1
|
Huang R, Zhou G, Cai J, Cao C, Zhu Z, Wu Q, Zhang F, Ding Y. Maternal consumption of urbanized diet compromises early-life health in association with gut microbiota. Gut Microbes 2025; 17:2483783. [PMID: 40176259 PMCID: PMC11988223 DOI: 10.1080/19490976.2025.2483783] [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: 11/25/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025] Open
Abstract
Urbanization has significantly transformed dietary habits worldwide, contributing to a globally increased burden of non-communicable diseases and altered gut microbiota landscape. However, it is often overlooked that the adverse effects of these dietary changes can be transmitted from the mother to offspring during early developmental stages, subsequently influencing the predisposition to various diseases later in life. This review aims to delineate the detrimental effects of maternal urban-lifestyle diet (urbanized diet) on early-life health and gut microbiota assembly, provide mechanistic insights on how urbanized diet mediates mother-to-offspring transfer of bioactive substances in both intrauterine and extrauterine and thus affects fetal and neonatal development. Moreover, we also further propose a framework for developing microbiome-targeted precision nutrition and diet strategies specifically for pregnant and lactating women. The establishment of such knowledge can help develop proactive preventive measures from the beginning of life, ultimately reducing the long-term risk of disease and improving public health outcomes.
Collapse
Affiliation(s)
- Rong Huang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Guicheng Zhou
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jie Cai
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Cha Cao
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhenjun Zhu
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Fen Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yu Ding
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Wei G, Shen FJ, Liu JL, Zhao JH, Yang FY, Feng RQ, Lu J, Zhang CY, Wang FW, Chen BD, Ding X, Yang JK. Uncoupling protein 1 deficiency leads to transcriptomic differences in livers of pregnancy female mice and aggravates hepatic steatosis. Arch Biochem Biophys 2025; 768:110395. [PMID: 40122441 DOI: 10.1016/j.abb.2025.110395] [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: 10/21/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Pregnancy requires the coordination of metabolically active organs to support maternal nutrition and fetal growth. However, the metabolic cross-talk between adipose tissue and liver in females during pregnancy is still less clear. In this study, we evaluated the metabolic adaptations and phenotypes of liver in response to pregnancy-associated metabolic stress, particularly in the context of genetic ablation of Uncoupling protein 1 (Ucp1)-mediated catabolic circuit. Our results revealed that Ucp1 deficiency (UCP1 knockout, KO) mice during late pregnancy exhibited significantly deteriorated metabolic phenotypes, including hepatic steatosis and whole-body glucose and lipid homeostasis, as compared to Ucp1 deficiency or normal pregnancy mice. However, non-pregnant Ucp1 deficiency mice displayed nearly normal metabolic phenotypes and structure alterations similar to those of littermate controls. Moreover, transcriptomic analyses by RNA sequencing (RNA-seq) clearly revealed that Ucp1 deficiency led to a significant liver metabolic remodeling of differentially express genes (DEGs) before and especially during pregnancy. Consistently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses demonstrated the potential altered functions and signaling pathways, including metabolic dysfunctions in ribosome, oxidative phosphorylation, etc. Importantly, as derived from trend analyses of DEGs, our results further revealed the distinct expression pattern of each subcluster, which coincided with potential biological functions and relevant signaling pathways. The findings in the present study might provide valuable insights into the molecular mechanism of metabolic dysfunction-associated fatty liver disease (MAFLD) during pregnancy. Additionally, our data may provide a novel animal model of MAFLD, thus facilitating its potential therapies. NEW & NOTEWORTHY: Genetic ablation of Ucp1 during pregnancy increases hepatic steatosis and deteriorated whole-body glucose and lipid homeostasis. Moreover, changes in hepatic gene expression are closely associated with metabolic dysfunctions in ribosome and oxidative phosphorylation. This work highlights the therapeutic potential of targeting UCP1- mediated catabolic circuit between adipose and liver during pregnancy, and the utility of RNA-seq analysis to reveal valuable information for the distinct expression pattern of each subcluster that contribute to pregnancy-dependent MASLD progression.
Collapse
Affiliation(s)
- Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China.
| | - Feng-Jie Shen
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jun-Li Liu
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Jian-Hua Zhao
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Fang-Yuan Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Ruo-Qi Feng
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Chen-Yang Zhang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Feng-Wei Wang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Bei-Dong Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100005, China.
| | - Xin Ding
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100020, China.
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
3
|
Abad C, Musilova I, Cifkova E, Portillo R, Kumnova F, Karahoda R, Sterba M, Lisa M, Kacerovsky M, Stranik J, Stuchlik A, Staud F. Impact of intraamniotic inflammation on tryptophan metabolism in the placenta-fetal brain axis in rats. Reproduction 2025; 169:e240378. [PMID: 40192828 DOI: 10.1530/rep-24-0378] [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: 10/06/2024] [Accepted: 04/07/2025] [Indexed: 04/09/2025]
Abstract
In brief Intrauterine inflammation disrupts tryptophan metabolism in both the placenta and the fetal brain, leading to a shift toward neurotoxic metabolites. These findings highlight the critical role of placental function in neurodevelopment and suggest that inflammation-induced metabolic changes may contribute to neurodevelopmental disorders. Abstract The placenta plays a crucial role beyond nutrient transfer, acting as a dynamic endocrine organ that significantly influences maternal physiology and fetal development. It responds rapidly to even slight changes in the in utero environment to promote fetal survival. Disruptions in placental function are increasingly recognized as key contributors to the origins of neurodevelopmental disorders. In this study, we employed advanced technology to induce intrauterine inflammation through ultrasound-guided administration of LPS into gestational sacs. We then evaluated its effects on the gene expression of enzymes involved in TRP metabolism and conducted a comprehensive LC/MS analysis of the metabolome in the placenta and fetal brain of Wistar rats. Our results show that intraamniotic injection of LPS induces a robust inflammatory response leading to significant alterations in TRP metabolism, including downregulation of tryptophan hydroxylase (TPH) in the placenta, resulting in a decrease in serotonin (5-HT) levels. Similarly, in the fetal brain, exposure to LPS led to reduced Tph expression and increased monoamine oxidase expression, suggesting a decrease in 5-HT synthesis and an increase in its degradation. Furthermore, an upregulation of the kynurenine pathway was observed in both the placenta and fetal brain. Moreover, we detected a shift toward neurotoxicity, evidenced by an imbalance between neuroprotective and neurotoxic metabolites, including decreased levels of kynurenic acid and upregulation of kynurenine monooxygenase in the fetal brain. In conclusion, our findings reveal significant alterations in TRP metabolism following intrauterine inflammation, potentially contributing to neurodevelopmental disorders.
Collapse
|
4
|
Pan H, Ouyang B, Zhang H, Zhao C. Non-coding RNAs: the architects of placental development and pregnancy success. Mol Genet Genomics 2025; 300:39. [PMID: 40159439 DOI: 10.1007/s00438-025-02244-8] [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: 01/10/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Noncoding RNAs (ncRNAs) constitute a significant portion of the transcriptome that lacks evident protein-coding functions; however, they have been confirmed to be crucial in various biological processes, including placental development. Notwithstanding the existence of various ncRNAs, research on their role in placental development and pregnancy has been constrained. The predominant category of identified ncRNAs specific to placental tissue is microRNAs (miRNAs). Given their prevalence, the significantly larger cohort of other non-coding RNAs, such as circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs), is anticipated to exert a considerably greater influence than miRNAs. Syncytiotrophoblast, a fetal-derived cell, serves as a conduit between the fetus and mother by secreting extracellular vesicles that contain fetal proteins and RNA. Alterations in ncRNAs within placental tissue, especially in trophoblast cells and extracellular vesicles, may be linked to placental dysfunction that leads to pregnancy complications, serving either as a causative factor or a result. This review encapsulates the existing understanding of ncRNAs in placental development, pregnancy success, pregnancy-related complications, extracellular vesicle conveyance, and their capacity as innovative diagnostic instruments and therapeutic strategies.
Collapse
Affiliation(s)
- Hongjuan Pan
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China
| | - Baisha Ouyang
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China
| | - Hui Zhang
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China
| | - Caizhen Zhao
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China.
| |
Collapse
|
5
|
Ponsuksili S, Murani E, Fuchs B, Galuska CE, Reyer H, Iqbal MA, Li S, Oster M, Wimmers K. Genetic regulation and variation of fetal plasma metabolome in the context of sex, paternal breeds and variable fetal weight. Open Biol 2025; 15:240285. [PMID: 40037532 DOI: 10.1098/rsob.240285] [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: 10/04/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
Metabolic processes in fetuses can significantly influence piglet weight at birth. Understanding the genetic determinants of systemic metabolism is crucial for uncovering how genetic and molecular pathways impact biological mechanisms, particularly during the fetal phase. We present data on 1112 plasma metabolites using untargeted ultra-high performance liquid chromatography-tandem mass spectrometry methods, of 260 backcross (BC) fetuses from two sires' breeds at 63 days post-conception. Eight chemical superclasses have been identified, with lipids accounting for the majority of metabolites. Genomic heritability (h²) was estimated for each metabolite, revealing that 50% had h² values below 0.2, with a higher average in the amino acid class compared with the lipid. We annotated 448 significant metabolite quantitative trait loci associated with 10 metabolites, primarily lipids, indicating strong genetic regulation. Additionally, metabolite associations with sex, fetal weight and sire's breed were explored, revealing significant associations for 354 metabolites. Fetal weight influenced the largest number of metabolites, particularly glycerophospholipids and sphingolipids, emphasizing the genetic and metabolic complexity underlying fetal development. These findings enhance our understanding of the genetic regulation of metabolite levels and their associations with key phenotypic traits in fetuses, providing insights into metabolic pathways, potential biomarkers and serving as a baseline dataset for metabolomics studies of fetuses.
Collapse
Affiliation(s)
- Siriluck Ponsuksili
- Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Eduard Murani
- Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Beate Fuchs
- Core Facility Metabolomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Christina E Galuska
- Core Facility Metabolomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Henry Reyer
- Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Muhammad Arsalan Iqbal
- Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Shuaichen Li
- Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Michael Oster
- Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
| | - Klaus Wimmers
- Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Dummerstorf 18196, Germany
- Faculty of Agricultural and Environmental Sciences, University of Rostock, Justus-von-Liebig-Weg 6b, Rostock 18059, Germany
| |
Collapse
|
6
|
Wu JJ, Zheng X, Wu C, Ma W, Wang Y, Wang J, Wei Y, Zeng X, Zhang S, Guan W, Chen F. Melatonin alleviates high temperature exposure induced fetal growth restriction via the gut-placenta-fetus axis in pregnant mice. J Adv Res 2025; 68:131-146. [PMID: 38382594 PMCID: PMC11785557 DOI: 10.1016/j.jare.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Global warming augments the risk of adverse pregnancy outcomes in vulnerable expectant mothers. Pioneering investigations into heat stress (HS) have predominantly centered on its direct impact on reproductive functions, while the potential roles of gut microbiota, despite its significant influence on distant tissues, remain largely unexplored. Our understanding of deleterious mechanisms of HS and the development of effective intervention strategies to mitigate the detrimental impacts are still limited. OBJECTIVES In this study, we aimed to explore the mechanisms by which melatonin targets gut microbes to alleviate HS-induced reproductive impairment. METHODS We firstly evaluated the alleviating effects of melatonin supplementation on HS-induced reproductive disorder in pregnant mice. Microbial elimination and fecal microbiota transplantation (FMT) experiments were then conducted to confirm the efficacy of melatonin through regulating gut microbiota. Finally, a lipopolysaccharide (LPS)-challenged experiment was performed to verify the mechanism by which melatonin alleviates HS-induced reproductive impairment. RESULTS Melatonin supplementation reinstated gut microbiota in heat stressed pregnant mice, reducing LPS-producing bacteria (Aliivibrio) and increasing beneficial butyrate-producing microflora (Butyricimonas). This restoration corresponded to decreased LPS along the maternal gut-placenta-fetus axis, accompanied by enhanced intestinal and placental barrier integrity, safeguarding fetuses from oxidative stress and inflammation, and ultimately improving fetal weight. Further pseudo-sterile and fecal microbiota transplantation trials confirmed that the protective effect of melatonin on fetal intrauterine growth under HS was partially dependent on gut microbiota. In LPS-challenged pregnant mice, melatonin administration mitigated placental barrier injury and abnormal angiogenesis via the inactivation of the TLR4/MAPK/VEGF signaling pathway, ultimately leading to enhanced nutrient transportation in the placenta and thereby improving the fetal weight. CONCLUSION Melatonin alleviates HS-induced low fetal weight during pregnancy via the gut-placenta-fetus axis, the first time highlighting the gut microbiota as a novel intervention target to mitigate the detrimental impact of global temperature rise on vulnerable populations.
Collapse
Affiliation(s)
- Jia-Jin Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyu Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Caichi Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Wen Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yibo Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yulong Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, PR China
| | - Shihai Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Fang Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
7
|
Van Willigen BG, van der Hout-van der Jagt MB, Huberts W, van de Vosse FN. A Multiscale Mathematical Model for Fetal Gas Transport and Regulatory Systems During Second Half of Pregnancy. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2025; 41:e3881. [PMID: 39625422 DOI: 10.1002/cnm.3881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/28/2024] [Accepted: 10/12/2024] [Indexed: 12/21/2024]
Abstract
Fetal asphyxia, a condition resulting from the combined effects of hypoxia and hypercapnia, leads to approximately 900,000 annual deaths worldwide. One cause is umbilical cord compression during labor-induced uterine contractions, disrupting the transport of metabolites to and from the placenta, and resulting in asphyxia. Current fetal well-being assessment relies on monitoring fetal heart rate and uterine contractions as indicators of oxygen delivery to the brain. To enhance our understanding of this complex relationship, this study aims to develop a modular mathematical model including fetal blood gas dynamics, the autonomic nervous system, and cerebral blood flow regulation. The novelty of this study lies in the capability of the model to simulate fetal growth. These submodels are part of a larger multiscale mathematical model describing fetal circulation in the second half of pregnancy. The blood gas model realistically replicates partial oxygen and carbon dioxide pressures in umbilical arteries and veins during healthy fetal development reported in the literature. An in silico experiment is conducted to simulate umbilical cord occlusion and is compared with lamb experiments to verify the realism of the regulation models during fetal growth. Our findings suggest that premature infants are more susceptible to umbilical cord occlusion, exhibiting elevated cerebral perfusion pressure and flow. This modular mathematical model may serve as a valuable tool for testing hypotheses related to the fetal regulatory system.
Collapse
Affiliation(s)
- Bettine G Van Willigen
- Cardiovascular Biomechanics, Biomedical Engineering, Eindhoven University of Technology, The Netherlands
- Obstetrics and Gynaecology, Máxima Medical Centre, Veldhoven, The Netherlands
| | - M Beatrijs van der Hout-van der Jagt
- Obstetrics and Gynaecology, Máxima Medical Centre, Veldhoven, The Netherlands
- Signal Processing Systems, Electrical Engineering, Eindhoven University of Technology, The Netherlands
| | - Wouter Huberts
- Cardiovascular Biomechanics, Biomedical Engineering, Eindhoven University of Technology, The Netherlands
| | - Frans N van de Vosse
- Cardiovascular Biomechanics, Biomedical Engineering, Eindhoven University of Technology, The Netherlands
| |
Collapse
|
8
|
Wilsterman K, Bautista AI, Butler CE, Juergens MY, Larson AM. Evolution of Litter Size: Proximate and Ultimate Mechanisms. Integr Comp Biol 2024; 64:1643-1660. [PMID: 38802126 PMCID: PMC11659681 DOI: 10.1093/icb/icae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Relative reproductive success and failure are the ultimate determinants of Darwinian fitness. As such, reproductive traits and variations therein have an immediate and considerable impact on the evolutionary trajectory of lineages. Historically, significant attention has been paid to the ecological and evolutionary processes (ultimate factors) that shape the diversity and canalization of reproductive traits within groups to better our understanding of organismal diversity and population or species resilience. In contrast, the physiological systems that mediate variation within and among species (i.e., the proximate factors) in reproductive traits remain a significant black box. To date, there is comparatively little information about how proximate mechanisms constrain or promote evolutionary potential in reproductive traits. In this mini-review, we focus on litter size in Eutherian mammals as a trait with relatively well-defined diversity (litter sizes are well-described both within and across species) and for which some genetic determinants have been identified. We discuss both the ultimate and potential proximate determinants of litter size with special attention to the breadth of physiological traits that may act as "toggle" switches for evolution of litter size. We close with a brief discussion of the role that physiological plasticity may play in the evolution of litter size and lay out several forward-looking areas for future research.
Collapse
Affiliation(s)
- Kathryn Wilsterman
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| | | | - Chloe E Butler
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| | - Makenna Y Juergens
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| | - Ashley M Larson
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| |
Collapse
|
9
|
Yu X, Xu J, Song B, Zhu R, Liu J, Liu YF, Ma YJ. The role of epigenetics in women's reproductive health: the impact of environmental factors. Front Endocrinol (Lausanne) 2024; 15:1399757. [PMID: 39345884 PMCID: PMC11427273 DOI: 10.3389/fendo.2024.1399757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
This paper explores the significant role of epigenetics in women's reproductive health, focusing on the impact of environmental factors. It highlights the crucial link between epigenetic modifications-such as DNA methylation and histones post-translational modifications-and reproductive health issues, including infertility and pregnancy complications. The paper reviews the influence of pollutants like PM2.5, heavy metals, and endocrine disruptors on gene expression through epigenetic mechanisms, emphasizing the need for understanding how dietary, lifestyle choices, and exposure to chemicals affect gene expression and reproductive health. Future research directions include deeper investigation into epigenetics in female reproductive health and leveraging gene editing to mitigate epigenetic changes for improving IVF success rates and managing reproductive disorders.
Collapse
Affiliation(s)
- Xinru Yu
- College Of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiawei Xu
- College Of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine School, Jinan, Shandong, China
| | - Bihan Song
- College Of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine School, Jinan, Shandong, China
| | - Runhe Zhu
- College Of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine School, Jinan, Shandong, China
| | - Jiaxin Liu
- College Of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yi Fan Liu
- Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ying Jie Ma
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
10
|
Yu X, Wu H, Su J, Liu X, Liang K, Li Q, Yu R, Shao X, Wang H, Wang YL, Shyh-Chang N. Acetyl-CoA metabolism maintains histone acetylation for syncytialization of human placental trophoblast stem cells. Cell Stem Cell 2024; 31:1280-1297.e7. [PMID: 39084220 DOI: 10.1016/j.stem.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/15/2023] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
During pregnancy, placental-fetal nutrient allocation is crucial for fetal and maternal health. However, the regulatory mechanisms for nutrient metabolism and allocation in placental trophoblasts have remained unclear. Here, we used human first-trimester placenta samples and human trophoblast stem cells (hTSCs) to discover that glucose metabolism is highly active in hTSCs and cytotrophoblasts, but during syncytialization, it decreases to basal levels, remaining necessary for fueling acetyl-CoA and differentiation potential. Acetate supplementation could rescue syncytiotrophoblast fusion from glycolysis deficiency by replenishing acetyl-CoA and maintaining histone acetylation, thus rescuing the activation of syncytialization genes. Even brief glycolysis deficiency could permanently inhibit differentiation potential and promote inflammation, which could also be permanently rescued by brief acetate supplementation in vivo. These results suggest that hTSCs retain only basal glycolytic acetyl-CoA metabolism during syncytialization to regulate cell fates via nutrient-responsive histone acetylation, with implications for our understanding of the balance between placental and fetal nutrition.
Collapse
Affiliation(s)
- Xin Yu
- Key Laboratory of Organ Regeneration and Reconstruction, 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 101408, China
| | - Hao Wu
- Key Laboratory of Organ Regeneration and Reconstruction, 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 101408, China
| | - Jiali Su
- Key Laboratory of Organ Regeneration and Reconstruction, 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 101408, China
| | - Xupeng Liu
- Key Laboratory of Organ Regeneration and Reconstruction, 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 101408, China
| | - Kun Liang
- Key Laboratory of Organ Regeneration and Reconstruction, 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 101408, China
| | - Qianqian Li
- Key Laboratory of Organ Regeneration and Reconstruction, 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 101408, China
| | - Ruoxuan Yu
- Key Laboratory of Organ Regeneration and Reconstruction, 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 101408, China
| | - Xuan Shao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Yan-Ling Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Ng Shyh-Chang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
11
|
Formisano E, Proietti E, Perrone G, Demarco V, Galoppi P, Stefanutti C, Pisciotta L. Characteristics, Physiopathology and Management of Dyslipidemias in Pregnancy: A Narrative Review. Nutrients 2024; 16:2927. [PMID: 39275243 PMCID: PMC11397408 DOI: 10.3390/nu16172927] [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: 07/26/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Dyslipidemia is a significant risk factor for atherosclerotic cardiovascular disease (ASCVD). During pregnancy, physiological changes elevate cholesterol and triglyceride levels to support fetal development, which can exacerbate pre-existing conditions and lead to complications such as pre-eclampsia, gestational diabetes, and increased ASCVD risk for both mother and child. Effective management strategies are necessary, especially for pregnant women with inherited forms of dyslipidemia (i.e., familial hypertriglyceridemia, hyperchylomicronemia), where personalized dietary adjustments are crucial for successful pregnancy outcomes. Pharmacological interventions and lipoprotein apheresis may be necessary for severe cases, though their use is often limited by factors such as cost, availability, and potential fetal risks. Despite the promise of advanced therapies, their widespread application remains constrained by limited studies and high costs. Thus, a personalized, multidisciplinary approach is essential for optimizing outcomes. This review provides a comprehensive overview of current strategies and evidence-based practices for managing dyslipidemia during pregnancy, emphasizing the balance of maternal and fetal health. Additionally, it discusses the physiological changes in lipid metabolism during pregnancy and their implications, particularly for women with inherited forms of dyslipidemia.
Collapse
Affiliation(s)
- Elena Formisano
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- Dietetics and Clinical Nutrition Unit, IRCCS Policlinic Hospital San Martino, 16132 Genoa, Italy
| | - Elisa Proietti
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Giuseppina Perrone
- Department of Maternal and Child Health and Urological Sciences, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Valentina Demarco
- Department of Maternal and Child Health and Urological Sciences, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Paola Galoppi
- Department of Maternal and Child Health and Urological Sciences, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Claudia Stefanutti
- Department of Molecular Medicine, Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Regional Centre for Rare Diseases, Immunohematology and Transfusion Medicine, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Livia Pisciotta
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- Dietetics and Clinical Nutrition Unit, IRCCS Policlinic Hospital San Martino, 16132 Genoa, Italy
| |
Collapse
|
12
|
Parenti M, Slupsky CM. Disrupted Prenatal Metabolism May Explain the Etiology of Suboptimal Neurodevelopment: A Focus on Phthalates and Micronutrients and their Relationship to Autism Spectrum Disorder. Adv Nutr 2024; 15:100279. [PMID: 39059765 PMCID: PMC11375317 DOI: 10.1016/j.advnut.2024.100279] [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: 05/13/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
Pregnancy is a time of high metabolic coordination, as maternal metabolism adapts to support the growing fetus. Many of these changes are coordinated by the placenta, a critical fetal endocrine organ and the site of maternal-fetal crosstalk. Dysregulation in maternal and placental metabolism during pregnancy has been linked to adverse outcomes, including altered neurodevelopment. Autism spectrum disorder (ASD) is a neurodevelopmental disorder linked to metabolic alterations in both children and their mothers. Prenatal environmental exposures have been linked to risk of ASD through dysregulated maternal, placental, and fetal metabolism. In this review, we focus on recent studies investigating the associations between prenatal metabolism in the maternal-placental-fetal unit and the impact of prenatal environmental exposures to phthalates and micronutrients on ASD risk. By identifying the mechanisms through which phthalates and other ubiquitous endocrine disrupting chemicals influence development, and how nutritional interventions can impact those mechanisms, we can identify promising ways to prevent suboptimal neurodevelopment.
Collapse
Affiliation(s)
- Mariana Parenti
- Department of Nutrition, University of California, Davis, CA, United States
| | - Carolyn M Slupsky
- Department of Nutrition, University of California, Davis, CA, United States; Department of Food Science and Technology, University of California, Davis, CA, United States.
| |
Collapse
|
13
|
Manoharan MM, Montes GC, Acquarone M, Swan KF, Pridjian GC, Nogueira Alencar AK, Bayer CL. Metabolic theory of preeclampsia: implications for maternal cardiovascular health. Am J Physiol Heart Circ Physiol 2024; 327:H582-H597. [PMID: 38968164 PMCID: PMC11442029 DOI: 10.1152/ajpheart.00170.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Preeclampsia (PE) is a multisystemic disorder of pregnancy that not only causes perinatal mortality and morbidity but also has a long-term toll on the maternal and fetal cardiovascular system. Women diagnosed with PE are at greater risk for the subsequent development of hypertension, ischemic heart disease, cardiomyopathy, cerebral edema, seizures, and end-stage renal disease. Although PE is considered heterogeneous, inefficient extravillous trophoblast (EVT) migration leading to deficient spiral artery remodeling and increased uteroplacental vascular resistance is the likely initiation of the disease. The principal pathophysiology is placental hypoxia, causing subsequent oxidative stress, leading to mitochondrial dysfunction, mitophagy, and immunological imbalance. The damage imposed on the placenta in turn results in the "stress response" categorized by the dysfunctional release of vasoactive components including oxidative stressors, proinflammatory factors, and cytokines into the maternal circulation. These bioactive factors have deleterious effects on systemic endothelial cells and coagulation leading to generalized vascular dysfunction and hypercoagulability. A better understanding of these metabolic factors may lead to novel therapeutic approaches to prevent and treat this multisystemic disorder. In this review, we connect the hypoxic-oxidative stress and inflammation involved in the pathophysiology of PE to the resulting persistent cardiovascular complications in patients with preeclampsia.
Collapse
Affiliation(s)
- Mistina M Manoharan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
| | - Guilherme C Montes
- Department of Pharmacology and Psychobiology, Roberto Alcântara Gomes Institute Biology (IBRAG), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Mariana Acquarone
- Department of Neurology, Tulane University, New Orleans, Louisiana, United States
| | - Kenneth F Swan
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | - Gabriella C Pridjian
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | | | - Carolyn L Bayer
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| |
Collapse
|
14
|
Luo Y, Wang Z, Zhao X, Xing J, Chen Z, Zhao W, Long X, Zhang Y, Shao Y. Combining the Vaginal Microbiome and Serum Metabolome to Screen for Potential Biomarkers of Early Pregnancy in Cows. Metabolites 2024; 14:469. [PMID: 39330476 PMCID: PMC11434538 DOI: 10.3390/metabo14090469] [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: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/24/2024] [Indexed: 09/28/2024] Open
Abstract
Early pregnancy diagnostic techniques are of significant importance in livestock farming, particularly in dairy farming. This study aimed to screen artificially inseminated cows for potential biomarkers at day 21 of pregnancy using microbiota-metabolomics analysis. The microbiome analysis revealed significant changes (p < 0.05) in the composition and abundance of the vaginal microbiota in cows after pregnancy. Notably, there was an increase in the abundance of [Eubacterium]_hallii_group (p < 0.05) associated with the production of short-chain fatty acids in the pregnant group compared with the non-pregnant group. Furthermore, significant alterations were observed in the serum metabolome, with notable changes in the concentrations of prolyl-hydroxyproline (Pro-Hyp) (p < 0.01) and bonactin (p < 0.01). The majority of differential metabolites clustered within the pathways of amino acid metabolism and lipid metabolism, with lipid metabolism exhibiting a higher proportion and playing a pivotal role in early pregnancy. An enzyme-linked immunosorbent assay was employed to quantify three key metabolites of the arachidonic acid pathway. The results demonstrated significant decreases in serum concentrations of leukotriene B4 (LTB4) (p < 0.05) and prostaglandin F2α (PGF2α) (p < 0.01) and no significant changes in arachidonic acid (AA) (NS) concentrations after 21 days of gestation in cows. Spearman's correlation analysis was utilized to investigate the interrelationship between the vaginal microbiota and serum metabolites. In conclusion, the present study demonstrated that biomaterials such as bonactin, Pro-hyp, LTB4, PGF2α in serum metabolites and [Eubacterium]_hallii_group in the vaginal flora of cows could be utilized as potential biomarkers for 21 days of gestation in cows.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yongbin Shao
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (Y.L.); (Z.W.); (X.Z.); (J.X.); (Z.C.); (W.Z.); (X.L.); (Y.Z.)
| |
Collapse
|
15
|
Lv W, Xie H, Wu S, Dong J, Jia Y, Ying H. Identification of key metabolism-related genes and pathways in spontaneous preterm birth: combining bioinformatic analysis and machine learning. Front Endocrinol (Lausanne) 2024; 15:1440436. [PMID: 39229380 PMCID: PMC11368757 DOI: 10.3389/fendo.2024.1440436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Background Spontaneous preterm birth (sPTB) is a global disease that is a leading cause of death in neonates and children younger than 5 years of age. However, the etiology of sPTB remains poorly understood. Recent evidence has shown a strong association between metabolic disorders and sPTB. To determine the metabolic alterations in sPTB patients, we used various bioinformatics methods to analyze the abnormal changes in metabolic pathways in the preterm placenta via existing datasets. Methods In this study, we integrated two datasets (GSE203507 and GSE174415) from the NCBI GEO database for the following analysis. We utilized the "Deseq2" R package and WGCNA for differentially expressed genes (DEGs) analysis; the identified DEGs were subsequently compared with metabolism-related genes. To identify the altered metabolism-related pathways and hub genes in sPTB patients, we performed multiple functional enrichment analysis and applied three machine learning algorithms, LASSO, SVM-RFE, and RF, with the hub genes that were verified by immunohistochemistry. Additionally, we conducted single-sample gene set enrichment analysis to assess immune infiltration in the placenta. Results We identified 228 sPTB-related DEGs that were enriched in pathways such as arachidonic acid and glutathione metabolism. A total of 3 metabolism-related hub genes, namely, ANPEP, CKMT1B, and PLA2G4A, were identified and validated in external datasets and experiments. A nomogram model was developed and evaluated with 3 hub genes; the model could reliably distinguish sPTB patients and term labor patients with an area under the curve (AUC) > 0.75 for both the training and validation sets. Immune infiltration analysis revealed immune dysregulation in sPTB patients. Conclusion Three potential hub genes that influence the occurrence of sPTB through shadow participation in placental metabolism were identified; these results provide a new perspective for the development and targeting of treatments for sPTB.
Collapse
Affiliation(s)
- Wenqi Lv
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Han Xie
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Shengyu Wu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Jiaqi Dong
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Yuanhui Jia
- Department of Clinical Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, sChina
| | - Hao Ying
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
- Department of Clinical Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, sChina
| |
Collapse
|
16
|
Hung SC, Chan TF, Chan HC, Wu CY, Chan ML, Jhuang JY, Tan JQ, Mei JB, Law SH, Ponnusamy VK, Chan HC, Ke LY. Lysophosphatidylcholine Impairs the Mitochondria Homeostasis Leading to Trophoblast Dysfunction in Gestational Diabetes Mellitus. Antioxidants (Basel) 2024; 13:1007. [PMID: 39199251 PMCID: PMC11351454 DOI: 10.3390/antiox13081007] [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: 07/05/2024] [Revised: 08/10/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is a common pregnancy disorder associated with an increased risk of pre-eclampsia and macrosomia. Recent research has shown that the buildup of excess lipids within the placental trophoblast impairs mitochondrial function. However, the exact lipids that impact the placental trophoblast and the underlying mechanism remain unclear. GDM cases and healthy controls were recruited at Kaohsiung Medical University Hospital. The placenta and cord blood were taken during birth. Confocal and electron microscopy were utilized to examine the morphology of the placenta and mitochondria. We determined the lipid composition using liquid chromatography-mass spectrometry in data-independent analysis mode (LC/MSE). In vitro studies were carried out on choriocarcinoma cells (JEG3) to investigate the mechanism of trophoblast mitochondrial dysfunction. Results showed that the GDM placenta was distinguished by increased syncytial knots, chorangiosis, lectin-like oxidized low-density lipoprotein (LDL) receptor-1 (LOX-1) overexpression, and mitochondrial dysfunction. Lysophosphatidylcholine (LPC) 16:0 was significantly elevated in the cord blood LDL of GDM patients. In vitro, we demonstrated that LPC dose-dependently disrupts mitochondrial function by increasing reactive oxygen species (ROS) levels and HIF-1α signaling. In conclusion, highly elevated LPC in cord blood plays a pivotal role in GDM, contributing to trophoblast impairment and pregnancy complications.
Collapse
Affiliation(s)
- Shao-Chi Hung
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Te-Fu Chan
- Graduate Institute of Medicine, College of Medicine & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
| | - Hsiu-Chuan Chan
- PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (H.-C.C.); (V.K.P.)
| | - Chia-Ying Wu
- The Master Program of AI Application in Health Industry, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Mei-Lin Chan
- Division of Thoracic Surgery, Department of Surgery, MacKay Memorial Hospital, MacKay Medical College, Taipei 104217, Taiwan;
- Department of Medicine, MacKay Medical College, New Taipei 252005, Taiwan;
| | - Jie-Yang Jhuang
- Department of Medicine, MacKay Medical College, New Taipei 252005, Taiwan;
- Department of Pathology, Mackay Memorial Hospital, Tamsui Branch, New Taipei 251404, Taiwan
| | - Ji-Qin Tan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Jia-Bin Mei
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Shi-Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
| | - Vinoth Kumar Ponnusamy
- PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (H.-C.C.); (V.K.P.)
- Department of Medicinal and Applied Chemistry & Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hua-Chen Chan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
- Department of Medical Laboratory Science, College of Medicine, I-Shou University, Kaohsiung 824005, Taiwan
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.-C.H.); (J.-Q.T.); (J.-B.M.); (S.-H.L.)
- Graduate Institute of Medicine, College of Medicine & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
- Center for Lipid Biosciences, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
| |
Collapse
|
17
|
Masserdotti A, Gasik M, Grillari-Voglauer R, Grillari J, Cargnoni A, Chiodelli P, Papait A, Magatti M, Romoli J, Ficai S, Di Pietro L, Lattanzi W, Silini AR, Parolini O. Unveiling the human fetal-maternal interface during the first trimester: biophysical knowledge and gaps. Front Cell Dev Biol 2024; 12:1411582. [PMID: 39144254 PMCID: PMC11322133 DOI: 10.3389/fcell.2024.1411582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 08/16/2024] Open
Abstract
The intricate interplay between the developing placenta and fetal-maternal interactions is critical for pregnancy outcomes. Despite advancements, gaps persist in understanding biomechanics, transport processes, and blood circulation parameters, all of which are crucial for safe pregnancies. Moreover, the complexity of fetal-maternal interactions led to conflicting data and methodological variations. This review presents a comprehensive overview of current knowledge on fetal-maternal interface structures, with a particular focus on the first trimester. More in detail, the embryological development, structural characteristics, and physiological functions of placental chorionic plate and villi, fetal membranes and umbilical cord are discussed. Furthermore, a description of the main structures and features of maternal and fetal fluid dynamic exchanges is provided. However, ethical constraints and technological limitations pose still challenges to studying early placental development directly, which calls for sophisticated in vitro, microfluidic organotypic models for advancing our understanding. For this, knowledge about key in vivo parameters are necessary for their design. In this scenario, the integration of data from later gestational stages and mathematical/computational simulations have proven to be useful tools. Notwithstanding, further research into cellular and molecular mechanisms at the fetal-maternal interface is essential for enhancing prenatal care and improving maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Alice Masserdotti
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- Institute of Molecular Biotechnology, BOKU University, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Paola Chiodelli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sara Ficai
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorena Di Pietro
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| |
Collapse
|
18
|
Hansen SSK, Krautz R, Rago D, Havelund J, Stigliani A, Færgeman NJ, Prézelin A, Rivière J, Couturier-Tarrade A, Akimov V, Blagoev B, Elfving B, Neess D, Vogel U, Khodosevich K, Hougaard KS, Sandelin A. Pulmonary maternal immune activation does not cross the placenta but leads to fetal metabolic adaptation. Nat Commun 2024; 15:4711. [PMID: 38830841 PMCID: PMC11148039 DOI: 10.1038/s41467-024-48492-x] [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: 02/10/2023] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.
Collapse
Affiliation(s)
- Signe Schmidt Kjølner Hansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
- National Research Centre for the Working Environment, Copenhagen, Denmark.
| | - Robert Krautz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Daria Rago
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Jesper Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Arnaud Stigliani
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Audrey Prézelin
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Julie Rivière
- Paris-Saclay University, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Vyacheslav Akimov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Ditte Neess
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Karin Sørig Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark.
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Albin Sandelin
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
19
|
Wang S, Cui Z, Yang H. Interactions between host and gut microbiota in gestational diabetes mellitus and their impacts on offspring. BMC Microbiol 2024; 24:161. [PMID: 38730357 PMCID: PMC11083820 DOI: 10.1186/s12866-024-03255-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/08/2024] [Indexed: 05/12/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is characterized by insulin resistance and low-grade inflammation, and most studies have demonstrated gut dysbiosis in GDM pregnancies. Overall, they were manifested as a reduction in microbiome diversity and richness, depleted short chain fatty acid (SCFA)-producing genera and a dominant of Gram-negative pathogens releasing lipopolysaccharide (LPS). The SCFAs functioned as energy substance or signaling molecules to interact with host locally and beyond the gut. LPS contributed to pathophysiology of diseases through activating Toll-like receptor 4 (TLR4) and involved in inflammatory responses. The gut microbiome dysbiosis was not only closely related with GDM, it was also vital to fetal health through vertical transmission. In this review, we summarized gut microbiota signature in GDM pregnancies of each trimester, and presented a brief introduction of microbiome derived SCFAs. We then discussed mechanisms of microbiome-host interactions in the physiopathology of GDM and associated metabolic disorders. Finally, we compared offspring microbiota composition from GDM with that from normal pregnancies, and described the possible mechanism.
Collapse
Affiliation(s)
- Shuxian Wang
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Zifeng Cui
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China.
| |
Collapse
|
20
|
Galineau L, Bourdin-Pintueles A, Bodard S, Busson J, Nadal-Desbarats L, Lefèvre A, Emond P, Mavel S. Temporal metabolomics state in pregnant rat: Analysis of amniotic fluid, placenta, and maternal plasma at embryonic and fetal time points. Placenta 2024; 150:22-30. [PMID: 38581971 DOI: 10.1016/j.placenta.2024.03.015] [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: 12/09/2023] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024]
Abstract
INTRODUCTION During pregnancy, the dynamic metabolic demands for fetal growth require a continuous supply of essential metabolites. Understanding maternal metabolome changes during gestation is crucial for predicting disease risks in neonates. METHODS The study aimed to characterize the placental and amniotic fluid (AF) metabolomes during gestation in rats at gestational days GD-13 and 19 reflecting the end of the embryonic and fetal periods, respectively, and the maternal plasma, using metabolomics (LC-MS) and chemometrics. The objective was to highlight, through univariate and multivariate analyses, the complementarity of the data obtained from these different biological matrices. RESULTS The biological matrix had more impact on the metabolome composition than the gestational stage. The placental and AF metabolomes showed specific metabolome evolving over the two gestational stages. Analyzing the three targeted metabolomes revealed evolving pathways in arginine and proline metabolism/glutathione metabolism and phenylalanine metabolism; purine metabolism; and carbohydrate metabolism. Significantly, lipid metabolism in the placenta exhibited substantial changes with higher levels of certain phosphatidylethanolamine and sphingomyelins at GD19 while some cholesteryl esters and some glycosphingolipids levels being in higher levels at GD13. DISCUSSION These data highlight the metabolic gradients (mainly in placenta, also in AF, but only a few in plasma) observed through embryonic patterning and organ development during mid-to late gestation.
Collapse
Affiliation(s)
- Laurent Galineau
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France
| | | | - Sylvie Bodard
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France
| | - Julie Busson
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France
| | - Lydie Nadal-Desbarats
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France; PST-ASB, Université de Tours, France
| | - Antoine Lefèvre
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France; PST-ASB, Université de Tours, France
| | - Patrick Emond
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France; PST-ASB, Université de Tours, France; Service de Médecine Nucléaire In Vitro, CHRU Tours, Tours, France
| | - Sylvie Mavel
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France.
| |
Collapse
|
21
|
Akinyemi AJ, Du XQ, Aguilan J, Sidoli S, Hirsch D, Wang T, Reznik S, Fuloria M, Charron MJ. Human cord plasma proteomic analysis reveals sexually dimorphic proteins associated with intrauterine growth restriction. Proteomics 2024; 24:e2300260. [PMID: 38059784 DOI: 10.1002/pmic.202300260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 12/08/2023]
Abstract
Intrauterine growth restriction (IUGR) is associated with increased risk of cardiometabolic disease later in life and has been shown to affect female and male offspring differently, but the mechanisms remain unclear. The purpose of this study was to identify proteomic differences and metabolic risk markers in IUGR male and female neonates when compared to appropriate for gestational age (AGA) babies that will provide a better understanding of IUGR pathogenesis and its associated risks. Our results revealed alterations in IUGR cord plasma proteomes with most of the differentially abundant proteins implicated in peroxisome pathways. This effect was evident in females but not in males. Furthermore, we observed that catalase activity, a peroxisomal enzyme, was significantly increased in females (p < 0.05) but unchanged in males. Finally, we identified risk proteins associated with obesity, type-2 diabetes, and glucose intolerance such as EGF containing fibulin extracellular matrix protein 1 (EFEMP1), proprotein convertase subtilisin/kexin type 9 (PCSK9) and transforming growth factor beta receptor 3 (TGFBR3) proteins unique to females while coagulation factor IX (C9) and retinol binding protein 4 (RBP4) are unique in males. In conclusion, IUGR may display sexual dimorphism which may be associated with differences in lifelong risk for cardiometabolic disease between males and females.
Collapse
Affiliation(s)
| | - Xiu Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jennifer Aguilan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David Hirsch
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Tao Wang
- Department of Epidemiology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sandra Reznik
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, Jamaica, New York, USA
| | - Mamta Fuloria
- Department of Pediatrics, Division of Neonatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Division of Endocrinology, Norman Fleisher Institute, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
22
|
Bonnet A, Bluy L, Gress L, Canario L, Ravon L, Sécula A, Billon Y, Liaubet L. Sex and fetal genome influence gene expression in pig endometrium at the end of gestation. BMC Genomics 2024; 25:303. [PMID: 38515025 PMCID: PMC10958934 DOI: 10.1186/s12864-024-10144-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND A fine balance of feto-maternal resource allocation is required to support pregnancy, which depends on interactions between maternal and fetal genetic potential, maternal nutrition and environment, endometrial and placental functions. In particular, some imprinted genes have a role in regulating maternal-fetal nutrient exchange, but few have been documented in the endometrium. The aim of this study is to describe the expression of 42 genes, with parental expression, in the endometrium comparing two extreme breeds: Large White (LW); Meishan (MS) with contrasting neonatal mortality and maturity at two days of gestation (D90-D110). We investigated their potential contribution to fetal maturation exploring genes-fetal phenotypes relationships. Last, we hypothesized that the fetal genome and sex influence their endometrial expression. For this purpose, pure and reciprocally crossbred fetuses were produced using LW and MS breeds. Thus, in the same uterus, endometrial samples were associated with its purebred or crossbred fetuses. RESULTS Among the 22 differentially expressed genes (DEGs), 14 DEGs were differentially regulated between the two days of gestation. More gestational changes were described in LW (11 DEGs) than in MS (2 DEGs). Nine DEGs were differentially regulated between the two extreme breeds, highlighting differences in the regulation of endometrial angiogenesis, nutrient transport and energy metabolism. We identified DEGs that showed high correlations with indicators of fetal maturation, such as ponderal index at D90 and fetal blood fructose level and placental weight at D110. We pointed out for the first time the influence of fetal sex and genome on endometrial expression at D90, highlighting AMPD3, CITED1 and H19 genes. We demonstrated that fetal sex affects the expression of five imprinted genes in LW endometrium. Fetal genome influenced the expression of four genes in LW endometrium but not in MS endometrium. Interestingly, both fetal sex and fetal genome interact to influence endometrial gene expression. CONCLUSIONS These data provide evidence for some sexual dimorphism in the pregnant endometrium and for the contribution of the fetal genome to feto-maternal interactions at the end of gestation. They suggest that the paternal genome may contribute significantly to piglet survival, especially in crossbreeding production systems.
Collapse
Affiliation(s)
- Agnes Bonnet
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, 31326, Castanet Tolosan, France.
| | - Lisa Bluy
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, 31326, Castanet Tolosan, France
| | - Laure Gress
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, 31326, Castanet Tolosan, France
| | - Laurianne Canario
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, 31326, Castanet Tolosan, France
| | - Laure Ravon
- GenESI, INRAE, Le Magneraud, 17700, Surgères, France
| | - Aurelie Sécula
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, 31326, Castanet Tolosan, France
- Present Address: IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | - Yvon Billon
- GenESI, INRAE, Le Magneraud, 17700, Surgères, France
| | - Laurence Liaubet
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, 31326, Castanet Tolosan, France
| |
Collapse
|
23
|
Fang H, Li Q, Wang H, Ren Y, Zhang L, Yang L. Maternal nutrient metabolism in the liver during pregnancy. Front Endocrinol (Lausanne) 2024; 15:1295677. [PMID: 38572473 PMCID: PMC10987773 DOI: 10.3389/fendo.2024.1295677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024] Open
Abstract
The liver plays pivotal roles in nutrient metabolism, and correct hepatic adaptations are required in maternal nutrient metabolism during pregnancy. In this review, hepatic nutrient metabolism, including glucose metabolism, lipid and cholesterol metabolism, and protein and amino acid metabolism, is first addressed. In addition, recent progress on maternal hepatic adaptations in nutrient metabolism during pregnancy is discussed. Finally, the factors that regulate hepatic nutrient metabolism during pregnancy are highlighted, and the factors include follicle-stimulating hormone, estrogen, progesterone, insulin-like growth factor 1, prostaglandins fibroblast growth factor 21, serotonin, growth hormone, adrenocorticotropic hormone, prolactin, thyroid stimulating hormone, melatonin, adrenal hormone, leptin, glucagon-like peptide-1, insulin glucagon and thyroid hormone. Our vision is that more attention should be paid to liver nutrient metabolism during pregnancy, which will be helpful for utilizing nutrient appropriately and efficiently, and avoiding liver diseases during pregnancy.
Collapse
Affiliation(s)
- Hongxu Fang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Qingyang Li
- College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Haichao Wang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ying Ren
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Leying Zhang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ling Yang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
24
|
Saadat N, Pallas B, Ciarelli J, Vyas AK, Padmanabhan V. Gestational testosterone excess early to mid-pregnancy disrupts maternal lipid homeostasis and activates biosynthesis of phosphoinositides and phosphatidylethanolamines in sheep. Sci Rep 2024; 14:6230. [PMID: 38486090 PMCID: PMC10940674 DOI: 10.1038/s41598-024-56886-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024] Open
Abstract
Gestational hyperandrogenism is a risk factor for adverse maternal and offspring outcomes with effects likely mediated in part via disruptions in maternal lipid homeostasis. Using a translationally relevant sheep model of gestational testosterone (T) excess that manifests maternal hyperinsulinemia, intrauterine growth restriction (IUGR), and adverse offspring cardiometabolic outcomes, we tested if gestational T excess disrupts maternal lipidome. Dimensionality reduction models following shotgun lipidomics of gestational day 127.1 ± 5.3 (term 147 days) plasma revealed clear differences between control and T-treated sheep. Lipid signatures of gestational T-treated sheep included higher phosphoinositides (PI 36:2, 39:4) and lower acylcarnitines (CAR 16:0, 18:0, 18:1), phosphatidylcholines (PC 38:4, 40:5) and fatty acids (linoleic, arachidonic, Oleic). Gestational T excess activated phosphatidylethanolamines (PE) and PI biosynthesis. The reduction in key fatty acids may underlie IUGR and activated PI for the maternal hyperinsulinemia evidenced in this model. Maternal circulatory lipids contributing to adverse cardiometabolic outcomes are modifiable by dietary interventions.
Collapse
Affiliation(s)
- Nadia Saadat
- Department of Pediatrics, 7510 MSRB, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI, 148019-5718, USA
| | - Brooke Pallas
- Unit Lab Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Joseph Ciarelli
- Department of Pediatrics, 7510 MSRB, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI, 148019-5718, USA
| | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University St. Louis, St. Louis, MO, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, 7510 MSRB, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI, 148019-5718, USA.
| |
Collapse
|
25
|
Nakano H, Nakano A. The role of metabolism in cardiac development. Curr Top Dev Biol 2024; 156:201-243. [PMID: 38556424 DOI: 10.1016/bs.ctdb.2024.01.005] [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] [Indexed: 04/02/2024]
Abstract
Metabolism is the fundamental process that sustains life. The heart, in particular, is an organ of high energy demand, and its energy substrates have been studied for more than a century. In recent years, there has been a growing interest in understanding the role of metabolism in the early differentiation of pluripotent stem cells and in cancer research. Studies have revealed that metabolic intermediates from glycolysis and the tricarboxylic acid cycle act as co-factors for intracellular signal transduction, playing crucial roles in regulating cell behaviors. Mitochondria, as the central hub of metabolism, are also under intensive investigation regarding the regulation of their dynamics. The metabolic environment of the fetus is intricately linked to the maternal metabolic status, and the impact of the mother's nutrition and metabolic health on fetal development is significant. For instance, it is well known that maternal diabetes increases the risk of cardiac and nervous system malformations in the fetus. Another notable example is the decrease in the risk of neural tube defects when pregnant women are supplemented with folic acid. These examples highlight the profound influence of the maternal metabolic environment on the fetal organ development program. Therefore, gaining insights into the metabolic environment within developing fetal organs is critical for deepening our understanding of normal organ development. This review aims to summarize recent findings that build upon the historical recognition of the environmental and metabolic factors involved in the developing embryo.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States; Cardiology Division, Department of Medicine, UCLA, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States; Molecular Biology Institute, UCLA, Los Angeles, CA, United States; Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
26
|
Dong Z, Liu H, Wan D, Wu X, Yin Y. Ferrous-sucrose complex supplementation regulates maternal plasma metabolism and the fecal microbiota composition and improves neonatal immunity and placental glucose transportation by activating the EGF/PI3K/AKT signaling pathways in sows. Food Funct 2024; 15:906-916. [PMID: 38168829 DOI: 10.1039/d3fo03733a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Pregnancy is a dynamic state involving rapid physiological changes in metabolism, affecting the health and development of the offspring. During pregnancy, the placenta constitutes a physical and immunological barrier to provide fetal nutrition through the maternal blood and prevent the exposure of the fetus to dangerous signals. Metabolic changes in the plasma, the fecal microbiota profile, and functional regulation in the placenta were studied in sows supplied with a ferrous-sucrose complex (FeSuc) from late gestation to parturition. The results revealed that maternal FeSuc supplementation enhanced arginine and proline metabolism, glutathione metabolism, with increased glutamic acid, beta-D-glucosamine, L-proline, 1-butylamine, and succinic acid and reduced sphingosine and chenodeoxycholic acid sulfate levels in the plasma. Moreover, significantly increased abundances of Christensenellaceae_R-7_group, Prevotellaceae_NK3B31_group, and Lachnospiraceae_NK4B4_group were detected in the feces of sows from the FeSuc group (P < 0.05). Spearman's correlation analysis indicated that Prevotellaceae_NK3B31_group abundances were positively correlated with glutamic acid, indoxyl sulfate, acetyl-DL-leucine, and beta-D-glucosamine, while Christensenellaceae_R-7_group was positively correlated with beta-D-glucosamine. Furthermore, maternal FeSuc supplementation significantly increased neonatal glucose (P < 0.01) and iron (P < 0.01) in the neonatal serum, significantly increased IL-10 and TGF-β1 levels in the neonatal liver (P < 0.01) and jejunum (P < 0.05), promoted the transcription of immune molecules in the placenta, and significantly increased the protein expressions of EGF (P < 0.05), PI3K (P < 0.01), p-PI3K (P < 0.001), p-AKT (P < 0.01), and glucose transporter 1 (GLUT1) (P < 0.001) in the placenta. The current study demonstrated that FeSuc supplementation regulated maternal metabolism processes by altering the fecal microbial composition and improved neonatal immunity and placental glucose transportation by activating the EGF/PI3K/AKT signaling pathways in sows.
Collapse
Affiliation(s)
- Zhenglin Dong
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, China.
| | - Hongwei Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, China.
| | - Dan Wan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, China.
| | - Xin Wu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, China.
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, China.
| |
Collapse
|
27
|
Yan H, Liang X, Luo H, Tang X, Xiao X. Association between gut microbiota, microbial network, and immunity in pregnancy with a focus on specific bacterial clusters. Front Microbiol 2023; 14:1314257. [PMID: 38156011 PMCID: PMC10753819 DOI: 10.3389/fmicb.2023.1314257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Background The community characteristics of the gut microbiota are not well defined and are not as widely studied as the functions of individual bacteria. This study aims to investigate the community composition of intestinal flora in women of childbearing age by conducting cluster analysis of gut microbiota and analyzing the relationship between different clusters and immune status. Methods A total of 45 women of childbearing age were recruited in the study, including 15 non-pregnant women and 30 women in late pregnancy, and stool samples were collected twice during the third trimester, specifically at 32 weeks and at full term. The gut microbiota data was analyzed using 16S rRNA amplicon sequencing. Partitioning Around Medoids algorithm was employed to assess microbial clustering patterns. Microbial network for each cluster was performed and plasm cytokines were measured to analyze the relationship between specific genera and immune state in clusters. Results There were three distinct clusters of intestinal community composition in women of childbearing age. Cluster 1 (PAM_1) was characterized by a high abundance of Bacteroides, while cluster 2 (PAM_2) showed higher levels of Bifidobacterium and Blautia, along with a significantly increased Firmicutes to Bacteroidota ratio. Cluster 3 (PAM_3) displayed a high abundance of Escherichia-shigella. PAM_1 was the most dominant cluster in non-pregnant women, and this dominant cluster was also one of the main in late pregnancy. At full term, the majority of subjects retained the same cluster as at 32 weeks, while a few experienced a shift. The microbial correlation networks differed across the three clusters, with PAM_1 exhibiting higher modularity and fewer connections. Analysis of the correlation between genera and plasma cytokines showed significant differences in their associations with cytokines between pregnancy and nonpregnancy within the same cluster, and the same genera had different effects in different clusters. Conclusion Women of childbearing age exhibit three distribution patterns of gut microbiota, and the intestinal clusters reshaped during late pregnancy in a small population. Different clusters may have diverse immunomodulatory effects in different physiological states. When studying the gut microbiome during pregnancy, it is crucial to consider the cluster differences within healthy women.
Collapse
Affiliation(s)
- Hao Yan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xinyuan Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Obstetrics, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Huijuan Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaomei Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
28
|
Zhu M, Sun R, Jin L, Yu D, Huang X, Zhu T, Gong Y, Chen Y, Shi J, Wang Q, Lu C, Wang D. Metabolomics profiling of maternal and umbilical cord blood in normoglycemia macrosomia. J Matern Fetal Neonatal Med 2023; 36:2270761. [PMID: 37848386 DOI: 10.1080/14767058.2023.2270761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023]
Abstract
Background: Macrosomia is a common disorder that occurs during pregnancy. We investigated the comprehensive metabolite profiles of pregnant maternal and fetal sera in normoglycemic macrosomia in a Chinese population. Methods: Forty pregnant women and their fetuses were included in the study (twenty macrosomia patients and twenty normal-weight controls). Maternal and umbilical cord serum metabolites were identified using ultra-performance liquid chromatography coupled with tandem mass spectrometry. Results: In total, 203 metabolites were identified. Lipids and lipid-like molecules were the predominant metabolites. Fifty-three metabolites with significant differences were obtained in the maternal samples. In the macrosomia group, the levels of docosahexaenoic acid, eicosapentaenoic acid, and arachidonic acid were significantly higher than those in the control group. Umbilical cord serum samples were obtained for 24 different metabolites. The maternal-fetal gradient of polyunsaturated fatty acids was decreased in the macrosomia group. Aconitic acid, citric acid, isocitric acid, 2-methylhexanoic acid, and 12-hydroxystearic acid were the common differential metabolites in the maternal and umbilical cord serum samples. Conclusion: There were obvious metabolic abnormalities in the sera of pregnant women and fetuses with macrosomia. Lipids and lipid-like molecules were the predominant differential metabolites but had different classifications in the maternal and umbilical cord serum. These results may provide new insights into the long-term metabolic disorders associated with macrosomia.
Collapse
Affiliation(s)
- Mianmian Zhu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Rongyue Sun
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Lixu Jin
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Dandan Yu
- Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Xiaoxia Huang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Ting Zhu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Yujing Gong
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Yuan Chen
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Jiamin Shi
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Qiu Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Chaosheng Lu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| | - Dan Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China
| |
Collapse
|
29
|
Song Y, Lu R, Yu G, Rahman ML, Chen L, Zhu Y, Tsai MY, Fiehn O, Chen Z, Zhang C. Longitudinal lipidomic profiles during pregnancy and associations with neonatal anthropometry: findings from a multiracial cohort. EBioMedicine 2023; 98:104881. [PMID: 38006745 PMCID: PMC10709105 DOI: 10.1016/j.ebiom.2023.104881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Maternal lipidomic profiling offers promise for characterizing lipid metabolites during pregnancy, but longitudinal data are limited. This study aimed to examine associations of longitudinal lipidomic profiles during pregnancy with multiple neonatal anthropometry using data from a multiracial cohort. METHODS We measured untargeted plasma lipidome profiles among 321 pregnant women from the NICHD Fetal Growth Study-Singletons using plasma samples collected longitudinally during four study visits at gestational weeks (GW) 10-14, 15-26, 23-31, and 33-39, respectively. We evaluated individual lipidomic metabolites at each study visit in association with neonatal anthropometry. We also evaluated the associations longitudinally by constructing lipid networks using weighted correlation network analysis and common networks using consensus network analysis across four visits using linear mixed-effects models with the adjustment of false discover rate. FINDINGS Multiple triglycerides (TG) were positively associated with birth weight (BW), BW Z-score, length and head circumference, while some cholesteryl ester (CE), phosphatidylcholine (PC), sphingomyelines (SM), phosphatidylethanolamines (PE), and lysophosphatidylcholines (LPC 20:3) families were inversely associated with BW, length, abdominal and head circumference at different GWs. Longitudinal trajectories of TG, PC, and glucosylcermides (GlcCer) were associated with BW, and CE (18:2) with BW z-score, length, and sum of skinfolds (SS), while some PC and PE were significantly associated with abdominal and head circumference. Modules of TG at GW 10-14 and 15-26 mainly were associated with BW. At GW 33-39, two networks of LPC (20:3) and of PC, TG, and CE, showed inverse associations with abdominal circumference. Distinct trajectories within two consensus modules with changes in TG, CE, PC, and LPC showed significant differences in BW and length. INTERPRETATION The results demonstrated that longitudinal changes of TGs during early- and mid-pregnancy and changes of PC, LPC, and CE during late-pregnancy were significantly associated with neonatal anthropometry. FUNDING National Institute of Child Health and Human Development intramural funding.
Collapse
Affiliation(s)
- Yiqing Song
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Ruijin Lu
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Guoqi Yu
- Global Center for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mohammad L Rahman
- National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Liwei Chen
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Yeiyi Zhu
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, 451 Health Sciences Drive, Davis, CA, USA
| | - Zhen Chen
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Cuilin Zhang
- Global Center for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
30
|
Wu Y, Wang Y, Zhang W, Peng J, Qin L, Zhang L, Chen R, Gu W, Sun Q, Liu C, Li R. Gestational exposure to ambient fine particulate matter disrupts maternal hepatic lipid metabolism. CHEMOSPHERE 2023; 344:140369. [PMID: 37802477 DOI: 10.1016/j.chemosphere.2023.140369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Pregnancy is a unique physiological stage for females as well as a vulnerable period for pollutant exposure. The effect of gestational ambient fine particulate matter (PM2.5) exposure on maternal lipid metabolism during pregnancy is rarely observed, and the mechanism is unknown. In the current study, pregnant C57BL/6 mice were randomly assigned to either ambient PM2.5 or filtered air exposure chambers since gestational day (GD) 0. Meanwhile, non-pregnant female mice were housed as controls in each exposure chamber. PM2.5 exposure exerted no significant effect on body weight gain or the body composition during pregnancy. Pregnant mice exposed to PM2.5 demonstrated improved glucose tolerance, whereas non-pregnant mice showed an increased fasting blood glucose level after PM2.5 exposure with no alterations in glucose tolerance. PM2.5 exposure exerted no significant effect on total lipid content in serum during pregnancy, while an increased serum total lipid level was found in non-pregnant mice exposed to PM2.5. PM2.5 exposure had no effect on total liver lipid levels, it increased several triacylglycerol (TAG) species and total cholesterol esters (CEs) in pregnant mice but lowered a considerable amount in non-pregnant mice' livers. Furthermore, gestational exposure to PM2.5 enhanced the expression of key enzymes in fatty acid uptake, de novo lipid synthesis, and β oxidation, and inhibited molecules for lipid export in mice liver. Conversely, PM2.5 exposure upregulated proteins involved in hepatic lipolysis and lipid export in non-pregnant mice. These results suggest that the interference of PM2.5 exposure during pregnancy on the lipid metabolism, particularly the hepatic lipid metabolism, differs from that during non-pregnancy. This study provides toxicological evidence that PM2.5 exposure during pregnancy disrupts the lipid metabolism of the liver and provides a basis for protecting vulnerable populations.
Collapse
Affiliation(s)
- Yunlu Wu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yirun Wang
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenhui Zhang
- Department of Environmental and Occupational Health, Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Jing Peng
- Zhuantang Community Healthcare Center, Hangzhou, Zhejiang, China
| | - Li Qin
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhang
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rucheng Chen
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weijia Gu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qinghua Sun
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ran Li
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
31
|
Cai L, Hyun SH, Kim E. Stem cell factor's role in enhancing the quality of fertilized and cloned porcine embryos for improved embryonic stem cell derivation. Front Vet Sci 2023; 10:1285530. [PMID: 38033636 PMCID: PMC10687439 DOI: 10.3389/fvets.2023.1285530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Stem cell factor (SCF), a cytokine growth factor, is expressed in various tissues of the male and female reproductive organs, including the testis, ovary, and endometrium. Its primary function involves cell survival, differentiation, and proliferation, achieved through its binding to the c-kit receptor. This study aimed to scrutinize the effects of SCF treatment during in vitro culture (IVC) on both the developmental potential and the efficiency of establishing embryonic stem cells (ESCs) from fertilized and cloned porcine embryos. The rates of cleavage and blastocyst formation exhibited no significant differences between fertilized and cloned embryos, even with the addition of SCF. However, it's worth noting that embryos cloned with Cloud eGFP as donor cells demonstrated notably increased rates of hatched blastocysts when treated with SCF, and this increase was statistically significant (p < 0.05). Furthermore, following the complete dissection of the blastocysts, although there was no significant difference in the SCF-treated group, the area of expansion was significantly reduced (p < 0.01) in the group treated with the antagonistic blocker (ACK2) compared to both the control and SCF-treated groups. These outcomes suggest that the SCF/c-kit signaling pathway might play a pivotal role in embryo implantation. As anticipated, the efficiency of deriving ESCs was significantly higher (p < 0.01) in the group subjected to SCF treatment (12.82 ± 1.02%) compared to the control group (5.41 ± 2.25%). In conclusion, this study highlights the crucial role of SCF in enhancing the quality of porcine embryos, a vital step in obtaining high-quality ESCs.
Collapse
Affiliation(s)
- Lian Cai
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Eunhye Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
32
|
Blanco Sequeiros E, Tuomaala AK, Tabassum R, Bergman PH, Koivusalo SB, Huvinen E. Early ascending growth is associated with maternal lipoprotein profile during mid and late pregnancy and in cord blood. Int J Obes (Lond) 2023; 47:1081-1087. [PMID: 37592059 PMCID: PMC10599999 DOI: 10.1038/s41366-023-01361-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
INTRODUCTION Intrauterine conditions and accelerating early growth are associated with childhood obesity. It is unknown, whether fetal programming affects the early growth and could alterations in the maternal-fetal metabolome be the mediating mechanism. Therefore, we aimed to assess the associations between maternal and cord blood metabolite profile and offspring early growth. METHODS The RADIEL study recruited 724 women at high risk for gestational diabetes mellitus (GDM) BMI ≥ 30 kg/m2 and/or prior GDM) before or in early pregnancy. Blood samples were collected once in each trimester, and from cord. Metabolomics were analyzed by targeted nuclear magnetic resonance (NMR) technique. Following up on offsprings' first 2 years growth, we discovered 3 distinct growth profiles (ascending n = 80, intermediate n = 346, and descending n = 146) by using latent class mixed models (lcmm). RESULTS From the cohort of mother-child dyads with available growth profile data (n = 572), we have metabolomic data from 232 mothers from 1st trimester, 271 from 2nd trimester, 277 from 3rd trimester and 345 from cord blood. We have data on 220 metabolites in each trimester and 70 from cord blood. In each trimester of pregnancy, the mothers of the ascending group showed higher levels of VLDL and LDL particles, and lower levels of HDL particles (p < 0.05). When adjusted for gestational age, birth weight, sex, delivery mode, and maternal smoking, there was an association with ascending profile and 2nd trimester total cholesterol in HDL2, 3rd trimester total cholesterol in HDL2 and in HDL, VLDL size and ratio of triglycerides to phosphoglycerides (TG/PG ratio) in cord blood (p ≤ 0.002). CONCLUSION Ascending early growth was associated with lower maternal total cholesterol in HDL in 2nd and 3rd trimester, and higher VLDL size and more adverse TG/PG ratio in cord blood. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, http://www. CLINICALTRIALS com , NCT01698385.
Collapse
Affiliation(s)
- Elina Blanco Sequeiros
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Soite Children's Hospital, Kokkola, Finland.
| | - Anna-Kaisa Tuomaala
- Department of Pediatrics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Rubina Tabassum
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Paula H Bergman
- Biostatistics Consulting, Department of Public Health, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Saila B Koivusalo
- Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, Turku, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Emilia Huvinen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
33
|
Martinez-Garza U, Choi J, Scafidi S, Wolfgang MJ. Proteomics identifies the developmental regulation of HKDC1 in liver of pigs and mice. Am J Physiol Regul Integr Comp Physiol 2023; 325:R389-R400. [PMID: 37545422 PMCID: PMC10639021 DOI: 10.1152/ajpregu.00253.2022] [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: 10/13/2022] [Revised: 06/01/2023] [Accepted: 07/18/2023] [Indexed: 08/08/2023]
Abstract
During the perinatal period, unique metabolic adaptations support energetic requirements for rapid growth. To gain insight into perinatal adaptations, quantitative proteomics was performed comparing the livers of Yorkshire pigs at postnatal day 7 and adult. These data revealed differences in the metabolic control of liver function including significant changes in lipid and carbohydrate metabolic pathways. Newborn livers showed an enrichment of proteins in lipid catabolism and gluconeogenesis concomitant with elevated liver carnitine and acylcarnitines levels. Sugar kinases were some of the most dramatically differentially enriched proteins compared with neonatal and adult pigs including galactokinase 1 (Galk1), ketohexokinase (KHK), hexokinase 1 (HK1), and hexokinase 4 (GCK). Interestingly, hexokinase domain containing 1 (HKDC1), a newly identified fifth hexokinase associated with glucose disturbances in pregnant women, was highly enriched in the liver during the prenatal and perinatal periods and continuously declined throughout postnatal development in pigs and mice. These changes were confirmed via Western blot and mRNA expression. These data provide new insights into the developmental and metabolic adaptations in the liver during the transition from the perinatal period to adulthood in multiple mammalian species.
Collapse
Affiliation(s)
- Ursula Martinez-Garza
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Joseph Choi
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Susana Scafidi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
34
|
de Lima Castro M, Dos Passos RR, Justina VD, do Amaral WN, Giachini FR. Physiological and pathological evidence of O-GlcNAcylation regulation during pregnancy related process. Placenta 2023; 141:43-50. [PMID: 37210277 DOI: 10.1016/j.placenta.2023.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/22/2023] [Accepted: 04/25/2023] [Indexed: 05/22/2023]
Abstract
O-GlcNAcylation is a dynamic and reversible post-translational modification (PTM) controlled by the enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Changes in its expression lead to a breakdown in cellular homeostasis, which is linked to several pathological processes. Placentation and embryonic development are periods of high cell activity, and imbalances in cell signaling pathways can result in infertility, miscarriage, or pregnancy complications. O-GlcNAcylation is involved in cellular processes such as genome maintenance, epigenetic regulation, protein synthesis/degradation, metabolic pathways, signaling pathways, apoptosis, and stress response. Trophoblastic differentiation/invasion and placental vasculogenesis, as well as zygote viability and embryonic neuronal development, are all dependent on O-GlcNAcylation. This PTM is required for pluripotency, which is a required condition for embryonic development. Further, this pathway is a nutritional sensor and cell stress marker, which is primarily measured by the OGT enzyme and its product, protein O-GlcNAcylation. Yet, this post-translational modification is enrolled in metabolic and cardiovascular adaptations during pregnancy. Finally, evidence of how O-GlcNAc impacts pregnancy during pathological conditions such as hyperglycemia, gestational diabetes, hypertension, and stress disorders are reviewed. Considering this scenario, progress in understanding the role of O- GlcNAcylation in pregnancy is required.
Collapse
Affiliation(s)
- Marta de Lima Castro
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Vanessa Dela Justina
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Waldemar Naves do Amaral
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Fernanda Regina Giachini
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil.
| |
Collapse
|
35
|
Lis-Kuberka J, Pupek M, Orczyk-Pawiłowicz M. The Mother-Child Dyad Adipokine Pattern: A Review of Current Knowledge. Nutrients 2023; 15:4059. [PMID: 37764842 PMCID: PMC10535905 DOI: 10.3390/nu15184059] [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: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
An important role in the network of interconnections between the mother and child is played by adipokines, which are adipose tissue hormones engaged in the regulation of metabolism. Alternations of maternal adipokines translate to the worsening of maternal insulin resistance as well as metabolic stress, altered placenta functions, and fetal development, which finally contribute to long-term metabolic unfavorable conditions. This paper is the first to summarize the current state of knowledge concerning the concentrations of individual adipokines in different biological fluids of maternal and cord plasma, newborn/infant plasma, milk, and the placenta, where it highlights the impact of adverse perinatal risk factors, including gestational diabetes mellitus, preeclampsia, intrauterine growth restriction, preterm delivery, and maternal obesity on the adipokine patterns in maternal-infant dyads. The importance of adipokine measurement and relationships in biological fluids during pregnancy and lactation is crucial for public health in the area of prevention of most diet-related metabolic diseases. The review highlights the huge knowledge gap in the field of hormones participating in the energy homeostasis and metabolic pathways during perinatal and postnatal periods in the mother-child dyad. An in-depth characterization is needed to confirm if the adverse outcomes of early developmental programming might be modulated via maternal lifestyle intervention.
Collapse
Affiliation(s)
- Jolanta Lis-Kuberka
- Department of Biochemistry and Immunochemistry, Division of Chemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| | | | - Magdalena Orczyk-Pawiłowicz
- Department of Biochemistry and Immunochemistry, Division of Chemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| |
Collapse
|
36
|
Kyrkou C, Fotakis C, Dimitropoulou A, Tsakoumaki F, Zoumpoulakis P, Menexes G, Biliaderis CG, Athanasiadis AP, Michaelidou AM. Maternal Dietary Protein Patterns and Neonatal Anthropometrics: A Prospective Study with Insights from NMR Metabolomics in Amniotic Fluid. Metabolites 2023; 13:977. [PMID: 37755257 PMCID: PMC10535439 DOI: 10.3390/metabo13090977] [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: 08/01/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
This study aimed to characterize dietary protein patterns (DPPs) in a sample pool of 298 well-nourished pregnant women and explore potential associations between DPPs and neonatal anthropometrics. Maternal dietary data were collected using a validated food frequency questionnaire. Neonatal anthropometrics were abstracted from health booklets. A hierarchical cluster analysis identified three DPPs: "Dairy-focused", "Med-fusion", and "Traditional-inspired". The "Dairy-focused" DPP exhibited the highest protein intake (p < 0.001), predominantly animal protein (p < 0.001), while the "Traditional-inspired" DPP presented higher plant protein (p < 0.001) and fiber intakes (p < 0.001), and, therefore, a reduced carbohydrate-to-fiber quotient (p < 0.001). The "Med-fusion" DPP had the lowest protein-to-fat ratio (p < 0.001). Infants of women following the "Dairy-focused" DPP had the highest birth height centiles (p = 0.007) and the lowest ponderal index (p = 0.003). The NMR-metabolomics approach was implemented on a subset of women that provided amniotic fluid (AF) specimens (n = 62) to elucidate distinct metabolic signatures associated with DPPs. PCA and OPLS-DA models verified the adherence to three DPPs, revealing that the levels of several amino acids (AAs) were the highest in "Dairy-focused", reflecting its protein-rich nature. The "Traditional-inspired" DPP showed decreased AAs and glucose levels. This knowledge may contribute to optimizing maternal dietary recommendations. Further research is needed to validate these findings and better understand the relationships between maternal diet, AF metabolic signature, and neonatal anthropometrics.
Collapse
Affiliation(s)
- Charikleia Kyrkou
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (C.K.); (A.D.); (F.T.); (C.G.B.)
| | - Charalambos Fotakis
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece; (C.F.); (P.Z.)
| | - Aristea Dimitropoulou
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (C.K.); (A.D.); (F.T.); (C.G.B.)
| | - Foteini Tsakoumaki
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (C.K.); (A.D.); (F.T.); (C.G.B.)
| | - Panagiotis Zoumpoulakis
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece; (C.F.); (P.Z.)
- Department of Food Science and Technology, University of West Attica, Ag. Spyridonos, 122 43 Egaleo, Greece
| | - Georgios Menexes
- Department of Field Crops and Ecology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Costas G. Biliaderis
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (C.K.); (A.D.); (F.T.); (C.G.B.)
| | - Apostolos P. Athanasiadis
- 3rd Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Alexandra-Maria Michaelidou
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (C.K.); (A.D.); (F.T.); (C.G.B.)
| |
Collapse
|
37
|
Li LJ, Du R, Ouidir M, Lu R, Chen Z, Weir NL, Tsai MY, Albert PS, Zhang C. Early Pregnancy Maternal Plasma Phospholipid Saturated Fatty Acids and Fetal Growth: Findings from a Multi-Racial/Ethnic Birth Cohort in US. Nutrients 2023; 15:3287. [PMID: 37571228 PMCID: PMC10420908 DOI: 10.3390/nu15153287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Saturated fatty acids (SFAs) during pregnancy are associated with disrupted metabolic programming among offspring at birth and later growth. We examined plasma phospholipid SFAs in early pregnancy and fetal growth throughout pregnancy. We enrolled 321 pregnant women from the NICHD Fetal Growth Studies-Singleton Cohort at gestational weeks 8-13. Ultrasonogram schedules were randomly assigned to capture weekly fetal growth. We measured plasma phospholipid SFAs at early pregnancy using blood samples and modeled fetal growth trajectories across tertiles of SFAs with cubic splines using linear mixed models after full adjustment. We then compared pairwise weekly fetal growth biometrics referencing the lowest tertile in each SFA using the Wald test. We found that even-chain and very long even-chain SFAs were inversely associated, whereas odd-chain SFAs were positively associated with fetal weight and size. Compared with the lowest tertile, the highest tertile of pentadecanoic acid (15:0) had a greater fetal weight and size, starting from week 13 until late pregnancy (at week 39: 3429.89 vs. 3269.08 g for estimated fetal weight; 328.14 vs. 323.00 mm for head circumference). Our findings could inspire future interventions using an alternative high-fat diet rich in odd-chain SFAs for optimal fetal growth.
Collapse
Affiliation(s)
- Ling-Jun Li
- Department of O&G, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore;
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- NUS Bia-Echo Asia Centre for Reproductive Longevity and Equality (ARCLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- Human Potential Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
| | - Ruochen Du
- Biostatics Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore;
| | - Marion Ouidir
- Institute for Advanced Biosciences, Grenoble Aples University, Site Santé, Allée des Alpes, 38700 La Tronche, France;
| | - Ruijin Lu
- Division of Biostatistics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA;
| | - Zhen Chen
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Natalie L. Weir
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (N.L.W.); (M.Y.T.)
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (N.L.W.); (M.Y.T.)
| | - Paul S. Albert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Cuilin Zhang
- Department of O&G, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore;
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- NUS Bia-Echo Asia Centre for Reproductive Longevity and Equality (ARCLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- Human Potential Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
| |
Collapse
|
38
|
Gkini V, Namba T. Glutaminolysis and the Control of Neural Progenitors in Neocortical Development and Evolution. Neuroscientist 2023; 29:177-189. [PMID: 35057642 PMCID: PMC10018057 DOI: 10.1177/10738584211069060] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple types of neural progenitor cells (NPCs) contribute to the development of the neocortex, a brain region responsible for our higher cognitive abilities. Proliferative capacity of NPCs varies among NPC types, developmental stages, and species. The higher proliferative capacity of NPCs in the developing human neocortex is thought to be a major contributing factor why humans have the most expanded neocortex within primates. Recent studies have shed light on the importance of cell metabolism in the neocortical NPC proliferative capacity. Specifically, glutaminolysis, a metabolic pathway that converts glutamine to glutamate and then to α-ketoglutarate, has been shown to play a critical role in human NPCs, both in apical and basal progenitors. In this review, we summarize our current knowledge of NPC metabolism, focusing especially on glutaminolysis, and discuss the role of NPC metabolism in neocortical development, evolution, and neurodevelopmental disorders, providing a broader perspective on a newly emerging research field.
Collapse
Affiliation(s)
- Vasiliki Gkini
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
- Takashi Namba, Neuroscience Center, HiLIFE
— Helsinki Institute of Life Science, University of Helsinki, PO 63,
Haartmaninkatu 8, Helsinki 00014, Finland.
| |
Collapse
|
39
|
Thangaraj SV, Kachman M, Halloran KM, Sinclair KD, Lea R, Bellingham M, Evans NP, Padmanabhan V. Developmental programming: Preconceptional and gestational exposure of sheep to a real-life environmental chemical mixture alters maternal metabolome in a fetal sex-specific manner. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 864:161054. [PMID: 36565874 PMCID: PMC10322214 DOI: 10.1016/j.scitotenv.2022.161054] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 05/21/2023]
Abstract
BACKGROUND Everyday, humans are exposed to a mixture of environmental chemicals some of which have endocrine and/or metabolism disrupting actions which may contribute to non-communicable diseases. The adverse health impacts of real-world chemical exposure, characterized by chronic low doses of a mixture of chemicals, are only recently emerging. Biosolids derived from human waste represent the environmental chemical mixtures humans are exposed to in real life. Prior studies in sheep have shown aberrant reproductive and metabolic phenotypes in offspring after maternal biosolids exposure. OBJECTIVE To determine if exposure to biosolids perturbs the maternal metabolic milieu of pregnant ewes, in a fetal sex-specific manner. METHODS Ewes were grazed on inorganic fertilizer (Control) or biosolids-treated pastures (BTP) from before mating and throughout gestation. Plasma from pregnant ewes (Control n = 15, BTP n = 15) obtained mid-gestation were analyzed by untargeted metabolomics. Metabolites were identified using Agilent MassHunter. Multivariate analyses were done using MetaboAnalyst 5.0 and confirmed using SIMCA. RESULTS Univariate and multivariate analysis of 2301 annotated metabolites identified 193 differentially abundant metabolites (DM) between control and BTP sheep. The DM primarily belonged to the super-class of lipids and organic acids. 15-HeTrE, oleamide, methionine, CAR(3:0(OH)) and pyroglutamic acid were the top DM and have been implicated in the regulation of fetal growth and development. Fetal sex further exacerbated differences in metabolite profiles in the BTP group. The organic acids class of metabolites was abundant in animals with male fetuses. Prenol lipid, sphingolipid, glycerolipid, alkaloid, polyketide and benzenoid classes showed fetal sex-specific responses to biosolids. DISCUSSION Our study illustrates that exposure to biosolids significantly alters the maternal metabolome in a fetal sex-specific manner. The altered metabolite profile indicates perturbations to fatty acid, arginine, branched chain amino acid and one‑carbon metabolism. These factors are consistent with, and likely contribute to, the adverse phenotypic outcomes reported in the offspring.
Collapse
Affiliation(s)
- S V Thangaraj
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - M Kachman
- MM BRCF Metabolomics Core, University of Michigan, Ann Arbor, MI, USA
| | - K M Halloran
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - K D Sinclair
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - R Lea
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - M Bellingham
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - N P Evans
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - V Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
40
|
[Hematologic and nutritional states are associated with the development of preeclampsia in a Mexican population]. NUTR HOSP 2023; 40:78-87. [PMID: 36602139 DOI: 10.20960/nh.04110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction Introduction: nutrient and trace element deficiency are risk factors for the development of preeclampsia; these nutrients induce changes in the hematologic state, which can be used to prevent complications during pregnancy. Objective: this research will analyze the nutritional and hematological status during the 3 trimesters of gestation in pregnant women with preeclampsia to evaluate its association with gestational alterations. Method: a cross-sectional, descriptive, and analytic study was conducted on 78 pregnant women who attended a prenatal control clinic, 11 of whom were diagnosed with preeclampsia. The hematological parameters were determined through hematometry. To get the nutritional intake profile of the pregnant women, a frequency of food consumption questionnaire was used. Results: the normotensive group´s arterial pressure (PA) was significantly lower than that of pregnant women with preeclampsia. The hematological profile changed during pregnancy, and platelet value was much higher in preeclamptic women. The nutritional status of pregnant women in the third trimester consumed more macronutrients and micronutrients than in the first and second trimesters, and this pattern was observed in pregnant women with preeclampsia. When compared to pregnant women with preeclampsia, normotensive women received more minerals and vitamins. Conclusion: hematological values are modified during each trimester of gestation, and pregnant women with preeclampsia had high platelet values compared to normotensive women. Our data show a greater intake of minerals and vitamins in normotensive pregnant women as compared to those who developed preeclampsia; therefore, it is recommended that pregnant women be provided with nutritional guidance in order to prevent pregnant complications.
Collapse
|
41
|
Gonzalez Fernandez J, Moncayo Arlandi J, Ochando A, Simon C, Vilella F. The role of extracellular vesicles in intercellular communication in human reproduction. Clin Sci (Lond) 2023; 137:281-301. [PMID: 36762584 DOI: 10.1042/cs20220793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/19/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Embryo-maternal cross-talk has emerged as a vitally important process for embryo development and implantation, which is driven by secreted factors and extracellular vesicles (EVs). The EV cargo of bioactive molecules significantly influences target cells and primes them for critical stages of reproductive biology, including embryo development, adhesion, and implantation. Recent research has suggested that EVs and their cargo represent a powerful non-invasive tool that can be leveraged to assess embryo and maternal tissue quality during assisted reproduction treatments. Here, we review the current scientific literature regarding the intercellular cross-talk between embryos and maternal tissues from fertilization to implantation, focusing on human biology and signaling mechanisms identified in animal models.
Collapse
Affiliation(s)
- Javier Gonzalez Fernandez
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Javier Moncayo Arlandi
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Ana Ochando
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Carlos Simon
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Felipe Vilella
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| |
Collapse
|
42
|
Kong J, Li S, Li Y, Chen M. Effects of Salvia miltiorrhiza active compounds on placenta-mediated pregnancy complications. Front Cell Dev Biol 2023; 11:1034455. [PMID: 36711034 PMCID: PMC9880055 DOI: 10.3389/fcell.2023.1034455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Placenta-mediated pregnancy complications (PMPCs), including preeclampsia (PE), fetal growth restriction (FGR), and recurrent spontaneous abortion (RSA), occur in approximately 5% of pregnancies and are caused by abnormal placenta development. The development of effective therapies for PMPCs is still challenging due to the complicated pathogenesis, such as disrupted vascular homeostasis and subsequent abnormal placentation. Synthetic drugs have been recommended for treating PMPCs; however, they tend to cause adverse reactions in the mother and fetus. Salvia miltiorrhiza (S. miltiorrhiza) has potential effects on PMPCs owing to its advantages in treating cardiovascular disorders. S. miltiorrhiza and its active compounds could attenuate the symptoms of PMPCs through anticoagulation, vasodilation, antioxidation, and endothelial protection. Thus, in this review, we summarize the literature and provide comprehensive insights on S. miltiorrhiza and its phytochemical constituents, pharmacological activities, and on PMPCs, which would be valuable to explore promising drugs.
Collapse
Affiliation(s)
- Jingyin Kong
- Department of Prenatal Diagnosis and Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Songjun Li
- Department of Reproduction Medical Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yingting Li
- Department of Prenatal Diagnosis and Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Chen
- Department of Prenatal Diagnosis and Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,*Correspondence: Min Chen,
| |
Collapse
|
43
|
Kannampuzha S, Ravichandran M, Mukherjee AG, Wanjari UR, Renu K, Vellingiri B, Iyer M, Dey A, George A, Gopalakrishnan AV. The mechanism of action of non-coding RNAs in placental disorders. Biomed Pharmacother 2022; 156:113964. [DOI: 10.1016/j.biopha.2022.113964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
|
44
|
Shao X, Yu W, Yang Y, Wang F, Yu X, Wu H, Ma Y, Cao B, Wang YL. The mystery of the life tree: the placenta. Biol Reprod 2022; 107:301-316. [PMID: 35552600 DOI: 10.1093/biolre/ioac095] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/20/2022] [Accepted: 05/21/2022] [Indexed: 11/13/2022] Open
Abstract
The placenta is the interface between the fetal and maternal environments during mammalian gestation, critically safeguarding the health of the developing fetus and the mother. Placental trophoblasts origin from embryonic trophectoderm that differentiates into various trophoblastic subtypes through villous and extravillous pathways. The trophoblasts actively interact with multiple decidual cells and immune cells at the maternal-fetal interface and thus construct fundamental functional units, which are responsible for blood perfusion, maternal-fetal material exchange, placental endocrine, immune tolerance, and adequate defense barrier against pathogen infection. Various pregnant complications are tightly associated with the defects in placental development and function maintenance. In this review, we summarize the current views and our recent progress on the mechanisms underlying the formation of placental functional units, the interactions among trophoblasts and various uterine cells, as well as the placental barrier against pathogen infections during pregnancy. The involvement of placental dysregulation in adverse pregnancy outcomes is discussed.
Collapse
Affiliation(s)
- Xuan Shao
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Wenzhe Yu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yun Yang
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Feiyang Wang
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Xin Yu
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Hongyu Wu
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Yeling Ma
- Medical College, Shaoxing University, Shaoxing, China
| | - Bin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
45
|
Conflict and the evolution of viviparity in vertebrates. Behav Ecol Sociobiol 2022. [DOI: 10.1007/s00265-022-03171-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
46
|
Umbilical cord blood metabolomics: association with intrauterine hyperglycemia. Pediatr Res 2022; 91:1530-1535. [PMID: 33980991 DOI: 10.1038/s41390-021-01516-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/11/2021] [Accepted: 03/20/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Intrauterine hyperglycemia can harm a fetus's growth and development, and this can be seen in the umbilical cord blood metabolism disorder. However, the metabolites and metabolic mechanisms involved in the condition remain unknown. METHODS Targeted metabolomics using liquid chromatography and MetaboAnalyst were conducted in this study to explore differences in metabolites and metabolic pathways between individuals with hyperglycemia or well-controlled gestational diabetes mellitus (GDM) and healthy controls. RESULTS Univariate analysis found that the hyperglycemic and healthy control groups differed in 30 metabolites, while the well-controlled GDM and the healthy control groups differed only in three metabolites-ursodeoxycholic acid, docosahexaenoic acid, and 8,11,14-eicosatrienoic acid. Most of these metabolic variations were negatively associated with neonatal weights. Further research showed that the variations in the metabolites were primarily associated with the metabolic pathways of linoleic acid (LA) and alpha-linolenic acid (ALA). CONCLUSION Gestational hyperglycemia and well-controlled GDM, which may play a major role by inhibiting the LA and ALA metabolic pathways, have detrimental effects on cord blood metabolism. IMPACT The main point of this paper is that intrauterine hyperglycemia has a negative effect on cord blood metabolism mainly through the linoleic acid and alpha-linolenic acid metabolic pathways. This is a study to report a new association between well-controlled GDM and cord blood metabolism. This study provides a possible explanation for the association between intrauterine hyperglycemia and neonatal adverse birth outcomes.
Collapse
|
47
|
Zhang L, Wang Z, Wu H, Gao Y, Zheng J, Zhang J. Maternal High-Fat Diet Impairs Placental Fatty Acid β-Oxidation and Metabolic Homeostasis in the Offspring. Front Nutr 2022; 9:849684. [PMID: 35495939 PMCID: PMC9050107 DOI: 10.3389/fnut.2022.849684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/22/2022] [Indexed: 12/30/2022] Open
Abstract
Maternal overnutrition can affect fetal growth and development, thus increasing susceptibility to obesity and diabetes in later life of the offspring. Placenta is the central organ connecting the developing fetus with the maternal environment. It is indicated placental fatty acid metabolism plays an essential role in affecting the outcome of the pregnancy and fetus. However, the role of placental fatty acid β-oxidation (FAO) in maternal overnutrition affecting glucose metabolism in the offspring remains unclear. In this study, C57BL/6J female mice were fed with normal chow or high-fat diet before and during pregnancy and lactation. The placenta and fetal liver were collected at gestation day 18.5, and the offspring's liver was collected at weaning. FAO-related genes and AMP-activated protein kinase (AMPK) signaling pathway were examined both in the placenta and in the human JEG-3 trophoblast cells. FAO-related genes were further examined in the liver of the fetuses and in the offspring at weaning. We found that dams fed with high-fat diet showed higher fasting blood glucose, impaired glucose tolerance at gestation day 14.5 and higher serum total cholesterol (T-CHO) at gestation day 18.5. The placental weight and lipid deposition were significantly increased in maternal high-fat diet group. At weaning, the offspring mice of high-fat diet group exhibited higher body weight, impaired glucose tolerance, insulin resistance and increased serum T-CHO, compared with control group. We further found that maternal high-fat diet downregulated mRNA and protein expressions of carnitine palmitoyltransferase 2 (CPT2), a key enzyme in FAO, by suppressing the AMPK/Sirt1/PGC1α signaling pathway in the placenta. In JEG-3 cells, protein expressions of CPT2 and CPT1b were both downregulated by suppressing the AMPK/Sirt1/PGC1α signaling pathway under glucolipotoxic condition, but were later restored by the AMPK agonist 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR). However, there was no difference in CPT2 and CPT1 gene expression in the liver of fetuses and offspring at weaning age. In conclusion, maternal high-fat diet can impair gene expression involved in FAO in the placenta by downregulating the AMPK signaling pathway, and can cause glucose and lipid dysfunction of offspring at weaning, indicating that placental FAO may play a crucial role in regulating maternal overnutrition and metabolic health in the offspring.
Collapse
|
48
|
Effects of Low Protein-High Carbohydrate Diet during Early and Late Pregnancy on Respiratory Quotient and Visceral Adiposity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3878581. [PMID: 35432727 PMCID: PMC9010209 DOI: 10.1155/2022/3878581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 12/05/2022]
Abstract
Background Low Protein-High Carbohydrate (LPHC) diet during pregnancy is considered a nutritional and health problem related to the development of maternal metabolic alterations, such as fatty liver and obesity in the perinatal and postnatal period. It is known that increase in visceral adiposity tissue (VAT) modulates maternal metabolic rate, with the respiratory quotient (RQ) being a parameter related to that variable; however, it is unknown whether LPHC intake during pregnancy affects the VAT and the RQ. In this study, we examine if consumption of LPHC during pregnancy modifies the VAT and RQ in early and late periods of pregnancy. Methods This is a longitudinal and cross-sectional study with Wistar rats during gestation (G) (3, 8, 15, and 20) and nonpregnant rats. Rats were fed with a control diet with 63/18% carbohydrate/protein and an experimental diet with 79/6% carbohydrate/protein. We studied water and food consumption and metabolic parameters such as RQ and energy expenditure (EE), calculated by indirect calorimetry. In the cross-sectional study, we determined visceral fat, as well as the concentration of free fatty acids, insulin, glucose, and lipid profile in serum. Results Nonpregnant rats with LPHC intake decreased significantly in VAT (86%) and the RQ (18%); in pregnant rats in early (8G) and late pregnancy (15G) in LPHC diet, both parameters (VAT and RQ) (25%-92%) increased during light time. When comparing time points during pregnancy in the control and LPHC groups, the RQ increased in 15G during daytime compared to 8G during the night period (17 and 5%, respectively). In late pregnancy, LPHC intake and triacylglyceride levels increased and cholesterol and glucose decreased (45 and 26%, respectively), in comparison to nonpregnant rats. Conclusions LPHC intake in nonpregnant rats decreases the RQ and VAT. Interestingly, the opposite occurs in early pregnancy: the RQ and VAT increased, and this correlates with free fatty acid (FFA) levels. The increase in RQ and VAT during light time in early pregnancy increased mobilization of carbohydrate and protein metabolism. These results suggest that LPHC intake during pregnancy increases the glucose metabolism as a compensatory mechanism for energy needs in the fetus and the mother in early pregnancy.
Collapse
|
49
|
Placental Development and Pregnancy-Associated Diseases. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
50
|
The metabolic landscape of decidua in recurrent pregnancy loss using a global metabolomics approach. Placenta 2021; 112:45-53. [PMID: 34273713 DOI: 10.1016/j.placenta.2021.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Maternal metabolism undergoes dynamic changes during pregnancy. A deviation from this physiological metabolic status might be involved in the pathogenesis of pregnancy complications, such as recurrent pregnancy loss (RPL). Decidua is an important uterine tissue, which provides immunological protection as well as nutritional support to the developing embryo during early pregnancy. Previous studies have shown that aberrant metabolism of the decidua is related to the etiology of RPL. However, the metabolic profile of the decidua in RPL has not yet been fully elucidated. METHODS In the current study, decidual samples from RPL patients (n = 23) and normal pregnancy (NP) women (n = 30) were collected, and hydrophilic and hydrophobic metabolites were extracted and measured using a liquid chromatography electrospray ionization tandem mass spectrometry system. Besides, the mRNA expression of several critical enzymes involved in sphingolipid metabolism and glycerophospholipid metabolism was detected. RESULTS The OSC-PLS-DA scores plot illustrates that metabolic differences are present in the decidual tissue of RPL patients compared with that of NP women. Combining multivariate analysis with univariate statistical analysis, a total of 62 metabolites related to RPL were selected, including carnitine, glycerophospholipids, sphingomyelin (SM), ceramide, organic acids and their derivatives, and amino acid metabolomics. KEGG analysis showed that abnormalities in multiple metabolic pathways occurred in RPL decidua, including vitamin digestion and absorption, tryptophan metabolism, citrate cycle, arginine biosynthesis, glycerophospholipid metabolism, sphingolipid metabolism, and sphingolipid signaling pathway. Increased SM synthase and decreased Phospholipase A2 Group IIE mRNA levels were detected in RPL compared with NP group. DISCUSSION Disruption of decidual metabolism, especially glycerophospholipid metabolism and sphingolipid metabolism, might be involved in the occurrence of RPL.
Collapse
|