1
|
Luo X, Wang H, Yin B, Huang B, Cao J, Qi H. β'-COP mediated loading of PPARγ into trophoblast-derived extracellular vesicles. Cell Mol Life Sci 2024; 81:464. [PMID: 39601826 PMCID: PMC11602898 DOI: 10.1007/s00018-024-05494-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/20/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024]
Abstract
Fetal growth restriction (FGR) is characterized by impaired fetal growth and dysregulated lipid metabolism. Extracellular vesicles (EVs) have been proved playing a crucial role in transporting biomolecules from the mother to the fetus. However, the mechanisms underlying cargo sorting and loading into trophoblastic EVs remain elusive. This study focuses on examining how the essential fatty acid regulator, peroxisome proliferator-activated receptor gamma (PPARγ), is sorted and loaded into EVs originating from trophoblasts. We conducted proteomic analysis on placenta-derived EVs from normal and FGR pregnancies. Interactions between PPARγ and coat protein complex I (COPI) subunit were evaluated using co-immunoprecipitation and bioinformatics simulation. Molecular dynamics simulations were conducted to identify critical binding sites between β'-coat protein complex I (β'-COP), a subunit of COPI, and PPARγ. lentivirus-mediated knockout and overexpression techniques were employed to elucidate the role of β'-COP in PPARγ loading into EVs. Our findings demonstrate that PPARγ protein levels are significantly decreased in EVs from FGR placentas. β'-COP subunit directly interacts with PPARγ in trophoblasts, mediating its sorting into early endosomes and multivesicular bodies for EVs incorporation. Knockout of β'-COP impaired PPARγ loading into EVs. Molecular dynamics simulations identified critical binding sites for the interaction between β'-COP and PPARγ. Mutation of these sites significantly weakened the β'-COP-PPARγ interaction and reduced PPARγ levels in trophoblastic EVs. In conclusion, β'-COP mediates sorting and loading of PPARγ into trophoblastic EVs. This study provides insights into regulating EVs cargo loading and potential strategies for targeted cargo delivery from the maternal to the fetal circulation.
Collapse
Affiliation(s)
- Xiaofang Luo
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
- Chongqing Municipal Health Commission Key Laboratory of Perinatal Medicine, Chongqing, 400016, China.
| | - Hao Wang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, China.
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, 401147, China.
- Chongqing Municipal Health Commission Key Laboratory of Perinatal Medicine, Chongqing, 400016, China.
| | - Biyang Yin
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, 401147, China
- Chongqing Municipal Health Commission Key Laboratory of Perinatal Medicine, Chongqing, 400016, China
| | - Biao Huang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, 401147, China
- Chongqing Municipal Health Commission Key Laboratory of Perinatal Medicine, Chongqing, 400016, China
| | - Jinfeng Cao
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- Chongqing Municipal Health Commission Key Laboratory of Perinatal Medicine, Chongqing, 400016, China
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, China.
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, 401147, China.
- Chongqing Municipal Health Commission Key Laboratory of Perinatal Medicine, Chongqing, 400016, China.
| |
Collapse
|
2
|
Ozawa R, Iwata H, Kuwayama T, Shirasuna K. Maternal hypertensive condition alters adipose tissue function and blood pressure sensitivity in offspring. Biochem Biophys Res Commun 2024; 707:149617. [PMID: 38520942 DOI: 10.1016/j.bbrc.2024.149617] [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: 01/24/2024] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 03/25/2024]
Abstract
Preeclampsia (PE) is characterized by hypertension, proteinuria, and fetal growth restriction during pregnancy, suggesting that the preeclamptic intrauterine environment may affect the growth and health of the offspring. This study aimed to how maternal hypertension affects male offspring growth, focusing on lipid metabolism and blood pressure in mice. Female mice were infused with angiotensin II (Ang II) on gestational day 12. Dysregulation and accumulation of lipid were observed in the placenta of Ang II-induced maternal hypertensive dams, associating with fetal growth restriction. Ang II-offspring showed lower birth weight than in the control-offspring. Isolated and differentiated adipocyte from neonatal mice of Ang II-dams showed higher Pparγ mRNA expression compared with the control group. Lower body weight tendency had continued in Ang II-offspring during long period, body weight of Ang II-offspring caught up the control-offspring at 16 weeks of age. The adipose tissue of Ang II-offspring in adult also showed higher Pparγ mRNA expression with the accumulation of neutrophils and inflammatory monocytes than in those control. In addition, Ang II-offspring had higher basal blood pressure and higher sensitivity to hypertensive stimuli than in the control-offspring. Taken together, maternal hypertension induced by Ang II changes placental function, causing a lower birth weight. These changes in the intrauterine environment may affect adipocyte function and blood pressure of offspring after growth.
Collapse
Affiliation(s)
- Ren Ozawa
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 234-0034, Japan
| | - Hisataka Iwata
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 234-0034, Japan
| | - Takehito Kuwayama
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 234-0034, Japan
| | - Koumei Shirasuna
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 234-0034, Japan.
| |
Collapse
|
3
|
Matsumoto A, Kawabata T, Kagawa Y, Shoji K, Kimura F, Miyazawa T, Tatsuta N, Arima T, Yaegashi N, Nakai K. Association of maternal blood and umbilical cord blood plasma fatty acid levels with the body size at birth of Japanese infants. Prostaglandins Leukot Essent Fatty Acids 2024; 202:102638. [PMID: 39216412 DOI: 10.1016/j.plefa.2024.102638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), mainly obtained from fish, have been implicated in fetal development. Because few studies have examined maternal and umbilical cord blood fatty acid levels and infant body size in Japan with a fish-eating culture, we examined differences in plasma fatty acid levels in pregnant women and infant size at birth. This study is a large birth cohort study of 1476 pairs of Japanese pregnant women and their infants. Maternal blood DHA levels and infant birth weight showed a positive relationship. However, analysis adjusted for gestational age did not reveal correlations. Negative relationships were found between cord blood DHA levels and infant body size, and between the difference in mother-to-child DHA levels and infant body size. Thus, the smaller the birth size, the higher the differences in umbilical cord blood DHA levels and mother-to-child DHA levels when considering gestational age.
Collapse
Affiliation(s)
- Azusa Matsumoto
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado, Saitama 350-0288, Japan; Department of Life and Culture, Faculty of Creative Life, Kawamura Gakuen Woman's University, 1133 Sageto, Abiko, Chiba 270-1138, Japan.
| | - Terue Kawabata
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado, Saitama 350-0288, Japan
| | - Yasuo Kagawa
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado, Saitama 350-0288, Japan
| | - Kumiko Shoji
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado, Saitama 350-0288, Japan
| | - Fumiko Kimura
- Faculty of Comprehensive Human Sciences, Shokei Gakuin University, 4-10-1 Yurigaoka, Natori, Miyagi 981-1295, Japan; Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Sciences, Tohoku University, 468-1 Aza-Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-0845, Japan
| | - Teruo Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, 6-6-10 Aza-Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | - Nozomi Tatsuta
- National Institute for Environmental Studies, Health and Environmental Risk Division, Environmental Epidemiology Section, 16-2 Onogawa, Tsukuba, Ibaraki, 305-8506, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Nobuo Yaegashi
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kunihiko Nakai
- School of Sport and Health Science, Tokai Gakuen University, Nishinohora 21-233, Miyoshi, Aichi 470-0207, Japan
| |
Collapse
|
4
|
Bolluk G, Oğlak SC, Kayaoğlu Yıldırım Z, Zengi O. Maternal serum fatty acid binding protein-4 level is upregulated in fetal growth restriction with abnormal Doppler flow patterns. J Obstet Gynaecol Res 2024; 50:430-437. [PMID: 38148278 DOI: 10.1111/jog.15868] [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: 08/09/2023] [Accepted: 12/11/2023] [Indexed: 12/28/2023]
Abstract
PURPOSE This study aimed to determine fatty acid binding protein-4 (FABP-4) concentrations in maternal serum of fetal growth restriction (FGR) pregnancies and controls of normal pregnancies. Furthermore, we hypothesized that the alterations in FABP-4 levels might correlate with FGR severity. METHODS We performed this prospective case-control study with 83 pregnant women. The study groups included 26 FGR pregnancies without abnormal fetal Doppler flow patterns and 25 pregnancies complicated with FGR accompanied by abnormal fetal Doppler flow patterns. RESULTS The median serum FABP-4 concentrations were significantly higher in the FGR cases with abnormal Doppler flow pattern group (2.09 ng/mL) than in the FGR cases without abnormal Doppler flow pattern group (1.62 ng/mL) and the control group (1.20 ng/mL, p < 0.001). A significant negative correlation was observed between maternal serum FABP-4 levels and time to birth from blood sample collection (r = -0.356 and p = 0.001), gestational week at birth (r = -0.386 and p < 0.001), and birth weight (r = -0.394 and p < 0.001). A 1.35 ng/mL cut-off value of serum FABP-4 level could be used to discriminate FGR cases with a 78.4% sensitivity and 60.6% specificity. The optimal cut-off value of FABP-4 levels as an indicator for the diagnosis of FGR with abnormal Doppler flow pattern was estimated to be 1.76 ng/mL, which yielded a sensitivity of 84.0% and a specificity of 75.8%. CONCLUSION FABP-4 is a crucial biomarker in the diagnosis and determining the severity of pregnancies with restricted fetal growth. We consider that FABP-4 is a powerful, reliable, and unique biomarker to diagnose FGR pregnancies.
Collapse
Affiliation(s)
- Gökhan Bolluk
- Department of Perinatology, Health Sciences University, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
| | - Süleyman Cemil Oğlak
- Department of Obstetrics and Gynecology, Health Sciences University, Gazi Yaşargil Training and Research Hospital, Diyarbakır, Turkey
| | - Zeynep Kayaoğlu Yıldırım
- Department of Perinatology, Health Sciences University, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
| | - Oğuzhan Zengi
- Department of Biochemistry, Health Sciences University, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Kramer AC, Jansson T, Bale TL, Powell TL. Maternal-fetal cross-talk via the placenta: influence on offspring development and metabolism. Development 2023; 150:dev202088. [PMID: 37831056 PMCID: PMC10617615 DOI: 10.1242/dev.202088] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Compelling epidemiological and animal experimental data demonstrate that cardiometabolic and neuropsychiatric diseases originate in a suboptimal intrauterine environment. Here, we review evidence suggesting that altered placental function may, at least in part, mediate the link between the maternal environment and changes in fetal growth and development. Emerging evidence indicates that the placenta controls the development and function of several fetal tissues through nutrient sensing, modulation of trophoblast nutrient transporters and by altering the number and cargo of released extracellular vesicles. In this Review, we discuss the development and functions of the maternal-placental-fetal interface (in humans and mice) and how cross-talk between these compartments may be a mechanism for in utero programming, focusing on mechanistic target of rapamycin (mTOR), adiponectin and O-GlcNac transferase (OGT) signaling. We also discuss how maternal diet and stress influences fetal development and metabolism and how fetal growth restriction can result in susceptibility to developing chronic disease later in life. Finally, we speculate how interventions targeting placental function may offer unprecedented opportunities to prevent cardiometabolic disease in future generations.
Collapse
Affiliation(s)
- Avery C. Kramer
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Thomas Jansson
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Tracy L. Bale
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Theresa L. Powell
- Departments of Obstetrics & Gynecology, Psychiatry and Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Lermant A, Rabussier G, Lanz HL, Davidson L, Porter IM, Murdoch CE. Development of a human iPSC-derived placental barrier-on-chip model. iScience 2023; 26:107240. [PMID: 37534160 PMCID: PMC10392097 DOI: 10.1016/j.isci.2023.107240] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Although recently developed placenta-on-chip systems opened promising perspectives for placental barrier modeling, they still lack physiologically relevant trophoblasts and are poorly amenable to high-throughput studies. We aimed to implement human-induced pluripotent stem cells (hiPSC)-derived trophoblasts into a multi-well microfluidic device to develop a physiologically relevant and scalable placental barrier model. When cultured in a perfused micro-channel against a collagen-based matrix, hiPSC-derived trophoblasts self-arranged into a 3D structure showing invasive behavior, fusogenic and endocrine activities, structural integrity, and expressing placental transporters. RNA-seq analysis revealed that the microfluidic 3D environment boosted expression of genes related to early placental structural development, mainly involved in mechanosensing and cell surface receptor signaling. These results demonstrated the feasibility of generating a differentiated primitive syncytium from hiPSC in a microfluidic platform. Besides expanding hiPSC-derived trophoblast scope of applications, this study constitutes an important resource to improve placental barrier models and boost research and therapeutics evaluation in pregnancy.
Collapse
Affiliation(s)
- Agathe Lermant
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | | | | | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Iain M. Porter
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, DD1 5EH, UK
| | - Colin E. Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
7
|
Yang Z, Luo X, Huang B, Jia X, Luan X, Shan N, An Z, Cao J, Qi H. Altered distribution of fatty acid exerting lipid metabolism and transport at the maternal-fetal interface in fetal growth restriction. Placenta 2023; 139:159-171. [PMID: 37406553 DOI: 10.1016/j.placenta.2023.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/17/2023] [Accepted: 05/31/2023] [Indexed: 07/07/2023]
Abstract
INTRODUCTION Fetal growth restriction (FGR) is a common complication of pregnancy. Lipid metabolism and distribution may contribute to the progression of FGR. However, the metabolism-related mechanisms of FGR remain unclear. The aim of this study was to identify metabolic profiles associated with FGR, as well as probable genes and signaling pathways. METHODS Metabolomic profiles at the maternal-fetal interface (including the placenta, maternal and fetal serum) from pregnant women with (n = 35) and without (n = 35) FGR were analyzed by gas chromatography-mass spectrometry (GC-MS). Combined with differentially expressed genes (DEGs) from the GSE35574 dataset, analysis was performed for differential metabolites, and identified by the Metabo Analyst dataset. Finally, the pathology and screened DEGs were further identified. RESULTS The results showed that fatty acids (FAs) accumulated in the placenta and decreased in fetal blood in FGR cases compared to controls. The linoleic acid metabolism was the focus of placental differential metabolites and genes enrichment analysis. In this pathway, phosphatidylcholine can interact with PLA2G2A and PLA2G4C, and 12(13)-EpOME can interact with CYP2J2. PLA2G2A and CYP2J2 were elevated, and PLA2G4C was decreased in the FGR placenta. DISCUSSION In conclusion, accumulation of FAs in the placental ischemic environments, may involve linoleic acid metabolism, which may be regulated by PLA2G2A, CYP2J2, and PLA2G4C. This study may contribute to understanding the underlying metabolic and molecular mechanisms of FGR.
Collapse
Affiliation(s)
- Zhongmei Yang
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; The Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| | - Xiaofang Luo
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China
| | - Biao Huang
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China
| | - Xiaoyan Jia
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China
| | - Xiaojin Luan
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China
| | - Nan Shan
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China
| | - Zhongling An
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China
| | - Jinfeng Cao
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China
| | - Hongbo Qi
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, China.
| |
Collapse
|
8
|
Guerrero-Santoro J, Morizane M, Oh SY, Mishima T, Goff JP, Bildirici I, Sadovsky E, Ouyang Y, Tyurin VA, Tyurina YY, Kagan VE, Sadovsky Y. The lipase cofactor CGI58 controls placental lipolysis. JCI Insight 2023; 8:168717. [PMID: 37212279 DOI: 10.1172/jci.insight.168717] [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: 01/10/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
In eutherians, the placenta plays a critical role in the uptake, storage, and metabolism of lipids. These processes govern the availability of fatty acids to the developing fetus, where inadequate supply has been associated with substandard fetal growth. Whereas lipid droplets are essential for the storage of neutral lipids in the placenta and many other tissues, the processes that regulate placental lipid droplet lipolysis remain largely unknown. To assess the role of triglyceride lipases and their cofactors in determining placental lipid droplet and lipid accumulation, we assessed the role of patatin like phospholipase domain containing 2 (PNPLA2) and comparative gene identification-58 (CGI58) in lipid droplet dynamics in the human and mouse placenta. While both proteins are expressed in the placenta, the absence of CGI58, not PNPLA2, markedly increased placental lipid and lipid droplet accumulation. These changes were reversed upon restoration of CGI58 levels selectively in the CGI58-deficient mouse placenta. Using co-immunoprecipitation, we found that, in addition to PNPLA2, PNPLA9 interacts with CGI58. PNPLA9 was dispensable for lipolysis in the mouse placenta yet contributed to lipolysis in human placental trophoblasts. Our findings establish a crucial role for CGI58 in placental lipid droplet dynamics and, by extension, in nutrient supply to the developing fetus.
Collapse
Affiliation(s)
- Jennifer Guerrero-Santoro
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Takuya Mishima
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie P Goff
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vladimir A Tyurin
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Department of Chemistry
- Department of Pharmacology and Chemical Biology
- Department of Radiation Oncology; and
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Wu Z, Hu G, Zhang Y, Ao Z. IGF2 May Enhance Placental Fatty Acid Metabolism by Regulating Expression of Fatty Acid Carriers in the Growth of Fetus and Placenta during Late Pregnancy in Pigs. Genes (Basel) 2023; 14:genes14040872. [PMID: 37107630 PMCID: PMC10137774 DOI: 10.3390/genes14040872] [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: 02/03/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Fatty acids (FAs) are essential substances for the growth and development of the fetus and placenta. The growing fetus and placenta must obtain adequate FAs received from the maternal circulation and facilitated by various placental FA carriers, including FA transport proteins (FATPs), FA translocase (FAT/CD36), and cytoplasmic FA binding proteins (FABPs). Placental nutrition transport was regulated by imprinted genes H19 and insulin-like growth factor 2 (IGF2). Nevertheless, the relationship between the expression patterns of H19/IGF2 and placental fatty acid metabolism throughout pig pregnancy remains poorly studied and unclear. We investigated the placental fatty acid profile, expression patterns of FA carriers, and H19/IGF2 in the placentae on Days 40 (D40), 65 (D65), and 95 (D95) of pregnancy. The results showed that the width of the placental folds and the number of trophoblast cells of D65 placentae were significantly increased than those of D40 placentae. Several important long-chain FAs (LCFAs), including oleic acid, linoleic acid, arachidonatic acid, eicosapentaenoic acid, and docosatetraenoic acid, in the pig placenta showed dramatically increased levels throughout pregnancy. The pig placenta possessed higher expression levels of CD36, FATP4, and FABP5 compared with other FA carriers, and their expression levels had significantly upregulated 2.8-, 5.6-, and 12.0-fold from D40 to D95, respectively. The transcription level of IGF2 was dramatically upregulated and there were corresponding lower DNA methylation levels in the IGF2 DMR2 in D95 placentae relative to D65 placentae. Moreover, in vitro experimentation revealed that the overexpression of IGF2 resulted in a significant increase in fatty acid uptake and expression levels of CD36, FATP4, and FABP5 in PTr2 cells. In conclusion, our results indicate that CD36, FATP4, and FABP5 may be important regulators that enhance the transport of LCFAs in the pig placenta and that IGF2 may be involved in FA metabolism by affecting the FA carriers expression to support the growth of the fetus and placenta during late pregnancy in pigs.
Collapse
Affiliation(s)
- Zhimin Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Guangling Hu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yiyu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zheng Ao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
10
|
Effects of maternal HF diet and absence of TRPC1 gene on mouse placental growth and fetal intrauterine growth retardation (IUGR). J Nutr Biochem 2023; 114:109162. [PMID: 36243380 DOI: 10.1016/j.jnutbio.2022.109162] [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: 06/08/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/06/2022]
Abstract
Placental tissue intracellular calcium (Ca2+) regulates placental development and growth. Maternal high-fat diet (HFD) results in placental lipid accumulation, increased inflammation, reduced nutrient transport expression, and intrauterine growth restriction (IUGR). Currently, whether maternal HFD differentially affects placental and fetal growth and development under reduced Ca2+ influx is not yet known. We hypothesized that maternal HFD feeding decreases placental growth and development resulting in IUGR and that reduction of Ca2+ influx in the placenta worsens maternal HFD-induced placental dysfunction and IUGR. Three-week-old female B6129SF2/J wild type (WT) and transient receptor potential canonical 1 (TRPC1) protein deficient (KO) mice were fed normal fat (NF, 16 kcal % fat) and high fat (HF, 45 kcal % fat) diets for 12 weeks prior to mating with NF diet fed male mice. Fetuses and placentae were examined at mid- (D12) and late- (D18) gestation. At D12, maternal HFD had no effects on placental or fetal weight changes in WT and TRPC1 KO mice while absence of TRPC1 resulted in decreased placental and fetal weights. At D18, maternal HFD increased placental weights in both TRPC1 KO and WT mice, in part, by moderately increasing placental tissue triacylglyceride (TAG, P=.0632). At D12, mRNA expression of key placental growth factors including IGF1, PLGF, and VEGF were increased in WT compared to TRPC1 KO mice while IGF2 and VEGF mRNA expression were increased at D18. Results presented in our study demonstrated that maternal HFD increased placental weight, in part, due to increased lipid concentration resulting in IUGR and via an additive adverse effect of genotype and maternal HFD. Future studies are needed to determine the signaling mechanism underlying Ca2+ influx reduction-induced placental dysfunction and IUGR.
Collapse
|
11
|
Tan S, Yang Y, Yang W, Han Y, Huang L, Yang R, Hu Z, Tao Y, Liu L, Li Y, Oyang L, Lin J, Peng Q, Jiang X, Xu X, Xia L, Peng M, Wu N, Tang Y, Cao D, Liao Q, Zhou Y. Exosomal cargos-mediated metabolic reprogramming in tumor microenvironment. J Exp Clin Cancer Res 2023; 42:59. [PMID: 36899389 PMCID: PMC9999652 DOI: 10.1186/s13046-023-02634-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of cancer. As nutrients are scarce in the tumor microenvironment (TME), tumor cells adopt multiple metabolic adaptations to meet their growth requirements. Metabolic reprogramming is not only present in tumor cells, but exosomal cargos mediates intercellular communication between tumor cells and non-tumor cells in the TME, inducing metabolic remodeling to create an outpost of microvascular enrichment and immune escape. Here, we highlight the composition and characteristics of TME, meanwhile summarize the components of exosomal cargos and their corresponding sorting mode. Functionally, these exosomal cargos-mediated metabolic reprogramming improves the "soil" for tumor growth and metastasis. Moreover, we discuss the abnormal tumor metabolism targeted by exosomal cargos and its potential antitumor therapy. In conclusion, this review updates the current role of exosomal cargos in TME metabolic reprogramming and enriches the future application scenarios of exosomes.
Collapse
Affiliation(s)
- Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yiqing Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Wenjuan Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Lisheng Huang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Ruiqian Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Zifan Hu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Yi Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Lin Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yun Li
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xuemeng Xu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Deliang Cao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
12
|
Altered Cord Blood Lipid Concentrations Correlate with Birth Weight and Doppler Velocimetry of Fetal Vessels in Human Fetal Growth Restriction Pregnancies. Cells 2022; 11:cells11193110. [PMID: 36231072 PMCID: PMC9562243 DOI: 10.3390/cells11193110] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Fetal growth restriction (FGR) is associated with short- and long-term morbidity, often with fetal compromise in utero, evidenced by abnormal Doppler velocimetry of fetal vessels. Lipids are vital for growth and development, but metabolism in FGR pregnancy, where fetuses do not grow to full genetic potential, is poorly understood. We hypothesize that triglyceride concentrations are increased in placentas and that important complex lipids are reduced in cord plasma from pregnancies producing the smallest babies (birth weight < 5%) and correlate with ultrasound Dopplers. Dopplers (umbilical artery, UA; middle cerebral artery, MCA) were assessed longitudinally in pregnancies diagnosed with estimated fetal weight (EFW) < 10% at ≥29 weeks gestation. For a subset of enrolled women, placentas and cord blood were collected at delivery, fatty acids were extracted and targeted lipid class analysis (triglyceride, TG; phosphatidylcholine, PC; lysophosphatidylcholine, LPC; eicosanoid) performed by LCMS. For this sub-analysis, participants were categorized as FGR (Fenton birth weight, BW ≤ 5%) or SGA "controls" (Fenton BW > 5%). FGRs (n = 8) delivered 1 week earlier (p = 0.04), were 29% smaller (p = 0.002), and had 133% higher UA pulsatility index (PI, p = 0.02) than SGAs (n = 12). FGR plasma TG, free arachidonic acid (AA), and several eicosanoids were increased (p < 0.05); docosahexaenoic acid (DHA)-LPC was decreased (p < 0.01). Plasma TG correlated inversely with BW (p < 0.05). Plasma EET, non-esterified AA, and DHA correlated inversely with BW and directly with UA PI (p < 0.05). Placental DHA-PC and AA-PC correlated directly with MCA PI (p < 0.05). In fetuses initially referred for inadequate fetal growth (EFW < 10%), those with BW ≤ 5% demonstrated distinctly different cord plasma lipid profiles than those with BW > 5%, which correlated with Doppler PIs. This provides new insights into fetal lipidomic response to the FGR in utero environment. The impact of these changes on specific processes of growth and development (particularly fetal brain) have not been elucidated, but the relationship with Doppler PI may provide additional context for FGR surveillance, and a more targeted approach to nutritional management of these infants.
Collapse
|
13
|
Ruebel ML, Martins LR, Schall PZ, Pursley JR, Latham KE. Effects of early lactation body condition loss in dairy cows on serum lipid profiles and on oocyte and cumulus cell transcriptomes. J Dairy Sci 2022; 105:8470-8484. [DOI: 10.3168/jds.2022-21919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/10/2022] [Indexed: 11/19/2022]
|
14
|
Lin J, Sun X, Dai X, Zhang S, Zhang X, Wang Q, Zheng Q, Huang M, He Y, Lin R. Integrated Proteomics and Metabolomics Analysis in Pregnant Rat Hippocampus After Circadian Rhythm Inversion. Front Physiol 2022; 13:941585. [PMID: 35936909 PMCID: PMC9355539 DOI: 10.3389/fphys.2022.941585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/07/2022] [Indexed: 11/15/2022] Open
Abstract
To investigate the changes in proteins, metabolites, and related mechanisms in the hypothalamus of pregnant rats after circadian rhythm inversion during the whole pregnancy cycle. A total of 12 Wistar female rats aged 7 weeks were randomly divided into control (six rats) and experimental (six rats) groups at the beginning of pregnancy. The control group followed a 12-h light and dark cycle (6 a.m. to 6 p.m. light, 6 p.m. to 6 a.m. dark the next day), and the experimental group followed a completely inverted circadian rhythm (6 p.m. to 6 a.m. light the next day, 6 a.m. to 6 p.m. dark). Postpartum data were collected until 7–24 h after delivery, and hypothalamus samples were collected in two groups for quantitative proteomic and metabolism analyses. The differential proteins and metabolites of the two groups were screened by univariate combined with multivariate statistical analyses, and the differential proteins and metabolites enriched pathways were annotated with relevant databases to analyze the potential mechanisms after circadian rhythm inversion. A comparison of postpartum data showed that circadian rhythm inversion can affect the number of offspring and the average weight of offspring in pregnant rats. Compared with the control group, the expression of 20 proteins and 37 metabolites was significantly changed in the experimental group. The integrated analysis between proteins and metabolites found that RGD1562758 and lysophosphatidylcholine acyltransferase 1 (LPCAT1) proteins were closely associated with carbon metabolism (choline, NAD+, L-glutamine, theobromine, D-fructose, and pyruvate) and glycerophospholipid metabolism (choline, NAD+, L-glutamine, phosphatidylcholine, theobromine, D-fructose, pyruvate, and arachidonate). Moreover, the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the differential metabolites enriched in adenosine triphosphate (ATP)–binding cassette (ABC) transporters. Our study suggested that circadian rhythm inversion in pregnant rats may affect the numbers, the average weight of offspring, and the expressions of proteins and metabolism in the hypothalamus, which may provide a comprehensive overview of the molecular profile of circadian rhythm inversion in pregnant groups.
Collapse
Affiliation(s)
- Jingjing Lin
- School of Nursing Fujian Medical University, Fuzhou City, China
| | - Xinyue Sun
- The First Affiliated Hospital of Fujian Medical University, Fuzhou City, China
| | - Xiaofeng Dai
- The First Affiliated Hospital of Fujian Medical University, Fuzhou City, China
| | | | - Xueling Zhang
- School of Nursing Fujian Medical University, Fuzhou City, China
- The First Affiliated Hospital of Fujian Medical University, Fuzhou City, China
| | - Qiaosong Wang
- School of Nursing Fujian Medical University, Fuzhou City, China
| | - Qirong Zheng
- School of Nursing Fujian Medical University, Fuzhou City, China
| | - Minfang Huang
- School of Nursing Fujian Medical University, Fuzhou City, China
- The First Affiliated Hospital of Fujian Medical University, Fuzhou City, China
| | - Yuanyuan He
- School of Nursing Fujian Medical University, Fuzhou City, China
| | - Rongjin Lin
- School of Nursing Fujian Medical University, Fuzhou City, China
- The First Affiliated Hospital of Fujian Medical University, Fuzhou City, China
- *Correspondence: Rongjin Lin,
| |
Collapse
|
15
|
Vaughan OR, Rosario FJ, Chan J, Cox LA, Ferchaud-Roucher V, Zemski-Berry KA, Reusch JEB, Keller AC, Powell TL, Jansson T. Maternal obesity causes fetal cardiac hypertrophy and alters adult offspring myocardial metabolism in mice. J Physiol 2022; 600:3169-3191. [PMID: 35545608 DOI: 10.1113/jp282462] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/04/2022] [Indexed: 01/09/2023] Open
Abstract
Obesity in pregnant women causes fetal cardiac dysfunction and increases offspring cardiovascular disease risk, but its effect on myocardial metabolism is unknown. We hypothesized that maternal obesity alters fetal cardiac expression of metabolism-related genes and shifts offspring myocardial substrate preference from glucose towards lipids. Female mice were fed control or obesogenic diets before and during pregnancy. Fetal hearts were studied in late gestation (embryonic day (E) 18.5; term ≈ E21), and offspring were studied at 3, 6, 9 or 24 months postnatally. Maternal obesity increased heart weight and peroxisome proliferator activated receptor gamma (Pparg) expression in female and male fetuses and caused left ventricular diastolic dysfunction in the adult offspring. Cardiac dysfunction worsened progressively with age in female, but not male, offspring of obese dams, in comparison to age-matched control animals. In 6-month-old offspring, exposure to maternal obesity increased cardiac palmitoyl carnitine-supported mitochondrial respiration in males and reduced myocardial 18 F-fluorodeoxyglucose uptake in females. Cardiac Pparg expression remained higher in adult offspring of obese dams than control dams and was correlated with contractile and metabolic function. Maternal obesity did not affect cardiac palmitoyl carnitine respiration in females or 18 F-fluorodeoxyglucose uptake in males and did not alter cardiac 3 H-oleic acid uptake, pyruvate respiration, lipid content or fatty acid/glucose transporter abundance in offspring of either sex. The results support our hypothesis and show that maternal obesity affects offspring cardiac metabolism in a sex-dependent manner. Persistent upregulation of Pparg expression in response to overnutrition in utero might underpin programmed cardiac impairments mechanistically and contribute to cardiovascular disease risk in children of women with obesity. KEY POINTS: Obesity in pregnant women causes cardiac dysfunction in the fetus and increases lifelong cardiovascular disease risk in the offspring. In this study, we showed that maternal obesity in mice induces hypertrophy of the fetal heart in association with altered expression of genes related to nutrient metabolism. Maternal obesity also alters cardiac metabolism of carbohydrates and lipids in the adult offspring. The results suggest that overnutrition in utero might contribute to increased cardiovascular disease risk in children of women with obesity.
Collapse
Affiliation(s)
- Owen R Vaughan
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Fredrick J Rosario
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jeannie Chan
- Department of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Laura A Cox
- Department of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Veronique Ferchaud-Roucher
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karin A Zemski-Berry
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jane E B Reusch
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
| | - Amy C Keller
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
| | - Theresa L Powell
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
16
|
Abstract
Fatty acids are critical bioactives for fetal and neonatal development. Premature delivery and current nutritional strategies pose several challenges in restoring fatty acid balance in the preterm infant. The impact on fatty acid balance and outcomes using lipid emulsions, enteral nutrition, and enteral supplements are reviewed, including a summary of the most recent large clinical trials of enteral fatty acid supplementation for the preterm infant. Research gaps remain in successfully implementing nutritional strategies to optimize fatty acid status in preterm infants.
Collapse
|
17
|
Maternal Blood Fatty Acid Levels in Small and Adequate for Gestational Age Pregnancies. J Obstet Gynaecol India 2022; 72:217-223. [PMID: 35928082 PMCID: PMC9343542 DOI: 10.1007/s13224-022-01632-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 02/01/2022] [Indexed: 10/18/2022] Open
Abstract
Background The aim is to assess the levels of fatty acids (FAs) in pregnancies with small for gestational age (SGA) and adequate for gestational age (AGA) fetuses, constituting an association between FAs and fetal growth; according to the role of FA, lower levels were expected in SGA. Materials and Methods This was an analytical cross-sectional study including pregnant women with gestational ages of 26-36 weeks with AGA and constitutionally SGA fetuses diagnosed by ultrasonography. The levels of saturated, trans, monounsaturated, and polyunsaturated fatty acids were measured using centrifugation and liquid chromatography. Student's t test and general linear model using gestational age as covariant were used to compare the levels of FAs and the groups (AGA and SGA). Chi-square was used to evaluate the association between groups and studied variables. Pearson correlation coefficient and linear regression were used to evaluate the correlation between the levels of FAs and gestational age. Results Peripheral blood was collected from 67 pregnant women, 3 of whom were excluded from the study. No significant statistical differences were observed between SGA (n = 40) and AGA (n = 24) in relation to saturated, trans, monounsaturated, and polyunsaturated fatty acids (p > 0.05). There was not significant correlation between saturated, trans, monounsaturated, and polyunsaturated FAs and gestational age (p > 0.05). Conclusion The levels of saturated, trans, monounsaturated, and polyunsaturated FAs were similar in constitutionally SGA and AGA fetuses.
Collapse
|
18
|
Nema J, Joshi N, Sundrani D, Joshi S. Influence of maternal one carbon metabolites on placental programming and long term health. Placenta 2022; 125:20-28. [DOI: 10.1016/j.placenta.2022.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/12/2022] [Accepted: 02/24/2022] [Indexed: 10/18/2022]
|
19
|
Ni LF, Han Y, Wang CC, Ye Y, Ding MM, Zheng T, Wang YH, Yan HT, Yang XJ. Relationships Between Placental Lipid Activated/Transport-Related Factors and Macrosomia in Healthy Pregnancy. Reprod Sci 2021; 29:904-914. [PMID: 34750770 DOI: 10.1007/s43032-021-00755-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/25/2021] [Indexed: 10/19/2022]
Abstract
To assess associations between infants with macrosomia and placental expression levels of lipid activated/transport-related factors and umbilical cord blood lipid concentrations in healthy pregnancy. We conducted a case-control study of 38 macrosomic neonates (MS group) and 39 normal-birth-weight newborns (NC group) in a healthy pregnancy. Cord blood lipid levels were measured by automatic biochemical analyzer, mRNA and protein expression levels of placental lipid activated/transport-related factors were determined by real-time polymerase chain reaction and western blot, respectively. Compared with NC group, cord blood total cholesterol (TC), low-density lipoprotein cholesterol (LDLC), and non-esterified fatty acid (NEFA) concentrations were decreased in the MS group. The mRNA and protein expression levels of placental peroxisome proliferator-activated receptors (PPARα, PPARγ), plasma membrane fatty acid-binding protein (FABPpm), and fatty acid translocase (FAT/CD36) were significantly higher in the MS group than the NC group. And there was a weak positive correlation between the expression of PPARγ, FABP4, and FABP3 mRNA in the placenta and the HDLC (rs = 0.439; P = 0.005), NEFA (rs = 0.342; P = 0.041), and TG (rs = 0.349; P = 0.034) levels in the cord blood in the MS group, respectively. After multivariate adjustment, the logistic regression analysis showed that high placental PPARα (adjusted odds ratio [AOR] = 3.022; 95% confidence interval [CI] 1.032-8.853) and FAT/CD36 (AOR=2.989; 95%CI 1.029-8.679) and low LDLC concentration in the cord blood (AOR=0.246; 95%CI 0.080-0.759) increased the risk of macrosomia. The increased PPARα and FAT/CD36 expression levels may influence the occurrence of fetal macrosomia through regulating placental lipid transport.
Collapse
Affiliation(s)
- Li-Fang Ni
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Han
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chen-Chen Wang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Ye
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Miao-Miao Ding
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tian Zheng
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu-Huan Wang
- Department of Obstetrics, The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hong-Tao Yan
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xin-Jun Yang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
20
|
Abstract
Almost 2 billion adults in the world are overweight, and more than half of them are classified as obese, while nearly one-third of children globally experience poor growth and development. Given the vast amount of knowledge that has been gleaned from decades of research on growth and development, a number of questions remain as to why the world is now in the midst of a global epidemic of obesity accompanied by the "double burden of malnutrition," where overweight coexists with underweight and micronutrient deficiencies. This challenge to the human condition can be attributed to nutritional and environmental exposures during pregnancy that may program a fetus to have a higher risk of chronic diseases in adulthood. To explore this concept, frequently called the developmental origins of health and disease (DOHaD), this review considers a host of factors and physiological mechanisms that drive a fetus or child toward a higher risk of obesity, fatty liver disease, hypertension, and/or type 2 diabetes (T2D). To that end, this review explores the epidemiology of DOHaD with discussions focused on adaptations to human energetics, placental development, dysmetabolism, and key environmental exposures that act to promote chronic diseases in adulthood. These areas are complementary and additive in understanding how providing the best conditions for optimal growth can create the best possible conditions for lifelong health. Moreover, understanding both physiological as well as epigenetic and molecular mechanisms for DOHaD is vital to most fully address the global issues of obesity and other chronic diseases.
Collapse
Affiliation(s)
- Daniel J Hoffman
- Department of Nutritional Sciences, Program in International Nutrition, and Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Theresa L Powell
- Department of Pediatrics and Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Daniel B Hardy
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
21
|
Sarli PM, Manousopoulou A, Efthymiou E, Zouridis A, Potiris A, Pervanidou P, Panoulis K, Vlahos N, Deligeoroglou E, Garbis SD, Eleftheriades M. Liver Proteome Profile of Growth Restricted and Appropriately Grown Newborn Wistar Rats Associated With Maternal Undernutrition. Front Endocrinol (Lausanne) 2021; 12:684220. [PMID: 34127923 PMCID: PMC8195994 DOI: 10.3389/fendo.2021.684220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022] Open
Abstract
Background Fetal growth restriction (FGR) has been associated with adverse perinatal outcomes and epigenetic modifications that impact gene expression leading to permanent changes of fetal metabolic pathways and thereby influence development of disease in childhood and adult life. In this study, we investigated the result of maternal food restriction on liver protein expression in Wistar male newborn pups. Materials & Methods Ten (n = 10) timed pregnant Wistar rats on their 14th day of gestation were randomly assigned to either control (n = 4) or food restricted group (n = 6). The control group had ad libitum access to food. In the food restricted group, maternal diet was limited in a moderate fashion (50%) from day 15 of pregnancy until delivery. All rats delivered spontaneously on day 21 and newborn pups were immediately weighed. Pups born to normally nourished mothers were considered as controls, while pups born to food restricted mothers were subdivided into two groups, based on their birth weight: growth restricted (FGR) and appropriately grown (non-FGR). Rats were euthanized immediately after birth and liver tissues of 11 randomly selected male offspring (FGR n = 4, non-FGR n = 4, control n = 3) were collected and analyzed using quantitative proteomics. Results In total 6,665 proteins were profiled. Of these, 451 and 751 were differentially expressed in FGR and non-FGR vs. control, respectively, whereas 229 proteins were commonly expressed. Bioinformatics analysis of the differentially expressed proteins (DEPs) in FGR vs. control revealed induction of the super-pathway of cholesterol biosynthesis and inhibition of thyroid hormone metabolism, fatty acid beta oxidation and apelin liver signaling pathway. Analysis of DEPs in non-FGR vs. control groups showed inhibition of thyroid hormone metabolism, fatty acid beta oxidation, and apelin liver signaling pathway. Conclusion This study demonstrates the impact of prenatal food restriction on the proteomic liver profile of FGR and non-FGR offspring underlying the importance of both prenatal adversities and birth weight on liver-dependent postnatal disease.
Collapse
Affiliation(s)
- Polyxeni-Maria Sarli
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antigoni Manousopoulou
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Elias Efthymiou
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Zouridis
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Potiris
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Pervanidou
- First Department of Paediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Panoulis
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Vlahos
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthymios Deligeoroglou
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Spiros D. Garbis
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Makarios Eleftheriades
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Barrett E, Loverin A, Wang H, Carlson M, Larsen TD, Almeida MM, Whitman J, Baack ML, Joss-Moore LA. Uteroplacental Insufficiency with Hypoxia Upregulates Placental PPARγ-KMT5A Axis in the Rat. Reprod Sci 2021; 28:1476-1488. [PMID: 33398850 PMCID: PMC8215892 DOI: 10.1007/s43032-020-00434-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/13/2020] [Indexed: 12/30/2022]
Abstract
The placenta represents a critical node in fetal lipid acquisition, yet the mechanisms by which the placenta handles lipids under normal and pathologic conditions are incompletely understood. A key player in placental lipid handling is peroxisome proliferator-activated receptor gamma (PPARγ). PPARγ influences global gene expression via its regulation of the epigenetic modifier lysine methyltransferase 5A (KMT5A), which places a methyl group on histone 4 lysine 20 (H4K20me) of target genes. Here we test the hypothesis that KMT5A is present in both the human and rat placentas and is affected by uteroplacental insufficiency (UPI) in the rat in association with increased placental lipid accumulation. We assessed levels and localization of KMT5A, as well as lipid droplet accumulation, in human placental tissue collected from maternal donors after delivery by planned cesarean section. Using a rat model of UPI, we also evaluated the effects of UPI on lipid accumulation, PPARγ, KMT5A, and H4K20me in the rat placenta. In this study, we show for the first time the presence and activity of KMT5A, in human and in rat placentas. We also demonstrate that in the rat placenta, UPI increases hypoxia, KMT5a expression, and activity in association with increased lipid accumulation in placenta supporting male fetuses. Placental PPARγ-KMT5A axis may be an important mediator of placental lipid handling.
Collapse
Affiliation(s)
- Emily Barrett
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, 84109, USA
| | - Amy Loverin
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, 84109, USA
| | - Haimei Wang
- Department of Pediatrics, University of Utah, 295 Chipeta Way, UT, 84108, Salt Lake City, USA
| | | | - Tricia D Larsen
- Environmental Influences on Health and Disease, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Mariana M Almeida
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jenna Whitman
- Department of Pediatrics, University of Utah, 295 Chipeta Way, UT, 84108, Salt Lake City, USA
| | - Michelle L Baack
- Environmental Influences on Health and Disease, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Lisa A Joss-Moore
- Department of Pediatrics, University of Utah, 295 Chipeta Way, UT, 84108, Salt Lake City, USA.
| |
Collapse
|
23
|
Zhao L, Li Y, Ding Q, Li Y, Chen Y, Ruan XZ. CD36 Senses Dietary Lipids and Regulates Lipids Homeostasis in the Intestine. Front Physiol 2021; 12:669279. [PMID: 33995128 PMCID: PMC8113691 DOI: 10.3389/fphys.2021.669279] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Dietary lipids absorbed in the intestine are closely related to the development of metabolic syndrome. CD36 is a multi-functional scavenger receptor with multiple ligands, which plays important roles in developing hyperlipidemia, insulin resistance, and metabolic syndrome. In the intestine, CD36 is abundant on the brush border membrane of the enterocytes mainly localized in proximal intestine. This review recapitulates the update and current advances on the importance of intestinal CD36 in sensing dietary lipids and regulating intestinal lipids uptake, synthesis and transport, and regulating intestinal hormones secretion. However, further studies are still needed to demonstrate the complex interactions between intestinal CD36 and dietary lipids, as well as its importance in diet associated metabolic syndrome.
Collapse
Affiliation(s)
- Lei Zhao
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yuqi Li
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qiuying Ding
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yanping Li
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yaxi Chen
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiong Z Ruan
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, University College London, London, United Kingdom
| |
Collapse
|
24
|
Godhamgaonkar AA, Wadhwani NS, Joshi SR. Exploring the role of LC-PUFA metabolism in pregnancy complications. Prostaglandins Leukot Essent Fatty Acids 2020; 163:102203. [PMID: 33227645 DOI: 10.1016/j.plefa.2020.102203] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/09/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022]
Abstract
Maternal nutrition during pregnancy plays a significant role in growth and development of the placenta and influencing pregnancy outcome. Suboptimal nutritional status during early gestational period compromises the normal course of pregnancy leading to adverse maternal and fetal outcomes. Omega-3 and omega-6 long chain polyunsaturated fatty acids (LC-PUFA) are important for the growth and development of the placenta. Maternal fatty acids and their metabolites influence the normal course of pregnancy by regulating cell growth and development, cell signaling, regulate angiogenesis, modulate inflammatory responses and influence various structural and functional processes. Alterations in LC-PUFA and their metabolites may result in inadequate spiral artery remodeling or placental angiogenesis leading to structural and functional deficiency of the placenta which contributes to several pregnancy complications like preeclampsia, gestational diabetes mellitus, intrauterine growth restriction, and results in adverse birth outcomes. In this review, we summarize studies examining the role of fatty acids and their metabolites in pregnancy. We also discuss the possible molecular mechanisms through which LC-PUFA influences placental growth and development. Studies have demonstrated that omega-3 fatty acid supplementation lowers the incidence of preterm births, but its effect on reducing pregnancy complications are inconclusive.
Collapse
Affiliation(s)
- Aditi A Godhamgaonkar
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India
| | - Nisha S Wadhwani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Pune 411043, India.
| |
Collapse
|
25
|
Abascal-Saiz A, Fuente-Luelmo E, Haro M, de la Calle M, Ramos-Álvarez MP, Perdomo G, Bartha JL. Placental Compartmentalization of Lipid Metabolism: Implications for Singleton and Twin Pregnancies. Reprod Sci 2020; 28:1150-1160. [PMID: 33171514 DOI: 10.1007/s43032-020-00385-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/01/2020] [Indexed: 11/27/2022]
Abstract
The study of placental lipid metabolism in uncomplicated pregnancies has not been developed in the literature to date. Its importance lies in expanding the knowledge of placental function to enable comparison with pathological pregnancies in future research. The aim of the present study was to compare the lipid metabolic activity and storage of the maternal and fetal sides of the placenta in healthy pregnancies. Moreover, we compare singleton vs. twin pregnancies to determine if placental metabolic needs differ. We analyzed placental explants from uncomplicated pregnancies, 20 from singleton and 8 from bichorial-biamniotic twin pregnancies (n = 28). Six cotyledon fragments were collected from each placenta at different distances from the umbilical cord, three close to the chorionic plate (hereinafter, we will refer to them as "fetal side") and another three close to the anchoring villi into the decidua basalis (referred to as "maternal side"). The samples were analyzed for quantitative assay placental fatty acid oxidation (FAO) and esterification (FAE) activities and triglyceride levels. The location of lipid storage in the chorionic villi was assessed by Oil red-O staining. Placental fatty acid oxidation did not show differences when comparing the maternal and fetal sides of the placenta or between single and twin pregnancies. When comparing placental sides, FAE was increased twofold in the maternal side compared to the fetal side of the placenta (P = 0.013). The tendency for lipogenesis in the placenta was exemplified by the FAE/FAO ratio, which was a 37.1% higher on the maternal side (P = 0.019). Despite this, triglyceride levels were five times higher in the fetal side than in the maternal one (P = 0.024). When analyzing singleton vs. twins, FAE was superior in the fetal side in multiple pregnancies (× 2.6, P = 0.007) and the FAE/FAO ratio was significantly higher in twins than in singleton pregnancies, on both sides of the placenta. Despite this finding, triglyceride levels were similar in twin and singleton pregnancies. Comparing the placentas of twins in the same pregnancy, there were no differences in lipid metabolism (FAO or FAE) or placental triglyceride levels between the two co-twins. Using Oil red-O staining, lipid storage in chorionic villi was found to be located on the syncytiotrophoblast cells and not in the connecting axis. The maternal side of the placenta is more active in the esterification of fatty acids, while the storage of neutral lipids concentrates on the fetal side. Moreover, multiple gestations have increased esterification without changes in the concentration of placental triglycerides, probably due to a higher transfer to the fetal circulation in response to the greater energy demand from twin fetuses.
Collapse
Affiliation(s)
- Alejandra Abascal-Saiz
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Eva Fuente-Luelmo
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María Haro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María de la Calle
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - María P Ramos-Álvarez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - Germán Perdomo
- Department of Health Sciences, University of Burgos, Burgos, Spain.,Institute of Molecular Biology and Genetic (IMBG), CSIC - University of Valladolid, Valladolid, Spain
| | - José L Bartha
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
26
|
Bidne KL, Rister AL, McCain AR, Hitt BD, Dodds ED, Wood JR. Maternal obesity alters placental lysophosphatidylcholines, lipid storage, and the expression of genes associated with lipid metabolism‡. Biol Reprod 2020; 104:197-210. [PMID: 33048132 DOI: 10.1093/biolre/ioaa191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/21/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Dyslipidemia is a characteristic of maternal obesity and previous studies have demonstrated abnormalities in fatty acid oxidation and storage in term placentas. However, there is little information about the effect of pre-pregnancy obesity on placental lipid metabolism during early pregnancy. The objective of this study was to determine the relationship between lipid profiles and markers of metabolism in placentas from obese and lean dams at midgestation. Mice were fed a western diet (WD) or normal diet (ND) and lysophosphatidylcholines (LPCs) and/or phosphatidylcholines (PCs) were measured in dam circulation and placenta sections using liquid chromatography-tandem mass spectrometry and mass spectrometry imaging, respectively. In WD dam, circulating LPCs containing 16:1, 18:1, 20:0, and 20:3 fatty acids were increased and 18:2 and 20:4 were decreased. In WD placenta from both sexes, LPC 18:1 and PC 36:1 and 38:3 were increased. Furthermore, there were moderate to strong correlations between LPC 18:1, PC 36:1, and PC 38:3. Treatment-, spatial-, and sex-dependent differences in LPC 20:1 and 20:3 were also detected. To identify genes that may regulate diet-dependent differences in placenta lipid profiles, the expression of genes associated with lipid metabolism and nutrient transport was measured in whole placenta and isolated labyrinth using droplet digital PCR and Nanostring nCounter assays. Several apolipoproteins were increased in WD placentas. However, no differences in nutrient transport or fatty acid metabolism were detected. Together, these data indicate that lipid storage is increased in midgestation WD placentas, which may lead to lipotoxicity, altered lipid metabolism and transport to the fetus later in gestation.
Collapse
Affiliation(s)
- Katie L Bidne
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Alana L Rister
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrea R McCain
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Brianna D Hitt
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Eric D Dodds
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, USA.,Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jennifer R Wood
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
27
|
Steinhauser CB, Askelson K, Lambo CA, Hobbs KC, Bazer FW, Satterfield MC. Lipid metabolism is altered in maternal, placental, and fetal tissues of ewes with small for gestational age fetuses†. Biol Reprod 2020; 104:170-180. [PMID: 33001151 DOI: 10.1093/biolre/ioaa180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/23/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Nutrient restriction (NR) has the potential to negatively impact birthweight, an indicator of neonatal survival and lifelong health. Those fetuses are termed as small for gestational age (SGA). Interestingly, there is a spectral phenotype of fetal growth rates in response to NR associated with changes in placental development, nutrient and waste transport, and lipid metabolism. A sheep model with a maternal diet, starting at Day 35, of 100% National Research Council (NRC) nutrient requirements (n = 8) or 50% NRC (n = 28) was used to assess alterations in fetuses designated NR SGA (n = 7) or NR NonSGA (n = 7) based on fetal weight at Day 135 of pregnancy. Allantoic fluid concentrations of triglycerides were greater in NR SGA fetuses than 100% NRC and NR NonSGA fetuses at Day 70 (P < 0.05). There was a negative correlation between allantoic fluid concentrations of triglycerides (R2 = 0.207) and bile acids (R2 = 0.179) on Day 70 and fetal weight at Day 135 for NR ewes (P < 0.05). Bile acids were more abundant in maternal and fetal blood for NR SGA compared to 100% NRC and NR NonSGA ewes (P < 0.05). Maternal blood concentrations of NEFAs increased in late pregnancy in NR NonSGA compared to NR SGA ewes (P < 0.05). Protein expression of fatty acid transporter SLC27A6 localized to placentomal maternal and fetal epithelia and decreased in Day 70 NR SGA compared to 100% NRC and NR NonSGA placentomes (P < 0.05). These results identify novel factors associated with an ability of placentae and fetuses in NR NonSGA ewes to adapt to, and overcome, nutritional hardship during pregnancy.
Collapse
Affiliation(s)
| | - Katharine Askelson
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Colleen A Lambo
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Kenneth C Hobbs
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - M Carey Satterfield
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
28
|
Mining of combined human placental gene expression data across pregnancy, applied to PPAR signaling pathway. Placenta 2020; 99:157-165. [PMID: 32805615 DOI: 10.1016/j.placenta.2020.07.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION To date, we have only an incomplete understanding of how gene expression in the human placenta changes at the genome-wide scale from very early in gestation to term. Our aim was to investigate the dynamic changes in gene expression throughout placentation. METHODS In our study, gene expression profiles were collected of human placentas from 4 to 40 gestational weeks of age. Simple linear regression and weighted correlation network analysis were applied to identify genes of interest. Analyses of gene enrichment, including gene ontology and pathways from the Kyoto Encyclopedia of Genes and Genomes, were performed using clusterProfiler. Finally, dynamic changes in the expression of individual genes were represented using line graphs of scaled and adjusted gene expression. RESULTS Our results highlighted a total of 5173 genes that are involved in different periods of placentation. Downstream annotation of these genes revealed the biological processes and pathways involved, from which we chose to further investigate the PPAR signaling pathway. We were able to detect changes over time in many genes involved in lipid storage/metabolism, including members of the FABP family and LPL. These patterns were corroborated by lipid staining of placental sections, which revealed a significant decrease in lipid droplet content in placentas from early in the first trimester to term. CONCLUSION Our study provides detailed information on the dynamics of biological processes and pathways across human placentation. These findings give us new clues for deciphering the normal functions of placentation and the ways in which the mis-regulation of these pathways may be linked to pregnancy-related diseases. As an example, our results show that the PPAR signaling pathway mediates a constant decrease in placental lipid content over the course of pregnancy.
Collapse
|
29
|
Chassen SS, Ferchaud-Roucher V, Palmer C, Li C, Jansson T, Nathanielsz PW, Powell TL. Placental fatty acid transport across late gestation in a baboon model of intrauterine growth restriction. J Physiol 2020; 598:2469-2489. [PMID: 32338384 PMCID: PMC7384518 DOI: 10.1113/jp279398] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Intrauterine growth restriction (IUGR) is associated with perinatal morbidity and increased risk of lifelong disease, including neurodevelopmental impairment. Fatty acids (FA) are critical for normal brain development, although their transport across the placenta in IUGR pregnancies is poorly understood. The present study used a baboon model of IUGR (maternal nutrient restriction, MNR) to investigate placental expression of FA transport and binding proteins, and to determine gestational age-related changes in maternal and fetal plasma FA concentrations. We found MNR to be associated with increased placental expression of FA binding and transport proteins in late gestation, with fetal plasma FA concentrations that were similar to those of control animals. The present study is the first to report a profile of fetal and maternal plasma FA concentrations in a baboon model of growth restriction with data that suggest adaptation of placental transport to maintain delivery of critically needed FA. ABSTRACT Intrauterine growth restriction (IUGR) is associated with specific changes in placental transport of amino acids, folate and ions. However, little is known about placental fatty acid (FA) transport in IUGR. We hypothesized that placental FA transport proteins (FATP) and FA binding proteins (FABP) are up-regulated and fetal plasma FA concentrations are decreased at term in a baboon model of IUGR. Pregnant baboons were fed control or maternal nutrient restricted (MNR) diet (70% of control calories) from gestation day (GD) 30 (term 184 days). Plasma and placental samples were collected at GD120 (control n = 8, MNR n = 9), GD140 (control n = 6, MNR n = 7) and GD170 (control n = 6, MNR n = 6). Placentas were homogenized, and syncytiotrophoblast microvillous plasma membrane (MVM) and basal plasma membranes (BM) were isolated. Protein expression of FABP1, 3, 4 and 5 (homogenate) and FATP2, 4, and 6 (MVM, BM) was determined by Western blotting. FA content in maternal and umbilical vein plasma was measured by gas chromatography-mass spectrometry. Placental FABP1 and FABP5 expression was increased in MNR compared to controls at GD170, as was MVM FATP2 and FATP6 expression at GD140 and FATP2 expression at GD170. BM FATP4 and FATP6 expression was increased in MNR at GD140. Fetal plasma FA concentrations were similar in controls and MNR. These data suggest the adaptation of placental transport when aiming to maintain delivery of critically needed FAs for fetal growth and brain development.
Collapse
Affiliation(s)
- Stephanie S Chassen
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Veronique Ferchaud-Roucher
- University of Nantes, CHU Nantes, INRA, UMR 1280 Physiology of Nutritional Adaptations, Nantes, France
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire Palmer
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cun Li
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
- Southwest National Primate Research Center, San Antonio, TX, USA
| | - Thomas Jansson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Peter W Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
- Southwest National Primate Research Center, San Antonio, TX, USA
| | - Theresa L Powell
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
30
|
Khaire A, Wadhwani N, Madiwale S, Joshi S. Maternal fats and pregnancy complications: Implications for long-term health. Prostaglandins Leukot Essent Fatty Acids 2020; 157:102098. [PMID: 32380367 DOI: 10.1016/j.plefa.2020.102098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022]
Abstract
Pregnancy imposes increased nutritional requirements for the well being of the mother and fetus. Maternal lipid metabolism is critical for fetal development and long-term health of the offspring as it plays a key role in energy storage, tissue growth and cell signaling. Maternal fat composition is considered as a modifiable risk for abnormal lipid metabolism and glucose tolerance during pregnancy. Data derived from observational studies demonstrate that higher intake of saturated fats during pregnancy is associated with pregnancy complications (preeclampsia, gestational diabetes mellitus and preterm delivery) and poor birth outcomes (intra uterine growth retardation and large for gestational age babies). On the other hand, prenatal long chain polyunsaturated fatty acids status is shown to improve birth outome. In this article, we discuss the role of maternal lipids during pregnancy on fetal growth and development and its consequences on the health of the offspring.
Collapse
Affiliation(s)
- Amrita Khaire
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Nisha Wadhwani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Shweta Madiwale
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India.
| |
Collapse
|
31
|
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: 2] [Impact Index Per Article: 0.5] [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
|
32
|
Duttaroy AK, Basak S. Maternal dietary fatty acids and their roles in human placental development. Prostaglandins Leukot Essent Fatty Acids 2020; 155:102080. [PMID: 32120190 DOI: 10.1016/j.plefa.2020.102080] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/24/2020] [Accepted: 02/18/2020] [Indexed: 12/26/2022]
Abstract
Fatty acids are essential for feto-placental growth and development. Maternal fatty acids and their metabolites are involved in every stage of pregnancy by supporting cell growth and development, cell signaling, and modulating other critical aspects of structural and functional processes. Early placentation process is critical for placental growth and function. Several fatty acids modulate angiogenesis as observed by increased tube formation and secretion of angiogenic growth factors in first-trimester human placental trophoblasts. Long-chain fatty acids stimulate angiogenesis in these cells via vascular endothelium growth factor (VEGF), angiopoietin-like protein 4 (ANGPTL4), fatty acid-binding proteins (FABPs), or eicosanoids. Inadequate placental angiogenesis and trophoblast invasion of the maternal decidua and uterine spiral arterioles leads to structural and functional deficiency of placenta, which contributes to preeclampsia, pre-term intrauterine growth restriction, and spontaneous abortion and also affects overall fetal growth and development. During the third trimester of pregnancy, placental preferential transport of maternal plasma long-chain polyunsaturated fatty acids is of critical importance for fetal growth and development. Fatty acids cross the placental microvillous and basal membranes by mainly via plasma membrane fatty acid transport system (FAT, FATP, p-FABPpm, & FFARs) and cytoplasmic FABPs. Besides, a member of the major facilitator superfamily-MFSD2a, present in the placenta is involved in the supply of DHA to the fetus. Maternal factors such as diet, obesity, endocrine, inflammation can modulate the expression and activity of the placental fatty acid transport activity and thereby impact feto-placental growth and development. In this review, we discuss the maternal dietary fatty acids, and placental transport and metabolism, and their roles in placental growth and development.
Collapse
Affiliation(s)
- Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Sanjay Basak
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway; National Institute of Nutrition, Hyderabad, India
| |
Collapse
|
33
|
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: 2.3] [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
|
34
|
Chassen S, Jansson T. Complex, coordinated and highly regulated changes in placental signaling and nutrient transport capacity in IUGR. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165373. [PMID: 30684642 PMCID: PMC6650384 DOI: 10.1016/j.bbadis.2018.12.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023]
Abstract
The most common cause of intrauterine growth restriction (IUGR) in the developed world is placental insufficiency, a concept often used synonymously with reduced utero-placental and umbilical blood flows. However, placental insufficiency and IUGR are associated with complex, coordinated and highly regulated changes in placental signaling and nutrient transport including inhibition of insulin and mTOR signaling and down-regulation of specific amino acid transporters, Na+/K+-ATPase, the Na+/H+-exchanger, folate and lactate transporters. In contrast, placental glucose transport capacity is unaltered and Ca2+-ATPase activity and the expression of proteins involved in placental lipid transport are increased in IUGR. These findings are not entirely consistent with the traditional view that the placenta is dysfunctional in IUGR, but rather suggest that the placenta adapts to reduce fetal growth in response to an inability of the mother to allocate resources to the fetus. This new model has implications for the understanding of the mechanisms underpinning IUGR and for the development of intervention strategies.
Collapse
Affiliation(s)
- Stephanie Chassen
- Department of Pediatrics, Division of Neonatology, University of Colorado, Anschutz Medical Campus, Aurora, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, USA.
| |
Collapse
|
35
|
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: 1.5] [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
|
36
|
Zhang H, Jin Y, Wang M, Loor JJ, Wang H. N-Carbamylglutamate and l-arginine supplementation improve hepatic antioxidant status in intrauterine growth-retarded suckling lambs. RSC Adv 2020; 10:11173-11181. [PMID: 35495302 PMCID: PMC9050450 DOI: 10.1039/c9ra09316h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/01/2020] [Indexed: 12/26/2022] Open
Abstract
The influence of dietary supplementation of l-arginine (Arg) or N-carbamylglutamate (NCG) on the hepatic antioxidant status in intrauterine-growth-retarded (IUGR) suckling lambs remains unclear. The current work aimed to investigate the regulatory mechanisms whereby dietary Arg or NCG alter hepatic antioxidant status in suckling lambs suffering from IUGR. Forty-eight newborn Hu lambs of normal birth weight (CON) and IUGR were allocated randomly into four groups of 12 animals each: CON (4.25 ± 0.14 kg), IUGR (3.01 ± 0.12 kg), IUGR + 1% Arg (2.99 ± 0.13 kg), or IUGR + 0.1% NCG (3.03 ± 0.11 kg). All lambs were raised for a period of 21 days from 7 to 28 days after birth. Compared with the IUGR suckling animals, glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), and reduced glutathione (GSH) content were greater (P < 0.05), and protein carbonyl and malondialdehyde (MDA) levels were reduced (P < 0.05) in the livers of both IUGR + 1% Arg and 0.1% NCG suckling animals. Relative to IUGR suckling lambs, supplementing with Arg or NCG markedly reduced (P < 0.05) reactive oxygen species (ROS) levels, apoptosis, and necrosis in liver. Relative to IUGR suckling lambs, protein and mRNA expression of GSH-Px1, SOD2, catalase (CAT), heme oxygenase-1 (HO-1), inducible nitric oxide (NO) synthase (iNOS), and epithelial NO synthase (eNOS) increased in IUGR animals receiving Arg or NCG (P < 0.05). Both Arg and NCG can protect neonates from IUGR-induced hepatic oxidative damage through promoting the expression of antioxidative enzymes (including SOD, CAT, and GSH-Px), phase II metabolizing enzymes, and activation of the NO pathway. The influence of dietary supplementation of l-arginine (Arg) or N-carbamylglutamate (NCG) on the hepatic antioxidant status in intrauterine-growth-retarded (IUGR) suckling lambs remains unclear.![]()
Collapse
Affiliation(s)
- Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| | - Yaqian Jin
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| | - Juan J. Loor
- Department of Animal Sciences and Division of Nutritional Sciences
- University of Illinois
- Urbana
- USA
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| |
Collapse
|
37
|
Miller CN, Kodavanti UP, Stewart EJ, Schladweiler MC, Richards JH, Snow SJ, Henriquez AR, Oshiro WM, Farraj AK, Hazari MS, Dye JA. Fetal growth outcomes following peri-implantation exposure of Long-Evans rats to noise and ozone differ by sex. Biol Sex Differ 2019; 10:54. [PMID: 31791410 PMCID: PMC6889602 DOI: 10.1186/s13293-019-0270-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/19/2019] [Indexed: 12/29/2022] Open
Abstract
Background Exposure to air pollution and high levels of noise have both been independently associated with the development of adverse pregnancy outcomes including low birth weight. However, exposure to such environmental stressors rarely occurs in isolation and is often co-localized, especially in large urban areas. Methods The purpose of this study was to compare the effects of combined exposure to noise (N) or ozone (O3), compared to either exposure alone. Long-Evans dams were exposed to air or 0.4 ppm ozone for 4 h on gestation day (GD) 5 and 6, coinciding with implantation receptivity. A subset of dams from each exposure group was further exposed to intermittent white noise (~ 85 dB) throughout the dark cycle following each inhalation exposure (n = 14 − 16/group). Uterine artery ultrasound was performed on GD 15 and 21. Fetal growth characteristics and indicators of placental nutrient status were measured at GD 21. Results Exposure to ozone + quiet (O3 + Q) conditions reduced uterine arterial resistance at GD 15 compared to air + quiet (A + Q) exposure, with no further reduction by GD 21. By contrast, exposure to air + noise (A + N) significantly increased uterine arterial resistance at both GD 15 and 21. Notably, while peri-implantation exposure to O3 + Q conditions reduced male fetal weight at GD 21, this effect was not observed in the air + noise (A + N) or the ozone + noise (O3 + N) exposure groups. Fetal weight in female offspring was not reduced by ozone exposure alone (O3 + Q), nor was it affected by air + noise (A + N) or by combined ozone + noise (O3 + N) exposure. Conclusions These data indicate that exposure to ozone and noise differentially impact uterine blood flow, particularly at mid-gestation, with only ozone exposure being associated with sex-dependent fetal growth retardation in male offspring.
Collapse
Affiliation(s)
- Colette N Miller
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA.
| | - Urmila P Kodavanti
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Erica J Stewart
- Oak Ridge Institute of Science and Education, Research Triangle Park, NC, USA
| | - Mette C Schladweiler
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Judy H Richards
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | | | - Andres R Henriquez
- Oak Ridge Institute of Science and Education, Research Triangle Park, NC, USA
| | - Wendy M Oshiro
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Aimen K Farraj
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Mehdi S Hazari
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Janice A Dye
- Cardiopulmonary Immunotoxicology Branch, Public Health & Integrated Toxicology Division, Center for Public Health & Environmental Assessment U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
38
|
Kappen C, Kruger C, Jones S, Herion NJ, Salbaum JM. Maternal diet modulates placental nutrient transporter gene expression in a mouse model of diabetic pregnancy. PLoS One 2019; 14:e0224754. [PMID: 31774824 PMCID: PMC6881028 DOI: 10.1371/journal.pone.0224754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 10/21/2019] [Indexed: 12/30/2022] Open
Abstract
Diabetes in the mother during pregnancy is a risk factor for birth defects and perinatal complications and can affect long-term health of the offspring through developmental programming of susceptibility to metabolic disease. We previously showed that Streptozotocin-induced maternal diabetes in mice is associated with altered cell differentiation and with smaller size of the placenta. Placental size and fetal size were affected by maternal diet in this model, and maternal diet also modulated the risk for neural tube defects. In the present study, we sought to determine the extent to which these effects might be mediated through altered expression of nutrient transporters, specifically glucose and fatty acid transporters in the placenta. Our results demonstrate that expression of several transporters is modulated by both maternal diet and maternal diabetes. Diet was revealed as the more prominent determinant of nutrient transporter expression levels, even in pregnancies with uncontrolled diabetes, consistent with the role of diet in placental and fetal growth. Notably, the largest changes in nutrient transporter expression levels were detected around midgestation time points when the placenta is being formed. These findings place the critical time period for susceptibility to diet exposures earlier than previously appreciated, implying that mechanisms underlying developmental programming can act on placenta formation.
Collapse
Affiliation(s)
- Claudia Kappen
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
- * E-mail:
| | - Claudia Kruger
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Sydney Jones
- Baton Rouge, Louisiana, United States of America Regulation of Gene Expression Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Nils J. Herion
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
- Baton Rouge, Louisiana, United States of America Regulation of Gene Expression Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - J. Michael Salbaum
- Baton Rouge, Louisiana, United States of America Regulation of Gene Expression Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
39
|
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: 0.8] [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
|
40
|
Xu P, Guo H, Wang H, Lee SC, Liu M, Pan Y, Zheng J, Zheng K, Wang H, Xie Y, Bai X, Liu Y, Zhao M, Wang L. Downregulations of placental fatty acid transporters during cadmium-induced fetal growth restriction. Toxicology 2019; 423:112-122. [PMID: 31152847 DOI: 10.1016/j.tox.2019.05.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/01/2019] [Accepted: 05/27/2019] [Indexed: 01/22/2023]
Abstract
Cadmium (Cd) is one of the environmental pollutants, which has multiple toxic effects on fetuses and placentas. Placental fatty acid (FA) uptake and transport are critical for the fetal and placental development. We aimed to analyze the triglyceride (TG) level, the expression patterns of several key genes involved in FA uptake and transport, and the molecular mechanisms for the altered gene expressions in placentas in response to Cd treatment. Our results showed that the placental TG level was significantly decreased in the Cd-exposed placentas. Fatty acid transporting protein 1 (FATP1), FATP6 and fatty acid binding protein 3 (FABP3) were significantly down-regulated in the placentas from Cd-exposed mice. The expression level of phospho-p38 MAPK was increased by Cd treatment, while the protein level of total p38 MAPK remained unchanged. The expression levels of peroxisome proliferator-activated receptor-γ (PPAR-γ) and the hypoxia-inducible factor-1α (HIF-1α) were significantly decreased in the Cd-exposed placentas. The methylation levels of the promoter regions of FATP1, FATP6 and FABP3 showed no significant differences between the treatment and control groups. In addition, the circulating non-esterified fatty acid (NEFA), total cholesterol (TC), and TG levels were not decreased in the maternal serum from the Cd-exposed mice. Therefore, our results suggest Cd exposure dose not reduce the maternal FA supply, but reduces the placental TG level. Cd treatment also downregulates the placental expressions of FATP1, FATP6 and FABP3, respectively associated with p38-MAPK, p38 MAPK/PPAR-γ and HIF-1α pathways.
Collapse
Affiliation(s)
- Peng Xu
- School of Life Science, Shanxi University, Taiyuan 030006, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Huiqin Guo
- School of Life Science, Shanxi University, Taiyuan 030006, China.
| | - Huan Wang
- School of Life Science, Shanxi University, Taiyuan 030006, China.
| | - Shao Chin Lee
- School of Life Science, Shanxi University, Taiyuan 030006, China; School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China.
| | - Ming Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yongliang Pan
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou 313000, China.
| | - Jian Zheng
- Department of Cardiopulmonary Function Examination, Shanxi Provincial Cancer Hospital, Taiyuan 030013, China.
| | - Kang Zheng
- Special Ward, Shanxi Provincial Cancer Hospital, Taiyuan 030013, China.
| | - Huihui Wang
- School of Life Science, Shanxi University, Taiyuan 030006, China.
| | - Yuxin Xie
- School of Life Science, Shanxi University, Taiyuan 030006, China.
| | - Xiaoxia Bai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China.
| | - Yun Liu
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai 201300, China.
| | - Meirong Zhao
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Lan Wang
- School of Life Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
41
|
Yen MC, Chou SK, Kan JY, Kuo PL, Hou MF, Hsu YL. New Insight on Solute Carrier Family 27 Member 6 (SLC27A6) in Tumoral and Non-Tumoral Breast Cells. Int J Med Sci 2019; 16:366-375. [PMID: 30911270 PMCID: PMC6428986 DOI: 10.7150/ijms.29946] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/17/2018] [Indexed: 12/25/2022] Open
Abstract
Long-chain fatty acids are the most abundant fatty acids and are essential for various physiological processes. Translocation of long-chain fatty acids across cell membrane is dependent on transport proteins. Solute carrier family 27 member 6 (SLC27A6) is a transport protein which mediates long-chain fatty acid uptake. The bioinformatic analysis revealed that the expression of SLC27A6 in non-tumoral breast tissue was higher than that in tumoral breast cancer in clinic samples. When SLC27A6 expression in non-tumorigenic cell H184B5F5/M10 was repressed, the fatty acids uptake capacity and cell proliferation was inhibited, and cell cycle was delayed. The protein expression of cell cycle regulators including cell division protein kinase 4 (CDK4), CDK6, and cyclin D1 was significantly decreased in SLC27A6-silenced H184B5F5/M10. By contrast, relatively low SLC27A6 expression in tumorigenic breast cancer cell Hs578T when compared to H184B5F5/M10. Repressing SLC27A6 expression did not affect these phenotypes in Hs578T. The interaction network of SLC27A6 was further investigated via STRING database. The function of these SLC27A6-associated proteins mainly involved in lipid biosynthesis, fatty acid metabolic process, and fatty acid transport. In conclusion, this study reveals inverse correlation between SLC27A6 expression and tumoral tissues and provides a new insight into SLC27A6-mediated cell growth and cell cycle regulation in non-tumorigenic breast cells.
Collapse
Affiliation(s)
- Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Kai Chou
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jung-Yu Kan
- Department of Breast Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Feng Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Department of Breast Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
42
|
Effects of Arachidonic and Docosohexahenoic Acid Supplementation during Gestation in Rats. Implication of Placental Oxidative Stress. Int J Mol Sci 2018; 19:ijms19123863. [PMID: 30518038 PMCID: PMC6321355 DOI: 10.3390/ijms19123863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/26/2022] Open
Abstract
Arachidonic and docosahexaenoic acids (ARA and DHA) are important during pregnancy. However, the effects of dietary supplementation on fetal growth and oxidative stress are inconclusive. We aimed to assess the effect of high ARA and DHA diet during rat gestation on: (1) ARA and DHA availability in plasma and placenta, (2) fetal growth, and (3) placental oxidative stress, analyzing the influence of sex. Experimental diet (ED) was prepared by substituting soybean oil in the control diet (CD) by a fungi/algae-based oil containing ARA and DHA (2:1). Rats were fed with CD or ED during gestation; plasma, placenta, and fetuses were obtained at gestational day 20. DHA, ARA, and their precursors were analyzed in maternal plasma and placenta by gas chromatography/mass spectrophotometry. Fetuses and placentas were weighed, the proportion of fetuses with intrauterine growth restriction (IUGR) determined, and placental lipid and protein oxidation analyzed. ED fetuses exhibited lower body weight compared to CD, being >40% IUGR; fetal weight negatively correlated with maternal plasma ARA, but not DHA. Only ED female placenta exhibited higher lipid and protein oxidation compared to its CD counterparts; lipid peroxidation is negatively associated with fetal weight. In conclusion, high ARA during gestation associates with IUGR, through placental oxidative stress, with females being more susceptible.
Collapse
|
43
|
Bildirici I, Schaiff WT, Chen B, Morizane M, Oh SY, O’Brien M, Sonnenberg-Hirche C, Chu T, Barak Y, Nelson DM, Sadovsky Y. PLIN2 Is Essential for Trophoblastic Lipid Droplet Accumulation and Cell Survival During Hypoxia. Endocrinology 2018; 159:3937-3949. [PMID: 30351430 PMCID: PMC6240902 DOI: 10.1210/en.2018-00752] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Trophoblast hypoxia and injury, key components of placental dysfunction, are associated with fetal growth restriction and other complications of pregnancy. Accumulation of lipid droplets has been found in hypoxic nonplacental cells. Unique to pregnancy, lipid accumulation in the placenta might perturb lipid transport to the fetus. We tested the hypothesis that hypoxia leads to accumulation of lipid droplets in human trophoblasts and that trophoblastic PLIN proteins play a key role in this process. We found that hypoxia promotes the accumulation of lipid droplets in primary human trophoblasts. A similar accretion of lipid droplets was found in placental villi in vivo from pregnancies complicated by fetal growth restriction. In both situations, these changes were associated with an increased level of cellular triglycerides. Exposure of trophoblasts to hypoxia led to reduced fatty acid efflux and oxidation with no change in fatty acid uptake or synthesis. We further found that hypoxia markedly stimulated PLIN2 mRNA synthesis and protein expression, which colocalized to lipid droplets. Knockdown of PLIN2, but not PLIN3, enhanced trophoblast apoptotic death, and overexpression of PLIN2 promoted cell viability. Collectively, our data indicate that hypoxia enhances trophoblastic lipid retention in the form of lipid droplets and that PLIN2 plays a key role in this process and in trophoblast defense against apoptotic death. These findings also imply that this protective mechanism may lead to diminished trafficking of lipids to the developing fetus.
Collapse
Affiliation(s)
- Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - W Timothy Schaiff
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Baosheng Chen
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew O’Brien
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tianjiao Chu
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yaacov Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - D Michael Nelson
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
- Correspondence: Yoel Sadovsky, MD, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213. E-mail:
| |
Collapse
|
44
|
Rodrigues DM, Manfro GG, Levitan RD, Steiner M, Meaney MJ, Silveira PP. Moderating effect of PLIN4 genetic variant on impulsivity traits in 5-year-old-children born small for gestational age. Prostaglandins Leukot Essent Fatty Acids 2018; 137:19-25. [PMID: 30293593 DOI: 10.1016/j.plefa.2018.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023]
Abstract
Poor fetal growth is associated with long-term behavioral, metabolic and psychiatric alterations, including impulsivity, insulin resistance, and mood disorders. However, the consumption of omega-3 polyunsaturated fatty acid (n-3 PUFA) seems to be protective for this population, improving inhibitory control and behavioral reactivity. We investigated whether the presence of the A allele of rs8887 SNP (PLIN4 gene), known to be associated with increased sensitivity to the consumption of n-3 PUFAs, interacts with fetal growth influencing inhibitory control. 152 five-year-old children were genotyped and performed the Stop Signal Task (SSRT). There was a significant interaction between birth weight and the presence of the A allele on SSRT performance, in which lower birth weight associated with poorer inhibitory control only in non-carriers. These results suggest that a higher responsiveness to n-3 PUFAS protects small for gestational age children from developing poor response inhibition, highlighting that optimizing n-3 PUFA intake may benefit this population.
Collapse
Affiliation(s)
- Danitsa Marcos Rodrigues
- Postgraduate Program in Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gisele Gus Manfro
- Postgraduate Program in Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Robert Daniel Levitan
- Department of Psychiatry, University of Toronto and Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Meir Steiner
- Department of Psychiatry and Behavioural Neurosciences, McMaster University. Hamilton, ON, Canada
| | - Michael Joseph Meaney
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; Sackler Program for Epigenetics & Psychobiology, McGill University, Canada
| | - Patrícia Pelufo Silveira
- Postgraduate Program in Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; Sackler Program for Epigenetics & Psychobiology, McGill University, Canada.
| |
Collapse
|