1
|
Valverde-Pérez E, Olea E, Rocher A, Aaronson PI, Prieto-Lloret J. Effects of gestational intermittent hypoxia on the respiratory system: A tale of the placenta, fetus, and developing offspring. J Sleep Res 2024:e14435. [PMID: 39675784 DOI: 10.1111/jsr.14435] [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: 09/13/2024] [Revised: 11/12/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024]
Abstract
Obstructive sleep apnea (OSA) is a common sleep disorder that is associated with a wide variety of health conditions, including cardiovascular, cerebrovascular, metabolic, neoplastic, and neurocognitive manifestations. OSA, as a chronic condition, is mainly characterised by repeated upper airway obstructions during sleep that cause episodes of intermittent hypoxia (IH), resulting in tissue hypoxia-reoxygenation cycles. Decreased arterial oxygen pressure (PaO2) and haemoglobin saturation (SatO2) stimulate reflex responses to overcome the obstruction. The prevalence of OSA is significant worldwide, and an underrated problem when focussing on women during pregnancy. The physiological changes associated with pregnancy, especially during its latest stages, are related to a higher prevalence of OSA events in pregnant mothers, and associated with an increased risk of hypertension, pre-eclampsia and diabetes, among other deleterious consequences. Furthermore, OSA during pregnancy can interfere with normal fetal development and is associated with growth retardation, preterm birth, or low birth weight. Carotid body overstimulation and hypoxia-reoxygenation episodes contribute to cardiovascular disease and oxidative stress, which can harm both mother and fetus and have long-lasting effects that can reach into adulthood. Because IH is the hallmark of OSA, this review examines the literature available about the impact of gestational intermittent hypoxia (GIH) on the respiratory system at maternal, fetal, and offspring levels. Offering the latest scientific data about OSA during pregnancy, we may help to tackle this condition with lifestyle changes and therapeutic approaches, that could influence the mothers, but also impact adult health problems, mostly unknown, inherited from these hypoxic episodes in the uterus.
Collapse
Affiliation(s)
- Esther Valverde-Pérez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM). Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Elena Olea
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM). Universidad de Valladolid-CSIC, Valladolid, Spain
- Departamento de Enfermería, Facultad de Enfermería, Universidad de Valladolid, Valladolid, Spain
| | - Asunción Rocher
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM). Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Philip I Aaronson
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jesús Prieto-Lloret
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM). Universidad de Valladolid-CSIC, Valladolid, Spain
| |
Collapse
|
2
|
Li S, Zhou L, Ren J, Zhang Q, Xiao X. Maternal exercise programs placental miR-495-5p-mediated Snx7 expression and kynurenic acid metabolic pathway induced by prenatal high-fat diet: Based on miRNA-seq, transcriptomics, and metabolomics. J Nutr Biochem 2024; 137:109830. [PMID: 39647668 DOI: 10.1016/j.jnutbio.2024.109830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Poor intrauterine environments increase the prevalence of chronic metabolic diseases in offspring, whereas maternal exercise is an effective measure to break this vicious intergenerational cycle. Placenta is increasingly being studied to explore its role in maternal-fetal metabolic cross-talk. The association between placental miRNA and offspring development trajectories has been established, yet the specific role and mechanism thereof in maternal exercise-induced metabolic protection remain elusive. Here, C57BL/6 female mice were subjected to either a normal control or a high-fat diet (HFD), half of the HFD-fed dams were housed with voluntary wheel running for 3 weeks before and during gestation. At embryonic day 18.5, we sacrificed parturient mice and then conducted miRNA-seq, transcriptomic, and metabolomic profiling of the placenta. Our data revealed that maternal HFD resulted in significant alterations in both miRNA and gene expressions, as well as metabolic pathways of the placenta, whereas prenatal exercise negated these perturbations. The common differentially expressed transcripts among three groups were enriched in multiple critical pathways involving energy expenditure, signal transduction, and fetal development. Through integrated analysis of multiomics data, we speculated that maternal exercise reversed the suppression of miR-495-5p induced by HFD, thereby inhibiting miR-495-5p-targeted Snx7 and modulating kynurenic acid production. These datasets provided novel mechanistic insight into how maternal exercise positively affects the metabolic homeostasis of offspring. The discovered important miRNAs, mRNAs, and metabolites could be promising predictive and therapeutic targets for protecting offspring metabolic health.
Collapse
Affiliation(s)
- Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
3
|
Ansar M, Ali MA, Ali N, Haider Z, Latif A, Tazeen A, Fatima Z, Anjum MN. Ultrasound shear wave elastography of the placenta: a potential tool for early detection of fetal growth restriction. Clin Imaging 2024; 116:110329. [PMID: 39461252 DOI: 10.1016/j.clinimag.2024.110329] [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: 06/29/2024] [Revised: 09/17/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Sonographic placental elastography has recently been employed as a non-invasive tool to investigate the structural alterations associated with various conditions such as pre-eclampsia, gestational diabetes and fetal growth restriction (FGR). The study was conducted based on the hypothesis that the placental elasticity might differ with varying severity of FGR and with that of appropriate for gestational age (AGA) pregnancies. METHODS This study involved 121 pregnant women, with 54 in the normal group and 67 in the FGR group, which was defined as the fetal weight below the 10th percentile for gestational age. The FGR pregnancies were sub grouped into different stages based on the presence and extent of Doppler abnormalities. Shear-wave elastography was carried out to investigate the placental elasticity values, which were compared using the Kruskal-Wallis test. A P value of ≤0.05 was considered significant. RESULTS The placental elasticity differed significantly between pregnancies with and without FGR and among the different stages of FGR. There was a significant difference in PE (kPa) and SWV (m/s) among groups, with a p-value of 0.000001. PE and SWV in FGR pregnancies were significantly higher compared to AGA as a whole using t-test with p values of <0.0001. Doppler indices of umbilical, uterine and fetal middle cerebral arteries also correlated significantly with these. CONCLUSION The study suggests that placental elasticity values reflect structural alterations associated with FGR and could serve as a valuable tool in the early detection and staging of this condition.
Collapse
Affiliation(s)
- Muhammad Ansar
- University Institute of Radiological Sciences and Medical Imaging Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | | | - Noraiz Ali
- University Institute of Radiological Sciences and Medical Imaging Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Zeshan Haider
- University Institute of Radiological Sciences and Medical Imaging Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Aruj Latif
- University Institute of Radiological Sciences and Medical Imaging Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Anjum Tazeen
- University Institute of Radiological Sciences and Medical Imaging Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Zareen Fatima
- University Institute of Radiological Sciences and Medical Imaging Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan.
| | - Muhammad Nawaz Anjum
- University Institute of Radiological Sciences and Medical Imaging Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| |
Collapse
|
4
|
Hourtovenko C, Sreetharan S, Tharmalingam S, Tai TC. Impact of Ionizing Radiation Exposure on Placental Function and Implications for Fetal Programming. Int J Mol Sci 2024; 25:9862. [PMID: 39337351 PMCID: PMC11432287 DOI: 10.3390/ijms25189862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/19/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Accidental exposure to high-dose radiation while pregnant has shown significant negative effects on the developing fetus. One fetal organ which has been studied is the placenta. The placenta performs all essential functions for fetal development, including nutrition, respiration, waste excretion, endocrine communication, and immunological functions. Improper placental development can lead to complications during pregnancy, as well as the occurrence of intrauterine growth-restricted (IUGR) offspring. IUGR is one of the leading indicators of fetal programming, classified as an improper uterine environment leading to the predisposition of diseases within the offspring. With numerous studies examining fetal programming, there remains a significant gap in understanding the placenta's role in irradiation-induced fetal programming. This review aims to synthesize current knowledge on how irradiation affects placental function to guide future research directions. This review provides a comprehensive overview of placental biology, including its development, structure, and function, and summarizes the placenta's role in fetal programming, with a focus on the impact of radiation on placental biology. Taken together, this review demonstrates that fetal radiation exposure causes placental degradation and immune function dysregulation. Given the placenta's crucial role in fetal development, understanding its impact on irradiation-induced IUGR is essential.
Collapse
Affiliation(s)
- Cameron Hourtovenko
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- School of Natural Sciences, Laurentian University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
| | - Shayen Sreetharan
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- Department of Medical Imaging, London Health Sciences Centre, 339 Windermere Rd., London, ON N6A 5A5, Canada
| | - Sujeenthar Tharmalingam
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- School of Natural Sciences, Laurentian University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
| | - T C Tai
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- School of Natural Sciences, Laurentian University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
5
|
Bai J, Wang Y, Deng S, Yang Y, Chen S, Wu Z. Microplastics caused embryonic growth retardation and placental dysfunction in pregnant mice by activating GRP78/IRE1α/JNK axis induced apoptosis and endoplasmic reticulum stress. Part Fibre Toxicol 2024; 21:36. [PMID: 39261835 PMCID: PMC11389422 DOI: 10.1186/s12989-024-00595-5] [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: 12/12/2023] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Microplastics (MPs), a brand-new class of worldwide environmental pollutant, have received a lot of attention. MPs are consumed by both humans and animals through water, food chain and other ways, which may cause potential health risks. However, the effects of MPs on embryonic development, especially placental function, and its related mechanisms still need to be further studied. We investigated the impact on fetal development and placental physiological function of pregnant mice by consecutive gavages of MPs at 0, 25, 50, 100 mg/kg body weight during gestational days (GDs 0-14). The results showed that continuous exposure to high concentrations of MP significantly reduced daily weight gain and impaired reproductive performance of pregnant mice. In addition, MPs could significantly induce oxidative stress and placental dysfunction in pregnant mice. On the other hand, MPs exposure significantly decreased placental barrier function and induced placental inflammation. Specifically, MPs treatment significantly reduced the expression of tight junction proteins in placentas, accompanied by inflammatory cell infiltration and increased mRNA levels of pro-inflammatory cytokines and chemokines in placentas. Finally, we found that MPs induced placental apoptosis and endoplasmic reticulum (ER) stress through the GRP78/IRE1α/JNK axis, leading to placental dysfunction and decreased reproductive performance in pregnant mice. We revealed for the first time that the effects of MPs on placental dysfunction in pregnant animals. Blocking the targets of MPs mediated ER stress will provide potential therapeutic ideas for the toxic effects of MPs on maternal pregnancy.
Collapse
Affiliation(s)
- Jun Bai
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yuzeng Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing, China
| | - Siwei Deng
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing, China
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hom Hung, Kowloon, Hong Kong, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing, China.
| |
Collapse
|
6
|
Camm EJ, Wong G, Pan Y, Wang JZ, Goldstein JA, Arcot A, Murphy CN, Hansji H, Mangwiro YT, Saffery R, Wlodek ME, Wyrwoll CS, Gernand AD, Kaitu'u-Lino TJ. Assessment of an AI-based tool for population-wide collection of placental morphological data. Eur J Obstet Gynecol Reprod Biol 2024; 299:110-117. [PMID: 38852316 DOI: 10.1016/j.ejogrb.2024.05.043] [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: 12/06/2023] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
OBJECTIVES Automated placental assessment could allow accurate and timely morphological/pathological measurements at scale. We undertook a pilot study using an artificial intelligence-based assessment system (AI-PLAX) to ascertain the potential of a state-wide rollout as part of Generation Victoria, assessing the impact of time post-delivery, user, and technology used for image capture, on a range of derived placental data. STUDY DESIGN Ten placentas were imaged by three different users and imaging technologies (iPad, iPhone, Samsung) at (0 h), 24 h, and 48 h post-delivery. Using AI-PLAX, disc size (short and long length, perimeter, area), shape (normal, abnormal), cord insertion type (central, eccentric), cord coiling, abruption (retroplacental hematoma), and meconium staining were determined. RESULTS When analysing the maternal surface of the placenta, time in cold storage post-delivery had modest effects on placental dimensions, with decreases in the short length (24-48 h: -3.7 %), disc area (0-24 h: 4.7 % and 0-48 h: -7.4 %), and perimeter (0-48 h: -3.8 %) observed. There was marginal impact on placental dimensions when the placenta was imaged by different users, including long length (+1.9 %), disc area (+2.9 %), and perimeter (+2.0 %). Measures of placental size were not impacted by the type of technology used to capture the images. When analysing the fetal surface of the placenta, more variance in placental size measures were observed between users. Abruption detection was not affected by any parameter. Time between delivery and imaging impacted apparent meconium staining - likely reflecting changes in fetal surface colour over time. Meconium staining was not affected by technology or user. CONCLUSIONS This study supports the feasibility of the collection of placenta images for later morphological analysis by AI-PLAX, with measures obtained minimally influenced by time in cold storage, user imaging the placenta, or technology to capture the images.
Collapse
Affiliation(s)
- E J Camm
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
| | - G Wong
- Translational Obstetrics Group, Mercy Hospital for Women, Dept. of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, VIC, Australia
| | - Y Pan
- College of Information Sciences and Technology, The Pennsylvania State University, University Park, PA, USA
| | - J Z Wang
- College of Information Sciences and Technology, The Pennsylvania State University, University Park, PA, USA
| | - J A Goldstein
- Department of Pathology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - A Arcot
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - C N Murphy
- Translational Obstetrics Group, Mercy Hospital for Women, Dept. of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, VIC, Australia
| | - H Hansji
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Y T Mangwiro
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia
| | - R Saffery
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - M E Wlodek
- Translational Obstetrics Group, Mercy Hospital for Women, Dept. of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, VIC, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia
| | - C S Wyrwoll
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - A D Gernand
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - T J Kaitu'u-Lino
- Translational Obstetrics Group, Mercy Hospital for Women, Dept. of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
7
|
Pereira-Carvalho D, Chagas Valim AC, Borba Vieira Andrade C, Bloise E, Fontes Dias A, Muller Oliveira Nascimento V, Silva Alves RK, Dos Santos RC, Lopes Brum F, Gomes Medeiros I, Antunes Coelho SV, Barros Arruda L, Regina Todeschini A, Barbosa Dias W, Ortiga-Carvalho TM. Sex-specific effect of antenatal Zika virus infection on murine fetal growth, placental nutrient transporters, and nutrient sensor signaling pathways. FASEB J 2024; 38:e23799. [PMID: 38979938 DOI: 10.1096/fj.202301951rr] [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: 09/22/2023] [Revised: 06/03/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
Maternal Zika virus (ZIKV) infection during pregnancy has been associated with severe intrauterine growth restriction (IUGR), placental damage, metabolism disturbances, and newborn neurological abnormalities. Here, we investigated the impact of maternal ZIKV infection on placental nutrient transporters and nutrient-sensitive pathways. Immunocompetent (C57BL/6) mice were injected with Low (103 PFU-ZIKVPE243) or High (5 × 107 PFU-ZIKVPE243) ZIKV titers at gestational day (GD) 12.5, and tissue was collected at GD18.5 (term). Fetal-placental growth was impaired in male fetuses, which exhibited higher placental expression of the ZIKV infective marker, eukaryotic translation initiation factor 2 (eIF2α), but lower levels of phospho-eIF2α. There were no differences in fetal-placental growth in female fetuses, which exhibited no significant alterations in placental ZIKV infective markers. Furthermore, ZIKV promoted increased expression of glucose transporter type 1 (Slc2a1/Glut1) and decreased levels of glucose-6-phosphate in female placentae, with no differences in amino acid transport potential. In contrast, ZIKV did not impact glucose transporters in male placentae but downregulated sodium-coupled neutral amino acid 2 (Snat2) transporter expression. We also observed sex-dependent differences in the hexosamine biosynthesis pathway (HBP) and O-GlcNAcylation in ZIKV-infected pregnancies, showing that ZIKV can disturb placental nutrient sensing. Our findings highlight molecular alterations in the placenta caused by maternal ZIKV infection, shedding light on nutrient transport, sensing, and availability. Our results also suggest that female and male placentae employ distinct coping mechanisms in response to ZIKV-induced metabolic changes, providing insights into therapeutic approaches for congenital Zika syndrome.
Collapse
Affiliation(s)
- Daniela Pereira-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Enrrico Bloise
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ariane Fontes Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Rakel Kelly Silva Alves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronan Christian Dos Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe Lopes Brum
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | | | | | - Luciana Barros Arruda
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriane Regina Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wagner Barbosa Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
8
|
Derisoud E, Jiang H, Zhao A, Chavatte-Palmer P, Deng Q. Revealing the molecular landscape of human placenta: a systematic review and meta-analysis of single-cell RNA sequencing studies. Hum Reprod Update 2024; 30:410-441. [PMID: 38478759 PMCID: PMC11215163 DOI: 10.1093/humupd/dmae006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 02/12/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND With increasing significance of developmental programming effects associated with placental dysfunction, more investigations are devoted to improving the characterization and understanding of placental signatures in health and disease. The placenta is a transitory but dynamic organ adapting to the shifting demands of fetal development and available resources of the maternal supply throughout pregnancy. Trophoblasts (cytotrophoblasts, syncytiotrophoblasts, and extravillous trophoblasts) are placental-specific cell types responsible for the main placental exchanges and adaptations. Transcriptomic studies with single-cell resolution have led to advances in understanding the placenta's role in health and disease. These studies, however, often show discrepancies in characterization of the different placental cell types. OBJECTIVE AND RATIONALE We aim to review the knowledge regarding placental structure and function gained from the use of single-cell RNA sequencing (scRNAseq), followed by comparing cell-type-specific genes, highlighting their similarities and differences. Moreover, we intend to identify consensus marker genes for the various trophoblast cell types across studies. Finally, we will discuss the contributions and potential applications of scRNAseq in studying pregnancy-related diseases. SEARCH METHODS We conducted a comprehensive systematic literature review to identify different cell types and their functions at the human maternal-fetal interface, focusing on all original scRNAseq studies on placentas published before March 2023 and published reviews (total of 28 studies identified) using PubMed search. Our approach involved curating cell types and subtypes that had previously been defined using scRNAseq and comparing the genes used as markers or identified as potential new markers. Next, we reanalyzed expression matrices from the six available scRNAseq raw datasets with cell annotations (four from first trimester and two at term), using Wilcoxon rank-sum tests to compare gene expression among studies and annotate trophoblast cell markers in both first trimester and term placentas. Furthermore, we integrated scRNAseq raw data available from 18 healthy first trimester and nine term placentas, and performed clustering and differential gene expression analysis. We further compared markers obtained with the analysis of annotated and raw datasets with the literature to obtain a common signature gene list for major placental cell types. OUTCOMES Variations in the sampling site, gestational age, fetal sex, and subsequent sequencing and analysis methods were observed between the studies. Although their proportions varied, the three trophoblast types were consistently identified across all scRNAseq studies, unlike other non-trophoblast cell types. Notably, no marker genes were shared by all studies for any of the investigated cell types. Moreover, most of the newly defined markers in one study were not observed in other studies. These discrepancies were confirmed by our analysis on trophoblast cell types, where hundreds of potential marker genes were identified in each study but with little overlap across studies. From 35 461 and 23 378 cells of high quality in the first trimester and term placentas, respectively, we obtained major placental cell types, including perivascular cells that previously had not been identified in the first trimester. Importantly, our meta-analysis provides marker genes for major placental cell types based on our extensive curation. WIDER IMPLICATIONS This review and meta-analysis emphasizes the need for establishing a consensus for annotating placental cell types from scRNAseq data. The marker genes identified here can be deployed for defining human placental cell types, thereby facilitating and improving the reproducibility of trophoblast cell annotation.
Collapse
Affiliation(s)
- Emilie Derisoud
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Hong Jiang
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Allan Zhao
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Pascale Chavatte-Palmer
- INRAE, BREED, Université Paris-Saclay, UVSQ, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden
| |
Collapse
|
9
|
Louwen F, Kreis NN, Ritter A, Yuan J. Maternal obesity and placental function: impaired maternal-fetal axis. Arch Gynecol Obstet 2024; 309:2279-2288. [PMID: 38494514 PMCID: PMC11147848 DOI: 10.1007/s00404-024-07462-w] [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: 11/07/2023] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
The prevalence of maternal obesity rapidly increases, which represents a major public health concern worldwide. Maternal obesity is characteristic by metabolic dysfunction and chronic inflammation. It is associated with health problems in both mother and offspring. Increasing evidence indicates that the placenta is an axis connecting maternal obesity with poor outcomes in the offspring. In this brief review, we have summarized the current data regarding deregulated placental function in maternal obesity. The data show that maternal obesity induces numerous placental defects, including lipid and glucose metabolism, stress response, inflammation, immune regulation and epigenetics. These placental defects affect each other and result in a stressful intrauterine environment, which transduces and mediates the adverse effects of maternal obesity to the fetus. Further investigations are required to explore the exact molecular alterations in the placenta in maternal obesity, which may pave the way to develop specific interventions for preventing epigenetic and metabolic programming in the fetus.
Collapse
Affiliation(s)
- Frank Louwen
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany
| | - Nina-Naomi Kreis
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany
| | - Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany
| | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany.
| |
Collapse
|
10
|
De Silva DA, Anderson EA, Kim J, Ting Lee ML, Thoma ME. The Association Between Prenatal Food Insecurity and Breastfeeding Initiation and Exclusive Breastfeeding Duration: A Longitudinal Study Using Oregon PRAMS and PRAMS-2, 2008-2015. Breastfeed Med 2024; 19:368-377. [PMID: 38506260 DOI: 10.1089/bfm.2023.0126] [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] [Indexed: 03/21/2024]
Abstract
Background: In the United States, 11.1% of households experience food insecurity; however, pregnant women are disproportionately affected. Maternal food insecurity may affect infant feeding practices, for example, through being a source of chronic stress that may alter the decision to initiate and continue breastfeeding. Thus, we sought to determine whether prenatal food insecurity was associated with breastfeeding (versus not) and exclusive breastfeeding duration among Oregon women. Method: The Oregon Pregnancy Risk Assessment Monitoring System (PRAMS) data of live births from 2008 to 2015 and the Oregon PRAMS-2 follow-up survey were used (n = 3,624) in this study. Associations with breastfeeding initiation and duration were modeled with multivariable logistic regression and accelerated failure time (AFT), respectively. Models were adjusted for maternal sociodemographic and pre-pregnancy health characteristics. Results: Nearly 10% of women experienced prenatal food insecurity. For breastfeeding initiation, unadjusted models suggested non-significant decreased odds (odds ratio (OR) 0.88 [confidence intervals (CI): 0.39, 1.99]), whereas adjusted models revealed a non-significant increased odds (OR 1.41 [CI: 0.58, 3.47]). Unadjusted AFT models suggested that food-insecure mothers had a non-significant decrease in exclusive breastfeeding duration (OR 0.76 [CI: 0.50, 1.17]), but adjustment for covariates attenuated results (OR 0.89 [CI: 0.57, 1.39]). Conclusions: Findings suggest minimal differences in breastfeeding practices when exploring food security status in the prenatal period, though the persistence of food insecurity may affect exclusive breastfeeding duration. Lower breastfeeding initiation may be due to other explanatory factors correlated with food insecurity and breastfeeding, such as education and marital status.
Collapse
Affiliation(s)
- Dane A De Silva
- Department of Family Science, School of Public Health, University of Maryland, College Park, Maryland, USA
| | - Elaine A Anderson
- Department of Family Science, School of Public Health, University of Maryland, College Park, Maryland, USA
| | - Jinhee Kim
- Department of Family Science, School of Public Health, University of Maryland, College Park, Maryland, USA
| | - Mei-Ling Ting Lee
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, Maryland, USA
| | - Marie E Thoma
- Department of Family Science, School of Public Health, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
11
|
Ray GW, Zeng Q, Kusi P, Zhang H, Shao T, Yang T, Wei Y, Li M, Che X, Guo R. Genetic and inflammatory factors underlying gestational diabetes mellitus: a review. Front Endocrinol (Lausanne) 2024; 15:1399694. [PMID: 38694942 PMCID: PMC11061502 DOI: 10.3389/fendo.2024.1399694] [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: 03/28/2024] [Indexed: 05/04/2024] Open
Abstract
Gestational diabetes mellitus (GDM) poses a significant global health concern, impacting both maternal and fetal well-being. Early detection and treatment are imperative to mitigate adverse outcomes during pregnancy. This review delves into the pivotal role of insulin function and the influence of genetic variants, including SLC30A8, CDKAL1, TCF7L2, IRS1, and GCK, in GDM development. These genetic variations affect beta-cell function and insulin activity in crucial tissues, such as muscle, disrupting glucose regulation during pregnancy. We propose a hypothesis that this variation may disrupt zinc transport, consequently impairing insulin production and secretion, thereby contributing to GDM onset. Furthermore, we discussed the involvement of inflammatory pathways, such as TNF-alpha and IL-6, in predisposing individuals to GDM. Genetic modulation of these pathways may exacerbate glucose metabolism dysregulation observed in GDM patients. We also discussed how GDM affects cardiovascular disease (CVD) through a direct correlation between pregnancy and cardiometabolic function, increasing atherosclerosis, decreased vascular function, dyslipidemia, and hypertension in women with GDM history. However, further research is imperative to unravel the intricate interplay between inflammatory pathways, genetics, and GDM. This understanding is pivotal for devising targeted gene therapies and pharmacological interventions to rectify genetic variations in SLC30A8, CDKAL1, TCF7L2, IRS1, GCK, and other pertinent genes. Ultimately, this review offers insights into the pathophysiological mechanisms of GDM, providing a foundation for developing strategies to mitigate its impact.
Collapse
Affiliation(s)
- Gyan Watson Ray
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
| | - Qiaoli Zeng
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
| | - Phidelia Kusi
- University of Ghana, Ministry of Fisheries and Aquaculture Development, Fisheries Commission, Accra, Ghana
| | - Hengli Zhang
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
| | - Taotao Shao
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, China
| | - Taili Yang
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
| | - Yue Wei
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
| | - Mianqin Li
- Department of Obstetric, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Xiaoqun Che
- Department of Obstetric, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Reproductive Medicine Center, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Runmin Guo
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, China
| |
Collapse
|
12
|
Dong J, Xu Q, Qian C, Wang L, DiSciullo A, Lei J, Lei H, Yan S, Wang J, Jin N, Xiong Y, Zhang J, Burd I, Wang X. Fetal growth restriction exhibits various mTOR signaling in different regions of mouse placentas with altered lipid metabolism. Cell Biol Toxicol 2024; 40:15. [PMID: 38451382 PMCID: PMC10920423 DOI: 10.1007/s10565-024-09855-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Fetal growth restriction (FGR) is a common complication of pregnancy and can have significant impact on obstetric and neonatal outcomes. Increasing evidence has shown that the inhibited mechanistic target of rapamycin (mTOR) signaling in placenta is associated with FGR. However, interpretation of existing research is limited due to inconsistent methodologies and varying understanding of the mechanism by which mTOR activity contributes to FGR. Hereby, we have demonstrated that different anatomic regions of human and mouse placentas exhibited different levels of mTOR activity in normal compared to FGR pregnancies. When using the rapamycin-induced FGR mouse model, we found that placentas of FGR pregnancies exhibited abnormal morphological changes and reduced mTOR activity in the decidual-junctional layer. Using transcriptomics and lipidomics, we revealed that lipid and energy metabolism was significantly disrupted in the placentas of FGR mice. Finally, we demonstrated that maternal physical exercise during gestation in our FGR mouse model was associated with increased fetal and placental weight as well as increased placental mTOR activity and lipid metabolism. Collectively, our data indicate that the inhibited placental mTOR signaling contributes to FGR with altered lipid metabolism in mouse placentas, and maternal exercise could be an effective method to reduce the occurrence of FGR or alleviate the adverse outcomes associated with FGR.
Collapse
Affiliation(s)
- Jie Dong
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China.
| | - Qian Xu
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Chenxi Qian
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Lu Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Alison DiSciullo
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, 22 S. Greene Street, Suite P6H302, Baltimore, MD, 21201, USA
| | - Jun Lei
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, 22 S. Greene Street, Suite P6H302, Baltimore, MD, 21201, USA
| | - Hui Lei
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Song Yan
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Jingjing Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Ni Jin
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Yujing Xiong
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China
| | - Jianhua Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Irina Burd
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, 22 S. Greene Street, Suite P6H302, Baltimore, MD, 21201, USA.
| | - Xiaohong Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, NO. 569, Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi Province, China.
| |
Collapse
|
13
|
Liu J, Yang T, Li Y, Li S, Li Y, Xu S, Xia W. Associations of maternal exposure to 2,4-dichlorophenoxyacetic acid during early pregnancy with steroid hormones among one-month-old infants. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169414. [PMID: 38114038 DOI: 10.1016/j.scitotenv.2023.169414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Exposure to 2,4-dichlorophenoxyacetic acid (2,4-D), a widely used hormonal herbicide, may disrupt steroid hormone homeostasis. However, evidence from population-based studies is limited, especially for one-month-old infants whose steroid hormones are in a state of adjustment to extrauterine life and can be important indicators of endocrine development. This study aimed to explore the associations between maternal 2,4-D exposure during early pregnancy and infant steroid hormone levels. METHODS The 885 mother-infant pairs were from a birth cohort in Wuhan, China. Maternal exposure to 2,4-D was determined in urine samples from early pregnancy, and nine steroid hormones were determined in infant urine. The associations of maternal 2,4-D exposure with infant steroid hormones and their product-to-precursor ratios were estimated based on generalized linear models, and bioinformatic analysis was conducted with public databases to explore the potential mechanisms involved. RESULTS The detection frequency of 2,4-D was 99.32 %, and the detection frequency of steroid hormones ranged from 98.42 % to 100.00 %. After adjusting for covariates, an interquartile range increase in 2,4-D concentrations was associated with a 7.84 % decrease in 11-deoxycortisol (95 % confidence interval, CI: -14.12 %, -1.10 %), an 8.09 % decrease in corticosterone (95 % CI: -14.56 %, -1.14 %), an 8.67 % decrease in cortisol (95 % CI: -14.43 %, -2.52 %), a 13.00 % decrease in cortisone (95 % CI: -20.64 %, -4.62 %), and an 11.17 % decrease in aldosterone (95 % CI: -19.62 %, -1.83 %). Maternal 2,4-D was also associated with lower infant cortisol/17α-OH-progesterone, cortisol/pregnenolone, and aldosterone/pregnenolone ratios. In bioinformatic analysis, pathways/biological processes related to steroid hormone synthesis and secretion were enriched from target genes of 2,4-D exposure. CONCLUSIONS Maternal urinary 2,4-D during early pregnancy was associated with lower infant urinary 11-deoxycortisol, corticosterone, cortisol, cortisone, and aldosterone, reflecting that 2,4-D exposure may interfere with infant steroid hormone homeostasis. Further efforts are still needed to study the relevant health effects of exposure to 2,4-D, particularly for vulnerable populations.
Collapse
Affiliation(s)
- Jiangtao Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Yang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Shulan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Cifkova E, Karahoda R, Stranik J, Abad C, Kacerovsky M, Lisa M, Staud F. Metabolomic analysis of the human placenta reveals perturbations in amino acids, purine metabolites, and small organic acids in spontaneous preterm birth. EXCLI JOURNAL 2024; 23:264-282. [PMID: 38487084 PMCID: PMC10938235 DOI: 10.17179/excli2023-6785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/31/2024] [Indexed: 03/17/2024]
Abstract
Spontaneous preterm delivery presents one of the most complex challenges in obstetrics and is a leading cause of perinatal morbidity and mortality. Although it is a common endpoint for multiple pathological processes, the mechanisms governing the etiological complexity of spontaneous preterm birth and the placental responses are poorly understood. This study examined placental tissues collected between May 2019 and May 2022 from a well-defined cohort of women who experienced spontaneous preterm birth (n = 72) and healthy full-term deliveries (n = 30). Placental metabolomic profiling of polar metabolites was performed using Ultra-High Performance Liquid Chromatography/Mass Spectrometry (UHPLC/MS) analysis. The resulting data were analyzed using multi- and univariate statistical methods followed by unsupervised clustering. A comprehensive metabolomic evaluation of the placenta revealed that spontaneous preterm birth was associated with significant changes in the levels of 34 polar metabolites involved in intracellular energy metabolism and biochemical activity, including amino acids, purine metabolites, and small organic acids. We found that neither the preterm delivery phenotype nor the inflammatory response explain the reported differential placental metabolome. However, unsupervised clustering revealed two molecular subtypes of placentas from spontaneous preterm pregnancies exhibiting differential enrichment of clinical parameters. We also identified differences between early and late preterm samples, suggesting distinct placental functions in early spontaneous preterm delivery. Altogether, we present evidence that spontaneous preterm birth is associated with significant changes in the level of placental polar metabolites. Dysregulation of the placental metabolome may underpin important (patho)physiological mechanisms involved in preterm birth etiology and long-term neonatal outcomes.
Collapse
Affiliation(s)
- Eva Cifkova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Sokolska 581, 50005, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Sokolska 581, 50005, Hradec Kralove, Czech Republic
| | - Miroslav Lisa
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| |
Collapse
|
15
|
Fuenzalida B, Yañez MJ, Mueller M, Mistry HD, Leiva A, Albrecht C. Evidence for hypoxia-induced dysregulated cholesterol homeostasis in preeclampsia: Insights into the mechanisms from human placental cells and tissues. FASEB J 2024; 38:e23431. [PMID: 38265294 PMCID: PMC10953329 DOI: 10.1096/fj.202301708rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024]
Abstract
Preeclampsia (PE) poses a considerable risk to the long-term cardiovascular health of both mothers and their offspring due to a hypoxic environment in the placenta leading to reduced fetal oxygen supply. Cholesterol is vital for fetal development by influencing placental function. Recent findings suggest an association between hypoxia, disturbed cholesterol homeostasis, and PE. This study investigates the influence of hypoxia on placental cholesterol homeostasis. Using primary human trophoblast cells and placentae from women with PE, various aspects of cholesterol homeostasis were examined under hypoxic and hypoxia/reoxygenation (H/R) conditions. Under hypoxia and H/R, intracellular total and non-esterified cholesterol levels were significantly increased. This coincided with an upregulation of HMG-CoA-reductase and HMG-CoA-synthase (key genes regulating cholesterol biosynthesis), and a decrease in acetyl-CoA-acetyltransferase-1 (ACAT1), which mediates cholesterol esterification. Hypoxia and H/R also increased the intracellular levels of reactive oxygen species and elevated the expression of hypoxia-inducible factor (HIF)-2α and sterol-regulatory-element-binding-protein (SREBP) transcription factors. Additionally, exposure of trophoblasts to hypoxia and H/R resulted in enhanced cholesterol efflux to maternal and fetal serum. This was accompanied by an increased expression of proteins involved in cholesterol transport such as the scavenger receptor class B type I (SR-BI) and the ATP-binding cassette transporter G1 (ABCG1). Despite these metabolic alterations, mitogen-activated-protein-kinase (MAPK) signaling, a key regulator of cholesterol homeostasis, was largely unaffected. Our findings indicate dysregulation of cholesterol homeostasis at multiple metabolic points in both the trophoblast hypoxia model and placentae from women with PE. The increased cholesterol efflux and intracellular accumulation of non-esterified cholesterol may have critical implications for both the mother and the fetus during pregnancy, potentially contributing to an elevated cardiovascular risk later in life.
Collapse
Affiliation(s)
- Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
| | - Maria Jose Yañez
- School of Medical Technology, Faculty of Medicine and ScienceUniversidad San SebastiánSantiagoChile
| | - Martin Mueller
- Division of Gynecology and ObstetricsLindenhofgruppeBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Hiten D. Mistry
- Department of Women and Children's HealthSchool of Life Course and Population Health Sciences, King's College LondonLondonUK
| | - Andrea Leiva
- School of Medical Technology, Faculty of Medicine and ScienceUniversidad San SebastiánSantiagoChile
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
- Swiss National Center of Competence in Research, NCCR TransCureUniversity of BernBernSwitzerland
| |
Collapse
|
16
|
Meng Q, Del Rosario I, Sung K, Janzen C, Devaskar SU, Carpenter CL, Ritz B. Maternal dietary patterns and placental outcomes among pregnant women in Los Angeles. Placenta 2024; 145:72-79. [PMID: 38100961 PMCID: PMC11419549 DOI: 10.1016/j.placenta.2023.12.002] [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: 08/04/2023] [Revised: 11/11/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Epidemiological studies have linked prenatal maternal diet to fetal growth, but whether diet affects placental outcomes is poorly understood. METHODS We collected past month dietary intake from 148 women in mid-pregnancy enrolled at University of California Los Angeles (UCLA) antenatal clinics from 2016 to 2019. We employed the food frequency Diet History Questionnaire II and generated the Healthy Eating Index-2015 (HEI-2015), the Alternate Healthy Eating Index for Pregnancy (AHEI-P), and the Alternate Mediterranean Diet (aMED). We conducted T2-weighted magnetic resonance imaging (MRI) in mid-pregnancy (1st during 14-17 and 2nd during 19-24 gestational weeks) to evaluate placental volume (cm3) and we measured placenta weight (g) at delivery. We estimated change and 95 % confidence interval (CI) in placental volume and associations of placenta weight with all dietary index scores and diet items using linear regression models. RESULTS Placental volume in mid-pregnancy was associated with an 18.9 cm3 (95 % CI 5.1, 32.8) increase per 100 gestational days in women with a higher HEI-2015 (≥median), with stronger results for placentas of male fetuses. We estimated positive associations between placental volume at the 1st and 2nd MRI and higher intake of vegetables, high-fat fish, dairy, and dietary intake of B vitamins. A higher aMED (≥median) score was associated with a 40.5 g (95 % CI 8.5, 72.5) increase in placenta weight at delivery, which was mainly related to protein intake. DISCUSSION Placental growth represented by volume in mid-pregnancy and weight at birth is influenced by the quality and content of the maternal diet.
Collapse
Affiliation(s)
- Qi Meng
- Department of Epidemiology, University of California, Los Angeles, CA, 90095, USA
| | - Irish Del Rosario
- Department of Epidemiology, University of California, Los Angeles, CA, 90095, USA
| | - Kyunghyun Sung
- Department of Radiological Sciences, University of California, Los Angeles, CA, 90095, USA
| | - Carla Janzen
- Department of Obstetrics & Gynecology, University of California, Los Angeles, CA, 90095, USA
| | - Sherin U Devaskar
- Department of Pediatrics, University of California, Los Angeles, CA, 90095, USA
| | | | - Beate Ritz
- Department of Epidemiology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
17
|
Chiorean DM, Cobankent Aytekin E, Mitranovici MI, Turdean SG, Moharer MS, Cotoi OS, Toru HS. Human Placenta and Evolving Insights into Pathological Changes of Preeclampsia: A Comprehensive Review of the Last Decade. Fetal Pediatr Pathol 2024; 43:33-46. [PMID: 37906285 DOI: 10.1080/15513815.2023.2274823] [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: 09/07/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023]
Abstract
The placenta, the foremost and multifaceted organ in fetal and maternal biology, is pivotal in facilitating optimal intrauterine fetal development. Remarkably, despite its paramount significance, the placenta remains enigmatic, meriting greater comprehension given its central influence on the health trajectories of both the fetus and the mother. Preeclampsia (PE) and intrauterine fetal growth restriction (IUGR), prevailing disorders of pregnancy, stem from compromised placental development. PE, characterized by heightened mortality and morbidity risks, afflicts 5-7% of global pregnancies, its etiology shrouded in ambiguity. Pertinent pathogenic hallmarks of PE encompass inadequate restructuring of uteroplacental spiral arteries, placental ischemia, and elevated levels of vascular endothelial growth factor receptor-1 (VEGFR-1), also recognized as soluble FMS-like tyrosine kinase-1 (sFlt-1). During gestation, the placental derivation of sFlt-1 accentuates its role as an inhibitory receptor binding to VEGF-A and placental growth factor (PlGF), curtailing target cell accessibility. This review expounds upon the placenta's defining cellular component of the trophoblast, elucidates the intricacies of PE pathogenesis, underscores the pivotal contribution of sFlt-1 to maternal pathology and fetal safeguarding, and surveys recent therapeutic strides witnessed in the past decade.
Collapse
Affiliation(s)
- Diana Maria Chiorean
- Department of Pathology, County Clinical Hospital of Targu Mures, Targu Mures, Romania
| | | | | | - Sabin Gligore Turdean
- Department of Pathology, County Clinical Hospital of Targu Mures, Targu Mures, Romania
| | | | - Ovidiu Simion Cotoi
- Department of Pathology, County Clinical Hospital of Targu Mures, Targu Mures, Romania
- Department Pathophysiology, "George Emil Palade" University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania, and
| | - Havva Serap Toru
- Department of Pathology, School of Medicine, Akdeniz University, Antalya Pınarbaşı, Konyaaltı/Antalya, Turkey
| |
Collapse
|
18
|
Jo S, Alejandro EU. RISING STARS: Mechanistic insights into maternal-fetal cross talk and islet beta-cell development. J Endocrinol 2023; 259:e230069. [PMID: 37855321 PMCID: PMC10692651 DOI: 10.1530/joe-23-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
The metabolic health trajectory of an individual is shaped as early as prepregnancy, during pregnancy, and lactation period. Both maternal nutrition and metabolic health status are critical factors in the programming of offspring toward an increased propensity to developing type 2 diabetes in adulthood. Pancreatic beta-cells, part of the endocrine islets, which are nutrient-sensitive tissues important for glucose metabolism, are primed early in life (the first 1000 days in humans) with limited plasticity later in life. This suggests the high importance of the developmental window of programming in utero and early in life. This review will focus on how changes to the maternal milieu increase offspring's susceptibility to diabetes through changes in pancreatic beta-cell mass and function and discuss potential mechanisms by which placental-driven nutrient availability, hormones, exosomes, and immune alterations that may impact beta-cell development in utero, thereby affecting susceptibility to type 2 diabetes in adulthood.
Collapse
Affiliation(s)
- Seokwon Jo
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Emilyn U Alejandro
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
19
|
Capobianco E, Pirrone I. Paternal programming of fetoplacental and offspring metabolic disorders. Placenta 2023; 141:71-77. [PMID: 37355440 DOI: 10.1016/j.placenta.2023.06.009] [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: 03/09/2023] [Revised: 06/08/2023] [Accepted: 06/11/2023] [Indexed: 06/26/2023]
Abstract
The alarming increase in the prevalence of metabolic pathologies is of worldwide concern and has been linked not only to genetic factors but also to a large number of non-genetic factors. In recent years, there has been increasing interest in the study of the programming of metabolic diseases, such as type 2 diabetes mellitus (T2DM) and obesity, by paternal exposure, a paradigm termed "Paternal Origins of Health and Disease" (POHaD). This term derives from the better known "Developmental Origins of Health and Disease" (DOHaD), which focuses on the involvement of the maternal intrauterine environment and complications during pregnancy associated with the health and disease of the offspring. Studies on paternal programming have documented environmentally induced epigenetic modifications in the male germline and in seminal plasma, which lead to intergenerational and transgenerational phenotypes, evident already during fetoplacental development. Studies with animal models at both ends of the nutritional spectrum (undernutrition or overnutrition) have been performed to understand the possible mechanisms and signaling pathways leading to the programming of metabolic disorders by exploring epigenetic changes throughout the life of the offspring. The aim of this review was to address the evidence of the programming of fetoplacental developmental alterations and metabolic pathologies in the offspring of males with metabolic disorders and unhealthy exposures, highlighting the mechanisms involved in such programming and looking for paternal interventions to reduce negative health outcomes in the offspring.
Collapse
Affiliation(s)
- Evangelina Capobianco
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina.
| | - Irune Pirrone
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| |
Collapse
|
20
|
Párraga-Ros E, Álvarez-Martín Ú, Seva J, Coy P, Romar R. The impact of in vitro embryo production on placental and umbilical cord vascularization is minimized by the addition of reproductive fluids. Theriogenology 2023; 208:149-157. [PMID: 37329589 DOI: 10.1016/j.theriogenology.2023.05.029] [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: 03/23/2023] [Revised: 05/10/2023] [Accepted: 05/29/2023] [Indexed: 06/19/2023]
Abstract
Animals born from in-vitro-produced (IVP) embryos show changes in the placenta and umbilical cord vascularization. This study compares the placental and umbilical vascular morphometry in pigs (n = 19) born through artificial insemination (AI group) or after transfer of IVP embryos cultured with (RF-IVP group) or without (C-IVP group) reproductive fluids. The relationship between vascular parameters and animal growth during the first year of life was also analyzed. Samples were collected at birth, fixed, paraffin-embedded, cut in sections, stained, and photographed for vascular and morphometric analysis with ImageJ® and Slide Viewer®. The average daily weight gain was individually scored from birth to the first year of life. No differences were found in placental vascular morphometry among groups, except for the vascular area of small vessels (arterioles, venules, and small vessels) that was higher in the C-IVP group. Regarding the umbilical cord, the values for perimeter (AI: 26.40 ± 3.93 mm; IVP: 30.51 ± 4.74 mm), diameter (AI: 8.35 ± 1.01 mm; IVP: 10.26 ± 1.85 mm), area (AI: 43.18 ± 12.87; IVP: 56.61 ± 14.89 mm2), and Wharton's jelly area (AI: 36.86 ± 12.04 mm2; IVP 48.88 ± 12.80 mm2) were higher in IVP-derived than AI-derived animals, whereas arterial and venous morphometric data were similar between groups. A correlation study showed that placental and umbilical cord vascular phenotypes affect the further growth of pigs. In conclusion, assisted reproductive technologies impact small caliber vessels in the placenta and morphometric parameters in the umbilical cord. The addition of reproductive fluids in IVP-embryo contributes to reduce the differences with in vivo-derived animals.
Collapse
Affiliation(s)
- Ester Párraga-Ros
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Úrsula Álvarez-Martín
- Department of Physiology, Faculty of Veterinary Medicine, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Juan Seva
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Pilar Coy
- Department of Physiology, Faculty of Veterinary Medicine, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain
| | - Raquel Romar
- Department of Physiology, Faculty of Veterinary Medicine, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain.
| |
Collapse
|
21
|
Pan M, Zhou J, Wang J, Cao W, Li L, Wang L. The role of placental aging in adverse pregnancy outcomes: A mitochondrial perspective. Life Sci 2023; 329:121924. [PMID: 37429418 DOI: 10.1016/j.lfs.2023.121924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
Premature placental aging is associated with placental insufficiency, which reduces the functional capacity of the placenta, leading to adverse pregnancy outcomes. Placental mitochondria are vital organelles that provide energy and play essential roles in placental development and functional maintenance. In response to oxidative stress, damage, and senescence, an adaptive response is induced to selectively remove mitochondria through the mitochondrial equivalent of autophagy. However, adaptation can be disrupted when mitochondrial abnormalities or dysfunctions persist. This review focuses on the adaptation and transformation of mitochondria during pregnancy. These changes modify placental function throughout pregnancy and can cause complications. We discuss the relationship between placental aging and adverse pregnancy outcomes from the perspective of mitochondria and potential approaches to improve abnormal pregnancy outcomes.
Collapse
Affiliation(s)
- Meijun Pan
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; The Second Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China; The Academy of Integrative Medicine of Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; The Academy of Integrative Medicine of Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; The Academy of Integrative Medicine of Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Wenli Cao
- Center for Reproductive Medicine, Zhoushan Women and Children Hospital, Zhejiang, China
| | - Lisha Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; The Academy of Integrative Medicine of Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; The Academy of Integrative Medicine of Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China.
| |
Collapse
|
22
|
Kent NL, Atluri SC, Moritz KM, Cuffe JSM. Maternal hypothyroidism in rats impairs placental nutrient transporter expression, increases labyrinth zone size, and impairs fetal growth. Placenta 2023; 139:148-158. [PMID: 37406552 DOI: 10.1016/j.placenta.2023.06.010] [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: 11/03/2022] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
INTRODUCTION Hypothyroidism during pregnancy is associated with fetal growth restriction (FGR). FGR is commonly caused by placental insufficiency and yet the role of hypothyroidism in placental regulation of fetal growth is unknown. This study aimed to investigate the effects of maternal hypothyroidism on placental nutrient transporter expression, placental morphology, and placental metabolism. METHODS Hypothyroidism was induced in female Sprague-Dawley rats by adding methimazole (MMI) to drinking water at moderate (MOD, MMI at 0.005% w/v) and severe (SEV, MMI at 0.02% w/v) doses from one week prior to pregnancy and throughout gestation. Maternal and fetal tissues were collected on embryonic day 20 (E20). RESULTS Hypothyroidism reduced fetal weight (PTrt<0.001) despite causing fetal hyperglycaemia (PTrt = 0.016). Placental weight was not affected by hypothyroidism however placental efficiency was reduced (PTrt<0.001), as was the junctional zone (JZ):labyrinth zone (LZ) weight ratio (PTrt = 0.005). LZ glycogen content was increased (PTrt = 0.029) and while mRNA expression of glucose transporters was reduced by hypothyroidism, only GLUT1 protein expression was reduced in male LZs. Maternal hypothyroidism reduced mitochondrial content (PTrt = 0.031), particularly in SEV males relative to CON males (P = 0.004). Protein expression of Complex V (P < 0.001) and Complex III (P = 0.002) of the electron transport chain were also reduced in males. Maternal hypothyroidism reduced LZ (PTrt<0.001) and fetal plasma triglycerides (P = 0.019) while fetal free fatty acids and the expression of LZ lipid transporters was not affected. DISCUSSION Overall, maternal hypothyroidism may lead to FGR through reduced maternal T4 availability, changes to placental morphology, altered nutrient transporter expression and sex-specific effects on placental metabolism. Changes to LZ glycogen and triglyceride stores as well as mitochondrial content suggest a metabolic shift from oxidative phosphorylation to anaerobic glycolysis in males. These changes also likely impact fetal substrate availability and therefore fetal growth.
Collapse
Affiliation(s)
- Nykola L Kent
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Sharat C Atluri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - James S M Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
23
|
Lopez-Tello J, Yong HEJ, Sandovici I, Dowsett GKC, Christoforou ER, Salazar-Petres E, Boyland R, Napso T, Yeo GSH, Lam BYH, Constancia M, Sferruzzi-Perri AN. Fetal manipulation of maternal metabolism is a critical function of the imprinted Igf2 gene. Cell Metab 2023; 35:1195-1208.e6. [PMID: 37437545 DOI: 10.1016/j.cmet.2023.06.007] [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: 04/06/2022] [Revised: 04/12/2023] [Accepted: 06/09/2023] [Indexed: 07/14/2023]
Abstract
Maternal-offspring interactions in mammals involve both cooperation and conflict. The fetus has evolved ways to manipulate maternal physiology to enhance placental nutrient transfer, but the mechanisms involved remain unclear. The imprinted Igf2 gene is highly expressed in murine placental endocrine cells. Here, we show that Igf2 deletion in these cells impairs placental endocrine signaling to the mother, without affecting placental morphology. Igf2 controls placental hormone production, including prolactins, and is crucial to establish pregnancy-related insulin resistance and to partition nutrients to the fetus. Consequently, fetuses lacking placental endocrine Igf2 are growth restricted and hypoglycemic. Mechanistically, Igf2 controls protein synthesis and cellular energy homeostasis, actions dependent on the placental endocrine cell type. Igf2 loss also has additional long-lasting effects on offspring metabolism in adulthood. Our study provides compelling evidence for an intrinsic fetal manipulation system operating in placenta that modifies maternal metabolism and fetal resource allocation, with long-term consequences for offspring metabolic health.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Hannah E J Yong
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A(∗)STAR), 30 Medical Drive, Singapore 117609, Singapore
| | - Ionel Sandovici
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0SW, UK; Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Georgina K C Dowsett
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Efthimia R Christoforou
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Esteban Salazar-Petres
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Rebecca Boyland
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Royal Devon and Exeter Hospital NHS Trust, Barrack Rd, Exeter EX2 5DW, UK
| | - Tina Napso
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Giles S H Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Brian Y H Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Miguel Constancia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0SW, UK; Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK.
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
24
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
25
|
Teo SM, Murrin CM, Mehegan J, Douglass A, Hébert JR, Segurado R, Kelleher CC, Phillips CM. Associations between the maternal healthy lifestyle score and its individual components during early pregnancy with placental outcomes. Placenta 2023; 139:75-84. [PMID: 37336158 DOI: 10.1016/j.placenta.2023.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/29/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023]
Abstract
INTRODUCTION The influence of maternal lifestyle behaviours on placental growth have been investigated individually, but with conflicting results, and their combined effect is under-researched. Therefore, we examined associations between a composite maternal healthy lifestyle score (HLS), and its individual components, during early pregnancy with placental outcomes. METHODS Participants included Lifeways Cross-Generational Cohort mother-child pairs (n = 202). A composite HLS based on a less inflammatory diet (bottom 40% of the energy-adjusted Dietary Inflammatory Index (E-DII™)), moderate-to-vigorous physical activity (MVPA), healthy pre-pregnancy BMI (18.5-24.9 kg/m2), never smoking, and non-/moderate alcohol intake was calculated. Quantile regression analysed HLS (and individual components) associations with measures of placental development (untrimmed placental weight (PW)) and function (birth weight:placental weight (BW:PW) ratio) at the 10th, 25th, 50th, 75th and 90th centiles. RESULTS A more pro-inflammatory diet was positively, and smoking and heavy alcohol consumption were negatively, associated with PW at median centiles (B: 41.97 g, CI: 3.71, 80.22, p < 0.05; B: -58.51 g, CI: -116.24, -0.77, p < 0.05; B: -120.20 g, CI: -177.97, -62.43, p < 0.05 respectively). Low MVPA was inversely associated with BW:PW ratio at the 10th and 90th centiles (B: -0.36, CI: -0.132, -0.29, p < 0.01 and B: -0.45, CI: -0.728, -0.182, p < 0.01, respectively). Heavy alcohol intake was positively associated with BW:PW ratio at the 10th centile (B: 0.54, CI: 0.24, 0.85, p < 0.01). Results of sex-stratified analysis provide evidence of sexual dimorphism. DISCUSSION Associations of certain lifestyle factors, but not the composite HLS, during early pregnancy with measures of placental development (PW) and function (BW:PW ratio) varied by quantiles and by sex.
Collapse
Affiliation(s)
- Shevaun M Teo
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, 4, Ireland.
| | - Celine M Murrin
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, 4, Ireland.
| | - John Mehegan
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, 4, Ireland.
| | - Alexander Douglass
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, 4, Ireland.
| | - James R Hébert
- Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States.
| | - Ricardo Segurado
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, 4, Ireland.
| | - Cecily C Kelleher
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, 4, Ireland.
| | - Catherine M Phillips
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, 4, Ireland.
| |
Collapse
|
26
|
Giovarelli M, Serati A, Zecchini S, Guelfi F, Clementi E, Mandò C. Cryopreserved placental biopsies maintain mitochondrial activity for high-resolution respirometry. Mol Med 2023; 29:45. [PMID: 37013473 PMCID: PMC10069067 DOI: 10.1186/s10020-023-00645-2] [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: 09/05/2022] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND High-resolution respirometry (HRR) of human biopsies can provide useful metabolic, diagnostic, and mechanistic insights for clinical research and comparative medical studies. Fresh tissues analysis offers the potential best condition, the drawback being the need to use them shortly after dissection for mitochondrial respiratory experiments. The development of effective long-term storage protocols for biopsies that allow the assessment of key Electron Transport System (ETS) parameters at later stages is thus a major need. METHODS We optimised a cryopreservation protocol that preserves mitochondrial membranes intactness, otherwise affected by direct tissue freezing. The protocol is based on a gradual freezing step from on-ice to liquid nitrogen and - 80 °C storage using a specific DMSO-based buffer. RESULTS Placenta is a suitable tissue to design and test the effectiveness of long-term storage protocols being metabolically active foetal tissue with mitochondrial dysfunctions contributing to placental disease and gestational disorders. Here we designed and tested the effectiveness of the cryopreservation protocol using human placenta biopsies; we measured the ETS activity by HRR of placenta specimens comparing fresh, cryopreserved, and snap frozen conditions. CONCLUSIONS By this protocol, Oxygen Consumption Rate (OCR) measurements of fresh and cryopreserved placental specimens are comparable whereas snap frozen procedure impairs mitochondrial activity.
Collapse
Affiliation(s)
- Matteo Giovarelli
- Department of Biomedical and Clinical Sciences, Università Degli Studi Di Milano, Via G.B. Grassi 74, 20157, Milan, Italy.
| | - Anais Serati
- Department of Biomedical and Clinical Sciences, Università Degli Studi Di Milano, Via G.B. Grassi 74, 20157, Milan, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences, Università Degli Studi Di Milano, Via G.B. Grassi 74, 20157, Milan, Italy
| | - Fabiola Guelfi
- Department of Woman Mother and Neonate 'V. Buzzi' Children Hospital, ASST Fatebenefratelli Sacco, 20154, Milan, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences, Università Degli Studi Di Milano, Via G.B. Grassi 74, 20157, Milan, Italy
- Scientific Institute, IRCCS Eugenio Medea, Via Don Luigi Monza 20, 23842, Bosisio Parini, Italy
| | - Chiara Mandò
- Department of Biomedical and Clinical Sciences, Università Degli Studi Di Milano, Via G.B. Grassi 74, 20157, Milan, Italy
| |
Collapse
|
27
|
Sferruzzi‐Perri AN, Lopez‐Tello J, Salazar‐Petres E. Placental adaptations supporting fetal growth during normal and adverse gestational environments. Exp Physiol 2023; 108:371-397. [PMID: 36484327 PMCID: PMC10103877 DOI: 10.1113/ep090442] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the topic of this review? How the placenta, which transports nutrients and oxygen to the fetus, may alter its support of fetal growth developmentally and with adverse gestational conditions. What advances does it highlight? Placental formation and function alter with the needs of the fetus for substrates for growth during normal gestation and when there is enhanced competition for substrates in species with multiple gestations or adverse gestational environments, and this is mediated by imprinted genes, signalling pathways, mitochondria and fetal sexomes. ABSTRACT The placenta is vital for mammalian development and a key determinant of life-long health. It is the interface between the mother and fetus and is responsible for transporting the nutrients and oxygen a fetus needs to develop and grow. Alterations in placental formation and function, therefore, have consequences for fetal growth and birthweight, which in turn determine perinatal survival and risk of non-communicable diseases for the offspring in later postnatal life. However, the placenta is not a static organ. As this review summarizes, research from multiple species has demonstrated that placental formation and function alter developmentally to the needs of the fetus for substrates for growth during normal gestation, as well as when there is greater competition for substrates in polytocous species and monotocous species with multiple gestations. The placenta also adapts in response to the gestational environment, integrating information about the ability of the mother to provide nutrients and oxygen with the needs of the fetus in that prevailing environment. In particular, placental structure (e.g. vascularity, surface area, blood flow, diffusion distance) and transport capacity (e.g. nutrient transporter levels and activity) respond to suboptimal gestational environments, namely malnutrition, obesity, hypoxia and maternal ageing. Mechanisms mediating developmentally and environmentally induced homeostatic responses of the placenta that help support normal fetal growth include imprinted genes, signalling pathways, subcellular constituents and fetal sexomes. Identification of these placental strategies may inform the development of therapies for complicated human pregnancies and advance understanding of the pathways underlying poor fetal outcomes and their consequences for health and disease risk.
Collapse
Affiliation(s)
- Amanda Nancy Sferruzzi‐Perri
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Jorge Lopez‐Tello
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Esteban Salazar‐Petres
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Facultad de CienciasDepartamento de Ciencias Básicas, Universidad Santo TomásValdiviaChile
| |
Collapse
|
28
|
McCoard S, Haack N, Heiser A, Maclean P. Effect of birth rank, and placentome subtype on expression of genes involved in placental nutrient transport in sheep. Theriogenology 2023; 203:109-117. [PMID: 37023492 DOI: 10.1016/j.theriogenology.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 02/23/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Placental function is a key determinant of fetal growth and development that can be influenced by maternal and fetal environmental factors. The molecular mechanisms by which the placenta senses and responds to environmental cues are poorly understood. This exploratory study aimed to characterize the effect of birth rank (single vs. twin) and placentome morphologic subtype on expression of genes involved in nutrient transport, angiogenesis, immunity and stress response. Cotyledonary tissue was collected from type A, B and C placentomes from five single and six twin fetuses at 140 days of gestation. GLUT1 and GLUT3 were the most highly expressed genes consistent with the high demand for glucose to support fetal growth. Expression of BCKDHβ and IGF-2 was 1.3- and 1.5-fold higher, respectively, and PCYT1A was 3-fold lower in singles compared to twins (P < 0.05) while no other differences in gene expression were observed between birth ranks. Expression of EAAT2 and LAT2 was higher while PCYT1A was lower in A compared to B type cotyledons. Expression of GUCY1B1/3 and IGF-1 was higher while CD98 and LAT2 were lower in type B compared to C cotyledons (P < 0.05). Compared to type C cotyledons, expression of EAAT2, IGF-1, IGF-2, LAT1 was higher, while TEK was lower in type A cotyledons. The effects of birth rank on placental gene expression in this study indicated that placental nutrient transport and/or function differs between single and twin pregnancies in sheep. Differences in gene expression between the placentome subtypes suggests that changes in placentome morphology are associated with shifts in amino acid transport and metabolism, oxidative stress and angiogenesis and/or blood flow. This study highlights that placental gene expression differs in response to birth rank and placentome morphologic subtype which suggests that both maternal and fetal factors may influence placental function in sheep. These associations provide insights into gene pathways for more targeted future investigations as well as potential adaptations to improve placental efficiency to support fetal growth in twin pregnancies.
Collapse
|
29
|
Lopez-Tello J, Sferruzzi-Perri AN. Characterization of placental endocrine function and fetal brain development in a mouse model of small for gestational age. Front Endocrinol (Lausanne) 2023; 14:1116770. [PMID: 36843585 PMCID: PMC9950515 DOI: 10.3389/fendo.2023.1116770] [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: 12/05/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Conditions such as small for gestational age (SGA), which is defined as birthweight less than 10th percentile for gestational age can predispose to neurodevelopmental abnormalities compared to babies with normal birthweight. Fetal growth and birthweight depend on placental function, as this organ transports substrates to the developing fetus and it acts as a source of endocrine factors, including steroids and prolactins that are required for fetal development and pregnancy maintenance. To advance our knowledge on the aetiology of fetal growth disorders, the vast majority of the research has been focused on studying the transport function of the placenta, leaving practically unexplored the contribution of placental hormones in the regulation of fetal growth. Here, using mice and natural variability in fetal growth within the litter, we compared fetuses that fell on or below the 10th percentile (classified as SGA) with those that had adequate weight for their gestational age (AGA). In particular, we compared placental endocrine metabolism and hormone production, as well as fetal brain weight and expression of developmental, growth and metabolic genes between SGA and AGA fetuses. We found that compared to AGA fetuses, SGA fetuses had lower placental efficiency and reduced capacity for placental production of hormones (e.g. steroidogenic gene Cyp17a1, prolactin Prl3a1, and pregnancy-specific glycoproteins Psg21). Brain weight was reduced in SGA fetuses, although this was proportional to the reduction in overall fetal size. The expression of glucose transporter 3 (Slc2a3) was reduced despite the abundance of AKT, FOXO and ERK proteins were similar. Developmental (Sv2b and Gabrg1) and microglia genes (Ier3), as well as the pregnancy-specific glycoprotein receptor (Cd9) were lower in the brain of SGA versus AGA fetuses. In this mouse model of SGA, our results therefore demonstrate that placental endocrine dysfunction is associated with changes in fetal growth and fetal brain development.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Centre for Trophoblast Research – Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research – Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
30
|
Kirlangic MM, Sade OS, Eraslan Sahin M. Effect of third trimester maternal vitamin D levels on placental weight to birth weight ratio in uncomplicated pregnancies. J Perinat Med 2022:jpm-2022-0432. [PMID: 36508611 DOI: 10.1515/jpm-2022-0432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Vitamin D has critical role for the fetal and placental development. Today, placental weight (PW), fetal birth weight (BW), and the PW/BW ratio are used as markers of fetal development. The aim of this study is to evaluate the relationship between vitamin D levels and these markers in uncomplicated pregnancies. METHODS This study included 108 women with uncomplicated pregnancies, defined as full-term and healthy pregnancies without perinatal complications. Vitamin D levels <12 ng/mL were classified as deficient, 12-20 ng/mL as insufficient, and >20 ng/mL as normal. Postnatal BW and PW were compared according to maternal serum vitamin D levels. RESULTS Maternal age, maternal height, maternal weight, body mass index, nulliparity, gestational age at delivery, mode of delivery, and fetal gender were similar between groups. Postnatal BW, PW, fetal height at birth, and fetal head circumference parameters were similar between the groups. The PW/BW ratio was 21.77±2.20 in the vitamin D deficient group, 21.20±2.40 in the insufficient group, and 19.98±2.37 in the normal group (p=0.012). In addition, there was a significant negative correlation between vitamin D level and the PW/BW ratio (p=0.012, r=0.031). CONCLUSIONS Our results indicated that PW/BW ratio which is the marker for prediction adverse perinatal outcomes were significantly increased in the presence of vitamin D deficiency and insufficiency.
Collapse
Affiliation(s)
- Mehmet Mete Kirlangic
- Department of Obstetrics and Gynecology, Kartal Dr. Lutfi Kirdar Research and Training Hospital, Istanbul, Turkiye
| | - Osman Sertac Sade
- Department of Obstetrics and Gynecology, Tuzla Government Hospital, Istanbul, Turkiye
| | | |
Collapse
|
31
|
Contreras-Correa ZE, Cochran T, Metcalfe A, Burnett DD, Lemley CO. Seasonal and temporal variation in the placenta during melatonin supplementation in a bovine compromised pregnancy model. J Anim Sci 2022; 100:skac372. [PMID: 36370127 PMCID: PMC9762882 DOI: 10.1093/jas/skac372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2023] Open
Abstract
Compromised pregnancies result in a poorly functioning placenta restricting the amount of oxygen and nutrient supply to the fetus resulting in intrauterine growth restriction (IUGR). Supplementing dietary melatonin during a compromised pregnancy increased uteroplacental blood flow and prevented IUGR in a seasonal-dependent manner. The objectives were to evaluate seasonal melatonin-mediated changes in temporal alterations of the bovine placental vascularity and transcript abundance of clock genes, angiogenic factors, and nutrient sensing genes in 54 underfed pregnant Brangus heifers (Fall, n = 29; Summer, n = 25). At day 160 of gestation, heifers were assigned to treatments consisting of adequately fed (ADQ-CON; 100% NRC; n = 13), nutrient restricted (RES-CON; 60% NRC; n = 13), and ADQ or RES supplemented with 20 mg/d of melatonin (ADQ-MEL, n = 13; RES-MEL, n = 15). The animals were fed daily at 0900 hours until day 240 where Cesarean sections were performed in the morning (0500 hours) or afternoon (1300 hours) for placentome collections. In both seasons, we observed a temporal alteration of the core clock genes in the cotyledonary tissue in a season-dependent manner. In the fall, ARNTL, CLOCK, NR1D1, and RORA transcript abundance were decreased (P ≤ 0.05) in the afternoon compared to the morning; whereas in the summer, ARNTL, PER2, and RORA expression were increased (P ≤ 0.05) in the afternoon. Interestingly, in both seasons, there was a concomitant temporal increase (P ≤ 0.05) of cotyledonary blood vessel perfusion and caruncular melatonin receptor 1A transcript abundance. Melatonin supplementation did not alter the melatonin receptor 1A transcript abundance (P > 0.05), however, in the summer, melatonin supplementation increased cotyledonary VEGFA, CRY1, and RORA (P ≤ 0.05) transcript abundance. In addition, during the summer the placentomes from underfed dams had increased average capillary size and HIF1α transcript abundance compared to those adequately fed (P ≤ 0.05). In conclusion, these data indicate increased cotyledonary blood vessel size and blood distribution after feeding to better facilitate nutrient transport. Interestingly, the maternal nutritional plane appears to play a crucial role in regulating the bovine placental circadian clock. Based on these findings, the regulation of angiogenic factors and clock genes in the bovine placenta appears to be an underlying mechanism of the therapeutic effect of dietary melatonin supplementation in the summer.
Collapse
Affiliation(s)
- Zully E Contreras-Correa
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Taylor Cochran
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Abbie Metcalfe
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Derris D Burnett
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Caleb O Lemley
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, 39762, USA
| |
Collapse
|
32
|
Xiao Y, Li M, Guo X, Zeng H, Shuai X, Guo J, Huang Q, Chu Y, Zhou B, Wen J, Liu J, Jiao H. Inflammatory Mechanism of Brucella Infection in Placental Trophoblast Cells. Int J Mol Sci 2022; 23:13417. [PMID: 36362199 PMCID: PMC9657658 DOI: 10.3390/ijms232113417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 01/03/2024] Open
Abstract
Brucellosis is a severe zoonotic infectious disease caused by the infection of the Brucella, which is widespread and causes considerable economic losses in underdeveloped areas. Brucella is a facultative intracellular bacteria whose main target cells for infection are macrophages, placental trophoblast cells and dendritic cells. The main clinical signs of Brucella infection in livestock are reproductive disorders and abortion. At present, the pathogenesis of placentitis or abortion caused by Brucella in livestock is not fully understood, and further research on the effect of Brucella on placental development is still necessary. This review will mainly introduce the research progress of Brucella infection of placental trophoblast cells as well as the inflammatory response caused by it, explaining the molecular regulation mechanism of Brucella leading to reproductive system disorders and abortion, and also to provide the scientific basis for revealing the pathogenesis and infection mechanism of Brucella.
Collapse
Affiliation(s)
- Yu Xiao
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Mengjuan Li
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Xiaoyi Guo
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Hui Zeng
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Xuehong Shuai
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Jianhua Guo
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Qingzhou Huang
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Yuefeng Chu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Bo Zhou
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Yujinxiang Street 573, Changchun 130102, China
| | - Jake Wen
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Jun Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Yujinxiang Street 573, Changchun 130102, China
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing 400715, China
- The Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 400715, China
| |
Collapse
|
33
|
Cindrova-Davies T, Sferruzzi-Perri AN. Human placental development and function. Semin Cell Dev Biol 2022; 131:66-77. [PMID: 35393235 DOI: 10.1016/j.semcdb.2022.03.039] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022]
Abstract
The placenta is a transient fetal organ that plays a critical role in the health and wellbeing of both the fetus and its mother. Functionally, the placenta sustains the growth of the fetus as it facilitates delivery of oxygen and nutrients and removal of waste products. Not surprisingly, defective early placental development is the primary cause of common disorders of pregnancy, including recurrent miscarriage, fetal growth restriction, pre-eclampsia and stillbirth. Adverse pregnancy conditions will also affect the life-long health of the fetus via developmental programming[1]. Despite its critical importance in reproductive success and life-long health, our understanding of placental development is not extensive, largely due to ethical limitations to studying early or chronological placental development, lack of long-term in vitro models, or comparative animal models. In this review, we examine current knowledge of early human placental development, discuss the critical role of the maternal endometrium and of the fetal-maternal dialogue in pregnancy success, and we explore the latest models of trophoblast and endometrial stem cells. In addition, we discuss the role of oxygen in placental formation and function, how nutrient delivery is mediated during the periods of histotrophic nutrition (uptake of uterine secretions) and haemotrophic nutrition (exchange between the maternal and fetal circulations), and how placental endocrine function facilitates fetal growth and development.
Collapse
Affiliation(s)
- Tereza Cindrova-Davies
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
34
|
Brombach C, Tong W, Giussani DA. Maternal obesity: new placental paradigms unfolded. Trends Mol Med 2022; 28:823-835. [PMID: 35760668 DOI: 10.1016/j.molmed.2022.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 01/24/2023]
Abstract
The prevalence of maternal obesity is increasing at an alarming rate, and is providing a major challenge for obstetric practice. Adverse effects on maternal and fetal health are mediated by complex interactions between metabolic, inflammatory, and oxidative stress signaling in the placenta. Endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) are common downstream pathways of cell stress, and there is evidence that this conserved homeostatic response may be a key mediator in the pathogenesis of placental dysfunction. We summarize the current literature on the placental cellular and molecular changes that occur in obese women. A special focus is cast onto placental ER stress in obese pregnancy, which may provide a novel link for future investigation.
Collapse
Affiliation(s)
| | - Wen Tong
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3EL, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Cambridge Strategic Research Initiative in Reproduction, Cambridge CB2 3EL, Cambridge UK.
| | - Dino A Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3EL, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Cambridge Strategic Research Initiative in Reproduction, Cambridge CB2 3EL, Cambridge UK; Cambridge Cardiovascular Centre for Research Excellence, Cambridge CB2 0QQ, UK.
| |
Collapse
|
35
|
Pereira-Carvalho D, Salazar-Petres E, Lopez-Tello J, Sferruzzi-Perri AN. Maternal and Fetal PI3K-p110α Deficiency Induces Sex-Specific Changes in Conceptus Growth and Placental Mitochondrial Bioenergetic Reserve in Mice. Vet Sci 2022; 9:vetsci9090501. [PMID: 36136716 PMCID: PMC9506205 DOI: 10.3390/vetsci9090501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Fetal growth is reliant on placental formation and function, which, in turn, requires the energy produced by the mitochondria. Prior work has shown that both mother and fetus operate via the phosphoinositol 3-kinase (PI3K)-p110α signalling pathway to modify placental development, function, and fetal growth outcomes. This study in mice used genetic inactivation of PI3K-p110α (α/+) in mothers and fetuses and high resolution respirometry to investigate the influence of maternal and fetal PI3K-p110α deficiency on fetal and placental growth, in relation to placental mitochondrial bioenergetics, for each fetal sex. The effect of PI3K-p110α deficiency on maternal body composition was also determined to understand more about the maternal-driven changes in feto-placental development. These data show that male fetuses were more sensitive than females to fetal PI3K-p110α deficiency, as they had greater reductions in fetal and placental weight, when compared to their WT littermates. Placental weight was also altered in males only of α/+ dams. In addition, α/+ male, but not female, fetuses showed an increase in mitochondrial reserve capacity, when compared to their WT littermates in α/+ dams. Finally, α/+ dams exhibited reduced adipose depot masses, compared to wild-type dams. These findings, thus, demonstrate that maternal nutrient reserves and ability to apportion nutrients to the fetus are reduced in α/+ dams. Moreover, maternal and fetal PI3K-p110α deficiency impacts conceptus growth and placental mitochondrial bioenergetic function, in a manner dependent on fetal sex.
Collapse
|
36
|
Lopez-Tello J, Jimenez-Martinez MA, Salazar-Petres E, Patel R, George AL, Kay RG, Sferruzzi-Perri AN. Identification of Structural and Molecular Signatures Mediating Adaptive Changes in the Mouse Kidney in Response to Pregnancy. Int J Mol Sci 2022; 23:6287. [PMID: 35682969 PMCID: PMC9181623 DOI: 10.3390/ijms23116287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
Pregnancy is characterized by adaptations in the function of several maternal body systems that ensure the development of the fetus whilst maintaining health of the mother. The renal system is responsible for water and electrolyte balance, as well as waste removal. Thus, it is imperative that structural and functional changes occur in the kidney during pregnancy. However, our knowledge of the precise morphological and molecular mechanisms occurring in the kidney during pregnancy is still very limited. Here, we investigated the changes occurring in the mouse kidney during pregnancy by performing an integrated analysis involving histology, gene and protein expression assays, mass spectrometry profiling and bioinformatics. Data from non-pregnant and pregnant mice were used to identify critical signalling pathways mediating changes in the maternal kidneys. We observed an expansion of renal medulla due to proliferation and infiltration of interstitial cellular constituents, as well as alterations in the activity of key cellular signalling pathways (e.g., AKT, AMPK and MAPKs) and genes involved in cell growth/metabolism (e.g., Cdc6, Foxm1 and Rb1) in the kidneys during pregnancy. We also generated plasma and urine proteomic profiles, identifying unique proteins in pregnancy. These proteins could be used to monitor and study potential mechanisms of renal adaptations during pregnancy and disease.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | | | - Esteban Salazar-Petres
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ritik Patel
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amy L George
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Richard G Kay
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
37
|
Alteration of LncRNA expression in mice placentae after frozen embryo transfer is associated with increased fetal weight. Reprod Biol 2022; 22:100646. [PMID: 35567965 DOI: 10.1016/j.repbio.2022.100646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/18/2022] [Accepted: 04/23/2022] [Indexed: 11/15/2022]
Abstract
The birthweight after frozen embryo transfer (FET) was significantly higher compared with fresh embryo transfer (fresh ET), while the mechanism remains unclear. In this study, we transferred vitrified-warmed or fresh mice blastocysts into pseudopregnant recipients (n = 11 each group) produced by natural mating to avoid the influence of superovulation. The fetal weight, placental weight, placental efficiency and placental architecture were studied at E18.5. Placental RNA-Seq analysis was used to identify candidate different lncRNAs and mRNAs between the FET group and the fresh ET group. We found that the fetal weight was increased in the FET group, with increased placental efficiency and the proportion of placental function related labyrinth zone area. 554 lncRNAs and 1012 mRNAs were differentially expressed. KEGG and GO enrichment analyses showed these differentially expressed lncRNAs and their targeted mRNAs might be related to placental morphogenesis. Furthermore, the most differentially expressed 15 lncRNAs and 15 mRNAs were validated by qRT-PCR, we found the LncRNA embryonic stem cells expressed 1 (Lncenc1) was significantly decreased, and Gjb5, which played an important role in labyrinth zone development, was increased. Gjb5 protein increase was further confirmed by Western blot. Lncenc1 and Gjb5 had 48 predicted co-targeted miRNAs, while the correlation analysis of Lncenc1 and Gjb5 mRNA showed a significant inverse correlation. The results showed that FET treatment might enhance the placental function to increase mouse fetal weight via the network diagram of Lncenc1-miRNA-Gjb5.
Collapse
|
38
|
Paternal periconception metabolic health and offspring programming. Proc Nutr Soc 2022; 81:119-125. [DOI: 10.1017/s0029665121003736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The association between maternal metabolic status at the time of conception and subsequent embryogenesis and offspring development has been studied in detail. However, less attention has been given to the significance of paternal nutrition and metabolism in directing offspring health. Despite this disparity, emerging evidence has begun to highlight an important connection between paternal metabolic well-being, semen quality, embryonic development and ultimately adult offspring health. This has established a new component within the Developmental Origins of Health and Disease hypothesis. Building on the decades of understanding and insight derived from the numerous models of maternal programming, attention is now becoming focused on defining the mechanisms underlying the links between paternal well-being, post-fertilisation development and offspring health. Understanding how the health and fitness of the father impact on semen quality is of fundamental importance for providing better information to intending fathers. Furthermore, assisted reproductive practices such as in vitro fertilisation rely on our ability to select the best quality sperm from a diverse and heterogeneous population. With considerable advances in sequencing capabilities, our understanding of the molecular and epigenetic composition of the sperm and seminal plasma, and their association with male metabolic health, has developed dramatically over recent years. This review will summarise our current understanding of how a father's metabolic status at the time of conception can affect sperm quality, post-fertilisation embryonic and fetal development and offspring health.
Collapse
|
39
|
Cleal JK, Poore KR, Lewis RM. The placental exposome, placental epigenetic adaptations and lifelong cardio-metabolic health. Mol Aspects Med 2022; 87:101095. [DOI: 10.1016/j.mam.2022.101095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 03/04/2022] [Accepted: 03/12/2022] [Indexed: 12/15/2022]
|
40
|
Napso T, Lean SC, Lu M, Mort EJ, Desforges M, Moghimi A, Bartels B, El‐Bacha T, Fowden AL, Camm EJ, Sferruzzi‐Perri AN. Diet-induced maternal obesity impacts feto-placental growth and induces sex-specific alterations in placental morphology, mitochondrial bioenergetics, dynamics, lipid metabolism and oxidative stress in mice. Acta Physiol (Oxf) 2022; 234:e13795. [PMID: 35114078 PMCID: PMC9286839 DOI: 10.1111/apha.13795] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023]
Abstract
AIM The current study investigated the impact of maternal obesity on placental phenotype in relation to fetal growth and sex. METHODS Female C57BL6/J mice were fed either a diet high in fat and sugar or a standard chow diet, for 6 weeks prior to, and during, pregnancy. At day 19 of gestation, placental morphology and mitochondrial respiration and dynamics were assessed using high-resolution respirometry, stereology, and molecular analyses. RESULTS Diet-induced maternal obesity increased the rate of small for gestational age fetuses in both sexes, and increased blood glucose concentrations in offspring. Placental weight, surface area, and maternal blood spaces were decreased in both sexes, with reductions in placental trophoblast volume, oxygen diffusing capacity, and an increased barrier to transfer in males only. Despite these morphological changes, placental mitochondrial respiration was unaffected by maternal obesity, although the influence of fetal sex on placental respiratory capacity varied between dietary groups. Moreover, in males, but not females, maternal obesity increased mitochondrial complexes (II and ATP synthase) and fission protein DRP1 abundance. It also reduced phosphorylated AMPK and capacity for lipid synthesis, while increasing indices of oxidative stress, specifically in males. In females only, placental mitochondrial biogenesis and capacity for lipid synthesis, were both enhanced. The abundance of uncoupling protein-2 was decreased by maternal obesity in both fetal sexes. CONCLUSION Maternal obesity exerts sex-dependent changes in placental phenotype in association with alterations in fetal growth and substrate supply. These findings may inform the design of personalized lifestyle interventions or therapies for obese pregnant women.
Collapse
Affiliation(s)
- Tina Napso
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Samantha C. Lean
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Minhui Lu
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Emily J. Mort
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Michelle Desforges
- Division of Developmental Biology and Medicine Maternal & Fetal Health Research Centre University of Manchester Manchester UK
| | - Ali Moghimi
- The Children’s Hospital at Westmead Westmead New South Wales Australia
- Department of Paediatrics Monash University Monash Victoria Australia
| | - Beverly Bartels
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Tatiana El‐Bacha
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Abigail L. Fowden
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Emily J. Camm
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| | - Amanda N. Sferruzzi‐Perri
- Department of Physiology Development and Neuroscience Centre for Trophoblast Research University of Cambridge Cambridge UK
| |
Collapse
|
41
|
Nema J, Joshi N, Sundrani D, Joshi S. Influence of maternal one carbon metabolites on placental programming and long term health. Placenta 2022; 125:20-28. [DOI: 10.1016/j.placenta.2022.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/12/2022] [Accepted: 02/24/2022] [Indexed: 10/18/2022]
|
42
|
Carvalho DP, Dias AF, Sferruzzi-Perri AN, Ortiga-Carvalho TM. Gaps in the knowledge of thyroid hormones and placental biology. Biol Reprod 2022; 106:1033-1048. [DOI: 10.1093/biolre/ioac006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Thyroid hormones (THs) are required for the growth and development of the foetus, stimulating anabolism and oxygen consumption from the early stages of pregnancy to the period of foetal differentiation close to delivery. Maternal changes in the hypothalamic–pituitary thyroid axis are also well known. In contrast, several open questions remain regarding the relationships between the placenta and the maternal and foetal TH systems. The exact mechanism by which the placenta participates in regulating the TH concentration in the foetus and mother and the role of TH in the placenta are still poorly studied. In this review, we aim to summarize the available data in the area and highlight significant gaps in our understanding of the ontogeny and cell-specific localization of TH transporters, TH receptors and TH metabolic enzymes in the placenta in both human and rodent models. Significant deficiencies also exist in knowledge of the contribution of genomic and nongenomic effects of TH on the placenta and finally how the placenta reacts during pregnancy when the mother has thyroid disease. By addressing these key knowledge gaps, improved pregnancy outcomes and management of women with thyroid alterations may be possible.
Collapse
Affiliation(s)
- Daniela Pereira Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Endocrinologia Translacional, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ariane Fontes Dias
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Endocrinologia Translacional, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda Nancy Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Tania Maria Ortiga-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Endocrinologia Translacional, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Aguilera N, Salas-Pérez F, Ortíz M, Álvarez D, Echiburú B, Maliqueo M. Rodent models in placental research. Implications for fetal origins of adult disease. Anim Reprod 2022; 19:e20210134. [PMID: 35493783 PMCID: PMC9037606 DOI: 10.1590/1984-3143-ar2021-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/21/2022] [Indexed: 11/22/2022] Open
|
44
|
Aykroyd BRL, Tunster SJ, Sferruzzi-Perri AN. Loss of imprinting of the Igf2-H19 ICR1 enhances placental endocrine capacity via sex-specific alterations in signalling pathways in the mouse. Development 2022; 149:dev199811. [PMID: 34982814 PMCID: PMC8783045 DOI: 10.1242/dev.199811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022]
Abstract
Imprinting control region (ICR1) controls the expression of the Igf2 and H19 genes in a parent-of-origin specific manner. Appropriate expression of the Igf2-H19 locus is fundamental for normal fetal development, yet the importance of ICR1 in the placental production of hormones that promote maternal nutrient allocation to the fetus is unknown. To address this, we used a novel mouse model to selectively delete ICR1 in the endocrine junctional zone (Jz) of the mouse placenta (Jz-ΔICR1). The Jz-ΔICR1 mice exhibit increased Igf2 and decreased H19 expression specifically in the Jz. This was accompanied by an expansion of Jz endocrine cell types due to enhanced rates of proliferation and increased expression of pregnancy-specific glycoprotein 23 in the placenta of both fetal sexes. However, changes in the endocrine phenotype of the placenta were related to sexually-dimorphic alterations to the abundance of Igf2 receptors and downstream signalling pathways (Pi3k-Akt and Mapk). There was no effect of Jz-ΔICR1 on the expression of targets of the H19-embedded miR-675 or on fetal weight. Our results demonstrate that ICR1 controls placental endocrine capacity via sex-dependent changes in signalling.
Collapse
Affiliation(s)
| | | | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
45
|
Song Y, Sun J, Li C, Lin L, Gao F, Yang M, Sun B, Wang Y. Long-term monitoring of blood biomarkers related to intrauterine growth restriction using AgNPs SERS tags-based lateral flow assay. Talanta 2021; 241:123128. [PMID: 35038640 DOI: 10.1016/j.talanta.2021.123128] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 01/12/2023]
Abstract
Sensitive and accurate early detection of fetal growth restriction (FGR) is of vital importance in the development of the fetus during pregnancy and even the health of future life. Here, an ultrasensitive and straightforward surface-enhanced Raman scattering (SERS)-based double targets detection in pregnancy is implemented by utilizing functionalized Ag nanoparticles (AgNPs). Through fabricating 4-MPA antibody-modified AgNPs on the lateral flow assay (LFA) strips as the SERS nanotags, the target proteins in blood samples from pregnancy were accurately captured, which further quantizing PI3K and CRAF in unprocessed blood. This strategy warrant excellent selectivity and sensitivity with the limits of detection (LODs) are 0.76 fg mL-1 for PI3K and 0.61 fg mL-1 for CRAF, leading reliable quantification for these two targets. Meanwhile, the feasibility of this assembly was testified by comparing with conventional ELISA method, and the results showed that a high degree of consistency was obtained in these two detection assays. This SERS-colorimetric dual-signal LFA strip can provide a novel strategy for early diagnosis of fetal-related disorders, which is essential for disease diagnosis and treatment guidance during pregnancy.
Collapse
Affiliation(s)
- Yanan Song
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; Qingdao Medical College of Qingdao University, Qingdao, China
| | - Jingyi Sun
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; Shandong Provincial Hospital Affiliated to Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, China
| | - Chunmei Li
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Li Lin
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Feng Gao
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Mingfeng Yang
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Baoliang Sun
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Ying Wang
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| |
Collapse
|
46
|
Placental mtDNA copy number and methylation in association with macrosomia in healthy pregnancy. Placenta 2021; 118:1-9. [PMID: 34972066 DOI: 10.1016/j.placenta.2021.12.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Fetal growth and development depend on metabolic energy from placental mitochondria. However, the impact of placental mitochondria on the occurrence of macrosomia remains unclear. We aimed to explore the association between macrosomia without gestational diabetes mellitus (non-GDM) and changes in placental mitochondrial DNA (mtDNA) copy number and methylation. METHODS Fifty-four newborns with macrosomia and 54 normal birthweight controls were enrolled in this study. Placental mtDNA copy number and mRNA expression of nuclear genes related to mitochondrial replication or ATP synthesis-related genes were measured by real-time quantitative polymerase chain reaction (qPCR). Methylation levels of the non-coding regulatory region D-loop and ATP synthesis-related genes were detected by targeted bisulfite sequencing. RESULTS Newborns with macrosomia had lower placental mtDNA copy number and higher methylation rates of the CpG15 site in the D-loop region (D-CpG15) and CpG6 site in the cytochrome C oxidase III (COX3) gene (COX3-CpG6) than normal birth weight newborns. After adjusting for potential covariates (gestational age, prepregnancy BMI, and infant sex), decreased placental mtDNA copy number (adjusted odds ratio [aOR] = 2.09, 95% confidence interval [CI] 1.03-4.25), elevated methylation rate of D-CpG15 (aOR = 2.06, 95% CI 1.03-4.09) and COX3-CpG6 (aOR = 2.13, 95% CI 1.08-4.20) remained significantly associated with a higher risk of macrosomia. DISCUSSION Reduced mtDNA copy number and increased methylation levels of specific loci at mtDNA would increase the risk of macrosomia. However, the detailed molecular mechanism needs further identification.
Collapse
|
47
|
Francis EC, Dabelea D, Boyle KE, Jansson T, Perng W. Maternal Diet Quality Is Associated with Placental Proteins in the Placental Insulin/Growth Factor, Environmental Stress, Inflammation, and mTOR Signaling Pathways: The Healthy Start ECHO Cohort. J Nutr 2021; 152:816-825. [PMID: 34850052 PMCID: PMC8891174 DOI: 10.1093/jn/nxab403] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/19/2021] [Accepted: 11/23/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Maternal nutritional status affects placental function, which may underlie the intrauterine origins of obesity and diabetes. The extent to which diet quality is associated with placental signaling and which specific pathways are impacted is unknown. OBJECTIVES To examine sex-specific associations of maternal diet quality according to the Healthy Eating Index (HEI)-developed to align with recommendations from the Dietary Guidelines for Americans-with placental proteins involved in metabolism and mediators of environmental stress, inflammation, and growth factors. METHODS Among 108 women from the Healthy Start cohort with a mean ± SD age of 29.0 ± 6.1 y and a prepregnancy BMI (in kg/m2) of 24.8 ± 5.3, we conducted multivariable linear regression analysis stratified by offspring sex. We adjusted for maternal race or ethnicity, age, education, prenatal smoking habits, and physical activity and tested for an association of maternal HEI >57 compared with ≤57 and the abundance and phosphorylation of key proteins involved in insulin/growth factor signaling; mediators of environmental stress, inflammation, and growth factors; mechanistic target of rapamycin signaling proteins; and energy sensing in placental villus samples. HEI >57 was chosen given its prior relevance among Healthy Start mother-child dyads. RESULTS In adjusted models, HEI >57 was associated with greater abundance of insulin receptor β (0.80; 95% CI: 0.11, 1.49) in placentas of females. In males, maternal HEI >57 was associated with greater activation and abundance of select placental nutrient-sensing proteins and environmental stress, inflammation, and growth factor proteins (S6K1Thr389/S6K1: 0.81; 95% CI: 0.21, 1.41; JNK1Thr183/Tyr185/JNK1: 0.82; 95% CI: 0.27, 1.37; JNK2Thr183/Tyr185/JNK2: 0.57; 95% CI: 0.02, 1.11). CONCLUSIONS Higher-quality diet had sex-specific associations with placental protein abundance/phosphorylation. Given that these proteins have been correlated with neonatal anthropometry, our findings provide insight into modifiable factors and placental pathways that should be examined in future studies as potential links between maternal diet and offspring metabolic health. This trial was registered at clinicaltrials.gov as NCT02273297.
Collapse
Affiliation(s)
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA,Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA,Department of Pediatrics, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen E Boyle
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA,Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Wei Perng
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA,Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA,Deptartment of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
48
|
Hsu CN, Hou CY, Hsu WH, Tain YL. Early-Life Origins of Metabolic Syndrome: Mechanisms and Preventive Aspects. Int J Mol Sci 2021; 22:11872. [PMID: 34769303 PMCID: PMC8584419 DOI: 10.3390/ijms222111872] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
One of the leading global public-health burdens is metabolic syndrome (MetS), despite the many advances in pharmacotherapies. MetS, now known as "developmental origins of health and disease" (DOHaD), can have its origins in early life. Offspring MetS can be programmed by various adverse early-life conditions, such as nutrition imbalance, maternal conditions or diseases, maternal chemical exposure, and medication use. Conversely, early interventions have shown potential to revoke programming processes to prevent MetS of developmental origins, namely reprogramming. In this review, we summarize what is currently known about adverse environmental insults implicated in MetS of developmental origins, including the fundamental underlying mechanisms. We also describe animal models that have been developed to study the developmental programming of MetS. This review extends previous research reviews by addressing implementation of reprogramming strategies to prevent the programming of MetS. These mechanism-targeted strategies include antioxidants, melatonin, resveratrol, probiotics/prebiotics, and amino acids. Much work remains to be accomplished to determine the insults that could induce MetS, to identify the mechanisms behind MetS programming, and to develop potential reprogramming strategies for clinical translation.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan;
| | - Wei-Hsuan Hsu
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Chen Kung University, Tainan 701, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
49
|
Iqbal K, Pierce SH, Kozai K, Dhakal P, Scott RL, Roby KF, Vyhlidal CA, Soares MJ. Evaluation of Placentation and the Role of the Aryl Hydrocarbon Receptor Pathway in a Rat Model of Dioxin Exposure. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:117001. [PMID: 34747641 PMCID: PMC8574979 DOI: 10.1289/ehp9256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND Our environment is replete with chemicals that can affect embryonic and extraembryonic development. Dioxins, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are compounds affecting development through the aryl hydrocarbon receptor (AHR). OBJECTIVES The purpose of this investigation was to examine the effects of TCDD exposure on pregnancy and placentation and to evaluate roles for AHR and cytochrome P450 1A1 (CYP1A1) in TCDD action. METHODS Actions of TCDD were examined in wild-type and genome-edited rat models. Placenta phenotyping was assessed using morphological, biochemical, and molecular analyses. RESULTS TCDD exposures were shown to result in placental adaptations and at higher doses, pregnancy termination. Deep intrauterine endovascular trophoblast cell invasion was a prominent placentation site adaptation to TCDD. TCDD-mediated placental adaptations were dependent upon maternal AHR signaling but not upon placental or fetal AHR signaling nor the presence of a prominent AHR target, CYP1A1. At the placentation site, TCDD activated AHR signaling within endothelial cells but not trophoblast cells. Immune and trophoblast cell behaviors at the uterine-placental interface were guided by the actions of TCDD on endothelial cells. DISCUSSION We identified an AHR regulatory pathway in rats activated by dioxin affecting uterine and trophoblast cell dynamics and the formation of the hemochorial placenta. https://doi.org/10.1289/EHP9256.
Collapse
Affiliation(s)
- Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Stephen H. Pierce
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Pramod Dhakal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Regan L. Scott
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Katherine F. Roby
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Anatomy and Cell Biology, KUMC, Kansas City, Kansas, USA
| | - Carrie A. Vyhlidal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy Kansas City, Kansas City, Missouri
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Michael J. Soares
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Obstetrics and Gynecology, KUMC, Kansas City, Kansas, USA
| |
Collapse
|
50
|
Abstract
Almost 2 billion adults in the world are overweight, and more than half of them are classified as obese, while nearly one-third of children globally experience poor growth and development. Given the vast amount of knowledge that has been gleaned from decades of research on growth and development, a number of questions remain as to why the world is now in the midst of a global epidemic of obesity accompanied by the "double burden of malnutrition," where overweight coexists with underweight and micronutrient deficiencies. This challenge to the human condition can be attributed to nutritional and environmental exposures during pregnancy that may program a fetus to have a higher risk of chronic diseases in adulthood. To explore this concept, frequently called the developmental origins of health and disease (DOHaD), this review considers a host of factors and physiological mechanisms that drive a fetus or child toward a higher risk of obesity, fatty liver disease, hypertension, and/or type 2 diabetes (T2D). To that end, this review explores the epidemiology of DOHaD with discussions focused on adaptations to human energetics, placental development, dysmetabolism, and key environmental exposures that act to promote chronic diseases in adulthood. These areas are complementary and additive in understanding how providing the best conditions for optimal growth can create the best possible conditions for lifelong health. Moreover, understanding both physiological as well as epigenetic and molecular mechanisms for DOHaD is vital to most fully address the global issues of obesity and other chronic diseases.
Collapse
Affiliation(s)
- Daniel J Hoffman
- Department of Nutritional Sciences, Program in International Nutrition, and Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Theresa L Powell
- Department of Pediatrics and Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Daniel B Hardy
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|