1
|
Schliefsteiner C, Wadsack C, Allerkamp HH. Exploring the Lifeline: Unpacking the Complexities of Placental Vascular Function in Normal and Preeclamptic Pregnancies. Compr Physiol 2024; 14:5763-5787. [PMID: 39699084 DOI: 10.1002/cphy.c230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
The proper development and function of the placenta are essential for the success of pregnancy and the well-being of both the fetus and the mother. Placental vascular function facilitates efficient fetal development during pregnancy by ensuring adequate gas exchange with low vascular resistance. This review focuses on how placental vascular function can be compromised in the pregnancy pathology preeclampsia, and conversely, how placental vascular dysfunction might contribute to this condition. While the maternal endothelium is widely recognized as a key focus in preeclampsia research, this review emphasizes the importance of understanding how this condition affects the development and function of the fetal placental vasculature. The placental vascular bed, consisting of microvasculature and macrovasculature, is discussed in detail, as well as structural and functional changes associated with preeclampsia. The complexity of placental vascular reactivity and function, its mediators, its impact on placental exchange and blood distribution, and how these factors are most affected in early-onset preeclampsia are further explored. These factors include foremost lipoproteins and their cargo, oxygen levels and oxidative stress, biomechanics, and shear stress. Challenges in studying placental pathophysiology are discussed, highlighting the necessity of innovative research methodologies, including ex vivo experiments, in vivo imaging tools, and computational modeling. Finally, an outlook on the potential of drug interventions targeting the placental endothelium to improve placental vascular function in preeclampsia is provided. Overall, this review highlights the need for further research and the development of models and tools to better understand and address the challenges posed by preeclampsia and its effects on placental vascular function to improve short- and long-term outcomes for the offspring of preeclamptic pregnancies. © 2024 American Physiological Society. Compr Physiol 14:5763-5787, 2024.
Collapse
Affiliation(s)
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Hanna H Allerkamp
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| |
Collapse
|
2
|
Zhou J, Jiang S, Liu D, Li X, Zhou Z, Wang Z, Wang H. Bidirectional Mendelian Randomization Analysis of Genetic Proxies of Plasma Fatty Acids and Pre-Eclampsia Risk. Nutrients 2024; 16:3748. [PMID: 39519582 PMCID: PMC11547509 DOI: 10.3390/nu16213748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Previous studies have reported associations between fatty acids and the risk of pre-eclampsia. However, the causality of these associations remains uncertain. This study postulates a causal relationship between specific plasma fatty acids and pre-eclampsia or other maternal hypertensive disorders (PE-HTPs). To test this hypothesis, two-sample bidirectional Mendelian randomization (MR) analyses were employed to determine the causality effects. METHODS Single-nucleotide polymorphisms associated with PE-HTPs and fatty acids were obtained from a genome-wide association study (GWAS) of European ancestry. Bidirectional MR analyses were conducted using methods such as inverse variance weighted, MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analyses, including tests for heterogeneity, horizontal pleiotropy, and co-localization, were conducted to assess the robustness of MR results. RESULTS The analyses revealed causal relationships between PE-HTPs and several fatty acids, including monounsaturated fatty acid (MUFA), omega-6 fatty acid (n-6 FA), linoleic acid (LA), docosahexaenoic acid (DHA), and the PUFA/MUFA ratio. Genetically predicted higher risk of PE-HTPs was significantly associated with lower plasma n-6 FA (OR = 0.96, 95% CI: 0.93-0.99), particularly LA (OR = 0.95, 95% CI: 0.92-0.98). Conversely, increased DHA (OR = 0.86, 95% CI: 0.78-0.96) and a higher PUFA/MUFA ratio (OR = 0.86, 95% CI: 0.76-0.98) were associated with a reduced risk of PE-HTPs. Elevated MUFA levels (OR = 1.12, 95% CI: 1.00-1.25) were related to an increased risk. CONCLUSIONS This study provides robust genetic evidence supporting bidirectional causal relationships between PE-HTPs and specific plasma fatty acids, underscoring the critical role of fatty acid metabolism in maternal hypertensive disorders.
Collapse
Affiliation(s)
- Jingqi Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.Z.); (S.J.); (X.L.); (Z.Z.)
| | - Shuo Jiang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.Z.); (S.J.); (X.L.); (Z.Z.)
| | - Dangyun Liu
- Department of Central Laboratory, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an 223300, China;
| | - Xinyi Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.Z.); (S.J.); (X.L.); (Z.Z.)
| | - Ziyi Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.Z.); (S.J.); (X.L.); (Z.Z.)
| | - Zhiheng Wang
- Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Hui Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.Z.); (S.J.); (X.L.); (Z.Z.)
| |
Collapse
|
3
|
Origüela V, Ferrer-Aguilar P, Gázquez A, Pérez-Cruz M, Gómez-Roig MD, Gómez-Llorente C, Larqué E. Placental MFSD2A expression in fetal growth restriction and maternal and fetal DHA status. Placenta 2024; 150:31-38. [PMID: 38583303 DOI: 10.1016/j.placenta.2024.04.002] [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: 01/22/2024] [Revised: 03/20/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Fetal growth restriction (FGR) may affect placental transfer of key nutrients to the fetus, such as the fatty acid docosahexaenoic acid (DHA). Major facilitator superfamily domain containing 2A (MFSD2A) has been described as a specific DHA carrier in placenta, but its expression has not been studied in FGR. The aim of this study was to evaluate for the first time the placental MFSD2A levels in late-FGR pregnancies and the maternal and cord plasma DHA. METHODS 87 pregnant women from a tertial reference center were classified into late-FGR (N = 18) or control (N = 69). Fatty acid profile was determined in maternal and cord venous plasma, as well as placental levels of MFSD2A and of insulin mediators like phospho-protein kinase B (phospho-AKT) and phospho-extracellular regulated kinase (phospho-ERK). RESULTS Maternal fatty acid profile did not differ between groups. Nevertheless, late-FGR cord vein presented higher content of saturated fatty acids than control, producing a concomitant decrease in the percentage of some unsaturated fatty acids. In the late-FGR group, a lower DHA fetal/maternal ratio was observed when using percentages, but not with concentrations. No alterations were found in the expression of MFSD2A in late-FGR placentas, nor in phospho-AKT or phospho-ERK. DISCUSSION MFSD2A protein expression was not altered in late-FGR placentas, in line with no differences in cord DHA concentration between groups. The increase in the saturated fatty acid content of late-FGR cord might be a compensatory mechanism to ensure fetal energy supply, decreasing other fatty acids percentage. Future studies are warranted to elucidate if altered saturated fatty acid profile in late-FGR fetuses might predispose them to postnatal catch-up and to long-term health consequences.
Collapse
Affiliation(s)
- Valentina Origüela
- Department of Physiology, Faculty of Biology, University of Murcia, Campus of Espinardo, 30100, Murcia, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120, Murcia, Spain
| | - Patricia Ferrer-Aguilar
- BCNatal, Barcelona Centre for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, University of Barcelona, 08950, Barcelona, Spain; Institute of Research Sant Joan de Déu, 08950, Barcelona, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS), RD21/0012/0003, Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain
| | - Antonio Gázquez
- Department of Physiology, Faculty of Biology, University of Murcia, Campus of Espinardo, 30100, Murcia, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120, Murcia, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS), RD21/0012/0003, Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain
| | - Miriam Pérez-Cruz
- BCNatal, Barcelona Centre for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, University of Barcelona, 08950, Barcelona, Spain; Institute of Research Sant Joan de Déu, 08950, Barcelona, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS), RD21/0012/0003, Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain
| | - María Dolores Gómez-Roig
- BCNatal, Barcelona Centre for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, University of Barcelona, 08950, Barcelona, Spain; Institute of Research Sant Joan de Déu, 08950, Barcelona, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS), RD21/0012/0003, Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain
| | - Carolina Gómez-Llorente
- Institute of Biosanitary Research ibs.GRANADA, 18012, Granada, Spain; Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, 18071, Granada, Spain; Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, 18100, Granada, Spain; Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERObn), CB12/03/30038, Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain
| | - Elvira Larqué
- Department of Physiology, Faculty of Biology, University of Murcia, Campus of Espinardo, 30100, Murcia, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120, Murcia, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS), RD21/0012/0003, Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain.
| |
Collapse
|
4
|
Rabinowich A, Avisdris N, Zilberman A, Link-Sourani D, Lazar S, Herzlich J, Specktor-Fadida B, Joskowicz L, Malinger G, Ben-Sira L, Hiersch L, Ben Bashat D. Reduced adipose tissue in growth-restricted fetuses using quantitative analysis of magnetic resonance images. Eur Radiol 2023; 33:9194-9202. [PMID: 37389606 DOI: 10.1007/s00330-023-09855-y] [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: 12/08/2022] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 07/01/2023]
Abstract
OBJECTIVES Fat-water MRI can be used to quantify tissues' lipid content. We aimed to quantify fetal third trimester normal whole-body subcutaneous lipid deposition and explore differences between appropriate for gestational age (AGA), fetal growth restriction (FGR), and small for gestational age fetuses (SGAs). METHODS We prospectively recruited women with FGR and SGA-complicated pregnancies and retrospectively recruited the AGA cohort (sonographic estimated fetal weight [EFW] ≥ 10th centile). FGR was defined using the accepted Delphi criteria, and fetuses with an EFW < 10th centile that did not meet the Delphi criteria were defined as SGA. Fat-water and anatomical images were acquired in 3 T MRI scanners. The entire fetal subcutaneous fat was semi-automatically segmented. Three adiposity parameters were calculated: fat signal fraction (FSF) and two novel parameters, i.e., fat-to-body volume ratio (FBVR) and estimated total lipid content (ETLC = FSF*FBVR). Normal lipid deposition with gestation and differences between groups were assessed. RESULTS Thirty-seven AGA, 18 FGR, and 9 SGA pregnancies were included. All three adiposity parameters increased between 30 and 39 weeks (p < 0.001). All three adiposity parameters were significantly lower in FGR compared with AGA (p ≤ 0.001). Only ETLC and FSF were significantly lower in SGA compared with AGA using regression analysis (p = 0.018-0.036, respectively). Compared with SGA, FGR had a significantly lower FBVR (p = 0.011) with no significant differences in FSF and ETLC (p ≥ 0.053). CONCLUSIONS Whole-body subcutaneous lipid accretion increased throughout the third trimester. Reduced lipid deposition is predominant in FGR and may be used to differentiate FGR from SGA, assess FGR severity, and study other malnourishment pathologies. CLINICAL RELEVANCE STATEMENT Fetuses with growth restriction have reduced lipid deposition than appropriately developing fetuses measured using MRI. Reduced fat accretion is linked with worse outcomes and may be used for growth restriction risk stratification. KEY POINTS • Fat-water MRI can be used to assess the fetal nutritional status quantitatively. • Lipid deposition increased throughout the third trimester in AGA fetuses. • FGR and SGA have reduced lipid deposition compared with AGA fetuses, more predominant in FGR.
Collapse
Affiliation(s)
- Aviad Rabinowich
- Sagol Brain Institute, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.
- Department of Radiology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Netanell Avisdris
- Sagol Brain Institute, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ayala Zilberman
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Obstetrics and Gynecology, Lis Hospital for Women, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | | | - Sapir Lazar
- Department of Radiology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jacky Herzlich
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Neonatal Intensive Care Unit, Dana Dwek Children's Hospital, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Bella Specktor-Fadida
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leo Joskowicz
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gustavo Malinger
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Obstetrics and Gynecology, Lis Hospital for Women, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Liat Ben-Sira
- Department of Radiology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Liran Hiersch
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Obstetrics and Gynecology, Lis Hospital for Women, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Dafna Ben Bashat
- Sagol Brain Institute, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
5
|
Gibbs RL, Swanson RM, Beard JK, Hicks ZM, Most MS, Beer HN, Grijalva PC, Clement SM, Marks-Nelson ES, Schmidt TB, Petersen JL, Yates DT. Daily injection of the β2 adrenergic agonist clenbuterol improved poor muscle growth and body composition in lambs following heat stress-induced intrauterine growth restriction. Front Physiol 2023; 14:1252508. [PMID: 37745251 PMCID: PMC10516562 DOI: 10.3389/fphys.2023.1252508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Intrauterine growth restriction (IUGR) is associated with reduced β2 adrenergic sensitivity, which contributes to poor postnatal muscle growth. The objective of this study was to determine if stimulating β2 adrenergic activity postnatal would rescue deficits in muscle growth, body composition, and indicators of metabolic homeostasis in IUGR offspring. Methods: Time-mated ewes were housed at 40°C from day 40 to 95 of gestation to produce IUGR lambs. From birth, IUGR lambs received daily IM injections of 0.8 μg/kg clenbuterol HCl (IUGR+CLEN; n = 11) or saline placebo (IUGR; n = 12). Placebo-injected controls (n = 13) were born to pair-fed thermoneutral ewes. Biometrics were assessed weekly and body composition was estimated by ultrasound and bioelectrical impedance analysis (BIA). Lambs were necropsied at 60 days of age. Results: Bodyweights were lighter (p ≤ 0.05) for IUGR and IUGR+CLEN lambs than for controls at birth, day 30, and day 60. Average daily gain was less (p ≤ 0.05) for IUGR lambs than controls and was intermediate for IUGR+CLEN lambs. At day 58, BIA-estimated whole-body fat-free mass and ultrasound-estimated loin eye area were less (p ≤ 0.05) for IUGR but not IUGR+CLEN lambs than for controls. At necropsy, loin eye area and flexor digitorum superficialis muscles were smaller (p ≤ 0.05) for IUGR but not IUGR+CLEN lambs than for controls. Longissimus dorsi protein content was less (p ≤ 0.05) and fat-to-protein ratio was greater (p ≤ 0.05) for IUGR but not IUGR+CLEN lambs than for controls. Semitendinosus from IUGR lambs had less (p ≤ 0.05) β2 adrenoreceptor content, fewer (p ≤ 0.05) proliferating myoblasts, tended to have fewer (p = 0.08) differentiated myoblasts, and had smaller (p ≤ 0.05) muscle fibers than controls. Proliferating myoblasts and fiber size were recovered (p ≤ 0.05) in IUGR+CLEN lambs compared to IUGR lambs, but β2 adrenoreceptor content and differentiated myoblasts were not recovered. Semitendinosus lipid droplets were smaller (p ≤ 0.05) in size for IUGR lambs than for controls and were further reduced (p ≤ 0.05) in size for IUGR+CLEN lambs. Conclusion: These findings show that clenbuterol improved IUGR deficits in muscle growth and some metabolic parameters even without recovering the deficit in β2 adrenoreceptor content. We conclude that IUGR muscle remained responsive to β2 adrenergic stimulation postnatal, which may be a strategic target for improving muscle growth and body composition in IUGR-born offspring.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Dustin T. Yates
- Stress Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
6
|
Chan M, Preston EV, Fruh V, Quinn MR, Hacker MR, Wylie BJ, O'Brien K, Williams PL, Hauser R, James-Todd T, Mahalingaiah S. Use of personal care products during pregnancy and birth outcomes - A pilot study. ENVIRONMENTAL RESEARCH 2023; 225:115583. [PMID: 36868449 PMCID: PMC10153796 DOI: 10.1016/j.envres.2023.115583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/10/2023] [Accepted: 02/25/2023] [Indexed: 05/04/2023]
Abstract
Prenatal exposure to endocrine disrupting chemicals (EDCs) from personal care products may be associated with birth outcomes including preterm birth and low birth weight. There is limited research examining the role of personal care product use during pregnancy on birth outcomes. Our pilot study consisted of 164 participants in the Environmental Reproductive and Glucose Outcomes (ERGO) study (Boston, MA), with data on self-reported personal care product use at four study visits throughout pregnancy (product use in the 48 h before a study visit and hair product use in the month before a study visit). We used covariate-adjusted linear regression models to estimate differences in mean gestational age at delivery, birth length, and sex-specific birth weight-for-gestational age (BW-for-GA) Z-score based on personal care product use. Hair product use in the past month prior to certain study visits was associated with decreased mean sex-specific BW-for-GA Z-scores. Notably, hair oil use in the month prior to study visit 1 was associated with a lower mean BW-for-GA Z-score (V1: -0.71, 95% confidence interval: -1.12, -0.29) compared to non-use. Across all study visits (V1-V4), increased mean birth length was observed among nail polish users vs. non-users. In comparison, decreased mean birth length was observed among shave cream users vs. non-users. Liquid soap, shampoo, and conditioner use at certain study visits were significantly associated with higher mean birth length. Suggestive associations were observed across study visits for other products including hair gel/spray with BW-for-GA Z-score and liquid/bar soap with gestational age. Overall, use of a variety of personal care products throughout pregnancy was observed to be associated with our birth outcomes of interest, notably hair oil use during early pregnancy. These findings may help inform future interventions/clinical recommendations to reduce exposures linked to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Marissa Chan
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| | - Emma V Preston
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Victoria Fruh
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, United States
| | - Marlee R Quinn
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Michele R Hacker
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Blair J Wylie
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Karen O'Brien
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Paige L Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Tamarra James-Todd
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Shruthi Mahalingaiah
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Obstetrics & Gynecology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
7
|
Grohmann RM, Marçal VMG, Corazza IC, Peixoto AB, Júnior EA, Nardozza LMM. Maternal Blood Fatty Acid Levels in Fetal Growth Restriction. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2023; 45:127-133. [PMID: 37105196 PMCID: PMC10139773 DOI: 10.1055/s-0043-1768455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
OBJECTIVE To assess the maternal blood levels of fatty acids (FAs) in pregnancies with fetal growth restriction (FGR). METHODS This prospective cross-sectional study included pregnant women with gestational age between 26 and 37 + 6 weeks with FGR and appropriate for gestational age (AGA) fetuses. The levels of saturated, trans, monounsaturated, and polyunsaturated FAs were measured using centrifugation and liquid chromatography. The Student's t-test, Mann-Whitney test, and general linear model, with gestational age and maternal weight as covariants, were used to compare FA levels and the FGR and AGA groups. The Chi-square was used to evaluate the association between groups and studied variables. RESULTS Maternal blood sample was collected from 64 pregnant women, being 24 FGR and 40 AGA. A weak positive correlation was found between the palmitoleic acid level and maternal weight (r = 0.285, p = 0.036). A weak negative correlation was found between the gamma-linoleic acid level and gestational age (r = - 0.277, p = 0.026). The median of the elaidic acid level (2.3 vs. 4.7 ng/ml, p = 0.045) and gamma-linoleic acid (6.3 vs. 6.6 ng/ml, p = 0.024) was significantly lower in the FGR than the AGA group. The palmitoleic acid level was significantly higher in the FGR than AGA group (50.5 vs. 47.6 ng/ml, p = 0.033). CONCLUSION Pregnant women with FGR had lower elaidic acid and gamma-linoleic acid levels and higher palmitoleic acid levels than AGA fetuses.
Collapse
Affiliation(s)
- Raquel Margiotte Grohmann
- Department of Obstetrics, Paulista School of Medicine-Federal University of São Paulo, São Paulo, SP, Brazil
| | - Vivian Macedo Gomes Marçal
- Department of Obstetrics, Paulista School of Medicine-Federal University of São Paulo, São Paulo, SP, Brazil
- Department of Obstetrics and Gynecology, Medical Science College of Santos, Santos, SP, Brazil
| | - Isabela César Corazza
- Department of Obstetrics, Paulista School of Medicine-Federal University of São Paulo, São Paulo, SP, Brazil
| | - Alberto Borges Peixoto
- Department of Gynecology and Obstetrics, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
- Gynecology and Obstetrics Service, Mário Palmério University Hospital, University of Uberaba, Uberaba, MG, Brazil
| | - Edward Araujo Júnior
- Department of Obstetrics, Paulista School of Medicine-Federal University of São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
8
|
Syring JG, Crouse MS, Neville TL, Ward AK, Dahlen CR, Reynolds LP, Borowicz PP, McLean KJ, Neville BW, Caton JS. Concentrations of vitamin B12 and folate in maternal serum and fetal fluids, metabolite interrelationships, and hepatic transcript abundance of key folate and methionine cycle genes: the impacts of maternal nutrition during the first 50 d of gestation. J Anim Sci 2023; 101:skad139. [PMID: 37129588 PMCID: PMC10199783 DOI: 10.1093/jas/skad139] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
Adequate maternal nutrition is key for proper fetal development and epigenetic programming. One-carbon metabolites (OCM), including vitamin B12, folate, choline, and methionine, play a role in epigenetic mechanisms associated with developmental programming. This study investigated the presence of B12 and folate in maternal serum, allantoic fluid (ALF), and amniotic fluid (AMF), as well as how those concentrations in all three fluids correlate to the concentrations of methionine-folate cycle intermediates in heifers receiving either a control (CON) or restricted (RES) diet for the first 50 d of gestation and fetal hepatic gene expression for methionine-folate cycle enzymes. Angus cross heifers (n = 43) were estrus synchronized, bred via artificial insemination with semen from a single sire, and randomly assigned to one of two nutrition treatments (CON = 20, RES = 23). Heifers were ovariohysterectomized on either day 16 (n = 14), 34 (n = 15), or 50 of gestation (n = 14), where samples of maternal serum (n = 42), ALF (n = 29), and AMF (n = 11) were collected and analyzed for concentrations of folate and B12. Concentrations of B12 and folate in ALF were greater (P < 0.05) in RES compared to CON. For ALF, folate concentrations were also greater (P < 0.01) on day 34 compared to day 50. There was a significant (P = 0.04) nutrition × fluid interaction for B12 concentrations where concentrations were greatest in restricted ALF, intermediate in control ALF, and lowest in CON and RES serum and AMF. Folate concentrations were greatest (P < 0.01) in ALF, intermediate in serum, and lowest in AMF. Additionally, positive correlations (P < 0.05) were found between ALF and AMF folate concentrations and AMF concentrations of methionine, serine, and glycine. Negative correlations (P < 0.05) between AMF folate and serum homocysteine were also observed. Both positive and negative correlations (P < 0.05) depending on the fluid evaluated were found between B12 and methionine, serine, and glycine concentrations. There was a downregulation (P = 0.05) of dihydrofolate reductase and upregulation (P = 0.03) of arginine methyltransferase 7 gene expression in RES fetal liver samples compared with CON fetal liver on day 50. Combined, these data show restricted maternal nutrition results in increased B12 and folate concentrations present in fetal fluids, and increased expression of genes for enzymes within one-carbon metabolism.
Collapse
Affiliation(s)
- Jessica G Syring
- Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Matthew S Crouse
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
| | - Tammi L Neville
- Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Alison K Ward
- Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Carl R Dahlen
- Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Lawrence P Reynolds
- Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Pawel P Borowicz
- Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Kyle J McLean
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Bryan W Neville
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
| | - Joel S Caton
- Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| |
Collapse
|
9
|
de Alwis N, Beard S, Binder NK, Pritchard N, Kaitu’u-Lino TJ, Walker SP, Stock O, Groom K, Petersen S, Henry A, Said JM, Seeho S, Kane SC, Tong S, Hui L, Hannan NJ. Placental OLAH Levels Are Altered in Fetal Growth Restriction, Preeclampsia and Models of Placental Dysfunction. Antioxidants (Basel) 2022; 11:1677. [PMID: 36139751 PMCID: PMC9495588 DOI: 10.3390/antiox11091677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Previously, we identified elevated transcripts for the gene Oleoyl-ACP Hydrolase (OLAH) in the maternal circulation of pregnancies complicated by preterm fetal growth restriction. As placental dysfunction is central to the pathogenesis of both fetal growth restriction and preeclampsia, we aimed to investigate OLAH levels and function in the human placenta. We assessed OLAH mRNA expression (qPCR) throughout pregnancy, finding placental expression increased as gestation progressed. OLAH mRNA and protein levels (Western blot) were elevated in placental tissue from cases of preterm preeclampsia, while OLAH protein levels in placenta from growth-restricted pregnancies were comparatively reduced in the preeclamptic cohort. OLAH expression was also elevated in placental explant tissue, but not isolated primary cytotrophoblast cultured under hypoxic conditions (as models of placental dysfunction). Further, we discovered that silencing cytotrophoblast OLAH reduced the expression of pro- and anti-apoptosis genes, BAX and BCL2, placental growth gene, IGF2, and oxidative stress gene, NOX4. Collectively, these findings suggest OLAH could play a role in placental dysfunction and may be a therapeutic target for mitigating diseases associated with this vital organ. Further research is required to establish the role of OLAH in the placenta, and whether these changes may be a maternal adaptation or consequence of disease.
Collapse
Affiliation(s)
- Natasha de Alwis
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sally Beard
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Natalie K. Binder
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Natasha Pritchard
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Tu’uhevaha J. Kaitu’u-Lino
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Susan P. Walker
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Owen Stock
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katie Groom
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Scott Petersen
- Centre for Maternal Fetal Medicine, Mater Mothers’ Hospital, South Brisbane, QLD 4101, Australia
| | - Amanda Henry
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Joanne M. Said
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Maternal Fetal Medicine, Joan Kirner Women’s & Children’s Sunshine Hospital, St Albans, VIC 3021, Australia
| | - Sean Seeho
- Women and Babies Research, Sydney Medical School-Northern, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW 2065, Australia
| | - Stefan C. Kane
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Maternal Fetal Medicine, Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | - Stephen Tong
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Lisa Hui
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Natalie J. Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
10
|
Maternal Fibroblast Growth Factor 21 Levels Decrease during Early Pregnancy in Normotensive Pregnant Women but Are Higher in Preeclamptic Women-A Longitudinal Study. Cells 2022; 11:cells11142251. [PMID: 35883694 PMCID: PMC9322099 DOI: 10.3390/cells11142251] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/11/2022] [Accepted: 07/17/2022] [Indexed: 12/04/2022] Open
Abstract
(1) Background: Fibroblast growth factor 21 (FGF-21) is an endocrine factor involved in glucose and lipid metabolism that exerts pleiotropic effects. The aim of this study was to investigate the serum FGF-21 profile in healthy and mild preeclamptic pregnant women at each trimester of pregnancy; (2) Methods: Serum FGF-21 levels were determined by ELISA in a nested case-control study within a longitudinal cohort study that included healthy (n = 54) and mild preeclamptic (n = 20) pregnant women, women at three months after delivery (n = 20) and eumenorrheic women during the menstrual cycle (n = 20); (3) Results: FGF-21 levels were significantly lower in the mid-luteal phase compared to the early follicular phase of the menstrual cycle in eumenorrheic women (p < 0.01). Maternal levels of FGF-21 were significantly lower in the first and second trimesters and peaked during the third trimester in healthy pregnant women (p < 0.01). Serum levels of FGF-21 in healthy pregnant were significantly lower in the first and second trimester of pregnancy compared with the follicular phase of the menstrual cycle and postpartum (p < 0.01). Serum FGF-21 levels were significantly higher in preeclamptic compared to healthy pregnant women during pregnancy (p < 0.01); (4) Conclusions: These results suggest that a peak of FGF-21 towards the end of pregnancy in healthy pregnancy and higher levels in preeclamptic women might play a critical role that contributes to protecting against the negatives effects of high concentrations of non-esterified fatty acids (NEFA) and hypertensive disorder. Furthermore, FGF-21 might play an important role in reproductive function in healthy eumenorrheic women during the menstrual cycle.
Collapse
|
11
|
Gopalakrishnan K, Mishra JS, Ross JR, Abbott DH, Kumar S. Hyperandrogenism diminishes maternal-fetal fatty acid transport by increasing FABP 4-mediated placental lipid accumulation. Biol Reprod 2022; 107:514-528. [PMID: 35357467 DOI: 10.1093/biolre/ioac059] [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: 01/18/2022] [Revised: 02/22/2022] [Accepted: 03/17/2022] [Indexed: 11/12/2022] Open
Abstract
Long-chain polyunsaturated fatty acids (LCPUFAs) are critical for fetal brain development. Infants born to preeclamptic mothers or those born growth restricted due to placental insufficiency have reduced LCPUFA, and are at higher risk for developing neurodevelopmental disorders. Since plasma levels of testosterone (T) and fatty acid-binding protein 4 (FABP4) are elevated in preeclampsia, we hypothesized that elevated T induces the expression of FABP4 in the placenta leading to compromised transplacental transport of LCPUFAs. Increased maternal T in pregnant rats significantly decreased n-3 and n-6 LCPUFA levels in maternal and fetal circulation, but increased their placental accumulation. Dietary LCPUFAs supplementation in T dams increased LCPUFA levels in the maternal circulation and further augmented placental storage, while failing to increase fetal levels. The placenta in T dams exhibited increased FABP4 mRNA and protein levels. In vitro, T dose-dependently upregulated FABP4 transcription in trophoblasts. T stimulated androgen receptor (AR) recruitment to the androgen response element and trans-activated FABP4 promoter activity, both of which were abolished by AR antagonist. T in pregnant rats and cultured trophoblasts significantly reduced transplacental transport of C14-docosahexaenoic acid (DHA) and increased C14-DHA accumulation in the placenta. Importantly, FABP4-overexpression by itself in pregnant rats and trophoblasts increased transplacental transport of C14-DHA with no significant placental accumulation. T exposure, in contrast, inhibited this FABP4-mediated effect by promoting C14-DHA placental accumulation. In summary, our studies show that maternal hyperandrogenism increases placental FABP4 expression via transcriptional upregulation and preferentially routes LCPUFAs toward cellular storage in the placenta leading to offspring lipid deficiency.
Collapse
Affiliation(s)
- Kathirvel Gopalakrishnan
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Jordan R Ross
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - David H Abbott
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA.,Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA.,Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA.,Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA
| |
Collapse
|
12
|
Mauro AK, Rengarajan A, Albright C, Boeldt DS. Fatty acids in normal and pathological pregnancies. Mol Cell Endocrinol 2022; 539:111466. [PMID: 34610360 DOI: 10.1016/j.mce.2021.111466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/26/2022]
Abstract
Long chain fatty acids, namely omega-3 and omega-6, are essential fatty acids and are necessary for proper pregnancy progression and fetal growth and development. Maternal fatty acid consumption and release of fatty acids from lipid stores provide increased availability of fatty acids for the placenta to transport to the growing fetus. Both omega-3 and omega-6 fatty acids are then utilized for generation of signaling molecules, such as eicosanoids, and for promoting of growth and developmental, most notably in the nervous system. Perturbations in fatty acid concentration and fatty acid signaling have been implicated in three major pregnancy complications - gestational diabetes, preeclampsia, and preterm birth. In this review we discuss the growing literature surrounding the role of fatty acids in normal and pathological pregnancies. Differences in maternal, placental, and fetal fatty acids and molecular regulation of fatty acid signaling and transport are presented. A look into novel fatty acid-based therapies for each of the highlighted disorders are discussed, and may present exciting bench to bedside alternatives to traditional pharmacological intervention.
Collapse
Affiliation(s)
- Amanda K Mauro
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Aishwarya Rengarajan
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Carly Albright
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Derek S Boeldt
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA.
| |
Collapse
|
13
|
Cheng JC, Fang L, Li Y, Thakur A, Hoodless PA, Guo Y, Wang Z, Wu Z, Yan Y, Jia Q, Gao Y, Han X, Yu Y, Sun YP. G protein-coupled estrogen receptor stimulates human trophoblast cell invasion via YAP-mediated ANGPTL4 expression. Commun Biol 2021; 4:1285. [PMID: 34773076 PMCID: PMC8589964 DOI: 10.1038/s42003-021-02816-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Insufficient invasion of trophoblast cells into the uterine decidua is associated with preeclampsia (PE). G protein-coupled estrogen receptor (GPER) is a membrane estrogen receptor involved in non-genomic estrogen signaling. GPER is expressed in human trophoblast cells and downregulated GPER levels are noted in PE. However, to date, the role of GPER in trophoblast cells remains largely unknown. Here, we applied RNA sequencing (RNA-seq) to HTR-8/SVneo human trophoblast cells in response to G1, an agonist of GPER, and identified angiopoietin-like 4 (ANGPTL4) as a target gene of GPER. Treatment of trophoblast cells with G1 or 17β-estradiol (E2) activated Yes-associated protein (YAP), the major downstream effector of the Hippo pathway, via GPER but in a mammalian STE20-like protein kinase 1 (MST1)-independent manner. Using pharmacological inhibitors as well as loss- and gain-of-function approaches, our results revealed that YAP activation was required for GPER-stimulated ANGPTL4 expression. Transwell invasion assays demonstrated that activation of GPER-induced ANGPTL4 promoted cell invasion. In addition, the expression levels of GPER, YAP, and ANGPTL4 were downregulated in the placenta of patients with PE. Our findings reveal a mechanism by which GPER exerts its stimulatory effect on human trophoblast cell invasion by upregulating YAP-mediated ANGPTL4 expression. Cheng, Fan, Li et al. identified ANGPTL4 as a G1-induced target gene of GPER/YAP in HRT8 cells using RNA-seq and highlighted its importance in regulating trophoblast cell invasion. The authors also reported GPER downregulation in the placenta and lower estradiol levels in patients who developed preeclampsia.
Collapse
Affiliation(s)
- Jung-Chien Cheng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| | - Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Yuxi Li
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Avinash Thakur
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada, V5Z 1L3.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Pamela A Hoodless
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada, V5Z 1L3.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Yanjie Guo
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Zhen Wang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Ze Wu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Yang Yan
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Qiongqiong Jia
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Yibo Gao
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Xiaoyu Han
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Yiping Yu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Ying-Pu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| |
Collapse
|
14
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
15
|
Yang C, Park S, Song G, Lim W. Inhibition of the cleaved half of tRNA Gly enhances palmitic acid-induced apoptosis in human trophoblasts. J Nutr Biochem 2021; 99:108866. [PMID: 34563666 DOI: 10.1016/j.jnutbio.2021.108866] [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: 02/05/2021] [Revised: 07/11/2021] [Accepted: 09/08/2021] [Indexed: 11/15/2022]
Abstract
Palmitic acid (PA) induces apoptosis in the human trophoblast cell line HTR8/SVneo. However, the molecular mechanism underlying this effect remains unclear. Although small noncoding RNAs are involved in trophoblast growth and invasion during early pregnancy, the functional roles of tRNA-derived species are currently unknown. Therefore, the purpose of this study was to examine the involvement of tRNA-derived species in PA-induced apoptosis in human trophoblasts. In this study, we investigate the expression and function of tRNA-derived stress-induced RNAs (tiRNAs) in HTR8/SVneo. We determined the expression of tiRNAs in HTR8/SVneo cells in response to PA. Then, we transfected inhibitor of target tiRNA in HTR8/SVneo with or without PA to examine the tRNA-derived species-regulated intracellular signal transduction by detecting calcium homeostasis, mitochondrial membrane potential, and signaling proteins. We found that the expression of tRNAGly-derived tiRNAs decreased in PA-treated human trophoblasts. Moreover, inhibition of tiRNAGlyCCC/GCC enhanced the PA-induced apoptosis along with the induction of DNA fragmentation and mitochondrial depolarization. Inhibition of tiRNAGlyCCC/GCC enhanced the expression of endoplasmic reticulum stress-related proteins and increased Ca2+ levels in the cytoplasm and mitochondria. Moreover, the levels of cytochrome c released from the mitochondria were synergistically affected by tiRNAGlyCCC/GCC inhibitor and PA. Furthermore, artificial regulation of ANG inhibited the expression of tiRNAGlyCCC/GCC and similar effects were observed upon the inhibition of tiRNAGlyCCC/GCC in human trophoblasts. These results suggest that tiRNAGlyCCC/GCC might be the molecule via which PA induces its effects in human trophoblasts.
Collapse
Affiliation(s)
- Changwon Yang
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Sunwoo Park
- Department of Plant & Biomaterials Science, Gyeongsang National University, Jinju-si, Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea.
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul, Republic of Korea.
| |
Collapse
|
16
|
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: 5.0] [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
|
17
|
Youssef L, Simões RV, Miranda J, García-Martín ML, Paules C, Crovetto F, Amigó N, Cañellas N, Gratacos E, Crispi F. Paired maternal and fetal metabolomics reveal a differential fingerprint in preeclampsia versus fetal growth restriction. Sci Rep 2021; 11:14422. [PMID: 34257400 PMCID: PMC8277896 DOI: 10.1038/s41598-021-93936-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023] Open
Abstract
Preeclampsia (PE) and fetal growth restriction (FGR) are both placenta-mediated disorders with unclear pathogenesis. Metabolomics of maternal and fetal pairs might help in understanding these disorders. We recruited prospectively pregnancies with normotensive FGR, PE without FGR, PE + FGR and uncomplicated pregnancies as controls. Nuclear magnetic resonance metabolomics were applied on plasma samples collected at delivery. Advanced lipoprotein, glycoprotein and choline profiling was performed using the Liposcale test. The software package Dolphin was used to quantify 24 low-molecular-weight metabolites. Statistical analysis comprised the comparison between each group of complicated pregnancies versus controls, considering 5% false discovery rate correction. Lipid profiles were altered in accordance with the clinical presentation of these disorders. Specifically, PE mothers and FGR fetuses (with or without FGR or PE, respectively) exhibited a pro-atherogenic and pro-inflammatory profile, with higher concentrations of triglycerides, remnant cholesterol (VLDL, IDL) and Glc/GalNAc-linked and lipid-associated glycoproteins compared to controls. Low-molecular-weight metabolites were extensively disturbed in preeclamptic mothers, with or without FGR. Growth restricted fetuses in the presence of PE showed changes in low-molecular-weight metabolites similar to their mothers (increased creatine and creatinine), while normotensive FGR fetuses presented scarce differences, consistent with undernutrition (lower isoleucine). Further research is warranted to clarify maternal and fetal adaptations to PE and FGR.
Collapse
Affiliation(s)
- Lina Youssef
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Rui V Simões
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.
- Champalimaud Research, Champalimaud Centre for the Unknown, Av Brasília, 1400-038, Lisbon, Portugal.
| | - Jezid Miranda
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - María Luisa García-Martín
- BIONAND, Andalusian Centre for Nanomedicine and Biotechnology (Junta de Andalucía- Universidad de Málaga), Málaga, Spain
| | - Cristina Paules
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Nuria Amigó
- Biosfer Teslab, Reus, Spain
- Department of Basic Medical Sciences, University Rovira I Virgili, CIBERDEM, Reus, Spain
| | - Nicolau Cañellas
- Universidad Rovira I Virgili, DEEEiA, IISPV, Tarragona, Spain
- CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Eduard Gratacos
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Fatima Crispi
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| |
Collapse
|
18
|
Gómez-Vilarrubla A, Mas-Parés B, Díaz M, Xargay-Torrent S, Carreras-Badosa G, Jové M, Martin-Gari M, Bonmatí-Santané A, de Zegher F, Ibañez L, López-Bermejo A, Bassols J. Fatty acids in the placenta of appropiate- versus small-for-gestational-age infants at term birth. Placenta 2021; 109:4-10. [PMID: 33895685 DOI: 10.1016/j.placenta.2021.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/24/2021] [Accepted: 04/15/2021] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Fatty acids are essential nutrients for the fetus and are supplied by the mother through the placenta. Desaturase and elongase enzymes play an important role in modulating the fatty acid composition of body tissues. We aimed to compare the fatty acid profile and the estimated desaturase and elongase activities in the placenta of appropriate (AGA) versus small-for-gestational-age (SGA), and to determine their relationship with the offspring size at birth. METHODS The placental fatty acid profile was analyzed by gas chromatography in 84 infants (45 AGA and 30 SGA) from a prenatal cohort study. The estimated desaturase and elongase activities were calculated from product-precursor fatty acid ratios. Results were associated with maternal (age, body mass index and weight gain during gestation) and neonatal (gestational age, sex, birth weight and birth length) parameters. RESULTS Differences in placental fatty acid composition between AGA and SGA infants rather than correlations thereof with neonatal parameters were observed. Placentas from SGA infants contained lower levels of omega-3 (ALA, EPA, DPA, and DHA) and high omega-6/omega-3 ratios (AA/DHA and LA/ALA), as well as low elongase (Elovl5) and high desaturase (D9Dn7 and D5Dn6) activity as compared to AGA infants (all p < 0.0001). DISCUSSION Placentas of AGA and SGA infants differed in fatty acids profile as well as in estimated desaturase and elongase activities. A striking feature of SGA placentas was the low availability of omega-3. Hence, omega-3 fatty acid status deserves further attention, as a potential target of prenatal interventions.
Collapse
Affiliation(s)
- Ariadna Gómez-Vilarrubla
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190, Salt, Spain
| | - Berta Mas-Parés
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190, Salt, Spain
| | - Marta Díaz
- Endocrinology, Pediatric Research Institute, Sant Joan de Déu Children's Hospital, 08950, Esplugues, Barcelona, Spain; CIBERDEM (Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders), ISCIII, 28029, Madrid, Spain
| | - Sílvia Xargay-Torrent
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190, Salt, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190, Salt, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, Spain
| | - Meritxell Martin-Gari
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, Spain
| | | | - Francis de Zegher
- Department of Development & Regeneration, University of Leuven, 3000, Leuven, Belgium
| | - Lourdes Ibañez
- Endocrinology, Pediatric Research Institute, Sant Joan de Déu Children's Hospital, 08950, Esplugues, Barcelona, Spain; CIBERDEM (Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders), ISCIII, 28029, Madrid, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190, Salt, Spain; Department of Pediatrics, Dr. Josep Trueta Hospital, 17007, Girona, Spain.
| | - Judit Bassols
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190, Salt, Spain.
| |
Collapse
|
19
|
Desoye G, Herrera E. Adipose tissue development and lipid metabolism in the human fetus: The 2020 perspective focusing on maternal diabetes and obesity. Prog Lipid Res 2020; 81:101082. [PMID: 33383022 DOI: 10.1016/j.plipres.2020.101082] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
During development, the human fetus accrues the highest proportion of fat of all mammals. Precursors of fat lobules can be found at week 14 of pregnancy. Thereafter, they expand, filling with triacylglycerols during pregnancy. The resultant mature lipid-filled adipocytes emerge from a developmental programme of embryonic stem cells, which is regulated differently than adult adipogenesis. Fetal triacylglycerol synthesis uses glycerol and fatty acids derived predominantly from glycolysis and lipogenesis in liver and adipocytes. The fatty acid composition of fetal adipose tissue at the end of pregnancy shows a preponderance of palmitic acid, and differs from the mother. Maternal diabetes mellitus does not influence this fatty acid profile. Glucose oxidation is the main source of energy for the fetus, but mitochondrial fatty acid oxidation also contributes. Indirect evidence suggests the presence of lipoprotein lipase in fetal adipose tissue. Its activity may be increased under hyperinsulinemic conditions as in maternal diabetes mellitus and obesity, thereby contributing to increased triacylglycerol deposition found in the newborns of such pregnancies. Fetal lipolysis is low. Changes in the expression of genes controlling metabolism in fetal adipose tissue appear to contribute actively to the increased neonatal fat mass found in diabetes and obesity. Many of these processes are under endocrine regulation, principally by insulin, and show sex-differences. Novel fatty acid derived signals such as oxylipins are present in cord blood with as yet undiscovered function. Despite many decades of research on fetal lipid deposition and metabolism, many key questions await answers.
Collapse
Affiliation(s)
- G Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria.
| | - E Herrera
- Faculties of Pharmacy and Medicine, University CEU San Pablo, Madrid, Spain.
| |
Collapse
|
20
|
Godhamgaonkar AA, Wadhwani NS, Joshi SR. Exploring the role of LC-PUFA metabolism in pregnancy complications. Prostaglandins Leukot Essent Fatty Acids 2020; 163:102203. [PMID: 33227645 DOI: 10.1016/j.plefa.2020.102203] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/09/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022]
Abstract
Maternal nutrition during pregnancy plays a significant role in growth and development of the placenta and influencing pregnancy outcome. Suboptimal nutritional status during early gestational period compromises the normal course of pregnancy leading to adverse maternal and fetal outcomes. Omega-3 and omega-6 long chain polyunsaturated fatty acids (LC-PUFA) are important for the growth and development of the placenta. Maternal fatty acids and their metabolites influence the normal course of pregnancy by regulating cell growth and development, cell signaling, regulate angiogenesis, modulate inflammatory responses and influence various structural and functional processes. Alterations in LC-PUFA and their metabolites may result in inadequate spiral artery remodeling or placental angiogenesis leading to structural and functional deficiency of the placenta which contributes to several pregnancy complications like preeclampsia, gestational diabetes mellitus, intrauterine growth restriction, and results in adverse birth outcomes. In this review, we summarize studies examining the role of fatty acids and their metabolites in pregnancy. We also discuss the possible molecular mechanisms through which LC-PUFA influences placental growth and development. Studies have demonstrated that omega-3 fatty acid supplementation lowers the incidence of preterm births, but its effect on reducing pregnancy complications are inconclusive.
Collapse
Affiliation(s)
- Aditi A Godhamgaonkar
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India
| | - Nisha S Wadhwani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India.
| |
Collapse
|
21
|
Metabolic reprogramming by Zika virus provokes inflammation in human placenta. Nat Commun 2020; 11:2967. [PMID: 32528049 PMCID: PMC7290035 DOI: 10.1038/s41467-020-16754-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
The recent outbreak of Zika virus (ZIKV) was associated with birth defects and pregnancy loss when maternal infection occurs in early pregnancy, but specific mechanisms driving placental insufficiency and subsequent ZIKV-mediated pathogenesis remain unclear. Here we show, using large scale metabolomics, that ZIKV infection reprograms placental lipidome by impairing the lipogenesis pathways. ZIKV-induced metabolic alterations provide building blocks for lipid droplet biogenesis and intracellular membrane rearrangements to support viral replication. Furthermore, lipidome reprogramming by ZIKV is paralleled by the mitochondrial dysfunction and inflammatory immune imbalance, which contribute to placental damage. In addition, we demonstrate the efficacy of a commercially available inhibitor in limiting ZIKV infection, provides a proof-of-concept for blocking congenital infection by targeting metabolic pathways. Collectively, our study provides mechanistic insights on how ZIKV targets essential hubs of the lipid metabolism that may lead to placental dysfunction and loss of barrier function. Zika virus (ZIKV) infection of pregnant women is associated with pregnancy loss and birth defects, but molecular insights for the aetiology are scarce. Here the authors show that ZIKV reprograms the host lipidome to facilitate viral replication, induce mitochondria dysfunction, and cause immune imbalance, thereby identifying a potential target for ZIKV therapy.
Collapse
|
22
|
Abstract
The composition of human milk is the result of the evolution of mammals over millions of years. Among the most important components of milk are fatty acids. Approximately 85% are saturated and monounsaturated fatty acids - the rest are polyunsaturated one. Their role is to provide energy and immunity and to serve as buildings blocks, as well as assisting the hormonal system and the metabolism of fats, carbohydrates and proteins. The structural differences between fatty acids determine their biodiversity and give them particular physiological importance. Correct development of the nervous system, retina and other structures depend on an adequate supply of both these fatty acids during intrauterine development and in the newborn and infant stages. The fats present in milk form milk fat globules - structures that do not appear in milk formula prepared using vegetable oils. Apart from the mother's diet, other sources of fatty acids are endogenous biosynthesis in the mammary gland and the fat deposits from which the fatty acids are released. Evolution of the mother's body has also created adaptive mechanisms that adjust the amount of fatty acids in milk to the state of health and needs of the child. These mechanisms go some way to creating a buffer with regard to dietary shortages experienced by pregnant/breastfeeding women, and optimalise the composition of milk fatty acids depending on the age of the pregnant woman, the birth weight of the infant and the efficiency of the placenta during pregnancy.
Collapse
Affiliation(s)
- Rafał Bobiński
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Bielsko-Biala, Poland
| | | |
Collapse
|
23
|
Ortega-Senovilla H, Schaefer-Graf U, Herrera E. Pregnant women with gestational diabetes and with well controlled glucose levels have decreased concentrations of individual fatty acids in maternal and cord serum. Diabetologia 2020; 63:864-874. [PMID: 31832744 DOI: 10.1007/s00125-019-05054-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS Both arachidonic acid (AA, 20:4 n-6) and docosahexaenoic acid (DHA,22:6 n-3), long-chain polyunsaturated fatty acids (LCPUFA), are involved in fetal development and, based on their percentage compositions, appear to be specifically accumulated in fetal circulation in a proposed phenomenon known as biomagnification. Discrepancies exist in the literature concerning the effect of gestational diabetes mellitus (GDM) on circulating fatty acids. Our objective was to analyse individual fatty acid concentrations in a large cohort of maternal and cord paired serum samples from pregnant women with and without GDM. METHODS Overnight fasted maternal and cord blood paired samples from 84 women with GDM and well controlled blood glucose levels and 90 healthy pregnant women (controls) were drawn at term. Individual fatty acids within total serum lipids were analysed by gas chromatography and expressed both as concentrations of fatty acid (mmol/l) and as a percentage of total fatty acids. RESULTS In the serum of overnight fasted pregnant women with GDM, the concentrations of most fatty acids were lower than in control women, except for AA and DHA, which remained the same. The concentrations of most fatty acids in cord serum were also lower in the GDM group than in the control group, except for α-linolenic acid (ALA,18:3 n-3), which was higher in the GDM group. In both groups, the concentrations of all fatty acids were lower in cord serum than in maternal serum. In GDM participants only, a positive and significant correlation between cord and maternal serum concentration of AA and DHA was observed. CONCLUSIONS/INTERPRETATION The expression of fatty acids in molar concentrations reveals that GDM decreases the concentration of most fatty acids in both maternal and cord serum. There is a high fetal dependence on maternal AA and DHA, but our findings do not support the existence of a fetal biomagnification of those two LCPUFA.
Collapse
Affiliation(s)
- Henar Ortega-Senovilla
- Dept. of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, Ctra. Boadilla del Monte km 5,3, E-28668, Madrid, Spain.
| | - Ute Schaefer-Graf
- Dept. of Obstetrics and Gynecology, St Joseph's Hospital Center for Diabetes in Pregnancy, Berlin, Germany
| | - Emilio Herrera
- Dept. of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, Ctra. Boadilla del Monte km 5,3, E-28668, Madrid, Spain
| |
Collapse
|
24
|
Weinheimer C, Wang H, Comstock JM, Singh P, Wang Z, Locklear BA, Goodwin KL, Maschek JA, Cox JE, Baack ML, Joss-Moore LA. Maternal Tobacco Smoke Exposure Causes Sex-Divergent Changes in Placental Lipid Metabolism in the Rat. Reprod Sci 2020; 27:631-643. [PMID: 32046449 PMCID: PMC7539808 DOI: 10.1007/s43032-019-00065-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/01/2019] [Indexed: 12/30/2022]
Abstract
Maternal tobacco smoke exposure (MTS) affects fetal acquisition of long-chain polyunsaturated fatty acids (LCPUFA) and increases the risk of obesity and cardio-metabolic disease in the offspring. Alterations in fetal LCPUFA acquisition in maternal smoking are mediated by the placenta. The handling of LCPUFA by the placenta involves protein-mediated transfer and storage. Molecular mediators of placental LCPUFA handling include PPARγ and the fatty acid transport proteins. We previously demonstrated, in a rat model, that MTS results in programming of adult-onset obesity and metabolic disease in male, but not female, offspring. In this study, we test the hypothesis that in utero MTS exposure alters placental structure, placental LCPUFA handling, and fetal fatty acid levels, in a sex-divergent manner. We exposed pregnant rats to tobacco smoke from embryonic day 11 to term gestation. We measured placental and fetal fatty acid profiles, the systolic/diastolic ratio (SD ratio), placental histology, and expression of molecular mediators in the placenta. Our primary finding is that MTS alters fatty acid profiles in male, but not female fetuses and placenta, including increasing the ratio of omega-6 to omega-3 fatty acids. MTS also increased SD ratio in male, but not female placenta. In contrast, the expression of PPARγ and FATPs was upregulated in female, but not male placenta. We conclude that MTS causes sex-divergent changes in placental handling of LCPUFA in the rat. We speculate that our results demonstrate an adaptive response to MTS by the female placenta.
Collapse
Affiliation(s)
- Claudia Weinheimer
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Haimei Wang
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | | | - Purneet Singh
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Zhengming Wang
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Brent A Locklear
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Kasi L Goodwin
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - J Alan Maschek
- Health Science Center Cores, University of Utah Health Sciences Center, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - James E Cox
- Health Science Center Cores, University of Utah Health Sciences Center, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | | | - Lisa A Joss-Moore
- Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA.
| |
Collapse
|
25
|
Li JW, Hu J, Wei M, Guo YY, Yan PS. The Effects of Maternal Obesity on Porcine Placental Efficiency and Proteome. Animals (Basel) 2019; 9:ani9080546. [PMID: 31408947 PMCID: PMC6720507 DOI: 10.3390/ani9080546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/26/2019] [Accepted: 08/08/2019] [Indexed: 01/19/2023] Open
Abstract
Maternal obesity is associated with impaired maternal metabolism and affects the developmental programming of the fetus. The placenta is dysfunctional when exposed to an obese intrauterine environment and can transduce and mediate detrimental maternal impacts to the fetus through mechanisms that remain largely unknown. The main objective of this study was to investigate the effects of maternal obesity on the porcine placental proteome and to analyze the deregulated proteins and potential pathways predicted to be disturbed in obese placentas, using sows with high backfat as a model of obese pregnancy. The sows were divided into two groups based on their backfat thickness: normal backfat (NBF, 17-22 mm; n = 30) and high backfat (HBF, ≥23 mm; n = 30) as the maternal obesity group. The placental tissues used for the proteomic and biochemical analyses were obtained through vaginal delivery, and the maternal blood samples used to determine the metabolic parameters were collected at day 107 of pregnancy. Our study demonstrated that HBF sows had significantly decreased placental efficiency, increased plasma-free fatty acids and triglyceride levels, and increased proinflammatory cytokines plasma levels (p < 0.05). HBF placentas had significantly higher malondialdehyde level, lower total antioxidant capacity and antioxidase activity, increased triglyceride content and enhanced proinflammatory tumor necrosis factor- α (TNF-α) and interleukin-6 (IL-6) contents (p < 0.05). Among the 4652 proteins identified using the proteomic method, 343 were quantified as differentially abundant proteins, which were involved in many vital biological processes. Based on our bioinformatic and placental biochemical analyses, we concluded that maternal obesity is associated with abnormal carbohydrate and lipid metabolism, mitochondrial dysfunction, decreased steroid hormone biosynthesis, and increased oxidative stress and inflammation in the placenta. The results of this study are undoubtedly valuable to other researchers.
Collapse
Affiliation(s)
- Ji-Wei Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jian Hu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ming Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ying-Ying Guo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Pei-Shi Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
26
|
El Beltagy NS, El Deen Sadek SS, Zidan MA, Abd El Naby RE. Can serum free fatty acids assessment predict severe preeclampsia? ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2011.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Nermeen Saad El Beltagy
- Department of Obstetrics and Gynaecology, El Shatby Maternity University Hospital, Alexandria, 21526, Egypt
| | - Sameh Saad El Deen Sadek
- Department of Obstetrics and Gynaecology, El Shatby Maternity University Hospital, Alexandria, 21526, Egypt
| | - Mohamed Abbas Zidan
- Department of Biochemistry, Alexandria Faculty of Medicine, Alexandria UniversityAlexandria, 21531,
Egypt
| | - Rania Emad Abd El Naby
- Department of Obstetrics and Gynaecology, El Shatby Maternity University Hospital, Alexandria, 21526, Egypt
| |
Collapse
|
27
|
Miranda J, Simões RV, Paules C, Cañueto D, Pardo-Cea MA, García-Martín ML, Crovetto F, Fuertes-Martin R, Domenech M, Gómez-Roig MD, Eixarch E, Estruch R, Hansson SR, Amigó N, Cañellas N, Crispi F, Gratacós E. Metabolic profiling and targeted lipidomics reveals a disturbed lipid profile in mothers and fetuses with intrauterine growth restriction. Sci Rep 2018; 8:13614. [PMID: 30206284 PMCID: PMC6134091 DOI: 10.1038/s41598-018-31832-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022] Open
Abstract
Fetal growth may be impaired by poor placental function or maternal conditions, each of which can influence the transfer of nutrients and oxygen from the mother to the developing fetus. Large-scale studies of metabolites (metabolomics) are key to understand cellular metabolism and pathophysiology of human conditions. Herein, maternal and cord blood plasma samples were used for NMR-based metabolic fingerprinting and profiling, including analysis of the enrichment of circulating lipid classes and subclasses, as well as the number of sub-fraction particles and their size. Changes in phosphatidylcholines and glycoproteins were prominent in growth-restricted fetuses indicating significant alterations in their abundance and biophysical properties. Lipoprotein profiles showed significantly lower plasma concentrations of cholesterol-intermediate density lipoprotein (IDL), triglycerides-IDL and high-density lipoprotein (HDL) in mothers of growth-restricted fetuses compared to controls (p < 0.05). In contrast, growth-restricted fetuses had significantly higher plasma concentrations of cholesterol and triglycerides transporting lipoproteins [LDL, IDL, and VLDL, (p < 0.005; all)], as well as increased VLDL particle types (large, medium and small). Significant changes in plasma concentrations of formate, histidine, isoleucine and citrate in growth-restricted fetuses were also observed. Comprehensive metabolic profiling reveals that both, mother and fetuses of pregnancies complicated with fetal growth restriction have a substantial disruption in lipid metabolism.
Collapse
Affiliation(s)
- Jezid Miranda
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rui V Simões
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Cristina Paules
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Daniel Cañueto
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
| | | | - María L García-Martín
- BIONAND, Andalusian Centre for Nanomedicine and Biotechnology, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | - Francesca Crovetto
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rocio Fuertes-Martin
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
- Biosfer Teslab, Reus, Spain
| | - Monica Domenech
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - María D Gómez-Roig
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Elisenda Eixarch
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Ramon Estruch
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Lund University, Sweden; Skåne University Hospital, Lund, Sweden
| | | | - Nicolau Cañellas
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
- CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Fatima Crispi
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain.
| | - Eduard Gratacós
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
28
|
Wadhwani N, Patil V, Joshi S. Maternal long chain polyunsaturated fatty acid status and pregnancy complications. Prostaglandins Leukot Essent Fatty Acids 2018; 136:143-152. [PMID: 28888333 DOI: 10.1016/j.plefa.2017.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 07/06/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022]
Abstract
Maternal nutrition plays a crucial role in influencing fetal growth and birth outcome. Any nutritional insult starting several weeks before pregnancy and during critical periods of gestation is known to influence fetal development and increase the risk for diseases during later life. Literature suggests that chronic adult diseases may have their origin during early life - a concept referred to as Developmental Origins of Health and Disease (DOHaD) which states that adverse exposures early in life "program" risks for later chronic disorders. Long chain polyunsaturated fatty acids (LCPUFA), mainly omega-6 and omega-3 fatty acids are known to have an effect on fetal programming. The placental supply of optimal levels of LCPUFA to the fetus during early life is extremely important for the normal growth and development of both placenta and fetus. Any alteration in placental development will result in adverse pregnancy outcome such as gestational diabetes mellitus (GDM), preeclampsia, and intrauterine growth restriction (IUGR). A disturbed materno-fetal LCPUFA supply is known to be linked with each of these pathologies. Further, a disturbed LCPUFA metabolism is reported to be associated with a number of metabolic disorders. It is likely that LCPUFA supplementation during early pregnancy may be beneficial in improving the health of the mother, improving birth outcome and thereby reducing the risk of diseases in later life.
Collapse
Affiliation(s)
- Nisha Wadhwani
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Vidya Patil
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Sadhana Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India.
| |
Collapse
|
29
|
Chavan-Gautam P, Rani A, Freeman DJ. Distribution of Fatty Acids and Lipids During Pregnancy. Adv Clin Chem 2018; 84:209-239. [PMID: 29478515 DOI: 10.1016/bs.acc.2017.12.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal fatty acid and lipid metabolism undergoes changes during pregnancy to facilitate fetal growth and development. Different types of fatty acids have different roles in maintaining a successful pregnancy and they are incorporated into different forms of lipids for the purpose of storage and transport. This chapter aims to provide an understanding of the distribution and metabolism of fatty acids and lipids in the maternal, placental, and fetal compartments. We further describe how this distribution is altered in maternal obesity, preterm birth, and pregnancy complications such as gestational diabetes mellitus, preeclampsia, and intrauterine growth restriction.
Collapse
Affiliation(s)
- Preeti Chavan-Gautam
- Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India.
| | - Alka Rani
- Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India
| | - Dilys J Freeman
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
30
|
Myatt L, Thornburg KL. Effects of Prenatal Nutrition and the Role of the Placenta in Health and Disease. Methods Mol Biol 2018; 1735:19-46. [PMID: 29380305 DOI: 10.1007/978-1-4939-7614-0_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Epidemiologic studies identified the linkage between exposures to stresses, including the type and plane of nutrition in utero with development of disease in later life. Given the critical roles of the placenta in mediating transport of nutrients between the mother and fetus and regulation of maternal metabolism, recent attention has focused on the role of the placenta in mediating the effect of altered nutritional exposures on the development of disease in later life. In this chapter we describe the mechanisms of nutrient transport in the placenta, the influence of placental metabolism on this, and how placental energetics influence placental function in response to a variety of stressors. Further the recent "recognition" that the placenta itself has a sex which affects its function may begin to help elucidate the mechanisms underlying the well-known dimorphism in development of disease in adult life.
Collapse
Affiliation(s)
- Leslie Myatt
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA. .,Bob and Charlee Moore Institute for Nutrition & Wellness, Oregon Health & Science University, Portland, OR, USA.
| | - Kent L Thornburg
- Bob and Charlee Moore Institute for Nutrition & Wellness, Oregon Health & Science University, Portland, OR, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
31
|
Miller CN, Dye JA, Ledbetter AD, Schladweiler MC, Richards JH, Snow SJ, Wood CE, Henriquez AR, Thompson LC, Farraj AK, Hazari MS, Kodavanti UP. Uterine Artery Flow and Offspring Growth in Long-Evans Rats following Maternal Exposure to Ozone during Implantation. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:127005. [PMID: 29269335 PMCID: PMC5963593 DOI: 10.1289/ehp2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Epidemiological studies suggest that increased ozone exposure during gestation may compromise fetal growth. In particular, the implantation stage of pregnancy is considered a key window of susceptibility for this outcome. OBJECTIVES The main goals of this study were to investigate the effects of short-term ozone inhalation during implantation on fetal growth outcomes and to explore the potential for alterations in uterine arterial flow as a contributing mechanism. METHODS Pregnant Long-Evans rats were exposed to filtered air, 0.4 ppm ozone, or 0.8 ppm ozone for 4 h/d during implantation, on gestation days (GD) 5 and 6. Tail cuff blood pressure and uterine artery Doppler ultrasound were measured on GD 15, 19, and 21. To assess whether peri-implantation ozone exposure resulted in sustained pulmonary or systemic health effects, bronchoalveolar lavage fluid, serum metabolic and inflammatory end points, and kidney histopathology were evaluated in dams at GD 21. Growth parameters assessed in GD 21 offspring included fetal weight, length, and body composition. RESULTS Measures of maternal uterine arterial flow, including resistance index and mean velocity, indicated that resistance increased between GD 15 and GD 21 in 0.8 ppm dams but decreased in controls, although absolute values were similar in both groups on GD 21. Ozone-exposed dams also had lower serum glucose and higher free fatty acid concentrations than controls on GD 21. On GD 21, both male and female offspring had lower body weight than controls, and pooled subsets of 3 male and 3 female fetuses from litters exposed to 0.8 ppm ozone had lower lean mass and fat mass than pooled control offspring. CONCLUSIONS Findings from our experimental model suggest that the offspring of dams exposed to ozone during implantation had reduced growth compared with controls, possibly as a consequence of ozone-induced vascular dysfunction. https://doi.org/10.1289/EHP2019.
Collapse
Affiliation(s)
- Colette N Miller
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Janice A Dye
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Allen D Ledbetter
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Mette C Schladweiler
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Judy H Richards
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Samantha J Snow
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Charles E Wood
- Integrated Systems Toxicology Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Andres R Henriquez
- Curriculum in Toxicology, University of North Carolina School of Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Leslie C Thompson
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Aimen K Farraj
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Mehdi S Hazari
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| | - Urmila P Kodavanti
- Environmental Public Health Division, National Health & Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina, USA
| |
Collapse
|
32
|
Zywicki M, Blohowiak SE, Magness RR, Segar JL, Kling PJ. Increasing fetal ovine number per gestation alters fetal plasma clinical chemistry values. Physiol Rep 2017; 4:4/16/e12905. [PMID: 27565903 PMCID: PMC5002913 DOI: 10.14814/phy2.12905] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/27/2016] [Indexed: 12/18/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is interconnected with developmental programming of lifelong pathophysiology. IUGR is seen in human multifetal pregnancies, with stepwise rises in fetal numbers interfering with placental nutrient delivery. It remains unknown whether fetal blood analyses would reflect fetal nutrition, liver, and excretory function in the last trimester of human or ovine IUGR. In an ovine model, we hypothesized that fetal plasma biochemical values would reflect progressive placental, fetal liver, and fetal kidney dysfunction as the number of fetuses per gestation rose. To determine fetal plasma biochemical values in singleton, twin, triplet, and quadruplet/quintuplet ovine gestation, we investigated morphometric measures and comprehensive metabolic panels with nutritional measures, liver enzymes, and placental and fetal kidney excretory measures at gestational day (GD) 130 (90% gestation). As anticipated, placental dysfunction was supported by a stepwise fall in fetal weight, fetal plasma glucose, and triglyceride levels as fetal number per ewe rose. Fetal glucose and triglycerides were directly related to fetal weight. Plasma creatinine, reflecting fetal renal excretory function, and plasma cholesterol, reflecting placental excretory function, were inversely correlated with fetal weight. Progressive biochemical disturbances and growth restriction accompanied the rise in fetal number. Understanding the compensatory and adaptive responses of growth‐restricted fetuses at the biochemical level may help explain how metabolic pathways in growth restriction can be predetermined at birth. This physiological understanding is important for clinical care and generating interventional strategies to prevent altered developmental programming in multifetal gestation.
Collapse
Affiliation(s)
- Micaela Zywicki
- Departments of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Sharon E Blohowiak
- Departments of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Ronald R Magness
- Departments of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA Obstetrics and Gynecology Perinatal Research Laboratories, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Jeffrey L Segar
- Department of Pediatrics, University of Iowa Children's Hospital, Iowa, IA, USA
| | - Pamela J Kling
- Departments of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
33
|
Assumpção RP, Mucci DB, Fonseca FCP, Marcondes H, Sardinha FLC, Citelli M, Tavares do Carmo MG. Fatty acid profile of maternal and fetal erythrocytes and placental expression of fatty acid transport proteins in normal and intrauterine growth restriction pregnancies. Prostaglandins Leukot Essent Fatty Acids 2017; 125:24-31. [PMID: 28987718 DOI: 10.1016/j.plefa.2017.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/19/2017] [Accepted: 08/21/2017] [Indexed: 02/02/2023]
Abstract
Long-chain polyunsaturated fatty acids (LC-PUFA), mainly docosahexaenoic (DHA) and arachidonic acids (AA), are critical for adequate fetal growth and development. We investigated mRNA expression of proteins involved in hydrolysis, uptake and/or transport of fatty acids in placenta of fifteen full term normal pregnancies and eleven pregnancies complicated by intrauterine growth restriction (IUGR) with normal umbilical blood flows. The mRNA expression of LPL, FATPs (-1, -2 and -4) and FABPs (-1 and -3) was increased in IUGR placentas, however, tissue profile of LC-PUFA was not different between groups. Erythrocytes from both mothers and fetuses of the IUGR group showed lower concentrations of AA and DHA and inferior DHA/ALA ratio compared to normal pregnancies (P < 0.05). We hypothesize that reduced circulating levels of AA and DHA could up-regulate mRNA expression of placental fatty acids transporters, as a compensatory mechanism, however this failed to sustain normal LC-PUFA supply to the fetus in IUGR.
Collapse
Affiliation(s)
- Renata P Assumpção
- Laboratório de Bioquímica Nutricional, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Daniela B Mucci
- Laboratório de Bioquímica Nutricional, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernanda C P Fonseca
- Laboratório de Bioquímica Nutricional, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Henrique Marcondes
- Laboratório de Bioquímica Nutricional, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fátima L C Sardinha
- Laboratório de Bioquímica Nutricional, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marta Citelli
- Departamento de Nutrição Básica e Experimental, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria G Tavares do Carmo
- Laboratório de Bioquímica Nutricional, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
34
|
Ravlić S, Škrobot Vidaček N, Nanić L, Laganović M, Slade N, Jelaković B, Rubelj I. Mechanisms of fetal epigenetics that determine telomere dynamics and health span in adulthood. Mech Ageing Dev 2017; 174:55-62. [PMID: 28847485 DOI: 10.1016/j.mad.2017.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 01/11/2023]
Abstract
Advances in epigenetics now enable us to better understand environmental influences on the genetic background of human diseases. This refers especially to fetal development where an adverse intrauterine environment impacts oxygen and nutrient supply to the fetus. Recently, differences in telomere length and telomere loss dynamics among individuals born with intrauterine growth restriction compared to normal controls have been described. In this paper we propose possible molecular mechanisms that (pre)program telomere epigenetics during pregnancy. This programming sets differences in telomere lengths and dynamics of telomere shortening in adulthood and therefore dictates the dynamics of aging and morbidity in later life.
Collapse
Affiliation(s)
- Sanda Ravlić
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, RBI, Zagreb, Croatia.
| | - Nikolina Škrobot Vidaček
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, RBI, Zagreb, Croatia.
| | - Lucia Nanić
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, RBI, Zagreb, Croatia.
| | - Mario Laganović
- Department for Nephrology, Hypertension, Dialysis and Transplantation, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Neda Slade
- Laboratory for Protein Dynamics, Division of Molecular Medicine, RBI, Zagreb, Croatia.
| | - Bojan Jelaković
- Department for Nephrology, Hypertension, Dialysis and Transplantation, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Ivica Rubelj
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, RBI, Zagreb, Croatia.
| |
Collapse
|
35
|
Tian L, Dong SS, Hu J, Yao JJ, Yan PS. The effect of maternal obesity on fatty acid transporter expression and lipid metabolism in the full-term placenta of lean breed swine. J Anim Physiol Anim Nutr (Berl) 2017; 102:e242-e253. [PMID: 28508539 DOI: 10.1111/jpn.12735] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 03/31/2017] [Indexed: 12/30/2022]
Abstract
This study was conducted to evaluate the influence of back-fat thickness (BF), at mating of sows, on the maternal and newborn circulating lipids, expression of placental fatty acids (FA) transporters and lipid accumulation in placenta. Full-term placentas were obtained by vaginal delivery from BFI (9-14 mm; n = 37), BFII (15-19 mm; n = 43) and BFIII (20-27 mm; n = 38) sows according to BF at mating, and frozen placental sections were analysed for fat accumulation. Blood samples were collected from the sows of day 105 pregnancy and from cord blood at delivery. mRNA and protein expression levels were evaluated with real-time RT-PCR and Western blotting. Our results demonstrated that BFII females had significantly increased litter weight and placental efficiency, decreased maternal triglyceride (TG) and non-esterified fatty acids (NEFA) levels, decreased maternal IL-6, TNFα and leptin levels compared to BFIII females (p < .05). BFIII sows were associated with significantly decreased newborn TG levels, increased newborn glucose, IL-6 and TNFα levels compared to BFI or BFII sows (p < .05). BFI and BFII females had significantly decreased placental TG, NEFA and cholesterol (CHOL) contents compared to BFIII females (p < .05). Moreover, decreased CD36, FATP1, FABP4, and FABP1 mRNA and protein and FATP4 protein expression, and increased LPL activity were also observed in BFIII group compared with BFII group (p < .05). PPARγ mRNA and protein and lipogenic genes such as SREBP-1c, ACSL1, ACCα, FAS and SCD mRNA expression were downregulated or upregulated, respectively, in the placentas of BFIII sows compared to BFI or BFII sows (p < .05). Overall, this study demonstrated that there is no advantage, in terms of litter live size, litter weight and placental FA transport and metabolism, in performing the mating of sows with BF>19 mm.
Collapse
Affiliation(s)
- L Tian
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - S S Dong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - J Hu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - J J Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - P S Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
36
|
Herrera E, Desoye G. Maternal and fetal lipid metabolism under normal and gestational diabetic conditions. Horm Mol Biol Clin Investig 2017; 26:109-27. [PMID: 26351960 DOI: 10.1515/hmbci-2015-0025] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/08/2015] [Indexed: 12/18/2022]
Abstract
Maternal lipids are strong determinants of fetal fat mass. Here we review the overall lipid metabolism in normal and gestational diabetes mellitus (GDM) pregnancies. During early pregnancy, the increase in maternal fat depots is facilitated by insulin, followed by increased adipose tissue breakdown and subsequent hypertriglyceridemia, mainly as a result of insulin resistance (IR) and estrogen effects. The response to diabetes is variable as a result of greater IR but decreased estrogen levels. The vast majority of fatty acids (FAs) in the maternal circulation are esterified and associated with lipoproteins. These are taken up by the placenta and hydrolyzed by lipases. The released FAs enter various metabolic routes and are released into fetal circulation. Although these determinants are modified in maternal GDM, the fetus does not seem to receive more FAs than in non-GDM pregnancies. Long-chain polyunsaturated FAs are essential for fetal development and are obtained from the mother. Mitochondrial FA oxidation occurs in fetal tissue and in placenta and contributes to energy production. Fetal fat accretion during the last weeks of gestation occurs very rapidly and is sustained not only by FAs crossing the placenta, but also by fetal lipogenesis. Fetal hyperinsulinemia in GDM mothers promotes excess accretion of adipose tissue, which gives rise to altered adipocytokine profiles. Fetal lipoproteins are low at birth, but the GDM effects are unclear. The increase in body fat in neonates of GDM women is a risk factor for obesity in early childhood and later life.
Collapse
|
37
|
Reyna-Villasmil E, Navarro-Briceño Y, Mejía-Montilla J, Reyna-Villasmil N, Torres-Cepeda D, Santos-Bolívar J, Fernández-Ramírez A. Lípidos y lipoproteínas en restricción intrauterina del crecimiento con velocimetría Doppler anormal de la arteria umbilical. PERINATOLOGÍA Y REPRODUCCIÓN HUMANA 2016. [DOI: 10.1016/j.rprh.2016.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
38
|
O'Tierney-Ginn PF, Gillingham M, Fowler J, Brass E, Marshall NE, Thornburg KL. Maternal Weight Gain Regulates Omega-3 Fatty Acids in Male, Not Female, Neonates. Reprod Sci 2016; 24:560-567. [PMID: 27470150 DOI: 10.1177/1933719116660843] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The fetus largely depends on maternal supply and placental transport for its source of long-chain polyunsaturated fatty acids (LCPUFAs), which are essential for proper neurological and cardiovascular development. Pregnancy complications such as diabetes reduces neonatal LCPUFA supply, but little is known of how fatty acid delivery is affected by maternal body type or weight gain in uncomplicated pregnancies. In a cross-sectional study of maternal-neonatal pairs at term, we sought to determine the effect of gestational weight gain on neonatal LCPUFA supply. Forty maternal-neonatal pairs of uncomplicated (no gestational hypertension or diabetes) term pregnancies were recruited upon admission to Oregon Health & Science University Labor & Delivery for scheduled cesarean section. Maternal and umbilical cord plasma fatty acid profiles were measured using gas chromatography-mass spectrophotometry. First trimester weight gain was negatively correlated with maternal n-3 LCPUFA ( r = -0.80, P = .0002), and this was not affected by fetal sex. High maternal weight gain in the first trimester was negatively associated with cord n-3 polyunsaturated fatty acid levels ( r = -0.70, P = .03) and placental thickness ( r = -0.69, P = .03) in male, but not female, offspring. High maternal weight gain in the first trimester is associated with a thinner placenta and low levels of n-3 LCPUFA in male offspring. Further study is required to confirm that male offspring are at a higher risk of poor outcomes associated with high maternal weight gain early in pregnancy.
Collapse
Affiliation(s)
- Perrie F O'Tierney-Ginn
- 1 Department of Reproductive Biology, Center for Reproductive Health, Case Western Reserve University, Cleveland, OH, USA
| | - Melanie Gillingham
- 2 Department of Molecular & Medical Genetics, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Jessica Fowler
- 3 Department of Obstetrics & Gynecology, UCLA, Los Angeles, CA, USA
| | - Elizabeth Brass
- 4 Department of Obstetrics & Gynecology, Kaiser Permanente, Hillsboro, OR, USA
| | - Nicole E Marshall
- 5 Department of Obstetrics & Gynecology, OHSU, Portland, OR, USA.,6 Center for Developmental Health, OHSU, Portland, OR, USA
| | - Kent L Thornburg
- 6 Center for Developmental Health, OHSU, Portland, OR, USA.,7 Department of Medicine (Cardiovascular Medicine), OHSU, Portland, OR, USA
| |
Collapse
|
39
|
Roifman M, Choufani S, Turinsky AL, Drewlo S, Keating S, Brudno M, Kingdom J, Weksberg R. Genome-wide placental DNA methylation analysis of severely growth-discordant monochorionic twins reveals novel epigenetic targets for intrauterine growth restriction. Clin Epigenetics 2016; 8:70. [PMID: 27330572 PMCID: PMC4915063 DOI: 10.1186/s13148-016-0238-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/12/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR), which refers to reduced fetal growth in the context of placental insufficiency, is etiologically heterogeneous. IUGR is associated not only with perinatal morbidity and mortality but also with adult-onset disorders, such as cardiovascular disease and diabetes, posing a major health burden. Placental epigenetic dysregulation has been proposed as one mechanism that causes IUGR; however, the spectrum of epigenetic pathophysiological mechanisms leading to IUGR remains to be elucidated. Monozygotic monochorionic twins are particularly affected by IUGR, in the setting of severe discordant growth. Because monozygotic twins have the same genotype at conception and a shared maternal environment, they provide an ideal model system for studying epigenetic dysregulation of the placenta. RESULTS We compared genome-wide placental DNA methylation patterns of severely growth-discordant twins to identify novel candidate genes for IUGR. Snap-frozen placental samples for eight severely growth-discordant monozygotic monochorionic twin pairs were obtained at delivery from each twin. A high-resolution DNA methylation array platform was used to identify methylation differences between IUGR and normal twins. Our analysis revealed differentially methylated regions in the promoters of eight genes: DECR1, ZNF300, DNAJA4, CCL28, LEPR, HSPA1A/L, GSTO1, and GNE. The largest methylation differences between the two groups were in the promoters of DECR1 and ZNF300. The significance of these group differences was independently validated by bisulfite pyrosequencing, implicating aberrations in fatty acid beta oxidation and transcriptional regulation, respectively. Further analysis of the array data identified methylation changes most prominently affecting the Wnt and cadherin pathways in the IUGR cohort. CONCLUSIONS Our results suggest that IUGR in monozygotic twins is associated with impairments in lipid metabolism and transcriptional regulation as well as cadherin and Wnt signaling. We show that monozygotic monochorionic twins discordant for growth provide a useful model to study one type of the epigenetic placental dysregulation that drives IUGR.
Collapse
Affiliation(s)
- Maian Roifman
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario Canada ; Department of Paediatrics, University of Toronto, Toronto, Ontario Canada ; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; The Prenatal and Medical Genetics Program, Department of Obstetrics and Gynaecology, Mount Sinai Hospital, Toronto, Ontario Canada
| | - Sanaa Choufani
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Andrei L Turinsky
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; Centre for Computational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Sascha Drewlo
- C.S. Mott Center for Human Growth and Development, Wayne State School of Medicine, Wayne State University, Detroit, MI USA
| | - Sarah Keating
- Maternal-Fetal Medicine Division, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Toronto, Ontario Canada ; Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario Canada
| | - Michael Brudno
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; Centre for Computational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada ; Department of Computer Science, University of Toronto, Toronto, Ontario Canada
| | - John Kingdom
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario Canada ; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario Canada
| | - Rosanna Weksberg
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario Canada ; Department of Paediatrics, University of Toronto, Toronto, Ontario Canada ; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario Canada ; Institute of Medical Science, University of Toronto, Toronto, Ontario Canada
| |
Collapse
|
40
|
Gaccioli F, Lager S. Placental Nutrient Transport and Intrauterine Growth Restriction. Front Physiol 2016; 7:40. [PMID: 26909042 PMCID: PMC4754577 DOI: 10.3389/fphys.2016.00040] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/28/2016] [Indexed: 01/30/2023] Open
Abstract
Intrauterine growth restriction refers to the inability of the fetus to reach its genetically determined potential size. Fetal growth restriction affects approximately 5–15% of all pregnancies in the United States and Europe. In developing countries the occurrence varies widely between 10 and 55%, impacting about 30 million newborns per year. Besides having high perinatal mortality rates these infants are at greater risk for severe adverse outcomes, such as hypoxic ischemic encephalopathy and cerebral palsy. Moreover, reduced fetal growth has lifelong health consequences, including higher risks of developing metabolic and cardiovascular diseases in adulthood. Numerous reports indicate placental insufficiency as one of the underlying causes leading to altered fetal growth and impaired placental capacity of delivering nutrients to the fetus has been shown to contribute to the etiology of intrauterine growth restriction. Indeed, reduced expression and/or activity of placental nutrient transporters have been demonstrated in several conditions associated with an increased risk of delivering a small or growth restricted infant. This review focuses on human pregnancies and summarizes the changes in placental amino acid, fatty acid, and glucose transport reported in conditions associated with intrauterine growth restriction, such as maternal undernutrition, pre-eclampsia, young maternal age, high altitude and infection.
Collapse
Affiliation(s)
- Francesca Gaccioli
- Department of Obstetrics and Gynaecology, University of Cambridge Cambridge, UK
| | - Susanne Lager
- Department of Obstetrics and Gynaecology, University of Cambridge Cambridge, UK
| |
Collapse
|
41
|
Meher A, Randhir K, Mehendale S, Wagh G, Joshi S. Maternal Fatty Acids and Their Association with Birth Outcome: A Prospective Study. PLoS One 2016; 11:e0147359. [PMID: 26815428 PMCID: PMC4729437 DOI: 10.1371/journal.pone.0147359] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/04/2016] [Indexed: 01/20/2023] Open
Abstract
Maternal nutrition, especially LCPUFA, is an important factor in determining fetal growth and development. Our earlier cross sectional study reports lower docosahexanoic acid (DHA) levels at the time of delivery in mothers delivering low birth weight (LBW) babies. This study was undertaken to examine the role of the maternal omega-3 and omega-6 fatty acid profile across the gestation in fetal growth. This is a hospital based study where women were recruited in early gestation. Maternal blood was collected at 3 time points, i.e., T1 = 16th–20th week, T2 = 26th–30th week and T3 = at delivery. Cord blood was collected at delivery. At delivery, these women were divided into 2 groups: those delivering at term a baby weighing >2.5kg [Normal birth weight (NBW) group] and those delivering at term a baby weighing <2.5kg [LBW group]. The study reports data on 111 women recruited at T1, out of which 60 women delivered an NBW baby at term and 51 women delivered an LBW baby at term. Fatty acids were analysed using gas chromatography. At T1 of gestation, maternal erythrocyte DHA levels were positively (p<0.05) associated with baby weight. Maternal plasma and erythrocyte arachidonic acid and total erythrocyte omega-6 fatty acid levels at T2 were higher (p<0.05 for both) in the LBW group. Total erythrocyte omega-3 fatty acid levels were lower (p<0.05) while total erythrocyte omega-6 fatty acid levels were higher (p<0.05) in the LBW group at delivery. Our data demonstrates the possible role of LCPUFA in the etiology of LBW babies right from early pregnancy.
Collapse
Affiliation(s)
- Akshaya Meher
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, 411043, India
| | - Karuna Randhir
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, 411043, India
| | - Savita Mehendale
- Dept of Obstetrics and Gynaecology, Bharati Medical College and Hospital, Bharati Vidyapeeth University, Pune, 411043, India
| | - Girija Wagh
- Dept of Obstetrics and Gynaecology, Bharati Medical College and Hospital, Bharati Vidyapeeth University, Pune, 411043, India
| | - Sadhana Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, 411043, India
- * E-mail:
| |
Collapse
|
42
|
Barrett HL, Kubala MH, Scholz Romero K, Denny KJ, Woodruff TM, McIntyre HD, Callaway LK, Dekker Nitert M. Placental lipase expression in pregnancies complicated by preeclampsia: a case-control study. Reprod Biol Endocrinol 2015; 13:100. [PMID: 26336959 PMCID: PMC4558775 DOI: 10.1186/s12958-015-0098-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/20/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preeclampsia (PE) is associated with maternal and neonatal morbidity and mortality. In PE, the physiological hyperlipidaemia of pregnancy is exaggerated. The purpose of this study was to examine the expression of adipose triglyceride lipase (ATGL), hormone sensitive lipase (HSL), lipoprotein lipase (LPL) and endothelial lipase (EL) in pregnancies complicated by PE. METHODS Placentae were collected from 16 women with PE and 20 women with uncomplicated pregnancies matched for maternal prepregnancy BMI and gestational age of delivery. Gene and protein expression of the placental lipases were measured by Q-PCR and Western blot. DNA methylation of the promoter of LPL was assessed by bisulfite sequencing. Lipase localisation and activity were analysed. RESULTS Gene expression of all lipases was significantly reduced, as was HSL protein level in women with PE. All lipases were localised to trophoblasts and endothelial cells in PE and control placentae. There was no difference in methylation of the LPL promoter between PE and control placentae. Lipase activity was not altered in placentae from women with PE. CONCLUSION These results suggest that the decreased placental lipase gene but not protein expression or lipase activity, which is associated with late-onset PE is not a major contributor to the abnormal lipids seen in PE.
Collapse
Affiliation(s)
- Helen L Barrett
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD, Australia.
- Obstetric Medicine, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.
- School of Medicine, The University of Queensland, Herston, QLD, Australia.
| | - Marta H Kubala
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD, Australia.
| | | | - Kerina J Denny
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.
| | - H David McIntyre
- School of Medicine, The University of Queensland, Herston, QLD, Australia.
- Mater Research Institute, The University of Queensland Brisbane, Brisbane, QLD, Australia.
| | - Leonie K Callaway
- Obstetric Medicine, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.
- School of Medicine, The University of Queensland, Herston, QLD, Australia.
| | - Marloes Dekker Nitert
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD, Australia.
- School of Medicine, The University of Queensland, Herston, QLD, Australia.
| |
Collapse
|
43
|
Yang X, Haghiac M, Glazebrook P, Minium J, Catalano PM, Hauguel-de Mouzon S. Saturated fatty acids enhance TLR4 immune pathways in human trophoblasts. Hum Reprod 2015. [PMID: 26202921 DOI: 10.1093/humrep/dev173] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
STUDY QUESTION What are the effects of fatty acids on placental inflammatory cytokine with respect to toll-like receptor-4/nuclear factor-kappa B (TLR4/NF-kB)? SUMMARY ANSWER Exogenous fatty acids induce a pro-inflammatory cytokine response in human placental cells in vitro via activation of TLR4 signaling pathways. WHAT IS KNOWN ALREADY The placenta is exposed to changes in circulating maternal fatty acid concentrations throughout pregnancy. Fatty acids are master regulators of innate immune pathways through recruitment of toll-like receptors and activation of cytokine synthesis. STUDY DESIGN, SIZE, DURATION Trophoblast cells isolated from 14 normal term human placentas were incubated with long chain fatty acids (FA) of different carbon length and degree of saturation. The expression and secretion of interleukin-6 (IL-6), IL-8 and tumor necrosis factor-alpha (TNF-α) were measured by reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. Antibodies against TLR4 ligand binding domain, downstream signaling and anti-p65 NFkB-inhibitor were used to characterize the pathways of FA action. PARTICIPANTS/MATERIALS, SETTING, METHODS General approach used primary human term trophoblast cell culture. Methods and end-points used real-time quantitative PCR, cytokine measurements, immunohistochemistry, western blots. MAIN RESULTS AND THE ROLE OF CHANCE The long chain saturated fatty acids, stearic and palmitic (PA), stimulated the synthesis as well as the release of TNF-α, IL-6 and IL-8 by trophoblast cells (2- to 6-fold, P < 0.001). In contrast, the unsaturated (palmitoleic, oleic, linoleic) acids did not modify cytokine expression significantly. Palmitate-induced inflammatory effects were mediated via TLR4 activation, NF-kB phosphorylation and nuclear translocation. LIMITATIONS, REASONS FOR CAUTION TNF-α protein level was close to the limit of detection in the culture medium even when cells were cultured with PA. WIDER IMPLICATIONS OF THE FINDINGS These mechanisms open the way to a better understanding of how changes in maternal lipid homeostasis may regulate placental inflammatory status. STUDY FUNDING/COMPETING INTERESTS X.Y. was recipient of fellowship award from West China Second University Hospital, Sichuan University (NIH HD 22965-19). The authors have nothing else to disclose. TRIAL REGISTRATION NUMBER None.
Collapse
Affiliation(s)
- Xiaohua Yang
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109-1998, USA
| | - Maricela Haghiac
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109-1998, USA
| | - Patricia Glazebrook
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109-1998, USA
| | - Judi Minium
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109-1998, USA
| | - Patrick M Catalano
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109-1998, USA
| | - Sylvie Hauguel-de Mouzon
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109-1998, USA
| |
Collapse
|
44
|
Abi Salloum B, Veiga-Lopez A, Abbott DH, Burant CF, Padmanabhan V. Developmental programming: exposure to testosterone excess disrupts steroidal and metabolic environment in pregnant sheep. Endocrinology 2015; 156:2323-37. [PMID: 25763641 PMCID: PMC4430607 DOI: 10.1210/en.2014-2006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Gestational exposure to excess T leads to intrauterine growth restriction, low birth weight, and adult metabolic/reproductive disorders in female sheep. We hypothesized that as early mediators of such disruptions, gestational T disrupts steroidal and metabolic homeostasis in both the mother and fetus by both androgenic and metabolic pathways. Maternal blood samples were measured weekly for levels of insulin, glucose, and progesterone from four groups of animals: control; gestational T (twice weekly im injections of 100 mg of T propionate from d 30 to d 90 of gestation); T plus an androgen antagonist, flutamide (15 mg/kg·d oral; T-Flutamide); and T plus the insulin sensitizer, rosiglitazone (0.11 mg/kg·d oral; T-Rosi) (n = 10-12/group). On day 90 of gestation, maternal and umbilical cord samples were collected after a 48-hour fast from a subset (n = 6/group) for the measurement of steroids, free fatty acids, amino acids, and acylcarnitines. Gestational T decreased maternal progesterone levels by 36.5% (P < .05), which was prevented by flutamide showing direct androgenic mediation. Gestational T also augmented maternal insulin levels and decreased medium chained acylcarnitines, suggesting increased mitochondrial fatty acid oxidation. These changes were prevented by rosiglitazone, suggesting alterations in maternal fuel use. Gestational T-induced increases in fetal estradiol were not prevented by either cotreatment. Gestational T disrupted associations of steroids with metabolites and progesterone with acylcarnitines, which was prevented either by androgen antagonist or insulin sensitizer cotreatment. These findings suggest a future combination of these treatments might be required to prevent alteration in maternal/fetal steroidal and metabolic milieu(s).
Collapse
Affiliation(s)
- B Abi Salloum
- Departments of Pediatrics (B.A.S., A.V.-L., V.P.) and Internal Medicine (C.F.B.) and Reproductive Sciences Program (V.P.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Obstetrics and Gynecology and Wisconsin National Primate Research Center (D.H.A.), University of Wisconsin, Madison, Wisconsin 53706
| | | | | | | | | |
Collapse
|
45
|
Effects of omega-3 and omega-6 fatty acids on human placental cytokine production. Placenta 2015; 36:34-40. [DOI: 10.1016/j.placenta.2014.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/07/2014] [Accepted: 10/28/2014] [Indexed: 11/20/2022]
|
46
|
Barrett HL, Dekker Nitert M, McIntyre HD, Callaway LK. Maternal lipids in pre-eclampsia: innocent bystander or culprit? Hypertens Pregnancy 2014; 33:508-23. [PMID: 25121342 DOI: 10.3109/10641955.2014.946614] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Pre-eclampsia continues to be a challenge--to understand the underlying pathogenesis and to prevent or treat in the clinical setting. One area of potential therapies opening up is treatment of maternal lipids and clinical trials are underway using statins in early pre-eclampsia. At present, most potential therapies to treat lipids cannot be recommended for general use in pregnancy and if we were to target maternal lipids to reduce rates of pre-eclampsia, very large numbers of women may need to be treated. Prior to reaching that point, we first need to understand whether maternal lipids are pathogenic in the processes underlying pre-eclampsia. The aim of this review is to examine the role of lipids in the pathogenesis and outcomes of pre-eclampsia, how abnormal lipid genes may be implicated and consider whether treatment of hyperlipidemia has a more general place in the prevention or treatment of pre-eclampsia.
Collapse
Affiliation(s)
- Helen L Barrett
- School of Medicine, The University of Queensland, St Lucia , Queensland , Australia
| | | | | | | |
Collapse
|
47
|
Metabolomic profile of umbilical cord blood plasma from early and late intrauterine growth restricted (IUGR) neonates with and without signs of brain vasodilation. PLoS One 2013; 8:e80121. [PMID: 24312458 PMCID: PMC3846503 DOI: 10.1371/journal.pone.0080121] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/30/2013] [Indexed: 12/04/2022] Open
Abstract
Objectives To characterize via NMR spectroscopy the full spectrum of metabolic changes in umbilical vein blood plasma of newborns diagnosed with different clinical forms of intrauterine growth restriction (IUGR). Methods 23 early IUGR cases and matched 23 adequate-for-gestational-age (AGA) controls and 56 late IUGR cases with 56 matched AGAs were included in this study. Early IUGR was defined as a birth weight <10th centile, abnormal umbilical artery (UA) Doppler and delivery <35 weeks. Late IUGR was defined as a birth weight <10th centile with normal UA Doppler and delivery >35 weeks. This group was subdivided in 18 vasodilated (VD) and 38 non-VD late IUGR fetuses. All AGA patients had a birth weight >10th centile. 1H nuclear magnetic resonance (NMR) metabolomics of the blood samples collected from the umbilical vein at delivery was obtained. Multivariate statistical analysis identified several metabolites that allowed the discrimination between the different IUGR subgroups, and their comparative levels were quantified from the NMR data. Results The NMR-based analysis showed increased unsaturated lipids and VLDL levels in both early and late IUGR samples, decreased glucose and increased acetone levels in early IUGR. Non-significant trends for decreased glucose and increased acetone levels were present in late IUGR, which followed a severity gradient when the VD and non-VD subgroups were considered. Regarding amino acids and derivatives, early IUGR showed significantly increased glutamine and creatine levels, whereas the amounts of phenylalanine and tyrosine were decreased in early and late-VD IUGR samples. Valine and leucine were decreased in late IUGR samples. Choline levels were decreased in all clinical subforms of IUGR. Conclusions IUGR is not associated with a unique metabolic profile, but important changes are present in different clinical subsets used in research and clinical practice. These results may help in characterizing comprehensively specific alterations underlying different IUGR subsets.
Collapse
|
48
|
Wadhwani NS, Pisal HR, Mehendale SS, Joshi SR. A prospective study of maternal fatty acids, micronutrients and homocysteine and their association with birth outcome. MATERNAL AND CHILD NUTRITION 2013; 11:559-73. [PMID: 23795920 DOI: 10.1111/mcn.12062] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Our earlier studies both in animals and in humans have indicated that micronutrients (folic acid, vitamin B12) and long-chain polyunsaturated fatty acids, especially docosahexaenoic acid (DHA), are interlinked in the one-carbon cycle, which plays an important role in fetal 'programming' of adult diseases. The present study examines the levels of maternal and cord plasma fatty acids, maternal folate, vitamin B12 and homocysteine in healthy mothers at various time points during pregnancy and also examine an association between them. A longitudinal study of 106 normal pregnant women was carried out, and maternal blood was collected at three time points, viz., T1 = 16-20th week, T2 = 26-30th week and T3 = at delivery. Cord blood was collected at delivery. Fatty acids were estimated using a gas chromatograph. Levels of folate, vitamin B12 and homocysteine were estimated by the chemiluminescent microparticle immunoassay (CMIA) technology. Maternal plasma folate (P < 0.05), vitamin B12 (P < 0.01) and DHA (P < 0.05) levels were lowest, while maternal homocysteine levels were highest (P < 0.01) at T3. There was a negative association between maternal DHA and homocysteine at T2 (P < 0.05) and T3 (P < 0.01). There was a positive association between plasma DHA in maternal blood at T3 and cord blood. Furthermore, there was a positive association between maternal folate and vitamin B12 at T3 and baby weight, whereas maternal homocysteine at T1 were inversely associated with baby weight at delivery. Our study provides evidence for the associations of folic acid, vitamin B12, homocysteine with DHA and baby weight, suggesting that a balanced dietary supplementation of folate-vitamin B12-DHA during pregnancy may be beneficial.
Collapse
Affiliation(s)
- Nisha S Wadhwani
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, India
| | - Hemlata R Pisal
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, India
| | - Savita S Mehendale
- Department of Obstetrics and Gynaecology, Bharati Medical College and Hospital, Charity Vidyapeeth University, Pune, India
| | - Sadhana R Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, India
| |
Collapse
|
49
|
van Vliet E, Eixarch E, Illa M, Arbat-Plana A, González-Tendero A, Hogberg HT, Zhao L, Hartung T, Gratacos E. Metabolomics reveals metabolic alterations by intrauterine growth restriction in the fetal rabbit brain. PLoS One 2013; 8:e64545. [PMID: 23724060 PMCID: PMC3664640 DOI: 10.1371/journal.pone.0064545] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/16/2013] [Indexed: 12/13/2022] Open
Abstract
Background Intrauterine Growth Restriction (IUGR) due to placental insufficiency occurs in 5–10% of pregnancies and is a major risk factor for abnormal neurodevelopment. The perinatal diagnosis of IUGR related abnormal neurodevelopment represents a major challenge in fetal medicine. The development of clinical biomarkers is considered a promising approach, but requires the identification of biochemical/molecular alterations by IUGR in the fetal brain. This targeted metabolomics study in a rabbit IUGR model aimed to obtain mechanistic insight into the effects of IUGR on the fetal brain and identify metabolite candidates for biomarker development. Methodology/Principal Findings At gestation day 25, IUGR was induced in two New Zealand rabbits by 40–50% uteroplacental vessel ligation in one horn and the contralateral horn was used as control. At day 30, fetuses were delivered by Cesarian section, weighed and brains collected for metabolomics analysis. Results showed that IUGR fetuses had a significantly lower birth and brain weight compared to controls. Metabolomics analysis using liquid chromatography-quadrupole time-of-flight mass spectrometry (LC-QTOF-MS) and database matching identified 78 metabolites. Comparison of metabolite intensities using a t-test demonstrated that 18 metabolites were significantly different between control and IUGR brain tissue, including neurotransmitters/peptides, amino acids, fatty acids, energy metabolism intermediates and oxidative stress metabolites. Principle component and hierarchical cluster analysis showed cluster formations that clearly separated control from IUGR brain tissue samples, revealing the potential to develop predictive biomarkers. Moreover birth weight and metabolite intensity correlations indicated that the extent of alterations was dependent on the severity of IUGR. Conclusions IUGR leads to metabolic alterations in the fetal rabbit brain, involving neuronal viability, energy metabolism, amino acid levels, fatty acid profiles and oxidative stress mechanisms. Overall findings identified aspargine, ornithine, N-acetylaspartylglutamic acid, N-acetylaspartate and palmitoleic acid as potential metabolite candidates to develop clinical biomarkers for the perinatal diagnosis of IUGR related abnormal neurodevelopment.
Collapse
Affiliation(s)
- Erwin van Vliet
- Department of Maternal-Fetal Medicine, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Hospital Clinic and Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Elisenda Eixarch
- Department of Maternal-Fetal Medicine, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Hospital Clinic and Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Miriam Illa
- Department of Maternal-Fetal Medicine, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Hospital Clinic and Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Ariadna Arbat-Plana
- Department of Maternal-Fetal Medicine, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Hospital Clinic and Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Anna González-Tendero
- Department of Maternal-Fetal Medicine, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Hospital Clinic and Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Helena T. Hogberg
- Johns Hopkins University, Bloomberg School of Public Health, Department of Environmental Health Science, Baltimore, Maryland, United States of America
| | - Liang Zhao
- Johns Hopkins University, Bloomberg School of Public Health, Department of Environmental Health Science, Baltimore, Maryland, United States of America
| | - Thomas Hartung
- Johns Hopkins University, Bloomberg School of Public Health, Department of Environmental Health Science, Baltimore, Maryland, United States of America
| | - Eduard Gratacos
- Department of Maternal-Fetal Medicine, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Hospital Clinic and Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
50
|
Hallows SE, Regnault TRH, Betts DH. The long and short of it: the role of telomeres in fetal origins of adult disease. J Pregnancy 2012; 2012:638476. [PMID: 23094159 PMCID: PMC3471439 DOI: 10.1155/2012/638476] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Accepted: 08/24/2012] [Indexed: 12/30/2022] Open
Abstract
Placental insufficiency, maternal malnutrition, and other causes of intrauterine growth restriction (IUGR) can significantly affect short-term growth and long-term health. Following IUGR, there is an increased risk for cardiovascular disease and Type 2 Diabetes. The etiology of these diseases is beginning to be elucidated, and premature aging or cellular senescence through increased oxidative stress and DNA damage to telomeric ends may be initiators of these disease processes. This paper will explore the areas where telomere and telomerase biology can have significant effects on various tissues in the body in IUGR outcomes.
Collapse
Affiliation(s)
- Stephanie E. Hallows
- Department of Physiology and Pharmacology, University of Western Ontario, Ontario, London, ON, Canada N6A 5C1
| | - Timothy R. H. Regnault
- Department of Physiology and Pharmacology, University of Western Ontario, Ontario, London, ON, Canada N6A 5C1
- Department of Obstetrics and Gynaecology, University of Western Ontario, Ontario, London, ON, Canada N6H 5W9
- Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada N6C 2V5
| | - Dean H. Betts
- Department of Physiology and Pharmacology, University of Western Ontario, Ontario, London, ON, Canada N6A 5C1
- Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada N6C 2V5
| |
Collapse
|