1
|
Jiang Y, Zhou X, He Z, Wei L, Zhu S, Zhang H, Zhang J, Chen Y, Du Y, Ding W, Wang S, Wang Z, Feng L. DHA Improves neurodevelopmental abnormalities in offspring of gestational diabetes mellitus patients via the PPAR-γ/FATP4 pathway. Biochem Pharmacol 2025; 232:116726. [PMID: 39716642 DOI: 10.1016/j.bcp.2024.116726] [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: 09/11/2024] [Revised: 12/03/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Offspring of women with gestational diabetes mellitus (GDM) face an increased risk of long-term neurodevelopmental abnormalities. This study explores the altered expression of key placental fatty acid transport proteins-FATP2, FATP4, FATP6, FABP4, and FAT/CD36-in GDM patients, and the potential of docosahexaenoic acid (DHA) to mitigate neurodevelopmental risks in offspring by enhancing their expression through activation of peroxisome proliferator-activated receptor γ (PPAR-γ). Our findings demonstrate that placental FATP4 expression is reduced in GDM patients. In HTR8/SVneo cells, PPAR-γ activation upregulated the expression of FATP4, FAT/CD36, and FABP4, while PPAR-γ inhibition only reduced FAT/CD36 expression. DHA treatment led to increased expression of FATP4, FATP/CD36 and FABP4, which was partially reversed by PPAR-γ inhibition. Consistent results were observed in an insulin-resistant cell model. Supplementing GDM mice with exogenous DHA restored placental FATP4 expression and improved offspring social behavior and cognitive function. These results suggest that DHA supplementation during pregnancy could reduce the adverse effects of GDM on placental FATP4 expression and support better neurodevelopmental outcomes in offspring by promoting essential fatty acid transport through the PPAR-γ/FATP4 pathway. This study highlights the therapeutic potential of DHA in improving fetal outcomes in GDM pregnancies.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xuan Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhenzhen He
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lijie Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shenglan Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Huiting Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jingyi Zhang
- Department of Obstetrics and Gynecology Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuting Chen
- Department of Obstetrics and Gynecology Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuanyuan Du
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wencheng Ding
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shaoshuai Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Zizhuo Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Bonnell V, White M, Connor K. Do nutritional interventions before or during pregnancy affect placental phenotype? Findings from a systematic review of human clinical trials. J Glob Health 2024; 14:04240. [PMID: 39700380 DOI: 10.7189/jogh.14.04240] [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: 12/21/2024] Open
Abstract
Background Maternal nutritional interventions aim to address nutrient deficiencies in pregnancy, a leading cause of maternal and neonatal morbidity and mortality worldwide. How these interventions influence the placenta, which plays a vital role in fetal growth and nutrient supply, is not well understood. This leaves a major gap in understanding how such interventions could influence pregnancy outcomes and fetal health. We hypothesised that nutritional interventions influence placental phenotype, and that these placental changes relate to how successful an intervention is in improving pregnancy outcomes. Methods We searched PubMed, ClinicalTrials.gov, and the World Health Organization (WHO) International Clinical Trials Registry Platform using pre-defined search terms for records published from January 2001 to September 2021 that reported on clinical trials in humans, which administered a maternal nutritional intervention during the periconceptional or pregnancy period and reported on placental phenotype (shape and form, function or placental disorders). These records were then screened by two reviewers for eligibility. Results Fifty-three eligible articles reported on (multiple) micronutrient- (n = 33 studies), lipid- (n = 11), protein- (n = 2), and diet-/lifestyle-based (n = 8) interventions. Of the micronutrient-based interventions, 16 (48%) were associated with altered placental function, namely altered nutrient transport/metabolism (n = 9). Nine (82%) of the lipid-based interventions were associated with altered placental phenotype, including elevated placental fatty acid levels (n = 5), altered nutrient transport/metabolism gene expression (n = 4), and decreased inflammatory biomarkers (n = 2). Of the protein-based interventions, two (66%) were associated with altered placental phenotype, including increased placental efficiency (n = 1) and decreased preeclampsia risk (n = 1). Three (38%) of diet and lifestyle-based interventions were associated with placental changes, namely placental gene expression (n = 1) and disease (n = 2). In studies with data on maternal (n = 30) or offspring (n = 20) outcomes, interventions that influenced placental phenotype were more likely to have also been associated with improved maternal outcomes (n/N = 11/15, 73%) and offspring birth outcomes (n/N = 6/11, 54%) compared to interventions that did not associate with placental changes (n/N = 2/15 (13%) and n/N = 1/9 (11%) respectively). Conclusions Periconceptional and prenatal nutritional interventions to improve maternal/pregnancy health associate with altered placental development and function. These placental adaptations likely benefit the pregnancy and improve offspring outcomes. Understanding the placenta's role in the success of interventions to combat nutrient deficiencies is critical for improving interventions and reducing maternal and neonatal morbidity and mortality globally.
Collapse
|
3
|
Li Q, Zhang Q, Su S, Yang S, Shao J, Guan W, Zhang S. Maternal fish oil supplementation enhances nutrient transport in the placenta and milk biosynthesis in the mammary gland via the GPR120 signaling pathway. J Adv Res 2024:S2090-1232(24)00607-6. [PMID: 39706333 DOI: 10.1016/j.jare.2024.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Maternal fish oil (FO) supplementation during pregnancy has been shown to improve pregnancy outcomes. FO is recognized as dietary source for n-3 polyunsaturated fatty acids (n-3 PUFAs). While early research has focused on the benefits of n-3 PUFAs for fetal neurodevelopment, retinal maturation and neonatal behavior, their roles in the placenta during late pregnancy and in the mammary gland during lactation still remain unknow. OBJECTIVES Here, we aim to clarify the mechanisms by which maternal supplementation with FO during pregnancy and lactation affects placental and mammary gland function. METHODS We evaluated the effects of FO on maternal placental nutrient transport, mammary gland milk synthesis and offspring growth. We then explored the molecular mechanisms by which docosahexaenoic acid (DHA) affects the biological function of placental trophoblast cells and mammary epithelial cells through in vitro experiments. Finally, a lipopolysaccharide-challenged experiment was performed to access the potential of maternal FO supplementation in alleviating offspring intestinal inflammation. RESULTS Maternal supplementation with FO during late pregnancy increased offspring birth weight, associated with enhanced maternal placental vascularization and nutrient transporter abundance. Additionally, maternal FO supplementation during lactation improved milk biosynthesis, increasing the fat, protein, and non-fat solids content in both colostrum and mature milk, thereby promoting offspring growth. The stimulatory effects of DHA on nutrient transportation in placental trophoblast cells and nutrient secretion in mammary gland epithelial cells were mediated by GPR120 signaling pathways. Furthermore, maternal FO supplementation strengthened the placental barrier, reduced placental inflammation, oxidative stress and alleviated lipopolysaccharide-induced intestinal inflammation in offspring. CONCLUSION Maternal FO supplementation during late pregnancy and lactation enhances offspring growth by increasing placental nutrient transport and milk biosynthesis, mediated by GPR120. Additionally, maternal FO supplementation reduces the susceptibility of offspring to intestinal inflammation.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Senlin Su
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
4
|
Blitek A, Szymanska M. Expression Profiles of Fatty Acid Transporters and the Role of n-3 and n-6 Polyunsaturated Fatty Acids in the Porcine Endometrium. Int J Mol Sci 2024; 25:11102. [PMID: 39456882 PMCID: PMC11507490 DOI: 10.3390/ijms252011102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Fatty acids (FAs) are important for cell membrane composition, eicosanoid synthesis, and metabolic processes. Membrane proteins that facilitate FA transport into cells include FA translocase (also known as CD36) and FA transporter proteins (encoded by SLC27A genes). The present study aimed to examine expression profiles of FA transporters in the endometrium of cyclic and early pregnant gilts on days 3 to 20 after estrus and the possible regulation by conceptus signals and polyunsaturated FAs (PUFAs). The effect of PUFAs on prostaglandin (PG) synthesis and transcript abundance of genes related to FA action and metabolism, angiogenesis, and immune response was also determined. Day after estrus and reproductive status of animals affected FA transporter expression, with greater levels of CD36, SLC27A1, and SLC27A4 observed in pregnant than in cyclic gilts. Conceptus-conditioned medium and/or estradiol-17β stimulated SLC27A1 and CD36 expression. Among PUFAs, linoleic acid decreased SLC27A1 and SLC27A6 mRNA expression, while arachidonic, docosahexaenoic, and eicosapentaenoic acids increased SLC27A4 transcript abundance. Moreover, arachidonic acid stimulated ACOX1, CPT1A, and IL1B expression and increased PGE2 and PGI2 secretion. In turn, α-linolenic acid up-regulated VEGFA, FGF2, FABP4, and PPARG mRNA expression. These results indicate the presence of an active transport of FAs in the porcine endometrium and the role of PUFAs as modulators of the uterine activity during conceptus implantation.
Collapse
Affiliation(s)
- Agnieszka Blitek
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland;
| | | |
Collapse
|
5
|
Cao M, Bai L, Wei H, Guo Y, Sun G, Sun H, Shi B. Dietary supplementation with pterostilbene activates the PI3K-AKT-mTOR signalling pathway to alleviate progressive oxidative stress and promote placental nutrient transport. J Anim Sci Biotechnol 2024; 15:133. [PMID: 39369257 PMCID: PMC11456245 DOI: 10.1186/s40104-024-01090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/14/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Progressive oxidative stress easily occurs as a result of a gradual increase in the intensity of maternal metabolism due to rapid foetal development and increased intensity of lactation. However, studies on the effects of processive oxidative stress on nutrient transport in the placenta have received little attention. The present study was conducted on sows at 85 days of gestation to study the effects of pterostilbene (PTE) on maternal oxidative stress status and placental nutrient transport. RESULTS PTE increased the antioxidant capacity and immunoglobulin content in mothers' blood and milk, reduced the level of inflammatory factors, and improved the nutrient content of milk. PTE also reduced sow backfat loss and the number of weak sons, and increased piglet weaning weight and total weaning litter weight. We subsequently found that PTE enhanced placental glucose and fatty acid transport and further affected glycolipid metabolism by increasing the expression of LAL, PYGM, and Gbe-1, which activated the PI3K phosphorylation pathway. Moreover, PTE addition altered the relative abundance of the Firmicutes, Proteobacteria, Parabacillus, and Bacteroidetes-like RF16 groups in sow faeces. PTE increased the levels of acetate, propionate, butyrate and isovalerate in the faeces. CONCLUSIONS These findings reveal that the addition of PTE during pregnancy and lactation mitigates the effects of processive oxidative stress on offspring development by altering maternal microbial and placental nutrient transport capacity.
Collapse
Affiliation(s)
- Mingming Cao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Liyun Bai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Haoyun Wei
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yantong Guo
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Guodong Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Haoyang Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
6
|
Masserdotti A, Gasik M, Grillari-Voglauer R, Grillari J, Cargnoni A, Chiodelli P, Papait A, Magatti M, Romoli J, Ficai S, Di Pietro L, Lattanzi W, Silini AR, Parolini O. Unveiling the human fetal-maternal interface during the first trimester: biophysical knowledge and gaps. Front Cell Dev Biol 2024; 12:1411582. [PMID: 39144254 PMCID: PMC11322133 DOI: 10.3389/fcell.2024.1411582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 08/16/2024] Open
Abstract
The intricate interplay between the developing placenta and fetal-maternal interactions is critical for pregnancy outcomes. Despite advancements, gaps persist in understanding biomechanics, transport processes, and blood circulation parameters, all of which are crucial for safe pregnancies. Moreover, the complexity of fetal-maternal interactions led to conflicting data and methodological variations. This review presents a comprehensive overview of current knowledge on fetal-maternal interface structures, with a particular focus on the first trimester. More in detail, the embryological development, structural characteristics, and physiological functions of placental chorionic plate and villi, fetal membranes and umbilical cord are discussed. Furthermore, a description of the main structures and features of maternal and fetal fluid dynamic exchanges is provided. However, ethical constraints and technological limitations pose still challenges to studying early placental development directly, which calls for sophisticated in vitro, microfluidic organotypic models for advancing our understanding. For this, knowledge about key in vivo parameters are necessary for their design. In this scenario, the integration of data from later gestational stages and mathematical/computational simulations have proven to be useful tools. Notwithstanding, further research into cellular and molecular mechanisms at the fetal-maternal interface is essential for enhancing prenatal care and improving maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Alice Masserdotti
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- Institute of Molecular Biotechnology, BOKU University, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Paola Chiodelli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sara Ficai
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorena Di Pietro
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| |
Collapse
|
7
|
Dos Santos Silva P, Kra G, Butenko Y, Daddam JR, Levin Y, Zachut M. Maternal supplementation with n-3 fatty acids affects placental lipid metabolism, inflammation, oxidative stress, the endocannabinoid system, and the neonate cytokine concentrations in dairy cows. J Anim Sci Biotechnol 2024; 15:74. [PMID: 38769527 PMCID: PMC11106909 DOI: 10.1186/s40104-024-01033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/06/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND The placenta plays a crucial role in supporting and influencing fetal development. We compared the effects of prepartum supplementation with omega-3 (n-3) fatty acid (FA) sources, flaxseed oil (FLX) and fish oil (FO), on the expression of genes and proteins related to lipid metabolism, inflammation, oxidative stress, and the endocannabinoid system (ECS) in the expelled placenta, as well as on FA profile and inflammatory response of neonates. Late-pregnant Holstein dairy cows were supplemented with saturated fat (CTL), FLX, or FO. Placental cotyledons (n = 5) were collected immediately after expulsion, and extracted RNA and proteins were analyzed by RT-PCR and proteomic analysis. Neonatal blood was assessed for FA composition and concentrations of inflammatory markers. RESULTS FO increased the gene expression of fatty acid binding protein 4 (FABP4), interleukin 10 (IL-10), catalase (CAT), cannabinoid receptor 1 (CNR1), and cannabinoid receptor 2 (CNR2) compared with CTL placenta. Gene expression of ECS-enzyme FA-amide hydrolase (FAAH) was lower in FLX and FO than in CTL. Proteomic analysis identified 3,974 proteins; of these, 51-59 were differentially abundant between treatments (P ≤ 0.05, |fold change| ≥ 1.5). Top canonical pathways enriched in FLX vs. CTL and in FO vs. CTL were triglyceride metabolism and inflammatory processes. Both n-3 FA increased the placental abundance of FA binding proteins (FABPs) 3 and 7. The abundance of CNR1 cannabinoid-receptor-interacting-protein-1 (CNRIP1) was reduced in FO vs. FLX. In silico modeling affirmed that bovine FABPs bind to endocannabinoids. The FLX increased the abundance of inflammatory CD44-antigen and secreted-phosphoprotein-1, whereas prostaglandin-endoperoxide synthase 2 was decreased in FO vs. CTL placenta. Maternal FO enriched neonatal plasma with n-3 FAs, and both FLX and FO reduced interleukin-6 concentrations compared with CTL. CONCLUSION Maternal n-3 FA from FLX and FO differentially affected the bovine placenta; both enhanced lipid metabolism and modulated oxidative stress, however, FO increased some transcriptional ECS components, possibly related to the increased FABPs. Maternal FO induced a unique balance of pro- and anti-inflammatory components in the placenta. Taken together, different sources of n-3 FA during late pregnancy enhanced placental immune and metabolic processes, which may affect the neonatal immune system.
Collapse
Affiliation(s)
- Priscila Dos Santos Silva
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel
| | - Gitit Kra
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel
- Department of Animal Science, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yana Butenko
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel
| | | | - Yishai Levin
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Maya Zachut
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel.
| |
Collapse
|
8
|
Carré C, Acar N, Daruich A, Grégoire S, Martine L, Buteau B, Aho S, Eid P, Arnould L, Bron AM, Driessen M, Kermorvant E, Simon E, Creuzot-Garcher C, Gabrielle PH. Study protocol of OmegaROP-2 prospective study: expression of placental fatty acid receptors in preterm newborns with retinopathy of prematurity. BMC Ophthalmol 2023; 23:404. [PMID: 37803473 PMCID: PMC10559396 DOI: 10.1186/s12886-023-03156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Incomplete vascularization of the retina in preterm infants carries a risk of retinopathy of prematurity (ROP). Progress in neonatal resuscitation in developing countries has led to the survival of an increasing number of premature infants, resulting in an increased rate of ROP and consequently in visual disability. Strategies to reduce ROP involve optimizing oxygen saturation, nutrition, and normalizing factors such as insulin-like growth factor 1 and n-3 long-chain polyunsaturated fatty acids (LC-PUFA). Our previous study, OmegaROP, showed that there is an accumulation or retention of docosahexaenoic acid (DHA) in mothers of infants developing ROP, suggesting abnormalities in the LC-PUFA placental transfer via fatty acid transporting proteins. The present study aims to better understand the LC-PUFA transport dysfunction in the fetoplacental unit during pregnancy and to find a novel target for the prevention of ROP development. METHODS The study protocol is designed to evaluate the correlation between the expression level of placental fatty acid receptors and ROP occurrence. This ongoing study will include 100 mother-infant dyads: mother-infant dyads born before 29 weeks of gestational age (GA) and mother-infant dyads with full-term pregnancies. Recruitment is planned over a period of 46 months. Maternal and cord blood samples as well as placental tissue samples will be taken following delivery. ROP screening will be performed using wide-field camera imaging according to the International Classification of ROP consensus statement. DISCUSSION The results of this study will have a tangible impact on public health. Indeed, if we show a correlation between the expression level of placental omega-3 receptors and the occurrence of ROP, it would be an essential step in discovering novel pathophysiological mechanisms involved in this retinopathy. TRIAL REGISTRATION NCT04819893.
Collapse
Affiliation(s)
- Chloé Carré
- Department of Ophthalmology, Dijon University Hospital, 14 Rue Paul Gaffarel, 21000, Dijon, France
| | - Niyazi Acar
- Eye and Nutrition Research Group, Centre Des Sciences du Goût Et de L'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | | | - Stéphane Grégoire
- Eye and Nutrition Research Group, Centre Des Sciences du Goût Et de L'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Lucy Martine
- Eye and Nutrition Research Group, Centre Des Sciences du Goût Et de L'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Bénédicte Buteau
- Eye and Nutrition Research Group, Centre Des Sciences du Goût Et de L'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Serge Aho
- Department of Epidemiology & Biostatistics, Dijon University Hospital, Dijon, France
| | - Petra Eid
- Department of Ophthalmology, Dijon University Hospital, 14 Rue Paul Gaffarel, 21000, Dijon, France
| | - Louis Arnould
- Department of Ophthalmology, Dijon University Hospital, 14 Rue Paul Gaffarel, 21000, Dijon, France
| | - Alain Marie Bron
- Department of Ophthalmology, Dijon University Hospital, 14 Rue Paul Gaffarel, 21000, Dijon, France
- Eye and Nutrition Research Group, Centre Des Sciences du Goût Et de L'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | | | | | - Emmanuel Simon
- Department of Gynecology, Dijon University Hospital, Dijon, France
| | - Catherine Creuzot-Garcher
- Department of Ophthalmology, Dijon University Hospital, 14 Rue Paul Gaffarel, 21000, Dijon, France
- Eye and Nutrition Research Group, Centre Des Sciences du Goût Et de L'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre-Henry Gabrielle
- Department of Ophthalmology, Dijon University Hospital, 14 Rue Paul Gaffarel, 21000, Dijon, France.
- Eye and Nutrition Research Group, Centre Des Sciences du Goût Et de L'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France.
| |
Collapse
|
9
|
Powell TL, Uhlson C, Madi L, Berry KZ, Chassen SS, Jansson T, Ferchaud-Roucher V. Fetal sex differences in placental LCPUFA ether and plasmalogen phosphatidylethanolamine and phosphatidylcholine contents in pregnancies complicated by obesity. Biol Sex Differ 2023; 14:66. [PMID: 37770949 PMCID: PMC10540428 DOI: 10.1186/s13293-023-00548-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND We have previously reported that maternal obesity reduces placental transport capacity for lysophosphatidylcholine-docosahexaenoic acid (LPC-DHA), a preferred form for transfer of DHA (omega 3) to the fetal brain, but only in male fetuses. Phosphatidylethanolamine (PE) and phosphatidylcholine (PC), have either sn-1 ester, ether or vinyl ether (plasmalogen) linkages to primarily unsaturated and monounsaturated fatty acids and DHA or arachidonic acid (ARA, omega 6) in the sn-2 position. Whether ether and plasmalogen PC and PE metabolism in placenta impacts transfer to the fetus is unexplored. We hypothesized that ether and plasmalogen PC and PE containing DHA and ARA are reduced in maternal-fetal unit in pregnancies complicated by obesity and these differences are dependent on fetal sex. METHODS In maternal, umbilical cord plasma and placentas from obese women (11 female/5 male infants) and normal weight women (9 female/7 male infants), all PC and PE species containing DHA and ARA were analyzed by LC-MS/MS. Placental protein expression of enzymes involved in phospholipid synthesis, were determined by immunoblotting. All variables were compared between control vs obese groups and separated by fetal sex, in each sample using the Benjamini-Hochberg false discovery rate adjustment to account for multiple testing. RESULTS Levels of ester PC containing DHA and ARA were profoundly reduced by 60-92% in male placentas of obese mothers, while levels of ether and plasmalogen PE containing DHA and ARA were decreased by 51-84% in female placentas. PLA2G4C abundance was lower in male placentas and LPCAT4 abundance was lower solely in females in obesity. In umbilical cord, levels of ester, ether and plasmalogen PC and PE with DHA were reduced by 43-61% in male, but not female, fetuses of obese mothers. CONCLUSIONS We found a fetal sex effect in placental PE and PC ester, ether and plasmalogen PE and PC containing DHA in response to maternal obesity which appears to reflect an ability of female placentas to adapt to maintain optimal fetal DHA transfer in maternal obesity.
Collapse
Affiliation(s)
- Theresa L Powell
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charis Uhlson
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lana Madi
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Karin Zemski Berry
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie S Chassen
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Veronique Ferchaud-Roucher
- Nantes Université, CHU Nantes, INRAE UMR 1280 PhAN, CRNH Ouest, 44000, Nantes, France.
- Nantes Université, INRAE, UMR 1280 PhAN, CHU Hôtel Dieu, HNB1, 1 place Alexis Ricordeau, 44093, Nantes, France.
| |
Collapse
|
10
|
Gómez-Vilarrubla A, Mas-Parés B, Carreras-Badosa G, Jové M, Berdún R, Bonmatí-Santané A, de Zegher F, Ibañez L, López-Bermejo A, Bassols J. Placental AA/EPA Ratio Is Associated with Obesity Risk Parameters in the Offspring at 6 Years of Age. Int J Mol Sci 2023; 24:10087. [PMID: 37373236 DOI: 10.3390/ijms241210087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
During pregnancy, maternal polyunsaturated fatty acids (PUFA) are transferred to the fetus through the placenta by specific FA transporters (FATP). A higher perinatal exposure to n-6 over n-3 PUFA could be linked to excess fat mass and obesity development later in life. In this context, we aimed to assess the associations between long chain PUFAs (LC-PUFAs) (n-6, n-3, and n-6/n-3 ratios) measured in the placenta at term birth with obesity-related parameters in the offspring at 6 years of age and assess whether these associations are dependent on the placental relative expression of fatty acid transporters. As results, the PUFAn-6/PUFAn-3 ratio was 4/1, which scaled up to 15/1 when considering only the arachidonic acid/eicosapentaenoic acid ratio (AA/EPA ratio). Positive associations between the AA/EPA ratio and offspring's obesity risk parameters were found with weight-SDS, BMI-SDS, percent fat mass-SDS, visceral fat, and HOMA-IR (r from 0.204 to 0.375; all p < 0.05). These associations were more noticeable in those subjects with higher expression of fatty acid transporters. Therefore, in conclusion, a higher placental AA/EPA ratio is positively associated with offspring's visceral adiposity and obesity risk parameters, which become more apparent in subjects with higher expressions of placental FATPs. Our results support the potential role of n-6 and n-3 LC-PUFA in the fetal programming of obesity risk in childhood. For the present study, 113 healthy pregnant women were recruited during the first trimester of pregnancy and their offspring were followed up at 6 years of age. The fatty acid profiles and the expression of fatty acid transporters (FATP1 and FATP4) were analyzed from placental samples at birth. Associations between LC-PUFA (n-6, n-3, and n-6/n-3 ratios) and obesity risk parameters (weight, body mass index (BMI), percent fat mass, visceral fat, and homeostatic model assessment of insulin resistance (HOMA-IR)) in the offspring at 6 years of age were examined.
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
| | - 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 (UdL-IRBLleida), 25008 Lleida, Spain
| | - Rebeca Berdún
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 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 de Llobregat, 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
- Department of Medical Sciences, University of Girona, 17003 Girona, Spain
| | - Judit Bassols
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| |
Collapse
|
11
|
Effect of the Supplementation Using an Herbal Mixture as a Choline Source during Early Gestation in Rambouillet Ewes. Animals (Basel) 2023; 13:ani13040645. [PMID: 36830432 PMCID: PMC9951712 DOI: 10.3390/ani13040645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Previous research indicates that adequate choline nutrition during late gestation improves fetal development. However, there is a lack of studies describing choline's role during early gestation. Thus, the current study hypothesizes that an herbal mixture as a source of choline (Biocholine) positively affects offspring development from ewes supplemented during early gestation. Therefore, the objectives were to evaluate the impact of biocholine on the programming of the offspring early in life through the evaluation of dams and newborn performance. Twenty-eight four-year-old Rambouillet ewes were assigned randomly to two treatments: non-supplementation and 4 gd-1 of biocholine during the early gestation. Compared with the dams without supplementation, the ewes supplemented using biocholine showed no increase in parameters such as birth and weaning weight (p > 0.05). Additionally, the milk yield and quality of colostrum and milk did not present statistical differences (p > 0.05). However, the placental membrane development was reduced in the ewes that received supplementation with biocholine; interestingly, those dams increased the weight of the newborns during the lambing period (p < 0.05). Finally, the current study proposes necessary elucidation of how placental size is programmed and if less placental development has potential benefits in the fetus's development.
Collapse
|
12
|
Garcia-Santillan JA, Lazo-de-la-Vega-Monroy ML, Rodriguez-Saldaña GC, Solis-Barbosa MA, Corona-Figueroa MA, Gonzalez-Dominguez MI, Gomez-Zapata HM, Malacara JM, Barbosa-Sabanero G. Placental Nutrient Transporters and Maternal Fatty Acids in SGA, AGA, and LGA Newborns From Mothers With and Without Obesity. Front Cell Dev Biol 2022; 10:822527. [PMID: 35399516 PMCID: PMC8990844 DOI: 10.3389/fcell.2022.822527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/25/2022] [Indexed: 12/01/2022] Open
Abstract
Adverse environmental factors in early life result in fetal metabolic programming and increased risk of adult diseases. Birth weight is an indirect marker of the intrauterine environment, modulated by nutrient availability and placental transport capacity. However, studies of placental transporters in idiopathic birth weight alterations and in maternal obesity in relation to neonatal metabolic outcomes are scarce. We aimed to analyze the placental nutrient transporter protein expression in small (SGA, n = 14), adequate (AGA, n = 18), and large (LGA n = 10) gestational age term for newborns from healthy or obese mothers (LGA-OB, n = 9) and their association with maternal fatty acids, metabolic status, placental triglycerides, and neonatal outcomes. The transporter expression was determined by Western blot. The fatty acid profile was evaluated by gas chromatography, and placental triglycerides were quantified by an enzymatic colorimetric method. GLUT1 was higher in LGA and lower in SGA and positively correlated with maternal HbA1c and placental weight (PW). SNAT2 was lower in SGA, while SNAT4 was lower in LGA-OB. FATP1 was lower in SGA and higher in LGA. SNAT4 correlated negatively and FATP1 correlated positively with the PW and birth anthropometry (BA). Placental triglycerides were higher in LGA and LGA-OB and correlated with pregestational BMI, maternal insulin, and BA. Maternal docosahexaenoic acid (DHA) was higher in SGA, specifically in male placentas, correlating negatively with maternal triglycerides, PW, cord glucose, and abdominal perimeter. Palmitic acid (PA) correlated positively with FATP4 and cord insulin, linoleic acid correlated negatively with PA and maternal cholesterol, and arachidonic acid correlated inversely with maternal TG and directly with FATP4. Our study highlights the importance of placental programming in birth weight both in healthy and obese pregnancies.
Collapse
Affiliation(s)
| | | | | | - Miguel-Angel Solis-Barbosa
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Campus Leon, Guanajuato, Mexico
| | | | | | | | - Juan-Manuel Malacara
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Campus Leon, Guanajuato, Mexico
| | - Gloria Barbosa-Sabanero
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Campus Leon, Guanajuato, Mexico
- *Correspondence: Gloria Barbosa-Sabanero,
| |
Collapse
|
13
|
Sanches APV, de Oliveira JL, Ferreira MS, Lima BDS, Miyamoto JÉ, Simino LADP, Torsoni MA, Torsoni AS, Milanski M, Ignácio-Souza LM. Obesity phenotype induced by high-fat diet leads to maternal-fetal constraint, placental inefficiency, and fetal growth restriction in mice. J Nutr Biochem 2022; 104:108977. [PMID: 35248701 DOI: 10.1016/j.jnutbio.2022.108977] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/20/2021] [Accepted: 02/09/2022] [Indexed: 11/28/2022]
Abstract
The aim of this study was to investigate certain parameters regarding the maternal-fetal outcomes in a diet-induced obesity model. Obese, glucose-intolerant females who were exposed to a high-fat diet prior to pregnancy had lower placental efficiency and lower birth weight pups compared to the controls. Simple linear regression analyses showed that maternal obesity disrupts the proportionality between maternal and fetal outcomes during pregnancy. Maternal obesity is correlated with fetal outcomes, perhaps because of problems with hormonal signaling and exacerbation of inflammation in the maternal metabolic environment. The maternal obese phenotype altered the thickness of the placental layer, the transport of fatty acids, and the expression of growth factors. For example, lower expression of epidermal growth factor receptor (EGFR) mRNA in the obesity-prone group may have contributed to the rupture of the placental layers, leading to adverse fetal outcomes. Furthermore, maintenance of maternal glucose homeostasis and overexpression of placental growth factor (PGF) in the obesity-resistant group likely protected the placenta and fetuses from morphological and functional damage.
Collapse
Affiliation(s)
- Ana Paula Varela Sanches
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Josilene Lopes de Oliveira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Maíra Schuchter Ferreira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Bruna de Souza Lima
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Josiane Érica Miyamoto
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Letícia Martins Ignácio-Souza
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.
| |
Collapse
|
14
|
Lee JG, Kim G, Park SG, Yon JM, Yeom J, Song HE, Cheong SA, Lim JS, Sung YH, Kim K, Yoo HJ, Hong EJ, Nam KH, Seong JK, Kim CJ, Nam SY, Baek IJ. Lipid signatures reflect the function of the murine primary placentation. Biol Reprod 2021; 106:583-596. [PMID: 34850819 DOI: 10.1093/biolre/ioab219] [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: 09/14/2021] [Revised: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
The placenta regulates maternal-fetal communication, and its defect leads to significant pregnancy complications. The maternal and embryonic circulations are primitively connected in early placentation, but the function of the placenta during this developmentally essential period is relatively unknown. We thus performed a comparative proteomic analysis of the placenta before and after primary placentation and found that the metabolism and transport of lipids were characteristically activated in this period. The placental fatty acid (FA) carriers in specific placental compartments were upregulated according to gestational age, and metabolomic analysis also showed that the placental transport of FAs increased in a time-dependent manner. Further analysis of two mutant mice models with embryonic lethality revealed that lipid-related signatures could reflect the functional state of the placenta. Our findings highlight the importance of the nutrient transport function of the primary placenta in the early gestational period and the role of lipids in embryonic development.
Collapse
Affiliation(s)
- Jong Geol Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea
| | - Globinna Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seul Gi Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.,Biomedical Mouse Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongwon-Gun, Republic of Korea
| | - Jung-Min Yon
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeonghun Yeom
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ha Eun Song
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-A Cheong
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea
| | - Joon Seo Lim
- Clinical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Hoon Sung
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyun Ju Yoo
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ki-Hoan Nam
- Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,Biomedical Mouse Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongwon-Gun, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Chong Jai Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Yoon Nam
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - In-Jeoung Baek
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Martinat M, Rossitto M, Di Miceli M, Layé S. Perinatal Dietary Polyunsaturated Fatty Acids in Brain Development, Role in Neurodevelopmental Disorders. Nutrients 2021; 13:1185. [PMID: 33918517 PMCID: PMC8065891 DOI: 10.3390/nu13041185] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/26/2022] Open
Abstract
n-3 and n-6 polyunsaturated fatty acids (PUFAs) are essential fatty acids that are provided by dietary intake. Growing evidence suggests that n-3 and n-6 PUFAs are paramount for brain functions. They constitute crucial elements of cellular membranes, especially in the brain. They are the precursors of several metabolites with different effects on inflammation and neuron outgrowth. Overall, long-chain PUFAs accumulate in the offspring brain during the embryonic and post-natal periods. In this review, we discuss how they accumulate in the developing brain, considering the maternal dietary supply, the polymorphisms of genes involved in their metabolism, and the differences linked to gender. We also report the mechanisms linking their bioavailability in the developing brain, their transfer from the mother to the embryo through the placenta, and their role in brain development. In addition, data on the potential role of altered bioavailability of long-chain n-3 PUFAs in the etiologies of neurodevelopmental diseases, such as autism, attention deficit and hyperactivity disorder, and schizophrenia, are reviewed.
Collapse
|
16
|
Rasool A, Alvarado-Flores F, O'Tierney-Ginn P. Placental Impact of Dietary Supplements: More Than Micronutrients. Clin Ther 2020; 43:226-245. [PMID: 33358257 DOI: 10.1016/j.clinthera.2020.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Maternal nutrition is a key modifier of fetal growth and development. However, many maternal diets in the United States do not meet nutritional recommendations. Dietary supplementation is therefore necessary to meet nutritional goals. The effects of many supplements on placental development and function are poorly understood. In this review, we address the therapeutic potential of maternal dietary supplementation on placental development and function in both healthy and complicated pregnancies. METHODS This is a narrative review of original research articles published between February 1970 and July 2020 on dietary supplements consumed during pregnancy and placental outcomes (including nutrient uptake, metabolism and delivery, as well as growth and efficiency). Impacts of placental changes on fetal outcomes were also reviewed. Both human and animal studies were included. FINDINGS We found evidence of a potential therapeutic benefit of several supplements on maternal and fetal outcomes via their placental impacts. Our review supports a role for probiotics as a placental therapeutic, with effects that include improved inflammation and lipid metabolism, which may prevent preterm birth and poor placental efficiency. Supplementation with omega-3 fatty acids (as found in fish oil) during pregnancy tempers the negative effects of maternal obesity but may have little placental impact in healthy lean women. The beneficial effects of choline supplementation on maternal health and fetal growth are largely attributable to its placental impacts. l-arginine supplementation has a potent provascularization effect on the placenta, which may underlie its fetal growth-promoting properties. IMPLICATIONS The placenta is exquisitely sensitive to dietary supplements. Pregnant women should consult their health care practitioner before continuing or initiating use of a dietary supplement. Because little is known about impacts of many supplements on placental and long-term offspring health, more research is required before robust clinical recommendations can be made.
Collapse
Affiliation(s)
- Aisha Rasool
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA
| | | | | |
Collapse
|
17
|
Maternal Docosahexaenoic Acid Status during Pregnancy and Its Impact on Infant Neurodevelopment. Nutrients 2020; 12:nu12123615. [PMID: 33255561 PMCID: PMC7759779 DOI: 10.3390/nu12123615] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
Dietary components are essential for the structural and functional development of the brain. Among these, docosahexaenoic acid, 22:6n-3 (DHA), is critically necessary for the structure and development of the growing fetal brain in utero. DHA is the major n-3 long-chain polyunsaturated fatty acid in brain gray matter representing about 15% of all fatty acids in the human frontal cortex. DHA affects neurogenesis, neurotransmitter, synaptic plasticity and transmission, and signal transduction in the brain. Data from human and animal studies suggest that adequate levels of DHA in neural membranes are required for maturation of cortical astrocyte, neurovascular coupling, and glucose uptake and metabolism. Besides, some metabolites of DHA protect from oxidative tissue injury and stress in the brain. A low DHA level in the brain results in behavioral changes and is associated with learning difficulties and dementia. In humans, the third trimester-placental supply of maternal DHA to the growing fetus is critically important as the growing brain obligatory requires DHA during this window period. Besides, DHA is also involved in the early placentation process, essential for placental development. This underscores the importance of maternal intake of DHA for the structural and functional development of the brain. This review describes DHA’s multiple roles during gestation, lactation, and the consequences of its lower intake during pregnancy and postnatally on the 2019 brain development and function.
Collapse
|
18
|
Easton ZJW, Regnault TRH. The Impact of Maternal Body Composition and Dietary Fat Consumption upon Placental Lipid Processing and Offspring Metabolic Health. Nutrients 2020; 12:nu12103031. [PMID: 33022934 PMCID: PMC7601624 DOI: 10.3390/nu12103031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022] Open
Abstract
The proportion of women of reproductive age who are overweight or obese is increasing globally. Gestational obesity is strongly associated in both human studies and animal models with early-onset development of adult-associated metabolic diseases including metabolic syndrome in the exposed offspring. However, animal model studies have suggested that gestational diet in obese pregnancies is an independent but underappreciated mediator of offspring risk for later life metabolic disease, and human diet consumption data have highlighted that many women do not follow nutritional guidelines prior to and during pregnancy. Thus, this review will highlight how maternal diet independent from maternal body composition impacts the risk for later-life metabolic disease in obesity-exposed offspring. A poor maternal diet, in combination with the obese metabolic state, are understood to facilitate pathological in utero programming, specifically through changes in lipid handling processes in the villous trophoblast layer of the placenta that promote an environment associated with the development of metabolic disease in the offspring. This review will additionally highlight how maternal obesity modulates villous trophoblast lipid processing functions including fatty acid transport, esterification and beta-oxidation. Further, this review will discuss how altering maternal gestational diet may ameliorate these functional changes in lipid metabolic processes in the obese placenta.
Collapse
Affiliation(s)
- Zachary J. W. Easton
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada;
- Correspondence: ; Tel.: +1-(519)-661-2111 (ext. 82869)
| | - Timothy R. H. Regnault
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada;
- Department of Obstetrics and Gynaecology, London Health Science Centre-Victoria Hospital, B2-401, London, ON N6H 5W9, Canada
- Children’s Health Research Institute, 800 Commissioners Road East, London, ON N6C 2V5, Canada
- Lawson Health Research Institute, 750 Base Line Rd E, London, ON N6C 2R5, Canada
| |
Collapse
|
19
|
Massari M, Novielli C, Mandò C, Di Francesco S, Della Porta M, Cazzola R, Panteghini M, Savasi V, Maggini S, Schaefer E, Cetin I. Multiple Micronutrients and Docosahexaenoic Acid Supplementation during Pregnancy: A Randomized Controlled Study. Nutrients 2020; 12:E2432. [PMID: 32823606 PMCID: PMC7468952 DOI: 10.3390/nu12082432] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022] Open
Abstract
Maternal dietary intake during pregnancy needs to meet increased nutritional demands to maintain metabolism and to support fetal development. Docosahexaenoic acid (DHA) is essential for fetal neuro-/visual development and in immunomodulation, accumulating rapidly within the developing brain and central nervous system. Levels available to the fetus are governed by the maternal diet. In this multicenter, parallel, randomized controlled trial, we evaluated once-daily supplementation with multiple micronutrients and DHA (i.e., multiple micronutrient supplementation, MMS) on maternal biomarkers and infant anthropometric parameters during the second and third trimesters of pregnancy compared with no supplementation. Primary efficacy endpoint: change in maternal red blood cell (RBC) DHA (wt% total fatty acids) during the study. Secondary variables: other biomarkers of fatty acid and oxidative status, vitamin D, and infant anthropometric parameters at delivery. Supplementation significantly increased RBC DHA levels, the omega-3 index, and vitamin D levels. Subscapular skinfold thickness was significantly greater with MMS in infants. Safety outcomes were comparable between groups. This first randomized controlled trial of supplementation with multiple micronutrients and DHA in pregnant women indicated that MMS significantly improved maternal DHA and vitamin D status in an industrialized setting-an important finding considering the essential roles of DHA and vitamin D.
Collapse
Affiliation(s)
- Maddalena Massari
- Department of Woman, Mother and Neonate, Buzzi Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy; (M.M.); (S.D.F.); (I.C.)
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy;
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Chiara Novielli
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Chiara Mandò
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Stefania Di Francesco
- Department of Woman, Mother and Neonate, Buzzi Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy; (M.M.); (S.D.F.); (I.C.)
| | - Matteo Della Porta
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Roberta Cazzola
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Mauro Panteghini
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Valeria Savasi
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy;
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | | | | | - Irene Cetin
- Department of Woman, Mother and Neonate, Buzzi Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy; (M.M.); (S.D.F.); (I.C.)
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy;
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| |
Collapse
|
20
|
Roque-Jimenez JA, Oviedo-Ojeda MF, Whalin M, Lee-Rangel HA, Relling AE. Eicosapentaenoic and docosahexaenoic acid supplementation during early gestation modified relative abundance on placenta and fetal liver tissue mRNA and concentration pattern of fatty acids in fetal liver and fetal central nervous system of sheep. PLoS One 2020; 15:e0235217. [PMID: 32574225 PMCID: PMC7310831 DOI: 10.1371/journal.pone.0235217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/10/2020] [Indexed: 12/21/2022] Open
Abstract
In sheep, polyunsaturated fatty acid (PUFA) supplementations in late gestation increases the growth of offspring; however, there is a lack of evidence on the effect of PUFA supplementation during early gestation. Thus, the objective of this study was to evaluate the effect of dietary supplementation of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in early gestation pregnant ewes on fatty acid concentration of fetal liver (FL) and fetal central nervous system (FCNS), and relative abundance of the mRNA for genes associated with transport and metabolism of fatty acids in FL and placenta. A total of 12 ewes, block for stage of gestation were fed a diet containing 1.6% (dry matter basis) monounsaturated fatty acids (MUFA) or EPA+DHA during the first 45 days of gestation. A cesarean section was conducted on day 45 of gestation to collect placenta (caruncle and cotyledon), FL, and FCNS. Relative abundance of mRNA in FL and FCNS and fatty acid concentration were analyzed using a 2x2 factorial arrangement of treatments considering fatty acid supplementation and tissue as the main factors. Concentrations of C18:1 isomers increase (P < 0.05) in FL and FCNS with MUFA supplementation; the FL and FCNS had a greater concentration of C20:3(n-6), C20:3(n-3), C22:1, C22:5 and C22:6 (P < 0.05) with EPA+DHA supplementation. In FL, the relative abundance of LPL mRNA was greater (P = 0.02) as a result of MUFA supplementation. In placenta, there was a FA x tissue interaction for relative abundance of DNMT3b and FFAR-4 mRNA (P < 0.05). Fetus from MUFA-supplemented dams had a greater relative abundance of FABP-4 mRNA (P < 0.05). Results indicate supplementation with EPA+DHA during early gestation increases the total EPA and DHA in FL. For the placenta, EPA+DHA supplementation led to an increase in the relative abundance of lipid mRNA for transport genes.
Collapse
Affiliation(s)
- José Alejandro Roque-Jimenez
- Universidad Autónoma de San Luis Potosí, Facultad de Agronomía, Soledad de Graciano Sánchez, San Luis Potosí, México
- Department of Animal Science, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH, United States of America
| | - Mario Francisco Oviedo-Ojeda
- Universidad Autónoma de San Luis Potosí, Facultad de Agronomía, Soledad de Graciano Sánchez, San Luis Potosí, México
- Department of Animal Science, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH, United States of America
| | - Megan Whalin
- Department of Animal Science, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH, United States of America
| | - Héctor Aaron Lee-Rangel
- Universidad Autónoma de San Luis Potosí, Facultad de Agronomía, Soledad de Graciano Sánchez, San Luis Potosí, México
| | - Alejandro Enrique Relling
- Department of Animal Science, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH, United States of America
- * E-mail:
| |
Collapse
|
21
|
Rani A, Chavan-Gautam P, Mehendale S, Wagh G, Mani NS, Joshi S. Region-specific changes in the mRNA and protein expression of LCPUFA biosynthesis enzymes and transporters in the placentae of women with preeclampsia. Placenta 2020; 95:33-43. [PMID: 32452400 DOI: 10.1016/j.placenta.2020.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022]
Abstract
The biosynthesis and transport of long chain polyunsaturated fatty acids (LCPUFA) require the activity of fatty acid desaturase (FADS) enzymes, fatty acid transport proteins (FATP) and fatty acid binding proteins (FABP). In a previous study we have demonstrated region-specific changes in the LCPUFA levels in preeclampsia (PE) as compared to the normotensive control (NC) placentae. AIM To understand the region-specific changes in the mRNA levels and protein expression of biosynthesis enzymes and transporters of LCPUFA in PE and NC placentae. METHODS In this cross-sectional study, 20 NC women and 44 women with PE (23 term (TPE) and 21 preterm PE (PTPE)) were recruited. The samples were collected from four regions of the placentae considering cord insertion as the center (CM, central maternal/basal; CF, central fetal/chorionic; PM, peripheral maternal/basal and PF, peripheral fetal/chorionic). The mRNA levels were estimated using qRT-PCR. Statistical analysis was done using both post hoc least significant difference (LSD) test and Benjamini Hochberg correction in the analysis of covariance. Preliminarily, localization and expression of proteins were studied by immunohistochemistry (n = 3/group). RESULTS The mRNA levels of FADS1, FADS2 and FATP1 were lower in the central regions (CM and CF) of the PE placentae (both TPE and PTPE) as compared to NC. These differences in the mRNA levels were observed by the LSD test and were not significant after the Benjamini Hochberg correction. Preliminary findings of IHC indicate that the protein expression of FADS1 and FATP4 was higher in the basal regions (CM and PM) of the PE placentae as compared to NC. FADS1, FADS2 and FATP4 proteins were localized in the syncytiotrophoblasts, cytotrophoblasts, mesenchymal cells, endothelial cells of the fetal capillaries and extravillous trophoblasts of the placenta. CONCLUSION FADS enzymes are detected in the placentae of Indian women. In PE placentae, there are region-specific alterations in the mRNA and protein levels of LCPUFA biosynthesis enzymes (FADS1 and FADS2) and transporters (FATP1, FATP4 and FABP3) as compared to term NC. These changes were more pronounced toward the basal side and region around the cord insertion.
Collapse
Affiliation(s)
- Alka Rani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Preeti Chavan-Gautam
- Interdisciplinary School of Health Science, Savitribai Phule Pune University, Pune, India
| | - Savita Mehendale
- Department of Obstetrics and Gynaecology, Bharati Vidyapeeth (Deemed to be University) Medical College and Bharati Hospital, Pune, India
| | - Girija Wagh
- Department of Obstetrics and Gynaecology, Bharati Vidyapeeth (Deemed to be University) Medical College and Bharati Hospital, Pune, India
| | | | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India.
| |
Collapse
|
22
|
James-Allan LB, Teal S, Powell TL, Jansson T. Changes in Placental Nutrient Transporter Protein Expression and Activity Across Gestation in Normal and Obese Women. Reprod Sci 2020; 27:1758-1769. [PMID: 32072607 DOI: 10.1007/s43032-020-00173-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 02/03/2020] [Indexed: 11/26/2022]
Abstract
Fetal growth and development are dependent on placental nutrient transport. The syncytiotrophoblast (ST) and its two polarized plasma membranes, the maternal-facing microvillous membrane (MVM) and fetal-facing basal membrane (BM), represent the primary barrier in the human placenta, controlling transplacental transfer of small solutes. MVM and BM nutrient transporter expression and activity are increased in obese mothers delivering large babies. However, placental nutrient transporter expression and activity in early gestation in normal and obese women are largely unknown. Placentas from normal BMI and obese women at 6-24 weeks of gestation, and term placentas from normal BMI women, were collected and ST plasma membranes isolated. The activity and protein expression of amino acid, glucose, and fatty acid transporters was assessed. No significant differences were observed in placental nutrient transporter protein expression between normal BMI and obese women in early pregnancy. In the MVM, system A amino acid activity (p = 0.02), SNAT2 (p < 0.0001), SNAT4 (p < 0.001), and GLUT1 (p = 0.01) protein expression were higher at term compared with early gestation. In contrast, MVM system L activity (p = 0.001), FATP4 (p = 0.03), and FATP6 (p = 0.009) protein expression were lower at term compared with early pregnancy. In the BM, there was no change in system L activity across gestation; however, BM FATP6 (p = 0.002) protein expression was lower at term compared with early pregnancy. These results suggest that placental transport of amino acids, glucose, and fatty acids are subjected to coordinated regulation across gestation to meet a fetal nutrient demand that changes with advancing pregnancy.
Collapse
Affiliation(s)
- Laura B James-Allan
- Department of Obstetrics/Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Stephanie Teal
- Department of Obstetrics/Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa L Powell
- Department of Obstetrics/Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics/Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
23
|
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
|
24
|
Yakah W, Singh P, Perides G, Brown J, Freedman SD, Martin CR. Developmental Accretion of Docosahexaenoic Acid Is Independent of Fatty Acid Transporter Expression in Brain and Lung Tissues of C57BL/6 and Fat1 Mice. J Nutr 2019; 149:1724-1731. [PMID: 31179494 PMCID: PMC6768804 DOI: 10.1093/jn/nxz074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/21/2018] [Accepted: 03/26/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Developmental expression of fatty acid transporters and their role in polyunsaturated fatty acid concentrations in the postnatal period have not been evaluated. OBJECTIVE We hypothesized that transporter expression is developmentally regulated, tissue-specific, and that expression can modulate fatty acid accretion independently of diet. METHODS Brain and lung transporter expression were quantified in C57BL/6 wild-type (WT) and Fat1 mice. Pups were dam-fed until day 21. Dams were fed AIN-76A 10% corn oil to represent a typical North American/European diet. After weaning, mice were fed the same diet as dams. Gene expression of Fatp1, Fatp4, Fabp5, and Fat/cd36 was quantified by quantitative reverse transcriptase-polymerase chain reaction. Fatty acid concentrations were measured by GC-MS. RESULTS Brain docosahexaenoic acid (DHA) concentrations increased from day 3 to day 28 in both genotypes, with higher concentrations at days 3 and 14 in Fat1 than in WT mice [median (IQR)]: 10.7 (10.6-11.2) mol% compared with 6.6 (6.4-7.2) mol% and 12.5 (12.4-12.9) mol% compared with 8.9 (8.7-9.1) mol%, respectively; P < 0.05). During DHA accrual, transporter expression decreased. Fold changes in brain Fatp4, Fabp5, and Fat/cd36 were inversely correlated with fold changes in brain DHA concentrations in Fat1 relative to WT mice (ρ = -0.85, -0.75, and -0.78, respectively; P ≤ 0.001). Lung DHA concentrations were unchanged across the 3 time points for both genotypes. Despite unchanging DHA concentrations, there was increased expression of Fatp1 at days 14 and 28 (5-fold), Fatp4 at day 14 (2.3-fold), and Fabp5 at day 14 (3.8-fold) relative to day 3 in Fat1 mice. In WT mice, Fatp1 increased almost 5-fold at day 28 relative to day 3. There was no correlation between lung transporters and DHA concentrations in Fat1 relative to WT mice. CONCLUSIONS Development of fatty acid transporter expression in C57BL/6 WT and Fat1 mice is genotype and tissue specific. Further, postnatal accretion of brain DHA appears independent of transporter status, with tissue concentrations representing dietary contributions.
Collapse
Affiliation(s)
- William Yakah
- Department of Neuroscience, Michigan State University, East Lansing, MI
| | | | | | | | - Steven D Freedman
- Division of Gastroenterology, Boston, MA,Division of Translational Research, Boston, MA
| | - Camilia R Martin
- Division of Translational Research, Boston, MA,Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA,Address correspondence to CRM (e-mail: )
| |
Collapse
|
25
|
Ao Z, Wu X, Zhou J, Gu T, Wang X, Shi J, Zhao C, Cai G, Zheng E, Liu D, Wu Z, Li Z. Cloned pig fetuses exhibit fatty acid deficiency from impaired placental transport. Mol Reprod Dev 2019; 86:1569-1581. [PMID: 31347235 DOI: 10.1002/mrd.23242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/08/2019] [Indexed: 01/23/2023]
Abstract
Cloned pig fetuses produced by somatic cell nuclear transfer show a high incidence of erroneous development in the uteri of surrogate mothers. The mechanisms underlying the abnormal intrauterine development of cloned pig fetuses are poorly understood. This study aimed to explore the potential causes of the aberrant development of cloned pig fetuses. The levels of numerous fatty acids in allantoic fluid and muscle tissue were lower in cloned pig fetuses than in artificial insemination-generated pig fetuses, thereby suggesting that cloned pig fetuses underwent fatty acid deficiency. Cloned pig fetuses also displayed trophoblast hypoplasia and a reduced expression of placental fatty acid transport protein 4 (FATP4), which is the predominant FATP family member expressed in porcine placentas. This result suggested that the placental fatty acid transport functions were impaired in cloned pig fetuses, possibly causing fatty acid deficiency in cloned pig fetuses. The present study provides useful information in elucidating the mechanisms underlying the abnormal development of cloned pig fetuses.
Collapse
Affiliation(s)
- Zheng Ao
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiao Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jun Zhou
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ting Gu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xingwang Wang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Junsong Shi
- Guangdong Wens Pig Breeding Technology Co. Ltd., Wens Foodstuff Group Co. Ltd., Yunfu, Guangdong, China
| | - Chengfa Zhao
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Gengyuan Cai
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Enqin Zheng
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Dewu Liu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Zhu Y, Tan Q, Zhang L, Yao J, Zhou H, Hu P, Liang X, Liu H. The migration of docosahexenoic acid (DHA) to the developing ovary of female zebrafish (Danio rerio). Comp Biochem Physiol A Mol Integr Physiol 2019; 233:97-105. [PMID: 30978471 DOI: 10.1016/j.cbpa.2019.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
Abstract
Fish selectively reserves docosahexenoic acid (DHA) in ovary during gonadal development. However, no direct proof supports this. The present study tried to elucidate the DHA migration to the developing ovary of female zebrafish. An injection study of 13C-labeled DHA for DHA tracing was conducted, and another injection study of unlabeled-DHA (DHA-injected group) and BSA-saline (control group) was conducted for lipid and DHA content detection, related gene expression analyses, and histological observation. The results showed that the rapid absorption of lipid occurred at stage III with a constant accumulation of DHA in the ovary. The proportion of oocytes at stage III on day 7 and 21, and at stage IV on day 3 and 21 in DHA-injected group was significantly higher than that in control group, respectively (P < .05). The injected 13C-labeled DHA was accumulated twice in the ovary respectively on day 1 and 7, and remained at a relatively high level. In DHA-injected group, the fatp4 expression was significantly higher in ovary on day 3, 5 and 7 (P < .05), and significantly lower (P < .05) in liver on day 5, 14 and in muscle on day 1, 5 and 7 than that in control group. In conclusion, the present study suggested a migration of DHA from the liver and muscle to the gonads when necessary.
Collapse
Affiliation(s)
- Yanhong Zhu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingsong Tan
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Leisen Zhang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Junpeng Yao
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Hai Zhou
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Pengcheng Hu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xufang Liang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Hong Liu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
27
|
Ferchaud-Roucher V, Kramer A, Silva E, Pantham P, Weintraub ST, Jansson T, Powell TL. A potential role for lysophosphatidylcholine in the delivery of long chain polyunsaturated fatty acids to the fetal circulation. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:394-402. [PMID: 30572119 DOI: 10.1016/j.bbalip.2018.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/10/2018] [Accepted: 12/15/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Véronique Ferchaud-Roucher
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Anita Kramer
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elena Silva
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Priyadarshini Pantham
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, IL, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center San Antonio, TX, USA
| | - Thomas Jansson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa L Powell
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
28
|
Neuroprotective and Antiaging Essential Oils and Lipids in Plants. BIOACTIVE MOLECULES IN FOOD 2019. [DOI: 10.1007/978-3-319-78030-6_89] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
29
|
Middleton P, Gomersall JC, Gould JF, Shepherd E, Olsen SF, Makrides M. Omega-3 fatty acid addition during pregnancy. Cochrane Database Syst Rev 2018; 11:CD003402. [PMID: 30480773 PMCID: PMC6516961 DOI: 10.1002/14651858.cd003402.pub3] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Higher intakes of foods containing omega-3 long-chain polyunsaturated fatty acids (LCPUFA), such as fish, during pregnancy have been associated with longer gestations and improved perinatal outcomes. This is an update of a review that was first published in 2006. OBJECTIVES To assess the effects of omega-3 LCPUFA, as supplements or as dietary additions, during pregnancy on maternal, perinatal, and neonatal outcomes and longer-term outcomes for mother and child. SEARCH METHODS For this update, we searched Cochrane Pregnancy and Childbirth's Trials Register, ClinicalTrials.gov, the WHO International Clinical Trials Registry Platform (ICTRP) (16 August 2018), and reference lists of retrieved studies. SELECTION CRITERIA Randomised controlled trials (RCTs) comparing omega-3 fatty acids (as supplements or as foods, stand-alone interventions, or with a co-intervention) during pregnancy with placebo or no omega-3, and studies or study arms directly comparing omega-3 LCPUFA doses or types. Trials published in abstract form were eligible for inclusion. DATA COLLECTION AND ANALYSIS Two review authors independently assessed study eligibility, extracted data, assessed risk of bias in trials and assessed quality of evidence for prespecified birth/infant, maternal, child/adult and health service outcomes using the GRADE approach. MAIN RESULTS In this update, we included 70 RCTs (involving 19,927 women at low, mixed or high risk of poor pregnancy outcomes) which compared omega-3 LCPUFA interventions (supplements and food) compared with placebo or no omega-3. Overall study-level risk of bias was mixed, with selection and performance bias mostly at low risk, but there was high risk of attrition bias in some trials. Most trials were conducted in upper-middle or high-income countries; and nearly half the trials included women at increased/high risk for factors which might increase the risk of adverse maternal and birth outcomes.Preterm birth < 37 weeks (13.4% versus 11.9%; risk ratio (RR) 0.89, 95% confidence interval (CI) 0.81 to 0.97; 26 RCTs, 10,304 participants; high-quality evidence) and early preterm birth < 34 weeks (4.6% versus 2.7%; RR 0.58, 95% CI 0.44 to 0.77; 9 RCTs, 5204 participants; high-quality evidence) were both lower in women who received omega-3 LCPUFA compared with no omega-3. Prolonged gestation > 42 weeks was probably increased from 1.6% to 2.6% in women who received omega-3 LCPUFA compared with no omega-3 (RR 1.61 95% CI 1.11 to 2.33; 5141 participants; 6 RCTs; moderate-quality evidence).For infants, there was a possibly reduced risk of perinatal death (RR 0.75, 95% CI 0.54 to 1.03; 10 RCTs, 7416 participants; moderate-quality evidence: 62/3715 versus 83/3701 infants) and possibly fewer neonatal care admissions (RR 0.92, 95% CI 0.83 to 1.03; 9 RCTs, 6920 participants; moderate-quality evidence - 483/3475 infants versus 519/3445 infants). There was a reduced risk of low birthweight (LBW) babies (15.6% versus 14%; RR 0.90, 95% CI 0.82 to 0.99; 15 trials, 8449 participants; high-quality evidence); but a possible small increase in large-for-gestational age (LGA) babies (RR 1.15, 95% CI 0.97 to 1.36; 6 RCTs, 3722 participants; moderate-quality evidence, for omega-3 LCPUFA compared with no omega-3. Little or no difference in small-for-gestational age or intrauterine growth restriction (RR 1.01, 95% CI 0.90 to 1.13; 8 RCTs, 6907 participants; moderate-quality evidence) was seen.For the maternal outcomes, there is insufficient evidence to determine the effects of omega-3 on induction post-term (average RR 0.82, 95% CI 0.22 to 2.98; 3 trials, 2900 participants; low-quality evidence), maternal serious adverse events (RR 1.04, 95% CI 0.40 to 2.72; 2 trials, 2690 participants; low-quality evidence), maternal admission to intensive care (RR 0.56, 95% CI 0.12 to 2.63; 2 trials, 2458 participants; low-quality evidence), or postnatal depression (average RR 0.99, 95% CI 0.56 to 1.77; 2 trials, 2431 participants; low-quality evidence). Mean gestational length was greater in women who received omega-3 LCPUFA (mean difference (MD) 1.67 days, 95% CI 0.95 to 2.39; 41 trials, 12,517 participants; moderate-quality evidence), and pre-eclampsia may possibly be reduced with omega-3 LCPUFA (RR 0.84, 95% CI 0.69 to 1.01; 20 trials, 8306 participants; low-quality evidence).For the child/adult outcomes, very few differences between antenatal omega-3 LCPUFA supplementation and no omega-3 were observed in cognition, IQ, vision, other neurodevelopment and growth outcomes, language and behaviour (mostly low-quality to very low-quality evidence). The effect of omega-3 LCPUFA on body mass index at 19 years (MD 0, 95% CI -0.83 to 0.83; 1 trial, 243 participants; very low-quality evidence) was uncertain. No data were reported for development of diabetes in the children of study participants. AUTHORS' CONCLUSIONS In the overall analysis, preterm birth < 37 weeks and early preterm birth < 34 weeks were reduced in women receiving omega-3 LCPUFA compared with no omega-3. There was a possibly reduced risk of perinatal death and of neonatal care admission, a reduced risk of LBW babies; and possibly a small increased risk of LGA babies with omega-3 LCPUFA.For our GRADE quality assessments, we assessed most of the important perinatal outcomes as high-quality (e.g. preterm birth) or moderate-quality evidence (e.g. perinatal death). For the other outcome domains (maternal, child/adult and health service outcomes) GRADE ratings ranged from moderate to very low, with over half rated as low. Reasons for downgrading across the domain were mostly due to design limitations and imprecision.Omega-3 LCPUFA supplementation during pregnancy is an effective strategy for reducing the incidence of preterm birth, although it probably increases the incidence of post-term pregnancies. More studies comparing omega-3 LCPUFA and placebo (to establish causality in relation to preterm birth) are not needed at this stage. A further 23 ongoing trials are still to report on over 5000 women, so no more RCTs are needed that compare omega-3 LCPUFA against placebo or no intervention. However, further follow-up of completed trials is needed to assess longer-term outcomes for mother and child, to improve understanding of metabolic, growth and neurodevelopment pathways in particular, and to establish if, and how, outcomes vary by different types of omega-3 LCPUFA, timing and doses; or by characteristics of women.
Collapse
Affiliation(s)
- Philippa Middleton
- Healthy Mothers, Babies and Children, South Australian Health and Medical Research InstituteWomen's and Children's Hospital72 King William RoadAdelaideSouth AustraliaAustralia5006
| | - Judith C Gomersall
- Healthy Mothers, Babies and Children, South Australian Health and Medical Research InstituteWomen's and Children's Hospital72 King William RoadAdelaideSouth AustraliaAustralia5006
| | - Jacqueline F Gould
- The University of AdelaideSchool of PsychologyNorth Terrace, AdelaideAdelaideSouth AustraliaAustralia5001
| | - Emily Shepherd
- The University of AdelaideARCH: Australian Research Centre for Health of Women and Babies, Robinson Research Institute, Discipline of Obstetrics and GynaecologyAdelaideSouth AustraliaAustralia5006
| | - Sjurdur F Olsen
- Statens Serum InstitutCentre for Fetal Programming, Department of EpidemiologyCopenhagenDenmark
| | - Maria Makrides
- Healthy Mothers, Babies and Children, South Australian Health and Medical Research InstituteWomen's and Children's Hospital72 King William RoadAdelaideSouth AustraliaAustralia5006
- The University of AdelaideSchool of Paediatrics and Reproductive HealthAdelaideAustraliaAustralia
- Women's and Children's Health Research InstituteNorth AdelaideAustralia
| | | |
Collapse
|
30
|
Kallol S, Moser-Haessig R, Ontsouka CE, Albrecht C. Comparative expression patterns of selected membrane transporters in differentiated BeWo and human primary trophoblast cells. Placenta 2018; 72-73:48-52. [PMID: 30501881 DOI: 10.1016/j.placenta.2018.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/25/2018] [Accepted: 10/27/2018] [Indexed: 02/07/2023]
Abstract
Although placental membrane transporters have an important impact on materno-fetal nutrient transfer, placental cell models are poorly characterized regarding transporter expression. We assessed the mRNA expression of 26 physiologically important solute carriers and ABC transporters in BeWo (b30 clone) and primary human trophoblast cells (PHT) before and after syncytialization. 77% of the transporters showed similar mRNA expression changes between BeWo and PHT after syncytialization. Selected transporters, however, were either lacking in BeWo or showed different trends after syncytialization. In conclusion, BeWo cells generally represent an apt model for transporter studies, but their suitability should be confirmed for each transporter by comparison with PHT.
Collapse
Affiliation(s)
- Sampada Kallol
- Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Switzerland
| | | | - Corneille Edgar Ontsouka
- Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Switzerland.
| |
Collapse
|
31
|
Maternal n-3 PUFA supplementation promotes fetal brown adipose tissue development through epigenetic modifications in C57BL/6 mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1488-1497. [PMID: 30266429 DOI: 10.1016/j.bbalip.2018.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/24/2018] [Accepted: 09/23/2018] [Indexed: 12/11/2022]
Abstract
Brown adipose tissue (BAT) is a crucial regulator of energy expenditure. Emerging evidence suggests that n-3 PUFA potentiate brown adipogenesis in vitro. Since the pregnancy and lactation is a critical time for brown fat formation, we hypothesized that maternal supplementation of n-3 PUFA promotes BAT development in offspring. Female C57BL/6 mice were fed a diet containing n-3 PUFA (3%) derived from fish oil (FO), or an isocaloric diet devoid of n-3 PUFA (Cont) during pregnancy and lactation. Maternal n-3 PUFA intake was delivered to the BAT of neonates significantly reducing the n-6/n-3 ratio. The maternal n-3 PUFA exposure was linked with upregulated brown-specific gene and protein profiles and the functional cluster of brown-specific miRNAs. In addition, maternal n-3 PUFA induced histone modifications in the BAT evidenced by 1) increased epigenetic signature of brown adipogenesis, i.e., H3K27Ac and H3K9me2, 2) modified chromatin-remodeling enzymes, and 3) enriched the H3K27Ac in the promoter region of Ucp1. The offspring received maternal n-3 PUFA nutrition exhibited a significant increase in whole-body energy expenditure and better maintenance of core body temperature against acute cold treatment. Collectively, our results suggest that maternal n-3 PUFA supplementation potentiates fetal BAT development via the synergistic action of miRNA production and histone modifications, which may confer long-lasting metabolic benefits to offspring.
Collapse
|
32
|
Zhang W, Chen R, Yang T, Xu N, Chen J, Gao Y, Stetler RA. Fatty acid transporting proteins: Roles in brain development, aging, and stroke. Prostaglandins Leukot Essent Fatty Acids 2018; 136:35-45. [PMID: 28457600 PMCID: PMC5650946 DOI: 10.1016/j.plefa.2017.04.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 04/16/2017] [Accepted: 04/20/2017] [Indexed: 12/18/2022]
Abstract
Polyunsaturated fatty acids are required for the brain development and significantly impact aging and stroke. Due to the hydrophobicity of fatty acids, fatty acids transportation related proteins that include fatty acid binding proteins (FABPs), long chain acyl-coA synthase (ACS), fatty acid transportation proteins (FATPs), fatty acid translocase (FAT/CD36) and newly reported major facilitator superfamily domain-containing protein (Mfsd2a) play critical roles in the uptake of various fatty acids, especially polyunsaturated fatty acids. They are not only involved in neurodevelopment, but also have great impact on neurological disease, such as aging related dementia and stroke.
Collapse
Affiliation(s)
- Wenting Zhang
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Ruiying Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Na Xu
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - R Anne Stetler
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA.
| |
Collapse
|
33
|
Tachikawa M, Akanuma SI, Imai T, Okayasu S, Tomohiro T, Hatanaka Y, Hosoya KI. Multiple Cellular Transport and Binding Processes of Unesterified Docosahexaenoic Acid in Outer Blood–Retinal Barrier Retinal Pigment Epithelial Cells. Biol Pharm Bull 2018; 41:1384-1392. [DOI: 10.1248/bpb.b18-00185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Shin-ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Tsubasa Imai
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Shun Okayasu
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Takenori Tomohiro
- Laboratory of Biorecognition Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Yasumaru Hatanaka
- Laboratory of Biorecognition Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Ken-ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| |
Collapse
|
34
|
Andraos S, de Seymour JV, O'Sullivan JM, Kussmann M. The Impact of Nutritional Interventions in Pregnant Women on DNA Methylation Patterns of the Offspring: A Systematic Review. Mol Nutr Food Res 2018; 62:e1800034. [PMID: 30035846 DOI: 10.1002/mnfr.201800034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/12/2018] [Indexed: 12/14/2022]
Abstract
Epidemiological studies have consistently demonstrated that environmental exposures in early life are associated with later-life health status and disease susceptibility. Epigenetic modifications, such as DNA methylation, have been suggested as potential mechanisms linking the intrauterine environment with offspring health status. The present systematic review compiles peer-reviewed randomized controlled trials assessing the impact of maternal nutritional interventions on DNA methylation patterns of the offspring. The results of the included trials are consistent with micronutrient supplementation not significantly affecting offspring tissue DNA methylation patterns, yet subgrouping by sex, BMI, and smoking status increased the significance of nutritional supplementation on DNA methylation. Maternal BMI and smoking status as well as offspring sex were factors influencing offspring DNA methylation responsiveness to nutritional interventions during pregnancy. Future research should aim at assessing the impact of nutritional interventions on DNA methylation patterns of neonates comparing single versus multi-micronutrient supplementation, within populations having high versus low baseline nutritional statuses.
Collapse
Affiliation(s)
- Stephanie Andraos
- The Liggins Institute, Faculty of Medical and Health Sciences, The University of Auckland, 1023, Auckland, New Zealand
| | - Jamie Violet de Seymour
- The Liggins Institute, Faculty of Medical and Health Sciences, The University of Auckland, 1023, Auckland, New Zealand
| | - Justin Martin O'Sullivan
- The Liggins Institute, Faculty of Medical and Health Sciences, The University of Auckland, 1023, Auckland, New Zealand
| | - Martin Kussmann
- The Liggins Institute, Faculty of Medical and Health Sciences, The University of Auckland, 1023, Auckland, New Zealand.,New Zealand National Science Challenge, High-Value Nutrition, The University of Auckland, 1023, Auckland, New Zealand
| |
Collapse
|
35
|
Differential Long-Chain Polyunsaturated Fatty Acids Status and Placental Transport in Adolescent Pregnancies. Nutrients 2018; 10:nu10020220. [PMID: 29462922 PMCID: PMC5852796 DOI: 10.3390/nu10020220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/22/2018] [Accepted: 02/12/2018] [Indexed: 12/11/2022] Open
Abstract
Adolescent pregnancy increases risk of adverse perinatal outcomes. Placental delivery of long-chain polyunsaturated fatty acids (LCPUFA) is essential for fetal growth and development. In this pilot study, we aimed to assess maternal and fetal status of fatty acids (FA) measured at birth and the expression of key genes involved in FA uptake, transport and metabolism in the placenta of fifteen adolescents and fifteen adults. FA were quantified by gas-liquid chromatography. Placental expression of FA transporters was assessed by quantitative real-time polymerase chain reaction (qRT-PCR) and peroxisome proliferator-activated receptor gamma (PPARγ) was quantified by Western Blot. Adolescents had lower docosahexaenoic acid (DHA, 22:6 n-3) and total n-3 FA levels in maternal erythrocytes and placenta, but these were not different in fetal erythrocytes. Arachidonic acid (AA, 20:4 n-6) concentration was increased in placenta but lower in fetal circulation. Plasma membrane fatty acid binding protein (FABPpm) and fatty acid transport protein (FATP) 4 mRNA expressions were not different, however FATP1, fatty acid translocase (FAT/CD36) and fatty acid binding protein 3 (FABP3) mRNA and PPARγ protein levels were decreased in placenta of adolescents. Despite significant downregulation of FATP1, CD36 and FABP3, there was only a modest decrease in LCPUFA (10%) and AA (12%) and no difference in DHA content in cord blood, suggesting that FA transfer to the fetus was partially protected by other factors in adolescents from this cohort.
Collapse
|
36
|
Das M, Pandima Devi K. Neuroprotective and Antiaging Essential Oils and Lipids in Plants. REFERENCE SERIES IN PHYTOCHEMISTRY 2018. [DOI: 10.1007/978-3-319-54528-8_89-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
37
|
Ostadrahimi A, Mohammad-Alizadeh S, Mirghafourvand M, Farshbaf-Khalili S, Jafarilar-Agdam N, Farshbaf-Khalili A. The effect of fish oil supplementation on maternal and neonatal outcomes: a triple-blind, randomized controlled trial. J Perinat Med 2017; 45:1069-1077. [PMID: 28141545 DOI: 10.1515/jpm-2016-0037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 12/09/2016] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To evaluate the effect of fish oil supplementation on pregnancy outcomes in mother and newborn. METHODS This randomized, triple-blind, placebo-controlled trial was conducted on 150 pregnant women aged 18-35 years from Feburary 2014 to April 2015 in Tabriz, Iran. Participants were assigned to receive either 1000 mg fish oil supplements containing 180 mg eicosapentaenoic acid (EPA) and 120 mg docosahexanoic acid or placebo from week 20 of gestation to birth. The primary outcome measure was birth weight. Gestational duration, preterm labor low birth weight (LBW), length, head circumference, and maternal serum docosahexaenoic acid (DHA) and EPA level at 35-37 weeks were also examined. The statistical analysis was on an intention-to-treat basis. RESULTS Demographic characteristics were similar in both groups (P>0.05). The mean (SD) birth weight values in the fish oil and placebo groups were 3256 (362) g and 3172 (447) g, respectively (adjusted mean difference (MD)=84.1 g, 95% confidence interval [CI]=-24.8 to 193.2). Five (7.6%) neonates in the placebo versus no case in the fish oil group were born with LBW (P=0.02). The rate of preterm labor was lower in the fish oil group (adjusted OR=0.74, 95% CI=0.16-3.42). However, there were no statistically significant differences in the maternal outcomes (P>0.05) with the exception of the proportion of maternal serum DHA fatty acid at 35-37 weeks (P<0.001). CONCLUSION Although low dose fish oil supplementation increased birth weight, its effect was not statistically significant. The frequency of LBW was significantly reduced in the intervention group, but the observed reduction needs to be confirmed in future larger investigations using different doses of omega-3.
Collapse
|
38
|
Lager S, Ramirez VI, Acosta O, Meireles C, Miller E, Gaccioli F, Rosario FJ, Gelfond JAL, Hakala K, Weintraub ST, Krummel DA, Powell TL. Docosahexaenoic Acid Supplementation in Pregnancy Modulates Placental Cellular Signaling and Nutrient Transport Capacity in Obese Women. J Clin Endocrinol Metab 2017; 102:4557-4567. [PMID: 29053802 PMCID: PMC5718695 DOI: 10.1210/jc.2017-01384] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 10/13/2017] [Indexed: 01/08/2023]
Abstract
Context Maternal obesity in pregnancy has profound impacts on maternal metabolism and promotes placental nutrient transport, which may contribute to fetal overgrowth in these pregnancies. The fatty acid docosahexaenoic acid (DHA) has bioactive properties that may improve outcomes in obese pregnant women by modulating placental function. Objective To determine the effects of DHA supplementation in obese pregnant women on maternal metabolism and placental function. Design Pregnant women were supplemented with DHA or placebo. Maternal fasting blood was collected at 26 and 36 weeks' gestation, and placentas were collected at term. Setting Academic health care institution. Subjects Thirty-eight pregnant women with pregravid body mass index ≥30 kg/m2. Intervention DHA (800 mg, algal oil) or placebo (corn/soy oil) daily from 26 weeks to term. Main Outcomes DHA content of maternal erythrocyte and placental membranes, maternal fasting blood glucose, cytokines, metabolic hormones, and circulating lipids were determined. Insulin, mTOR, and inflammatory signaling were assessed in placental homogenates, and nutrient transport capacity was determined in isolated syncytiotrophoblast plasma membranes. Results DHA supplementation increased erythrocyte (P < 0.0001) and placental membrane DHA levels (P < 0.0001) but did not influence maternal inflammatory status, insulin sensitivity, or lipids. DHA supplementation decreased placental inflammation, amino acid transporter expression, and activity (P < 0.01) and increased placental protein expression of fatty acid transporting protein 4 (P < 0.05). Conclusions Maternal DHA supplementation in pregnancy decreases placental inflammation and differentially modulates placental nutrient transport capacity and may mitigate adverse effects of maternal obesity on placental function.
Collapse
Affiliation(s)
- Susanne Lager
- Department of Obstetrics and Gynaecology, University of Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, United Kingdom
| | - Vanessa I Ramirez
- Department of Obstetrics and Gynecology, University of Texas Health Science Center
| | - Ometeotl Acosta
- Department of Obstetrics and Gynecology, University of Texas Health Science Center
| | - Christiane Meireles
- Department of Obstetrics and Gynecology, University of Texas Health Science Center
| | - Evelyn Miller
- Department of Obstetrics and Gynecology, University of Texas Health Science Center
| | - Francesca Gaccioli
- Department of Obstetrics and Gynaecology, University of Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, United Kingdom
| | - Fredrick J Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus
| | - Jonathan A L Gelfond
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center
| | - Kevin Hakala
- Department of Biochemistry, University of Texas Health Science Center
| | - Susan T Weintraub
- Department of Biochemistry, University of Texas Health Science Center
| | | | - Theresa L Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus
- Department of Pediatrics, University of Colorado Anschutz Medical Campus
| |
Collapse
|
39
|
Büyükuslu N, Ovalı S, Altuntaş ŞL, Batırel S, Yiğit P, Garipağaoğlu M. Supplementation of docosahexaenoic acid (DHA) / Eicosapentaenoic acid (EPA) in a ratio of 1/1.3 during the last trimester of pregnancy results in EPA accumulation in cord blood. Prostaglandins Leukot Essent Fatty Acids 2017; 125:32-36. [PMID: 28987719 DOI: 10.1016/j.plefa.2017.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 07/21/2017] [Accepted: 08/21/2017] [Indexed: 01/18/2023]
Abstract
Omega-3 fatty acids (n-3 FA), specifically DHA, are associated with fetal growth and development. We aimed to determine the levels of DHA and EPA in cord serum after n-3 FA supplementation during the last trimester of pregnancy. Among 55 women, 23 were administered daily one capsule of n-3 FA supplement, involving DHA/EPA in a ratio of 1/1.3. Twenty nine women were enrolled as control group. Blood samples were collected at 22-24 weeks of gestation and at delivery. Fatty acids were analyzed with the method of GC-MS. Cord DHA level increased and EPA level decreased in both groups between the days of 22-24 and delivery. However, decrease in cord EPA level was significant in control group (p < 0.001) but not in supplement group (p > 0.05). Supplementation of DHA/EPA in a ratio of 1/1.3 during the last trimester of pregnancy caused higher cord EPA level compared to control group indicating an accumulation in umbilical cord.
Collapse
Affiliation(s)
- Nihal Büyükuslu
- Department of Nutrition and Dietetics, School of Health Sciences, Istanbul Medipol University, Kavacık Mah. Ekinciler Cad. No.19 Kavacık Kavşağı-Beykoz, 34810 Istanbul, Turkey.
| | - Sema Ovalı
- Medipol Koşuyolu Hospital, Obstetrics and Gynecology Clinic, Istanbul, Turkey
| | | | - Saime Batırel
- Department of Medical Biochemistry, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Pakize Yiğit
- Department of Statistics and Medical Informatics, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Muazzez Garipağaoğlu
- Department of Nutrition and Dietetics, School of Health Sciences, Istanbul Medipol University, Kavacık Mah. Ekinciler Cad. No.19 Kavacık Kavşağı-Beykoz, 34810 Istanbul, Turkey
| |
Collapse
|
40
|
Echeverría F, Valenzuela R, Catalina Hernandez-Rodas M, Valenzuela A. Docosahexaenoic acid (DHA), a fundamental fatty acid for the brain: New dietary sources. Prostaglandins Leukot Essent Fatty Acids 2017; 124:1-10. [PMID: 28870371 DOI: 10.1016/j.plefa.2017.08.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/10/2017] [Accepted: 08/09/2017] [Indexed: 01/25/2023]
Abstract
Docosahexaenoic acid (C22: 6n-3, DHA) is a long-chain polyunsaturated fatty acid of marine origin fundamental for the formation and function of the nervous system, particularly the brain and the retina of humans. It has been proposed a remarkable role of DHA during human evolution, mainly on the growth and development of the brain. Currently, DHA is considered a critical nutrient during pregnancy and breastfeeding due their active participation in the development of the nervous system in early life. DHA and specifically one of its derivatives known as neuroprotectin D-1 (NPD-1), has neuroprotective properties against brain aging, neurodegenerative diseases and injury caused after brain ischemia-reperfusion episodes. This paper discusses the importance of DHA in the human brain given its relevance in the development of the tissue and as neuroprotective agent. It is also included a critical view about the ways to supply this noble fatty acid to the population.
Collapse
Affiliation(s)
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | | | - Alfonso Valenzuela
- Lipid Center, Institute of Nutrition and Food Technology (INTA), University of Chile and Faculty of Medicine,, University de Los Andes, Santiago, Chile
| |
Collapse
|
41
|
Metherel AH, Kitson AP, Domenichiello AF, Lacombe RJS, Hopperton KE, Trépanier MO, Alashmali SM, Lin L, Bazinet RP. Docosahexaenoic acid (DHA) accretion in the placenta but not the fetus is matched by plasma unesterified DHA uptake rates in pregnant Long Evans rats. Placenta 2017; 58:90-97. [PMID: 28962703 DOI: 10.1016/j.placenta.2017.08.072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/31/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022]
Abstract
Maternal delivery of docosahexaenoic acid (DHA, 22:6n-3) to the developing fetus via the placenta is required for fetal neurodevelopment, and is the only mechanism by which DHA can be accreted in the fetus. The aim of the current study was to utilize a balance model of DHA accretion combined with kinetic measures of serum unesterified DHA uptake to better understand the mechanism by which maternal DHA is delivered to the fetus via the placenta. Female rats maintained on a 2% α-linolenic acid diet free of DHA for 56 days were mated, and for balance analysis, sacrificed at 18 days of pregnancy, and fetus, placenta and maternal carcass fatty acid concentration were determined. For tissue DHA uptake, pregnant dams (14-18 days) were infused for 5 min with radiolabeled 14C-DHA and kinetic modeling was used to determine fetal and placental serum unesterified DHA uptake rates. DHA accretion rates in the fetus were determined to be 38 ± 2 nmol/d/g, 859 ± 100 nmol/d/litter and 74 ± 3 nmol/d/pup, which are all higher (P < 0.05) than the fetal serum unesterified DHA uptake rates of 16 ± 6 nmol/d/g, 239 ± 145 nmol/d/litter and 14 ± 8 nmol/d/pup. No differences (p > 0.05) in placental DHA accretion rates versus serum unesterified DHA uptake rates were observed as values varied only 6-35% between studies. No differences in placental accretion and uptake rates suggests that serum unesterified DHA is a significant pool for the maternal-placental transfer of DHA, and lower fetal DHA uptake compared to accretion supports remodeling of placental DHA for delivery to the fetus.
Collapse
Affiliation(s)
- Adam H Metherel
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada.
| | - Alex P Kitson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| | - Anthony F Domenichiello
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| | - R J Scott Lacombe
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| | - Kathryn E Hopperton
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| | - Marc-Olivier Trépanier
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| | - Shoug M Alashmali
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| | - Lin Lin
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College St., Toronto, Ontario M5S 3E2, Canada
| |
Collapse
|
42
|
Dubé C, Aguer C, Adamo K, Bainbridge S. A role for maternally derived myokines to optimize placental function and fetal growth across gestation. Appl Physiol Nutr Metab 2017; 42:459-469. [DOI: 10.1139/apnm-2016-0446] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Exercise during pregnancy is associated with improved health outcomes for both mother and baby, including a reduced risk of future obesity and susceptibility to chronic diseases. Overwhelming evidence demonstrates a protective effect of maternal exercise against fetal birth weight extremes, reducing the rates of both large- and small-for-gestational-age infants. It is speculated that this protective effect is mediated in part through exercise-induced regulation of maternal physiology and placental development and function. However, the specific mechanisms through which maternal exercise regulates these changes remain to be discovered. We hypothesize that myokines, a collection of peptides and cytokines secreted from contracting skeletal muscles during exercise, may be an important missing link in the story. Myokines are known to reduce inflammation, improve metabolism and enhance macronutrient transporter expression and activity in various tissues of nonpregnant individuals. Little research to date has focused on the specific roles of the myokine secretome in the context of pregnancy; however, it is likely that myokines secreted from exercising skeletal muscles may modulate the maternal milieu and directly impact the vital organ of pregnancy—the placenta. In the current review, data in strong support of this potential role of myokines will be presented, suggesting myokine secretion as a key mechanism through which maternal exercise optimizes fetal growth trajectories. It is clear that further research is warranted in this area, as knowledge of the biological roles of myokines in the context of pregnancy would better inform clinical recommendations for exercise during pregnancy and contribute to the development of important therapeutic interventions.
Collapse
Affiliation(s)
- Chantal Dubé
- Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Céline Aguer
- Institut de recherche de l’Hôpital Montfort, Ottawa, ON K1K 0T1, Canada
- Biochemistry, Microbiology and Immunology department, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Kristi Adamo
- School of Human Kinetics, University of Ottawa, Ottawa, ON K1N 1A2, Canada
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Shannon Bainbridge
- Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
43
|
Neonatal fatty acid profiles are correlated with infant growth measures at 6 months. J Dev Orig Health Dis 2017; 8:474-482. [PMID: 28300011 DOI: 10.1017/s2040174417000150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Rapid weight gain in infancy and low levels of n-3 long chain polyunsaturated fatty acids (LCPUFA) at birth are associated with increased adiposity later in life. The association between placental LCPUFA delivery and weight gain in infancy is poorly understood. We sought to determine the relationships between maternal phenotype, placental fatty acid transporter expression and offspring growth patterns over the first 6 months. Placental tissue and cord blood were collected at term delivery from women with uncomplicated pregnancies. Offspring body composition measurements were recorded 1 day and 6 months after birth. Body mass index (BMI) z-scores were determined using World Health Organization 2006 reference data. Body phenotype patterns were compared among offspring who had an increase in BMI z-score and those who had a decrease. High skinfold thickness at birth and positive change in BMI z-scores during infancy were associated with low neonatal n-3 LCPUFA plasma levels (r=-0.46, P=0.046) and high saturated fatty acids levels (r=0.49, P=0.034). Growth of skinfolds over 6 months of age was associated with placental fatty acid transporter gene expression. Change in BMI z-score in the first 6 months of life correlated with arm muscle area growth, a measure of lean mass (r=0.62, P=0.003), but not with growth in skinfold thickness. Early infancy weight gain was associated with poor plasma LCPUFA status at birth, and fat deposition in infancy was related to changes in placental lipid handling. Thus, neonatal fatty acid profiles may influence the trajectory of infant growth and fat and lean mass deposition.
Collapse
|
44
|
Salehi R, Ambrose DJ. Prepartum maternal diets supplemented with oilseeds alter the fatty acid profile in bovine neonatal plasma possibly through reduced placental expression of fatty acid transporter protein 4 and fatty acid translocase. Reprod Fertil Dev 2017; 29:1846-1855. [DOI: 10.1071/rd15476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 10/06/2016] [Indexed: 11/23/2022] Open
Abstract
In the present study, we determined the effects of maternal dietary fat and the type of fat on plasma fatty acids and the expression of placental fatty acid transporter genes. In Experiment 1, Holstein cows in the last 35 days of gestation received diets containing sunflower seed (n = 8; high in linoleic acid (LA)), canola seed (n = 7; high in oleic acid (OLA)) or no oilseed (n = 7; control). Fatty acids were quantified in dam and neonate plasma at calving. In Experiment 2, placental cotyledons were collected (LA: n = 4; OLA: n = 4; control: n = 5) to quantify gene expression. Maternal long-chain polyunsaturated fatty acids, neonatal total n-3 fatty acids and eicosapentaenoic acid (EPA) declined, whereas docosahexaenoic acid (DHA) and total fat tended to decline following fat supplementation prepartum. Feeding of LA versus OLA prepartum tended to increase peroxisome proliferator-activated receptor α (PPARA) expression, whereas peroxisome proliferator-activated receptor δ (PPARD) and peroxisome proliferator-activated receptor γ (PPARG) expression tended to be higher in OLA- than LA-fed cows. Expression of fatty acid transporter protein 4 (FATP4) and fatty acid translocase (FAT/CD36) expression was lower in placental tissue of cows fed fat compared with control cows. Reduced total n-3 fatty acids, EPA and DHA in neonates born of dams fed fat prepartum is likely due to changes in PPARs and reduced expression of placental FATP4 and FAT/CD36.
Collapse
|
45
|
Garcia-Rodriguez CE, Olza J, Mesa MD, Aguilera CM, Miles EA, Noakes PS, Vlachava M, Kremmyda LS, Diaper ND, Godfrey KM, Calder PC, Gil A. Fatty acid status and antioxidant defense system in mothers and their newborns after salmon intake during late pregnancy. Nutrition 2017; 33:157-162. [DOI: 10.1016/j.nut.2016.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/18/2016] [Accepted: 05/25/2016] [Indexed: 11/24/2022]
|
46
|
Léveillé P, Ardilouze JL, Pasquier JC, Deacon C, Whittingstall K, Plourde M. Fatty acid profile in cord blood of neonates born to optimally controlled gestational diabetes mellitus. Prostaglandins Leukot Essent Fatty Acids 2016; 115:48-52. [PMID: 27914513 DOI: 10.1016/j.plefa.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate the fatty acid profile of cord blood phospholipids (PL), cholesteryl esters (CE), triglycerides (TG) and non-esterified fatty acids (NEFA) in neonates born to mothers with gestational diabetes mellitus (GDM) compared to non-diabetic mothers. METHODS The offspring of 30 pregnant women (15 non-diabetic controls, 15 with diet- or insulin-controlled GDM) were recruited before delivery. Cord blood was collected. After lipid extraction, PL, CE, TG and NEFA were separated by thin layer chromatography and analysed by gas chromatography. RESULTS In GDM vs. control mothers, maternal glycated haemoglobin (A1C, mean±SD) was not different between groups: 5.3±0.5% vs. 5.3±0.3% (p=0.757), respectively. Cord plasma fatty acids were not different in TG, CE and NEFA between GDM and non-diabetic mothers. However, in PL, levels of palmitate, palmitoleate, oleate, vaccinate and di-homo-gamma-linolenate were significantly lower, with a trend for lower arachidonate (p=0.078), in neonates born to GDM mothers compared to controls. CONCLUSION In contrast to other studies on cord blood docosahexaenoic acid (DHA) levels in GDM mothers, we did not found lower levels of DHA in cord PL, CE, TG or NEFA in neonates born to GDM compared to non-diabetic mothers.
Collapse
Affiliation(s)
- Pauline Léveillé
- Research Center on Aging, Health and Social Services Centre - University Institute of Geriatrics of Sherbrooke, 1036 Belvédère Sud Sherbrooke, Québec, Canada J1H 4C4; Department of Physiology, Université de Sherbrooke, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4
| | - Jean-Luc Ardilouze
- Department of Physiology, Université de Sherbrooke, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4; Department of Medicine, Université de Sherbrooke, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4; Centre de recherche du CHUS, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4
| | - Jean-Charles Pasquier
- Department of Medicine, Université de Sherbrooke, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4; Centre de recherche du CHUS, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4
| | - Charles Deacon
- Department of Medicine, Université de Sherbrooke, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4
| | - Kevin Whittingstall
- Department of Medicine, Université de Sherbrooke, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4
| | - Mélanie Plourde
- Research Center on Aging, Health and Social Services Centre - University Institute of Geriatrics of Sherbrooke, 1036 Belvédère Sud Sherbrooke, Québec, Canada J1H 4C4; Department of Medicine, Université de Sherbrooke, 3001, 12e avenue Nord Sherbrooke, Québec, Canada J1H 5N4.
| |
Collapse
|
47
|
Leghi GE, Muhlhausler BS. The effect of n-3 LCPUFA supplementation on oxidative stress and inflammation in the placenta and maternal plasma during pregnancy. Prostaglandins Leukot Essent Fatty Acids 2016; 113:33-39. [PMID: 27720038 DOI: 10.1016/j.plefa.2016.08.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 12/22/2022]
Abstract
Pregnancy represents a state of heightened oxidative stress and inflammation, and these processes are further increased in pregnancy complications. The quality of the maternal diet is directly associated with maternal health and wellbeing, pregnancy and fetal outcomes, as well as the risk of pregnancy complications. Long chain polyunsaturated fatty acids (LCPUFAs) have significant potential to modify placental and fetal lipid environments and thereby modulate health outcomes. The omega-3 (n-3) LCPUFA in particular have been shown to exhibit both antioxidant and anti-inflammatory properties, and have potential therapeutic applications in reducing oxidative damage and inflammation during pregnancy. The purpose of this review is to provide an overview of our current understanding of the impact of maternal n-3 LCPUFA supplementation on oxidative stress and inflammation during pregnancy, with a particular focus on effects on the mother and the placenta.
Collapse
Affiliation(s)
- G E Leghi
- FOODplus Research Centre, School of Agriculture, Food and Wine, The University of Adelaide, Waite Campus, Urrbrae, South Australia 5064, Australia
| | - B S Muhlhausler
- FOODplus Research Centre, School of Agriculture, Food and Wine, The University of Adelaide, Waite Campus, Urrbrae, South Australia 5064, Australia; Women's and Children's Health Research Institute, Women's and Children's Hospital, Adelaide, Australia.
| |
Collapse
|
48
|
Berti C, Cetin I, Agostoni C, Desoye G, Devlieger R, Emmett PM, Ensenauer R, Hauner H, Herrera E, Hoesli I, Krauss-Etschmann S, Olsen SF, Schaefer-Graf U, Schiessl B, Symonds ME, Koletzko B. Pregnancy and Infants' Outcome: Nutritional and Metabolic Implications. Crit Rev Food Sci Nutr 2016; 56:82-91. [PMID: 24628089 DOI: 10.1080/10408398.2012.745477] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pregnancy is a complex period of human growth, development, and imprinting. Nutrition and metabolism play a crucial role for the health and well-being of both mother and fetus, as well as for the long-term health of the offspring. Nevertheless, several biological and physiological mechanisms related to nutritive requirements together with their transfer and utilization across the placenta are still poorly understood. In February 2009, the Child Health Foundation invited leading experts of this field to a workshop to critically review and discuss current knowledge, with the aim to highlight priorities for future research. This paper summarizes our main conclusions with regards to maternal preconceptional body mass index, gestational weight gain, placental and fetal requirements in relation to adverse pregnancy and long-term outcomes of the fetus (nutritional programming). We conclude that there is an urgent need to develop further human investigations aimed at better understanding of the basis of biochemical mechanisms and pathophysiological events related to maternal-fetal nutrition and offspring health. An improved knowledge would help to optimize nutritional recommendations for pregnancy.
Collapse
Affiliation(s)
- C Berti
- a Unit of Obstetrics & Gynecology, Department of Biomedical and Clinical Sciences, Hospital 'L. Sacco', and Center for Fetal Research Giorgio Pardi , University of Milan , Milan , Italy
| | - I Cetin
- a Unit of Obstetrics & Gynecology, Department of Biomedical and Clinical Sciences, Hospital 'L. Sacco', and Center for Fetal Research Giorgio Pardi , University of Milan , Milan , Italy.,b Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - C Agostoni
- b Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - G Desoye
- c Department of Obstetrics & Gynaecology , Medical University of Graz , Graz , Austria
| | - R Devlieger
- d Department of Obstetrics & Gynaecology , University Hospitals K.U. Leuven , Leuven , Belgium
| | - P M Emmett
- e Centre for Child & Adolescent Health, School of Social & Community Medicine , University of Bristol , Bristol , United Kingdom
| | - R Ensenauer
- f Research Center, Dr. von Hauner Children's Hospital, Klinikum der Ludwig-Maximilians-Universität München , Munich , Germany
| | - H Hauner
- g Else Kroener-Fresenius-Centre for Nutritional Medicine, Klinikum rechts der Isar , Technical University of Munich , Munich , Germany
| | - E Herrera
- h Unit of Biochemistry & Molecular Biology, Universidad San Pablo CEU , Madrid , Spain
| | - I Hoesli
- i Department of Obstetrics and Gynaecology , University Hospital of Basel , Basel , Switzerland
| | - S Krauss-Etschmann
- j Comprehensive Pneumology Center, Ludwig Maximilians University Hospital , Munich , Germany.,k Helmholtz Zentrum München , Munich , Germany
| | - S F Olsen
- l Centre for Fetal Programming, Statens Serum Institut , Copenhagen , Denmark
| | - U Schaefer-Graf
- m Department of Obstetrics & Gynecology , Berlin Center for Diabetes & Pregnancy, St. Joseph Hospital , Berlin , Germany
| | - B Schiessl
- n Fetal Maternal Unit, Department of Obstetrics & Gynecology , University of Munich , Munich , Germany
| | - M E Symonds
- o The Early Life Nutrition Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital of Nottingham , Nottingham , United Kingdom
| | - B Koletzko
- p Dr. von Hauner Children's Hospital , University of Munich Medical Centre , Munich , Germany
| |
Collapse
|
49
|
The effect of gestational age on expression of genes involved in uptake, trafficking and synthesis of fatty acids in the rat placenta. Gene 2016; 591:403-10. [PMID: 27317891 DOI: 10.1016/j.gene.2016.06.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/11/2016] [Accepted: 06/14/2016] [Indexed: 12/30/2022]
Abstract
Gestation triggers a tight coordination among maternal tissues to provide fatty acids (FA) to the fetus through placental transport; however, there is insufficient evidence regarding regulation of proteins involved in placental transport of FA according to gestational age. The aim of this study was to determine the role of gestational age on the expression of genes involved in FA uptake, trafficking and synthesis in the rat placenta to support fetal demands. Gene expression of encoding proteins for placental transport and synthesis of FA was measured in placenta. Also, FA composition was measured in placenta, fetuses and newborns. mRNA expression of lipoprotein lipase (lpl) and fatp-1 (for uptake) was 4.4- and 1.43-fold higher, respectively, during late gestation than at P14, but expression of p-fabp-pm decreased 0.37-fold at late pregnancy in comparison with P14. Only mRNA fabp-4 member for trafficking of FA was 2.95-fold higher at late gestation than at P14. mRNA of fasn and elovl-6 participating in saturated FA and enzymes for the polyunsaturated FA synthesis were downregulated during late gestation and their regulator srebf-1c increased at P16. This study suggests that gestational age has an effect on expression of some genes involved in uptake, trafficking and synthesis of FA in the rat placenta; mRNA expression of lpl and, fatp-1 for uptake and fabp-4 implicated in trafficking was expressed at high levels at late gestation. In addition, placenta expresses the mRNAs involved in FA synthesis; these genes were expressed at low levels at late gestation. Additionally, mRNAs of Srebf-1c transcriptional regulator of desaturases and elongases was highly expressed during late gestation. Finally, these changes in the rat placenta allowed the placenta to partially supply saturated and monounsaturated FA to the fetus.
Collapse
|
50
|
Rani A, Wadhwani N, Chavan-Gautam P, Joshi S. Altered development and function of the placental regions in preeclampsia and its association with long-chain polyunsaturated fatty acids. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:582-97. [DOI: 10.1002/wdev.238] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/18/2016] [Accepted: 03/29/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Alka Rani
- Department of Nutritional Medicine; Interactive Research School for Health Affairs, Bharati Vidyapeeth University; Pune India
| | - Nisha Wadhwani
- Department of Nutritional Medicine; Interactive Research School for Health Affairs, Bharati Vidyapeeth University; Pune India
| | - Preeti Chavan-Gautam
- Department of Nutritional Medicine; Interactive Research School for Health Affairs, Bharati Vidyapeeth University; Pune India
| | - Sadhana Joshi
- Department of Nutritional Medicine; Interactive Research School for Health Affairs, Bharati Vidyapeeth University; Pune India
| |
Collapse
|