiac disease in a high-risk population (PreventCD), 569 human milk samples were donated by women from five European countries between 16 and 163 days postpartum. Some 202 mothers provided two samples at different time points. Protein, carbohydrates, fat and fatty acids, insulin, adiponectin, and insulin-like growth factor II (IGF-II) were analyzed. Milk protein and n-6 long chain polyunsaturated fatty acids decreased during the first three months of lactation. Fatty acid composition was significantly influenced by the country of residence. IGF-II and adiponectin concentrations correlated with protein content (r = 0.24 and r = 0.35), and IGF-II also correlated with fat content (r = 0.36), suggesting a possible regulatory role of IGF in milk macronutrient synthesis. Regarding the impact of celiac disease, only the level in palmitic acid was influenced by this disease, suggesting that breastfeeding by celiac disease mothers should not be discouraged.
Collapse
Affiliation(s)
- Maria Grunewald
- Ludwig-Maximilians-Universität, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany
| | - Christian Hellmuth
- Ludwig-Maximilians-Universität, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany
| | - Franca F Kirchberg
- Ludwig-Maximilians-Universität, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany
| | - Maria Luisa Mearin
- Department of Paediatrics, Leiden University Medical Center, 2300 Leiden, The Netherlands
| | - Renata Auricchio
- Department of Medical Translational Sciences and European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, 80131 Naples, Italy
| | - Gemma Castillejo
- Department of Pediatric Gastroenterology Unit, Hospital Universitari Sant Joan de Reus, URV, IIPV, 43201 Reus, Spain
| | | | - Isabel Polanco
- Department of Pediatric Gastroenterology and Nutrition, La Paz University Hospital, 28033 Madrid, Spain
| | - Maria Roca
- U. Enfermedad Celiaca e Inmunopatología Digestiva, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Sabine L Vriezinga
- Department of Paediatrics, Leiden University Medical Center, 2300 Leiden, The Netherlands
| | - Katharina Werkstetter
- Ludwig-Maximilians-Universität, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany
| | - Berthold Koletzko
- Ludwig-Maximilians-Universität, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany.
| | - Hans Demmelmair
- Ludwig-Maximilians-Universität, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany.
| |
Collapse
|
45
|
Polyphenols and IUGR Pregnancies: Effects of Maternal Hydroxytyrosol Supplementation on Hepatic Fat Accretion and Energy and Fatty Acids Profile of Fetal Tissues. Nutrients 2019; 11:nu11071534. [PMID: 31284510 PMCID: PMC6682965 DOI: 10.3390/nu11071534] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
Maternal supplementation with hydroxytyrosol, a polyphenol present in olive leaves and fruits, is a highly promising strategy to improve the oxidative and metabolic status of fetuses at risk of intrauterine growth restriction, which may diminish the appearance of low-birth-weight neonates. The present study aimed to determine whether hydroxytyrosol, by preventing lipid peroxidation, may influence the fat accretion and energy homeostasis in the liver, as well as the fatty acid composition in the liver and muscle. The results indicate that hydroxytyrosol treatment significantly decreased the energy content of the fetal liver, without affecting fat accretion, and caused significant changes in the availability of fatty acids. There were significant increases in the amount of total polyunsaturated fatty acids, omega-3 and omega-6, which are highly important for adequate fetal tissue development. However, there were increases in the omega-6/omega-3 ratio and the desaturation index, which make further studies necessary to determine possible effects on the pro/anti-inflammatory status of the fetuses.
Collapse
|
46
|
Xue Y, Guo C, Hu F, Zhu W, Mao S. Maternal undernutrition induces fetal hepatic lipid metabolism disorder and affects the development of fetal liver in a sheep model. FASEB J 2019; 33:9990-10004. [PMID: 31167079 DOI: 10.1096/fj.201900406r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Undernutrition accelerates body fat mobilization to alleviate negative energy balance, which disrupts homeostasis of lipid metabolism in maternal liver. However, little is known about its effect on fetal metabolism and development. Here, a sheep model was used to explore whether maternal undernutrition induces fetal lipid metabolism disorder and further inhibits fetal hepatic development. Twenty pregnant ewes were either fed normally or restricted to 30% level for 15 d, after which fetal hepatic samples were collected to conduct transcriptome, metabolome, histomorphology, and biochemical analysis. Results showed that maternal undernutrition altered the general transcriptome profile and metabolic mode in fetal liver. Fatty acid oxidation and ketogenesis were enhanced in fetal livers of undernourished ewes, which might be promoted by the activated peroxisome proliferator-activated receptor α signaling pathway, whereas cholesterol, steroid, and fatty acid synthesis were repressed. Maternal undernutrition increased triglyceride synthesis, decreased triglyceride degradation, and inhibited phospholipid degradation and synthesis in fetal liver. In addition, our data revealed that maternal undernutrition extremely inhibited DNA replication, cell cycle progression, and antiapoptosis and broke the balance between cell proliferation and apoptosis in fetal liver, indicating that maternal undernutrition affects the growth and development of fetal liver. Generally, these findings provide evidence that maternal undernutrition during pregnancy disturbs fetal lipid metabolism and inhibits fetal hepatic development in sheep, which greatly contribute to the further study of fetal metabolism and development in human beings.-Xue, Y., Guo, C., Hu, F., Zhu, W., Mao, S. Maternal undernutrition induces fetal hepatic lipid metabolism disorder and affects the development of fetal liver in a sheep model.
Collapse
Affiliation(s)
- Yanfeng Xue
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Changzheng Guo
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Fan Hu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Shengyong Mao
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
47
|
Ferchaud-Roucher V, Barner K, Jansson T, Powell TL. Maternal obesity results in decreased syncytiotrophoblast synthesis of palmitoleic acid, a fatty acid with anti-inflammatory and insulin-sensitizing properties. FASEB J 2019; 33:6643-6654. [PMID: 30811959 PMCID: PMC6463919 DOI: 10.1096/fj.201802444r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022]
Abstract
The fetus is dependent on delivery of fatty acids (FAs) by the syncytiotrophoblast, the transporting epithelium of the human placenta. Obese pregnant women have dyslipidemia; however, whether obesity impacts placental lipid transport and metabolism remains to be fully established. Palmitoleic acid (POA), an FA with anti-inflammatory and insulin-sensitizing properties, is synthesized from palmitic acid (PA) catalyzed by stearoyl-coenzyme A desaturase (SCD) activity. We hypothesized that the uptake and incorporation of FAs and POA synthesis are reduced in primary human trophoblasts (PHTs) isolated from pregnancies complicated by maternal obesity. Villous cytotrophoblasts were isolated from 7 placentas of obese [body mass index (BMI) = 37.5 ± 1.9] and 12 normal (BMI = 23.6 ± 0.6) mothers. FA uptake and incorporation were assessed using uniformly labeled (U[13C])-FA mixtures of PA, oleic acid (OA), linoleic acid, and docosahexaenoic acid. Cellular [13C] FAs were quantified both in total cellular lipids and in lipid classes by GC-MS. Uptake and incorporation of [13C] FAs in total cellular lipids were not different in PHTs isolated from obese mothers compared with normal mothers. Only the concentration of OA was increased in the triglyceride fraction (P < 0.05) if the mother was obese. We found an isotopic enrichment of POA after U[13C]-PA treatment, demonstrating SCD activity in PHT cells. Labeled POA content and the POA:PA ratio were significantly lower in PHTs isolated from placentas of obese mothers compared with normal, healthy controls. Decreased syncytiotrophoblast POA synthesis may contribute to insulin resistance and low-grade inflammation in the mother, placenta, or fetus (or a combination of the 3) in pregnancies complicated by obesity.-Ferchaud-Roucher, V., Barner, K., Jansson, T., Powell, T. L. Maternal obesity results in decreased syncytiotrophoblast synthesis of palmitoleic acid, a fatty acid with anti-inflammatory and insulin-sensitizing properties.
Collapse
Affiliation(s)
- Véronique Ferchaud-Roucher
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kelsey Barner
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Theresa L. Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
48
|
Abstract
Background: Although pregestational obesity has been associated with increased risk of adverse fetal outcome, the mechanisms behind are not known. We aimed to investigate the influence of the maternal metabolic state on fetal outcome in rats exposed to either a high-fat diet (HFD) or a control diet (CD). We also investigated the impact of serum collected from HFD/CD pregnant rats on CD embryonic development in whole-embryo cultures. Material and methods: On gestational day 0, 9, 10, or 20 maternal plasma/serum samples were collected as pregnancies were terminated for the estimations of maternal metabolic state and embryo-fetal development. We measured embryonic gene expression of ROS scavenger enzymes as well as genes involved in inflammation in maternal adipose tissue. Results: In HFD maternal plasma/serum, concentrations of glucose, β-hydroxybutyrate, branched-chain amino acids, and leptin were increased, whereas those of triacylglycerol, cholesterol, and palmitic, oleic, linoleic, and α-linolenic acids were decreased. Gene expression of CuZnSOD, IL-6, IL-10, and resistin was increased in HFD maternal adipose tissue, whereas that of CuZnSOD and MnSOD was decreased in HFD-exposed embryos. HFD caused retention of most fatty acids in the maternal liver as well. Conclusion: HFD alters the maternal metabolic state, increases fetal resorptions in vivo, and increases the rate of fetal/embryonic malformations both in vivo and in vitro. These findings suggest that metabolic disturbances in HFD pregnant rats have profound adverse developmental effects in the offspring.
Collapse
Affiliation(s)
- Parri Wentzel
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- CONTACT Parri Wentzel Department of Medical Cell Biology, Uppsala University, PO Box 571, Biomedical Center, SE-751 23Uppsala, Sweden
| | - Ulf J. Eriksson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Emilio Herrera
- Department of Biochemistry, CEU San Pablo University, Madrid, Spain
| |
Collapse
|
49
|
Kadakia R, Nodzenski M, Talbot O, Kuang A, Bain JR, Muehlbauer MJ, Stevens RD, Ilkayeva OR, O'Neal SK, Lowe LP, Metzger BE, Newgard CB, Scholtens DM, Lowe WL. Maternal metabolites during pregnancy are associated with newborn outcomes and hyperinsulinaemia across ancestries. Diabetologia 2019; 62:473-484. [PMID: 30483859 PMCID: PMC6374187 DOI: 10.1007/s00125-018-4781-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/26/2018] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS We aimed to determine the association of maternal metabolites with newborn adiposity and hyperinsulinaemia in a multi-ethnic cohort of mother-newborn dyads. METHODS Targeted and non-targeted metabolomics assays were performed on fasting and 1 h serum samples from a total of 1600 mothers in four ancestry groups (Northern European, Afro-Caribbean, Mexican American and Thai) who participated in the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study, underwent an OGTT at ~28 weeks gestation and whose newborns had anthropometric measurements at birth. RESULTS In this observational study, meta-analyses demonstrated significant associations of maternal fasting and 1 h metabolites with birthweight, cord C-peptide and/or sum of skinfolds across ancestry groups. In particular, maternal fasting triacylglycerols were associated with newborn sum of skinfolds. At 1 h, several amino acids, fatty acids and lipid metabolites were associated with one or more newborn outcomes. Network analyses revealed clusters of fasting acylcarnitines, amino acids, lipids and fatty acid metabolites associated with cord C-peptide and sum of skinfolds, with the addition of branched-chain and aromatic amino acids at 1 h. CONCLUSIONS/INTERPRETATION The maternal metabolome during pregnancy is associated with newborn outcomes. Maternal levels of amino acids, acylcarnitines, lipids and fatty acids and their metabolites during pregnancy relate to fetal growth, adiposity and cord C-peptide, independent of maternal BMI and blood glucose levels.
Collapse
Affiliation(s)
- Rachel Kadakia
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Ann and Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Avenue, Box 54, Chicago, IL, 60611, USA.
| | - Michael Nodzenski
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Octavious Talbot
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alan Kuang
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
- Duke University School of Medicine, Durham, NC, USA
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
- Duke University School of Medicine, Durham, NC, USA
| | - Robert D Stevens
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
- Duke University School of Medicine, Durham, NC, USA
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
- Duke University School of Medicine, Durham, NC, USA
| | - Sara K O'Neal
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
- Duke University School of Medicine, Durham, NC, USA
| | - Lynn P Lowe
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Boyd E Metzger
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
- Duke University School of Medicine, Durham, NC, USA
| | | | - William L Lowe
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | |
Collapse
|
50
|
Layton J, Powe C, Allard C, Battista MC, Doyon M, Bouchard L, Perron P, Wessel J, Hivert MF. Maternal lipid profile differs by gestational diabetes physiologic subtype. Metabolism 2019; 91:39-42. [PMID: 30468781 PMCID: PMC8058811 DOI: 10.1016/j.metabol.2018.11.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/02/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023]
Abstract
AIM To characterize lipid profiles in women with different gestational diabetes mellitus (GDM) physiologic subtypes. METHODS We measured seven lipid markers (total cholesterol, LDL, HDL, triglycerides, non-esterified fatty acids (NEFA), ApoA, ApoB) in fasting plasma collected in a prospective cohort of 805 pregnant women during second trimester. We estimated insulin sensitivity and secretion using oral glucose tolerance test-based validated indices. We categorized GDM physiologic subtypes by insulin sensitivity and secretion defects defined as values below the 25th percentile among women with normal glucose tolerance (NGT), as previously established. We compared lipid markers across NGT and GDM subtypes. We explored associations between lipid markers and newborn anthropometry in the overall group and stratified by glucose tolerance status. RESULTS Among 805 women, 67 (8.3%) developed GDM. Women with GDM had higher body mass index (BMI; 29.3 vs. 26.6 kg/m2), while ethnicity (97.3% vs. 97.0% European ancestry) and age (28 vs. 29 years) were similar. In comparison to women with NGT, women with GDM characterized by a predominant insulin sensitivity defect had significantly higher triglycerides (2.20 vs. 1.82, P = 0.002), lower HDL (1.64 vs. 1.90, P = 0.01) and higher NEFA (0.34 vs. 0.24, P < 0.0001). GDM women with a predominant insulin secretion defect differed from women with NGT with respect to NEFA (0.32 vs. 0.24, P = 0.003) while other lipid markers were similar. These associations remained significant after adjusting for maternal age and BMI. Greater maternal levels of NEFA were associated with higher birth weight z-scores in women with an insulin secretion defect (BMI-adjusted r = 0.58, P = 0.01). We did not find significant associations between other lipid markers and newborn anthropometry in other groups. CONCLUSION Women with GDM have distinct lipid profiles based on their GDM physiologic subtype which may not be apparent when investigating GDM as a single group.
Collapse
Affiliation(s)
- Jill Layton
- Department of Epidemiology, Indiana University Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Camille Powe
- Diabetes Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Catherine Allard
- Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CHUS), Québec, Canada
| | - Marie-Claude Battista
- Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CHUS), Québec, Canada
| | - Myriam Doyon
- Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CHUS), Québec, Canada
| | - Luigi Bouchard
- Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CHUS), Québec, Canada
| | - Patrice Perron
- Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CHUS), Québec, Canada
| | - Jennifer Wessel
- Department of Epidemiology, Indiana University Fairbanks School of Public Health, Indianapolis, IN, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Diabetes Translational Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marie-France Hivert
- Diabetes Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA, USA; Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CHUS), Québec, Canada; Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|