1
|
Guleroglu FY, Cetin A, Coskun GP, Caliskan M, Karaduman F, Bilginer C, Misirlioglu R, Tekin S, Al MN, Caklili T, Tutar Y. The role of 1-Deoxysphingolipids and Polyamines in the pathogenesis of placental syndrome. BMC Pregnancy Childbirth 2025; 25:51. [PMID: 39844083 PMCID: PMC11753022 DOI: 10.1186/s12884-025-07175-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Placental syndrome, mainly composed of preeclampsia and fetal growth restriction, has an impact on the health of mother and baby dyads. While impaired placentation is central to their pathophysiology, the underlying molecular mechanisms remain incompletely understood. This study investigates the association between placental syndrome and metabolic alterations in 1-deoxysphingolipids (1-deoxySLs) and polyamines, along with their regulatory enzymes. METHODS This prospective case-control study involved 26 healthy pregnant women and 17 with placental syndrome. Blood samples were collected from maternal, uterine venous, and umbilical cord veins. Levels of 1-deoxySL, spermine, and spermidine, as well as related enzymes of polyamine metabolism such as ornithine decarboxylase (ODC), spermidine/spermine N1-acetyltransferase (SSAT), polyamine oxidase (PAO), and spermine oxidase (SMO), were measured using the techniques of LC-MS and ELISA, respectively. RESULTS Women with placental syndrome had significantly higher levels of 1-deoxySL, spermine, and spermidine in all blood samples compared to the healthy pregnancy group. Additionally, ODC and SSAT levels were reduced significantly in the placental syndrome group, while PAO and SMO levels showed no significant differences. Strong positive correlations were found between the studied enzymes and biomolecules in healthy pregnancies, which were notably weaker in the placental syndrome group. CONCLUSION This study demonstrates significantly altered levels of 1-deoxySL and polyamines, with corresponding enzyme activity changes, in placental syndrome compared to healthy pregnancies. The disrupted correlations between these biomolecules suggest alterations in their metabolic pathways and potential utility as biomarkers. Further mechanistic studies are warranted to elucidate their role in placental syndrome pathophysiology.
Collapse
Affiliation(s)
- Filiz Yarsilikal Guleroglu
- Department of Obstetrics and Gynecology, Haseki Training and Research Hospital, Health Sciences University, Istanbul, Turkey.
| | - Ali Cetin
- Department of Obstetrics and Gynecology, Haseki Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| | - Goknil Pelin Coskun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Meltem Caliskan
- Department of Obstetrics and Gynecology, Haseki Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| | - Fulya Karaduman
- Department of Obstetrics and Gynecology, Haseki Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| | - Can Bilginer
- Department of Obstetrics and Gynecology, Haseki Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| | - Resat Misirlioglu
- Department of Obstetrics and Gynecology, Haseki Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| | - Sinem Tekin
- Department of Obstetrics and Gynecology, Haseki Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| | - Merve Nur Al
- Department of Basic Pharmaceutical Sciences, Division of Biochemistry, Health Sciences University, Istanbul, Turkey
| | - Tugce Caklili
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Yusuf Tutar
- Department of Basic Pharmaceutical Sciences, Division of Biochemistry, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
2
|
Abuaish S, Babineau V, Lee S, Tycko B, Champagne FA, Werner E, Monk C. Maternal high BMI: Sex-dimorphic alterations in maternal and offspring stress indices. Psychoneuroendocrinology 2025; 171:107196. [PMID: 39341002 PMCID: PMC11568910 DOI: 10.1016/j.psyneuen.2024.107196] [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: 02/28/2024] [Revised: 07/30/2024] [Accepted: 09/24/2024] [Indexed: 09/30/2024]
Abstract
Maternal body mass index (BMI) influences pregnancy and birth outcomes along with child metabolic and neurodevelopmental health and fetal sex may be a moderating factor in these effects. Alternations in autonomic nervous system (ANS) functioning, identified in heart rate (HR) measurements, could present early markers of these prenatal programming effects in both the mother and the developing fetus. This study examines the associations between pre-pregnancy BMI and maternal and fetal ANS functioning and infant postnatal behavioral outcomes stratified by fetal sex. Pregnant women (N=176) were recruited at gestational week (GW) T1: 12-22 and categorized into Normal (BMI< 25) or High BMI (BMI > 25). Women attended laboratory sessions at T2: GW 23-28, and T3: GW 34-36 to assess maternal and fetal HR and HR variability (HRV) at baseline and after a stressor at T3. Infant behavior was assessed at 4 months using the Infant Behavior Questionnaire-Revised. Women with high BMI bearing female fetuses had higher HR and lower HRV at both gestational time points. Later in the third trimester, female fetuses of high BMI women exhibited lower HRV when challenged with a stressor. At 4 months, female infants were rated as having lower scores on the Orienting/Regulatory scale. Our findings provide evidence of female sex-specific programming of maternal pre-pregnancy BMI on maternal ANS regulation and neurodevelopment identified in-utero and continuing into early infancy.
Collapse
Affiliation(s)
- Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O.Box 84428, Riyadh 11671, Saudi Arabia.
| | - Vanessa Babineau
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Seonjoo Lee
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA
| | - Benjamin Tycko
- Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, NJ 07110, USA
| | | | - Elizabeth Werner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Catherine Monk
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
3
|
Di Giorgio E, Xodo S, Orsaria M, Mariuzzi L, Picco R, Tolotto V, Cortolezzis Y, D'Este F, Grandi N, Driul L, Londero A, Xodo LE. The central role of creatine and polyamines in fetal growth restriction. FASEB J 2024; 38:e70222. [PMID: 39614665 DOI: 10.1096/fj.202401946r] [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: 08/20/2024] [Revised: 10/23/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024]
Abstract
Placental insufficiency often correlates with fetal growth restriction (FGR), a condition that has both short- and long-term effects on the health of the newborn. In our study, we analyzed placental tissue from infants with FGR and from infants classified as small for gestational age (SGA) or appropriate for gestational age (AGA), performing comprehensive analyses that included transcriptomics and metabolomics. By examining villus tissue biopsies and 3D trophoblast organoids, we identified significant metabolic changes in placentas associated with FGR. These changes include adaptations to reduced oxygen levels and modifications in arginine metabolism, particularly within the polyamine and creatine phosphate synthesis pathways. Specifically, we found that placentas with FGR utilize arginine to produce phosphocreatine, a crucial energy reservoir for ATP production that is essential for maintaining trophoblast function. In addition, we found polyamine insufficiency in FGR placentas due to increased SAT1 expression. SAT1 facilitates the acetylation and subsequent elimination of spermine and spermidine from trophoblasts, resulting in a deficit of polyamines that cannot be compensated by arginine or polyamine supplementation alone, unless SAT1 expression is suppressed. Our study contributes significantly to the understanding of metabolic adaptations associated with placental dysfunction and provides valuable insights into potential therapeutic opportunities for the future.
Collapse
Affiliation(s)
| | - Serena Xodo
- Clinic of Obstetrics and Gynecology, Santa Maria della Misericordia Hospital, ASUFC, Udine, Italy
| | - Maria Orsaria
- Institute of Pathology, Department of Medicine, University of Udine, Udine, Italy
| | - Laura Mariuzzi
- Institute of Pathology, Department of Medicine, University of Udine, Udine, Italy
| | | | | | | | | | - Nicole Grandi
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Lorenza Driul
- Department of Medicine, University of Udine, Udine, Italy
- Clinic of Obstetrics and Gynecology, Santa Maria della Misericordia Hospital, ASUFC, Udine, Italy
| | - Ambrogio Londero
- Obstetrics and Gynecology Unit, IRCCS Institute Giannina Gaslini, Genova, Italy
| | - Luigi E Xodo
- Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
4
|
Li Y, Ma L, He R, Teng F, Qin X, Liang X, Wang J. Pregnancy Metabolic Adaptation and Changes in Placental Metabolism in Preeclampsia. Geburtshilfe Frauenheilkd 2024; 84:1033-1042. [PMID: 39524034 PMCID: PMC11543110 DOI: 10.1055/a-2403-4855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/24/2024] [Indexed: 11/16/2024] Open
Abstract
Pregnancy is a unique physiological state in which the maternal body undergoes a series of changes in the metabolism of glucose, lipids, amino acids, and other nutrients in order to adapt to the altered state of pregnancy and provide adequate nutrients for the fetus' growth and development. The metabolism of various nutrients is regulated by one another in order to maintain homeostasis in the body. Failure to adapt to the altered physiological conditions of pregnancy can lead to a range of pregnancy issues, including fetal growth limitation and preeclampsia. A failure of metabolic adaptation during pregnancy is linked to the emergence of preeclampsia. The treatment of preeclampsia by focusing on metabolic changes may provide new therapeutic alternatives.
Collapse
Affiliation(s)
- Yaxi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ling Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Fei Teng
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xue Qin
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| | - Jing Wang
- The First Clinical Medical College of Lanzhou University, the First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China
| |
Collapse
|
5
|
Schibalski RS, Shulha AS, Tsao BP, Palygin O, Ilatovskaya DV. The role of polyamine metabolism in cellular function and physiology. Am J Physiol Cell Physiol 2024; 327:C341-C356. [PMID: 38881422 PMCID: PMC11427016 DOI: 10.1152/ajpcell.00074.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Polyamines are molecules with multiple amino groups that are essential for cellular function. The major polyamines are putrescine, spermidine, spermine, and cadaverine. Polyamines are important for posttranscriptional regulation, autophagy, programmed cell death, proliferation, redox homeostasis, and ion channel function. Their levels are tightly controlled. High levels of polyamines are associated with proliferative pathologies such as cancer, whereas low polyamine levels are observed in aging, and elevated polyamine turnover enhances oxidative stress. Polyamine metabolism is implicated in several pathophysiological processes in the nervous, immune, and cardiovascular systems. Currently, manipulating polyamine levels is under investigation as a potential preventive treatment for several pathologies, including aging, ischemia/reperfusion injury, pulmonary hypertension, and cancer. Although polyamines have been implicated in many intracellular mechanisms, our understanding of these processes remains incomplete and is a topic of ongoing investigation. Here, we discuss the regulation and cellular functions of polyamines, their role in physiology and pathology, and emphasize the current gaps in knowledge and potential future research directions.
Collapse
Affiliation(s)
- Ryan S Schibalski
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Anastasia S Shulha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Betty P Tsao
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
6
|
van Roekel M, Verhoeven CJ, Kamphof HD, Gordijn SJ, Ganzevoort W, Franx A, van Wieringen W, de Jonge A, Henrichs J. Unisex and Sex-Specific Prescriptive Fetal Growth Charts for Improved Detection of Small-for-Gestational-Age Babies in a Low-Risk Population: A post hoc Analysis of a Cluster-Randomized Study. Fetal Diagn Ther 2024; 51:571-582. [PMID: 39068914 DOI: 10.1159/000540554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Our aim was to develop and evaluate the performance of population-based sex-specific and unisex prescriptive fetal abdominal circumference growth charts in predicting small-for-gestational-age (SGA) birthweight, severe SGA (sSGA) birthweight, and severe adverse perinatal outcomes (SAPO) in a low-risk population. METHODS This is a post hoc analysis of the Dutch nationwide cluster-randomized IRIS study, encompassing ultrasound data of 7,704 low-risk women. IRIS prescriptive unisex and IRIS sex-specific abdominal circumference (AC) fetal growth charts were derived using quantile regression. As a comparison, we used the descriptive unisex Verburg chart, which is commonly applied in the Netherlands. Diagnostic parameters were calculated based on the 34-36 weeks' ultrasound. RESULTS Sensitivity rates for predicting SGA and sSGA birthweights were more than twofold higher based on the IRIS prescriptive sex-specific (respectively SGA 43%; sSGA 59%) and unisex (SGA 39%; sSGA 55%) charts, compared to the Verburg chart (SGA 16%; sSGA 23% both p < 0.01). Specificity rates were highest for Verburg (SGA 99%; sSGA 98%) and lowest for IRIS sex-specific (SGA 94%; sSGA 92%). Results for predicting SGA with SAPO were similar for the prescriptive charts (44%), and again higher than the Verburg chart (20%). The IRIS sex-specific chart identified significantly more males as SGA and sSGA (respectively, 42%; 60%, p < 0.001) than the IRIS unisex chart (respectively, 35%; 53% p < 0.01). CONCLUSION Our study demonstrates improved performance of both the IRIS sex-specific and unisex prescriptive fetal growth compared to the Verburg descriptive chart, doubling detection rates of SGA, sSGA, and SGA with SAPO. Additionally, the sex-specific chart outperformed the unisex chart in detecting SGA and sSGA. Our findings suggest the potential benefits of using prescriptive AC fetal growth charts in low-risk populations and emphasize the importance of considering customizing fetal growth charts for sex. Nevertheless, the increased sensitivity of these charts should be weighed against the decrease in specificity.
Collapse
Affiliation(s)
- Mariëlle van Roekel
- Amsterdam UMC location Vrije Universiteit Amsterdam, Midwifery Science, Amsterdam, The Netherlands
- Midwifery Academy Amsterdam Groningen, Inholland, Amsterdam, The Netherlands
| | - Corine J Verhoeven
- Amsterdam UMC location Vrije Universiteit Amsterdam, Midwifery Science, Amsterdam, The Netherlands
- Division of Midwifery, School of Health Sciences, University of Nottingham, Nottingham, UK
| | - Hester D Kamphof
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sanne J Gordijn
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wessel Ganzevoort
- Department of Obstetrics and Gynaecology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Arie Franx
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wessel van Wieringen
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, VU Medical Center, Amsterdam, The Netherlands
- Department of Mathematics, VU University Amsterdam, Amsterdam, The Netherlands
| | - Ank de Jonge
- Amsterdam UMC location Vrije Universiteit Amsterdam, Midwifery Science, Amsterdam, The Netherlands
- Department of Primary and Long-Term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jens Henrichs
- Amsterdam UMC location Vrije Universiteit Amsterdam, Midwifery Science, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Quality of Care, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Varghese B, Babu S, Jala A, Das P, Raju R, Borkar RM, Adela R. Integrative Placental Multi-Omics Analysis Reveals Perturbed Pathways and Potential Prognostic Biomarkers in Gestational Hypertension. Arch Med Res 2024; 55:102909. [PMID: 37984232 DOI: 10.1016/j.arcmed.2023.102909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Gestational hypertension (GH) is a severe complication that occurs after 20 weeks of pregnancy; however, its molecular mechanisms are not yet fully understood. OBJECTIVE Through this case-control discovery phase study, we aimed to find disease-specific candidate placental microRNAs (miRNAs) and metabolite markers for differentiating GH by integrating next-generation sequencing and metabolomics multi-omics analysis of placenta. Using small RNA sequencing and metabolomics of placental tissues of healthy pregnant (HP, n = 24) and GH subjects (n = 20), the transcriptome and metabolome were characterized in both groups. RESULTS The study identified a total of 44 downregulated placental miRNAs which includes three novel, three mature and 38 precursor miRNAs. Six miRNAs including three mature (hsa-miR-181a-5p, hsa-miR-498-5p, and hsa-miR-26b-5p) and three novel (NC_000016.10_1061, NC_000005.10_475, and NC_000001.11_53) were considered for final target prediction and functional annotation. Integrative analysis of differentially expressed miRNAs and metabolites yielded five pathways such as purine, glutathione, glycerophospholipid, inositol phosphate and β-alanine to be significantly perturbed in GH. We present fourteen genes (LPCAT1, LPCAT2, DGKH, PISD, GPAT2, PTEN, SACM1L, PGM2, AMPD3, AK7, AK3, CNDP1, IDH2, and ODC1) and eight metabolites (xanthosine, xanthine, spermine, glycine, CDP-Choline, glyceraldehyde 3-phosphate, β-alanine, and histidine) with potential to distinguish GH and HP. CONCLUSION The differential expression of miRNAs, their target genes, altered metabolites and metabolic pathways in GH patients were identified for the first time in our study. Further, the altered miRNAs and metabolites were integrated to build their inter-connectivity network. The findings obtained from our study may be used as a valuable source to further unravel the molecular pathways associated with GH and also for the evaluation of prognostic markers.
Collapse
Affiliation(s)
- Bincy Varghese
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Sreeranjini Babu
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India; Centre for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Aishwarya Jala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Panchanan Das
- Department of Obstetrics and Gynecology, Gauhati Medical College, Guwahati, Assam, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India.
| |
Collapse
|
8
|
Saddiki H, Zhang X, Colicino E, Wilson A, Kloog I, Wright RO, Wright RJ, Lesseur C. DNA methylation profiles reveal sex-specific associations between gestational exposure to ambient air pollution and placenta cell-type composition in the PRISM cohort study. Clin Epigenetics 2023; 15:188. [PMID: 38041176 PMCID: PMC10693032 DOI: 10.1186/s13148-023-01601-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Gestational exposure to ambient air pollution has been associated with adverse health outcomes for mothers and newborns. The placenta is a central regulator of the in utero environment that orchestrates development and postnatal life via fetal programming. Ambient air pollution contaminants can reach the placenta and have been shown to alter bulk placental tissue DNA methylation patterns. Yet the effect of air pollution on placental cell-type composition has not been examined. We aimed to investigate whether the exposure to ambient air pollution during gestation is associated with placental cell types inferred from DNA methylation profiles. METHODS We leveraged data from 226 mother-infant pairs in the Programming of Intergenerational Stress Mechanisms (PRISM) longitudinal cohort in the Northeastern US. Daily concentrations of fine particulate matter (PM2.5) at 1 km spatial resolution were estimated from a spatiotemporal model developed with satellite data and linked to womens' addresses during pregnancy and infants' date of birth. The proportions of six cell types [syncytiotrophoblasts, trophoblasts, stromal, endothelial, Hofbauer and nucleated red blood cells (nRBCs)] were derived from placental tissue 450K DNA methylation array. We applied compositional regression to examine overall changes in placenta cell-type composition related to PM2.5 average by pregnancy trimester. We also investigated the association between PM2.5 and individual cell types using beta regression. All analyses were performed in the overall sample and stratified by infant sex adjusted for covariates. RESULTS In male infants, first trimester (T1) PM2.5 was associated with changes in placental cell composition (p = 0.03), driven by a decrease [per one PM2.5 interquartile range (IQR)] of 0.037 in the syncytiotrophoblasts proportion (95% confidence interval (CI) [- 0.066, - 0.012]), accompanied by an increase in trophoblasts of 0.033 (95% CI: [0.009, 0.064]). In females, second and third trimester PM2.5 were associated with overall changes in placental cell-type composition (T2: p = 0.040; T3: p = 0.049), with a decrease in the nRBC proportion. Individual cell-type analysis with beta regression showed similar results with an additional association found for third trimester PM2.5 and stromal cells in females (decrease of 0.054, p = 0.024). CONCLUSION Gestational exposure to air pollution was associated with placenta cell composition. Further research is needed to corroborate these findings and evaluate their role in PM2.5-related impact in the placenta and consequent fetal programming.
Collapse
Affiliation(s)
- Hachem Saddiki
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA
| | - Xueying Zhang
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, The Kravis Children's Hospital, New York, NY, USA
- Institute of Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA
- Institute of Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ander Wilson
- Department of Statistics, Colorado State University, Fort Collins, CO, USA
| | - Itai Kloog
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA
- Institute of Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, The Kravis Children's Hospital, New York, NY, USA
- Institute of Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA.
- Institute of Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Thomas G, Syngelaki A, Hamed K, Perez-Montaño A, Panigassi A, Tuytten R, Nicolaides KH. Preterm preeclampsia screening using biomarkers: combining phenotypic classifiers into robust prediction models. Am J Obstet Gynecol MFM 2023; 5:101110. [PMID: 37752025 DOI: 10.1016/j.ajogmf.2023.101110] [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: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Preeclampsia screening is a critical component of antenatal care worldwide. Currently, the most developed screening test for preeclampsia at 11 to 13 weeks' gestation integrates maternal demographic characteristics and medical history with 3 biomarkers-serum placental growth factor, mean arterial pressure, and uterine artery pulsatility index-to identify approximately 75% of women who develop preterm preeclampsia with delivery before 37 weeks of gestation. It is generally accepted that further improvements to preeclampsia screening require the use of additional biomarkers. We recently reported that the levels of specific metabolites and metabolite ratios are associated with preterm preeclampsia. Notably, for several of these markers, preterm preeclampsia prediction varied according to maternal body mass index class. These findings motivated us to study whether patient classification allowed for combining metabolites with the current biomarkers more effectively to improve prediction of preterm preeclampsia. OBJECTIVE This study aimed to investigate whether metabolite biomarkers can improve biomarker-based preterm preeclampsia prediction in 3 screening resource scenarios according to the availability of: (1) placental growth factor, (2) placental growth factor+mean arterial pressure, and (3) placental growth factor+mean arterial pressure+uterine artery pulsatility index. STUDY DESIGN This was an observational case-control study, drawn from a large prospective screening study at 11 to 13 weeks' gestation on the prediction of pregnancy complications, conducted at King's College Hospital, London, United Kingdom. Maternal blood samples were also collected for subsequent research studies. We used liquid chromatography-mass spectrometry to quantify levels of 50 metabolites previously associated with pregnancy complications in plasma samples from singleton pregnancies. Biomarker data, normalized using multiples of medians, on 1635 control and 106 preterm preeclampsia pregnancies were available for model development. Modeling was performed using a methodology that generated a prediction model for preterm preeclampsia in 4 consecutive steps: (1) z-normalization of predictors, (2) combinatorial modeling of so-called (weak) classifiers in the unstratified patient set and in discrete patient strata based on body mass index and/or race, (3) selection of classifiers, and (4) aggregation of the selected classifiers (ie, bagging) into the final prediction model. The prediction performance of models was evaluated using the area under the receiver operating characteristic curve, and detection rate at 10% false-positive rate. RESULTS First, the predictor development methodology itself was evaluated. The patient set was split into a training set (2/3) and a test set (1/3) for predictor model development and internal validation. A prediction model was developed for each of the 3 different predictor panels, that is, placental growth factor+metabolites, placental growth factor+mean arterial pressure+metabolites, and placental growth factor+mean arterial pressure+uterine artery pulsatility index+metabolites. For all 3 models, the area under the receiver operating characteristic curve in the test set did not differ significantly from that of the training set. Next, a prediction model was developed using the complete data set for the 3 predictor panels. Among the 50 metabolites available for modeling, 26 were selected across the 3 prediction models; 21 contributed to at least 2 out of the 3 prediction models developed. Each time, area under the receiver operating characteristic curve and detection rate were significantly higher with the new prediction model than with the reference model. Markedly, the estimated detection rate with the placental growth factor+mean arterial pressure+metabolites prediction model in all patients was 0.58 (95% confidence interval, 0.49-0.70), a 15% increase (P<.001) over the detection rate of 0.43 (95% confidence interval, 0.33-0.55) estimated for the reference placental growth factor+mean arterial pressure. The same prediction model significantly improved detection in Black (14%) and White (19%) patients, and in the normal-weight group (18.5≤body mass index<25) and the obese group (body mass index≥30), with respectively 19% and 20% more cases detected, but not in the overweight group, when compared with the reference model. Similar improvement patterns in detection rates were found in the other 2 scenarios, but with smaller improvement amplitudes. CONCLUSION Metabolite biomarkers can be combined with the established biomarkers of placental growth factor, mean arterial pressure, and uterine artery pulsatility index to improve the biomarker component of early-pregnancy preterm preeclampsia prediction tests. Classification of the pregnant women according to the maternal characteristics of body mass index and/or race proved instrumental in achieving improved prediction. This suggests that maternal phenotyping can have a role in improving the prediction of obstetrical syndromes such as preeclampsia.
Collapse
Affiliation(s)
- Grégoire Thomas
- SQU4RE, Lokeren, Belgium (Dr Thomas); Metabolomic Diagnostics, Cork, Ireland (Drs Thomas, Panigassi, and Tuytten)
| | - Argyro Syngelaki
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| | - Karam Hamed
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| | - Anais Perez-Montaño
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| | - Ana Panigassi
- Metabolomic Diagnostics, Cork, Ireland (Drs Thomas, Panigassi, and Tuytten)
| | - Robin Tuytten
- Metabolomic Diagnostics, Cork, Ireland (Drs Thomas, Panigassi, and Tuytten).
| | - Kypros H Nicolaides
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| |
Collapse
|
10
|
O'Brien K, Wang Y. The Placenta: A Maternofetal Interface. Annu Rev Nutr 2023; 43:301-325. [PMID: 37603428 DOI: 10.1146/annurev-nutr-061121-085246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
The placenta is the gatekeeper between the mother and the fetus. Over the first trimester of pregnancy, the fetus is nourished by uterine gland secretions in a process known as histiotrophic nutrition. During the second trimester of pregnancy, placentation has evolved to the point at which nutrients are delivered to the placenta via maternal blood (hemotrophic nutrition). Over gestation, the placenta must adapt to these variable nutrient supplies, to alterations in maternal physiology and blood flow, and to dynamic changes in fetal growth rates. Numerous questions remain about the mechanisms used to transport nutrients to the fetus and the maternal and fetal determinants of this process. Growing data highlight the ability of the placenta to regulate this process. As new technologies and omics approaches are utilized to study this maternofetal interface, greater insight into this unique organ and its impact on fetal development and long-term health has been obtained.
Collapse
Affiliation(s)
- Kimberly O'Brien
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA; ,
| | - Yiqin Wang
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA; ,
| |
Collapse
|
11
|
Gong S, Gaccioli F, Aye ILMH, Avellino G, Cook E, Lawson ARJ, Harvey LMR, Smith GCS, Charnock-Jones DS. The human placenta exhibits a unique transcriptomic void. Cell Rep 2023; 42:112800. [PMID: 37453066 DOI: 10.1016/j.celrep.2023.112800] [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: 09/22/2022] [Revised: 02/08/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
The human placenta exhibits a unique genomic architecture with an unexpectedly high mutation burden and many uniquely expressed genes. The aim of this study is to identify transcripts that are uniquely absent or depleted in the placenta. Here, we show that 40 of 46 of the other organs have no selectively depleted transcripts and that, of the remaining six, the liver has the largest number, with 26. In contrast, the term placenta has 762 depleted transcripts. Gene Ontology analysis of this depleted set highlighted multiple pathways reflecting known unique elements of placental physiology. For example, transcripts associated with neuronal function are in the depleted set-as expected given the lack of placental innervation. However, this demonstrated overrepresentation of genes involved in mitochondrial function (p = 5.8 × 10-10), including PGC-1α, the master regulator of mitochondrial biogenesis, and genes involved in polyamine metabolism (p = 2.1 × 10-4).
Collapse
Affiliation(s)
- Sungsam Gong
- Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Francesca Gaccioli
- Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Irving L M H Aye
- Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Giulia Avellino
- Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emma Cook
- Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | | | | | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
12
|
Tsompanidis A, Blanken L, Broere-Brown ZA, van Rijn BB, Baron-Cohen S, Tiemeier H. Sex differences in placenta-derived markers and later autistic traits in children. Transl Psychiatry 2023; 13:256. [PMID: 37443170 DOI: 10.1038/s41398-023-02552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Autism is more prevalent in males and males on average score higher on measures of autistic traits. Placental function is affected significantly by the sex of the fetus. It is unclear if sex differences in placental function are associated with sex differences in the occurrence of autistic traits postnatally. To assess this, concentrations of angiogenesis-related markers, placental growth factor (PlGF) and soluble fms-like tyrosine kinase (sFlt-1) were assessed in maternal plasma of expectant women in the late 1st (mean= 13.5 [SD = 2.0] weeks gestation) and 2nd trimesters (mean=20.6 [SD = 1.2] weeks gestation), as part of the Generation R Study, Rotterdam, the Netherlands. Subsequent assessment of autistic traits in the offspring at age 6 was performed with the 18-item version of the Social Responsiveness Scale (SRS). Associations of placental protein concentrations with autistic traits were tested in sex-stratified and cohort-wide regression models. Cases with pregnancy complications or a later autism diagnosis (n = 64) were also assessed for differences in placenta-derived markers. sFlt-1 levels were significantly lower in males in both trimesters but showed no association with autistic traits. PlGF was significantly lower in male pregnancies in the 1st trimester, and significantly higher in the 2nd trimester, compared to female pregnancies. Higher PlGF levels in the 2nd trimester and the rate of PlGF increase were both associated with the occurrence of higher autistic traits (PlGF-2nd: n = 3469,b = 0.24 [SE = 0.11], p = 0.03) in both unadjusted and adjusted linear regression models that controlled for age, sex, placental weight and maternal characteristics. Mediation analyses showed that higher autistic traits in males compared to females were partly explained by higher PlGF or a faster rate of PlGF increase in the second trimester (PlGF-2nd: n = 3469, ACME: b = 0.005, [SE = 0.002], p = 0.004). In conclusion, higher PlGF levels in the 2nd trimester and a higher rate of PlGF increase are associated with both being male, and with a higher number of autistic traits in the general population.
Collapse
Affiliation(s)
- A Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.
| | - L Blanken
- The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Z A Broere-Brown
- The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - B B van Rijn
- The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - S Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - H Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Harvard University, Boston, USA
| |
Collapse
|
13
|
Unger HW, Hadiprodjo AJ, Gutman JR, Briand V, Fievet N, Valea I, Tinto H, D'Alessandro U, Landis SH, Ter Kuile F, Ouma P, Oneko M, Mwapasa V, Slutsker L, Terlouw DJ, Kariuki S, Ayisi J, Nahlen B, Desai M, Madanitsa M, Kalilani-Phiri L, Ashorn P, Maleta K, Tshefu-Kitoto A, Mueller I, Stanisic D, Cates J, Van Eijk AM, Ome-Kaius M, Aitken EH, Rogerson SJ. Fetal sex and risk of pregnancy-associated malaria in Plasmodium falciparum-endemic regions: a meta-analysis. Sci Rep 2023; 13:10310. [PMID: 37365258 DOI: 10.1038/s41598-023-37431-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/21/2023] [Indexed: 06/28/2023] Open
Abstract
In areas of moderate to intense Plasmodium falciparum transmission, malaria in pregnancy remains a significant cause of low birth weight, stillbirth, and severe anaemia. Previously, fetal sex has been identified to modify the risks of maternal asthma, pre-eclampsia, and gestational diabetes. One study demonstrated increased risk of placental malaria in women carrying a female fetus. We investigated the association between fetal sex and malaria in pregnancy in 11 pregnancy studies conducted in sub-Saharan African countries and Papua New Guinea through meta-analysis using log binomial regression fitted to a random-effects model. Malaria infection during pregnancy and delivery was assessed using light microscopy, polymerase chain reaction, and histology. Five studies were observational studies and six were randomised controlled trials. Studies varied in terms of gravidity, gestational age at antenatal enrolment and bed net use. Presence of a female fetus was associated with malaria infection at enrolment by light microscopy (risk ratio 1.14 [95% confidence interval 1.04, 1.24]; P = 0.003; n = 11,729). Fetal sex did not associate with malaria infection when other time points or diagnostic methods were used. There is limited evidence that fetal sex influences the risk of malaria infection in pregnancy.
Collapse
Affiliation(s)
- Holger W Unger
- Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Darwin, NT, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Anastasia Jessica Hadiprodjo
- Department of Medicine (RMH), Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Valerie Briand
- Université de Paris, UMR261, IRD, Paris, France
- Epicentre MSF, Paris, France
| | | | - Innocent Valea
- Unite de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de La Santé-DRCO, Nanoro, Burkina Faso
- Departement de Recherche Clinique, Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Halidou Tinto
- Unite de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de La Santé-DRCO, Nanoro, Burkina Faso
- Departement de Recherche Clinique, Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Umberto D'Alessandro
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
- London School of Hygiene and Tropical Medicine, London, UK
| | | | - Feiko Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Peter Ouma
- Kenya Medical Research Institute (KEMRI)/Centre for Global Health Research, Kisumu, Kenya
| | - Martina Oneko
- Kenya Medical Research Institute (KEMRI)/Centre for Global Health Research, Kisumu, Kenya
| | - Victor Mwapasa
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Laurence Slutsker
- Malaria and Neglected Tropical Diseases, Center for Malaria Control and Elimination, PATH, Seattle, WA, USA
| | - Dianne J Terlouw
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Simon Kariuki
- Kenya Medical Research Institute (KEMRI)/Centre for Global Health Research, Kisumu, Kenya
| | - John Ayisi
- Kenya Medical Research Institute (KEMRI)/Centre for Global Health Research, Kisumu, Kenya
| | | | - Meghna Desai
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Linda Kalilani-Phiri
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Per Ashorn
- Faculty of Medicine and Health Technology, Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
- Department for Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Kenneth Maleta
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | | | - Ivo Mueller
- Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Danielle Stanisic
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Jordan Cates
- Department of Epidemiology, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Anna Maria Van Eijk
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Maria Ome-Kaius
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Elizabeth H Aitken
- Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Medicine (RMH), Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Tsompanidis A, Warrier V, Baron-Cohen S. The genetics of autism and steroid-related traits in prenatal and postnatal life. Front Endocrinol (Lausanne) 2023; 14:1126036. [PMID: 37223033 PMCID: PMC10200920 DOI: 10.3389/fendo.2023.1126036] [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: 12/17/2022] [Accepted: 04/18/2023] [Indexed: 05/25/2023] Open
Abstract
Background Autism likelihood is a largely heritable trait. Autism prevalence has a skewed sex ratio, with males being diagnosed more often than females. Steroid hormones play a mediating role in this, as indicated by studies of both prenatal biology and postnatal medical conditions in autistic men and women. It is currently unclear if the genetics of steroid regulation or production interact with the genetic liability for autism. Methods To address this, two studies were conducted using publicly available datasets, which focused respectively on rare genetic variants linked to autism and neurodevelopmental conditions (study 1) and common genetic variants (study 2) for autism. In Study 1 an enrichment analysis was conducted, between autism-related genes (SFARI database) and genes that are differentially expressed (FDR<0.1) between male and female placentas, in 1st trimester chorionic villi samples of viable pregnancies (n=39). In Study 2 summary statistics of genome wide association studies (GWAS) were used to investigate the genetic correlation between autism and bioactive testosterone, estradiol and postnatal PlGF levels, as well as steroid-related conditions such as polycystic ovaries syndrome (PCOS), age of menarche, and androgenic alopecia. Genetic correlation was calculated based on LD Score regression and results were corrected for multiple testing with FDR. Results In Study 1, there was significant enrichment of X-linked autism genes in male-biased placental genes, independently of gene length (n=5 genes, p<0.001). In Study 2, common genetic variance associated with autism did not correlate to the genetics for the postnatal levels of testosterone, estradiol or PlGF, but was associated with the genotypes associated with early age of menarche in females (b=-0.109, FDR-q=0.004) and protection from androgenic alopecia for males (b=-0.135, FDR-q=0.007). Conclusion The rare genetic variants associated with autism appear to interact with placental sex differences, while the common genetic variants associated with autism appear to be involved in the regulation of steroid-related traits. These lines of evidence indicate that the likelihood for autism is partly linked to factors mediating physiological sex differences throughout development.
Collapse
Affiliation(s)
- Alex Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
15
|
Tsompanidis A, Hampton S, Aydin E, Allison C, Holt R, Baron-Cohen S. Mini-puberty testosterone and infant autistic traits. Front Endocrinol (Lausanne) 2023; 14:1126023. [PMID: 37091846 PMCID: PMC10113441 DOI: 10.3389/fendo.2023.1126023] [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: 12/16/2022] [Accepted: 02/24/2023] [Indexed: 04/08/2023] Open
Abstract
Background Levels of steroid hormones in the first three months of life, a period referred to as 'mini-puberty', are one of the earliest physiological differences between typical males and females postnatally. Autistic traits also show consistent typical sex differences in later infancy, after the 18th month of life. Previous studies have shown prenatal testosterone is associated with later levels of autistic traits. Studies testing if postnatal testosterone levels are associated with autistic traits have reported null results. No studies to date have investigated mini-puberty longitudinally or tested for interactions with baseline sex differences or familial likelihood of autism. Methods The 'Cambridge Human Imaging and Longitudinal Development Study' (CHILD) is a prospective enriched cohort study in Cambridge, UK. It includes physiological measurements in early infancy, as well as neurodevelopmental follow-ups over the first two years of life. A subset of the cohort also includes children with a family history of autism (a diagnosed parent or sibling). Salivary testosterone levels were assessed at two time-points, just after the 2nd and 6th month of life. Autistic traits were measured using the Quantitative Checklist of Autism in Toddlers (Q-CHAT) when the children were 18 months of age. Results Salivary testosterone levels were significantly higher during 'mini-puberty' in the 2nd and 3rd month of life, compared to after the 6th month of life, in both males and females. There was no significant sex difference at either time-point. Log-transformed testosterone levels were not associated with autistic traits (Q-CHAT). There was no interaction effect with infant sex, autism family history or baseline testosterone levels after mini-puberty (at >6 months of age). Conclusion Both male and female infants have elevated levels of salivary testosterone during mini-puberty but in this relatively small sample this was not associated with their later autistic traits at 18 months or their family history of autism. This suggests that prenatal rather than postnatal testosterone levels are more relevant for understanding the causes of autism. Future studies should test these relationships in larger samples.
Collapse
Affiliation(s)
- Alex Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Hampton
- York Trials Unit, University of York, York, United Kingdom
| | - Ezra Aydin
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Carrie Allison
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Rosemary Holt
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
Thangaraj SV, Kachman M, Halloran KM, Sinclair KD, Lea R, Bellingham M, Evans NP, Padmanabhan V. Developmental programming: Preconceptional and gestational exposure of sheep to a real-life environmental chemical mixture alters maternal metabolome in a fetal sex-specific manner. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 864:161054. [PMID: 36565874 PMCID: PMC10322214 DOI: 10.1016/j.scitotenv.2022.161054] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 05/21/2023]
Abstract
BACKGROUND Everyday, humans are exposed to a mixture of environmental chemicals some of which have endocrine and/or metabolism disrupting actions which may contribute to non-communicable diseases. The adverse health impacts of real-world chemical exposure, characterized by chronic low doses of a mixture of chemicals, are only recently emerging. Biosolids derived from human waste represent the environmental chemical mixtures humans are exposed to in real life. Prior studies in sheep have shown aberrant reproductive and metabolic phenotypes in offspring after maternal biosolids exposure. OBJECTIVE To determine if exposure to biosolids perturbs the maternal metabolic milieu of pregnant ewes, in a fetal sex-specific manner. METHODS Ewes were grazed on inorganic fertilizer (Control) or biosolids-treated pastures (BTP) from before mating and throughout gestation. Plasma from pregnant ewes (Control n = 15, BTP n = 15) obtained mid-gestation were analyzed by untargeted metabolomics. Metabolites were identified using Agilent MassHunter. Multivariate analyses were done using MetaboAnalyst 5.0 and confirmed using SIMCA. RESULTS Univariate and multivariate analysis of 2301 annotated metabolites identified 193 differentially abundant metabolites (DM) between control and BTP sheep. The DM primarily belonged to the super-class of lipids and organic acids. 15-HeTrE, oleamide, methionine, CAR(3:0(OH)) and pyroglutamic acid were the top DM and have been implicated in the regulation of fetal growth and development. Fetal sex further exacerbated differences in metabolite profiles in the BTP group. The organic acids class of metabolites was abundant in animals with male fetuses. Prenol lipid, sphingolipid, glycerolipid, alkaloid, polyketide and benzenoid classes showed fetal sex-specific responses to biosolids. DISCUSSION Our study illustrates that exposure to biosolids significantly alters the maternal metabolome in a fetal sex-specific manner. The altered metabolite profile indicates perturbations to fatty acid, arginine, branched chain amino acid and one‑carbon metabolism. These factors are consistent with, and likely contribute to, the adverse phenotypic outcomes reported in the offspring.
Collapse
Affiliation(s)
- S V Thangaraj
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - M Kachman
- MM BRCF Metabolomics Core, University of Michigan, Ann Arbor, MI, USA
| | - K M Halloran
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - K D Sinclair
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - R Lea
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - M Bellingham
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - N P Evans
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - V Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
17
|
Ten-Doménech I, Cascant-Vilaplana MM, Navarro-Esteve V, Felderer B, Moreno-Giménez A, Rienda I, Gormaz M, Moreno-Torres M, Pérez-Guaita D, Quintás G, Kuligowski J. Metabolomic Diversity of Human Milk Cells over the Course of Lactation-A Preliminary Study. Nutrients 2023; 15:nu15051100. [PMID: 36904100 PMCID: PMC10005050 DOI: 10.3390/nu15051100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Human milk (HM) is a complex biofluid containing a wide cell variety including epithelial cells and leukocytes. However, the cellular compositions and their phenotypic properties over the course of lactation are poorly understood. The aim of this preliminary study was to characterize the cellular metabolome of HM over the course of lactation. Cells were isolated via centrifugation and the cellular fraction was characterized via cytomorphology and immunocytochemical staining. Cell metabolites were extracted and analyzed using ultra-performance liquid chromatography coupled to quadrupole time-of-flight mass spectrometry (UPLC-QqTOF-MS) in the positive and negative electrospray ionization modes. Immunocytochemical analysis revealed a high variability of the number of detected cells with relative median abundances of 98% of glandular epithelial cells, 1% of leukocytes, and 1% of keratinocytes. Significant correlations between the milk postnatal age with percentage of epithelial cells and leukocytes, and with total cell count were observed. Results from the Hierarchical Cluster Analysis of immunocytochemical profiles were very similar to those observed in the analysis of the metabolomic profiles. In addition, metabolic pathway analysis showed alterations in seven metabolic pathways correlating with postnatal age. This work paves the way for future investigations on changes in the metabolomic fraction of the cellular compartment of HM.
Collapse
Affiliation(s)
- Isabel Ten-Doménech
- Neonatal Research Group, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Mari Merce Cascant-Vilaplana
- Neonatal Research Group, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Víctor Navarro-Esteve
- Neonatal Research Group, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Spain
| | - Birgit Felderer
- Neonatal Research Group, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
- Master Program Biotechnical Processes, Austrian Biotech University of Applied Sciences, Konrad Lorenz-Strasse 10, 3430 Tulln, Austria
| | - Alba Moreno-Giménez
- Neonatal Research Group, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Iván Rienda
- Servicio de Anatomía Patológica, University & Polytechnic Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - María Gormaz
- Neonatal Research Group, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
- Division of Neonatology, University & Polytechnic Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Marta Moreno-Torres
- Unidad de Hepatología Experimental y Trasplante Hepático, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
- Department of Biochemistry and Molecular Biology, University of Valencia, C/Blasco Ibáñez 15, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - David Pérez-Guaita
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Spain
| | - Guillermo Quintás
- Health and Biomedicine, Leitat Technological Center, Carrer de la Innovació, 2, 08225 Terrassa, Spain
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute Hospital La Fe, Avda Fernando Abril Martorell 106, 46026 Valencia, Spain
- Correspondence: ; Tel.: +34-96-1246661
| |
Collapse
|
18
|
Gaccioli F, Sovio U, Gong S, Cook E, Charnock-Jones DS, Smith GC. Increased Placental sFLT1 (Soluble fms-Like Tyrosine Kinase Receptor-1) Drives the Antiangiogenic Profile of Maternal Serum Preceding Preeclampsia but Not Fetal Growth Restriction. Hypertension 2023; 80:325-334. [PMID: 35866422 PMCID: PMC9847691 DOI: 10.1161/hypertensionaha.122.19482] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Preeclampsia and fetal growth restriction (FGR) are both associated with an increased ratio of sFLT1 (soluble fms-like tyrosine kinase-1) to PlGF (placenta growth factor) in maternal serum. In preeclampsia, it is assumed that increased placental release of sFLT1 results in PlGF being bound and inactivated. However, direct evidence for this model is incomplete, and it is unclear whether the same applies in FGR. METHODS We conducted a prospective cohort study where we followed 4212 women having first pregnancies from their dating ultrasound, obtained blood samples serially through the pregnancy, and performed systematic sampling of the placenta after delivery. The aim of the present study was to determine the relationship between protein levels of sFLT1 and PlGF in maternal serum measured at ≈36 weeks and placental tissue lysates obtained after term delivery in 82 women with preeclampsia, 50 women with FGR, and 132 controls. RESULTS The sFLT1:PlGF ratio was increased in both preeclampsia and FGR in both the placenta and maternal serum. However, in preeclampsia, the maternal serum ratio of sFLT1:PlGF was strongly associated with placental sFLT1 level (r=0.45; P<0.0001) but not placental PlGF level (r=-0.17; P=0.16). In contrast, in FGR, the maternal serum ratio of sFLT1:PlGF was strongly associated with placental PlGF level (r=-0.35; P=0.02) but not sFLT1 level (r=0.04; P=0.81). CONCLUSIONS We conclude that the elevated sFLT1:PlGF ratio is primarily driven by increased placental sFLT1 in preeclampsia, whereas in FGR, it is primarily driven by decreased placental PlGF.
Collapse
Affiliation(s)
- Francesca Gaccioli
- Department of Obstetrics and Gynaecology (F.G., U.S., S.G., E.C., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
- Centre for Trophoblast Research (F.G., U.S., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
| | - Ulla Sovio
- Department of Obstetrics and Gynaecology (F.G., U.S., S.G., E.C., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
- Centre for Trophoblast Research (F.G., U.S., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
| | - Sungsam Gong
- Department of Obstetrics and Gynaecology (F.G., U.S., S.G., E.C., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
| | - Emma Cook
- Department of Obstetrics and Gynaecology (F.G., U.S., S.G., E.C., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
| | - D. Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology (F.G., U.S., S.G., E.C., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
- Centre for Trophoblast Research (F.G., U.S., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
| | - Gordon C.S. Smith
- Department of Obstetrics and Gynaecology (F.G., U.S., S.G., E.C., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
- Centre for Trophoblast Research (F.G., U.S., D.S.C.-J., G.C.S.S.), University of Cambridge, United Kingdom
| |
Collapse
|
19
|
Kedziora SM, Obermayer B, Sugulle M, Herse F, Kräker K, Haase N, Langmia IM, Müller DN, Staff AC, Beule D, Dechend R. Placental Transcriptome Profiling in Subtypes of Diabetic Pregnancies Is Strongly Confounded by Fetal Sex. Int J Mol Sci 2022; 23:ijms232315388. [PMID: 36499721 PMCID: PMC9740420 DOI: 10.3390/ijms232315388] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The placenta is a temporary organ with a unique structure and function to ensure healthy fetal development. Placental dysfunction is involved in pre-eclampsia (PE), fetal growth restriction, preterm birth, and gestational diabetes mellitus (GDM). A diabetic state affects maternal and fetal health and may lead to functional alterations of placental metabolism, inflammation, hypoxia, and weight, amplifying the fetal stress. The placental molecular adaptations to the diabetic environment and the adaptive spatio-temporal consequences to elevated glucose or insulin are largely unknown (2). We aimed to identify gene expression signatures related to the diabetic placental pathology of placentas from women with diabetes mellitus. Human placenta samples (n = 77) consisting of healthy controls, women with either gestational diabetes mellitus (GDM), type 1 or type 2 diabetes, and women with GDM, type 1 or type 2 diabetes and superimposed PE were collected. Interestingly, gene expression differences quantified by total RNA sequencing were mainly driven by fetal sex rather than clinical diagnosis. Association of the principal components with a full set of clinical patient data identified fetal sex as the single main explanatory variable. Accordingly, placentas complicated by type 1 and type 2 diabetes showed only few differentially expressed genes, while possible effects of GDM and diabetic pregnancy complicated by PE were not identifiable in this cohort. We conclude that fetal sex has a prominent effect on the placental transcriptome, dominating and confounding gene expression signatures resulting from diabetes mellitus in settings of well-controlled diabetic disease. Our results support the notion of placenta as a sexual dimorphic organ.
Collapse
Affiliation(s)
- Sarah M. Kedziora
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Benedikt Obermayer
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Core Unit Bioinformatics, 10117 Berlin, Germany
| | - Meryam Sugulle
- Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, 0424 Oslo, Norway
| | - Florian Herse
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Immaculate M. Langmia
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
| | - Dominik N. Müller
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Anne Cathrine Staff
- Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, 0424 Oslo, Norway
| | - Dieter Beule
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Core Unit Bioinformatics, 10117 Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
- HELIOS Clinic, Department of Cardiology and Nephrology, 13125 Berlin, Germany
- Correspondence: ; Tel.: +49-30-4505-40301
| |
Collapse
|
20
|
Chai N, Zheng H, Zhang H, Li L, Yu X, Wang L, Bi X, Yang L, Niu T, Liu X, Zhao Y, Dong L. Spermidine Alleviates Intrauterine Hypoxia-Induced Offspring Newborn Myocardial Mitochondrial Damage in Rats by Inhibiting Oxidative Stress and Regulating Mitochondrial Quality Control. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e133776. [PMID: 36945337 PMCID: PMC10024813 DOI: 10.5812/ijpr-133776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/24/2023] [Accepted: 02/01/2023] [Indexed: 03/05/2023]
Abstract
Background Intrauterine hypoxia (IUH) increases the risk of cardiovascular diseases in offspring. As a reactive oxygen species (ROS) scavenger, polyamine spermidine (SPD) is essential for embryonic and fetal survival and growth. However, further studies on the SPD protection and mechanisms for IUH-induced heart damage in offspring are required. Objectives This study aimed to investigate the preventive effects of prenatal SPD treatment on IUH-induced heart damage in newborn offspring rats and its underlying mitochondrial-related mechanism. Methods The rat model of IUH was established by exposure to 10% O2 seven days before term. Meanwhile, for seven days, the pregnant rats were given SPD (5 mg.kg-1.d-1; ip). The one-day offspring rats were sacrificed to assess several parameters, including growth development, heart damage, cardiomyocytes proliferation, myocardial oxidative stress, cell apoptosis, and mitochondrial function, and have mitochondrial quality control (MQC), including mitophagy, mitochondrial biogenesis, and mitochondrial fusion/fission. In in vitro experiments, primary cardiomyocytes were subjected to hypoxia with or without SPD for 24 hours. Results IUH decreased body weight, heart weight, cardiac Ki67 expression, the activity of SOD, and the CAT and adenosine 5'-triphosphate (ATP) levels and increased the BAX/BCL2 expression, and TUNEL-positive nuclei numbers. Furthermore, IUH also caused mitochondrial structure abnormality, dysfunction, and decreased mitophagy (decreased number of mitophagosomes), declined mitochondrial biogenesis (decreased expression of SIRT-1, PGC-1α, NRF-2, and TFAM), and led to fission/fusion imbalance (increased percentage of mitochondrial fragments, increased DRP1 expression, and decreased MFN2 expression) in the myocardium. Surprisingly, SPD treatment normalized the variations in the IUH-induced parameters. Furthermore, SPD also prevented hypoxia-induced ROS accumulation, mitochondrial membrane potential decay, and the mitophagy decrease in cardiomyocytes. Conclusion Maternal SPD treatment caused IUH-induced heart damage in newborn offspring rats by improving the myocardial mitochondrial function via anti-oxidation and anti-apoptosis, and regulating MQC.
Collapse
Affiliation(s)
- Nannan Chai
- College of Nursing, Chifeng University, Chifeng, China
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Haihong Zheng
- The Second Affiliated Hospital Department of the Laboratory Animal, Harbin Medical University, Harbin, China
| | - Hao Zhang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Lingxu Li
- Department of Nephrology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xue Yu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Liyi Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Bi
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lihong Yang
- College of Nursing, Chifeng University, Chifeng, China
| | - Tongxu Niu
- College of Nursing, Chifeng University, Chifeng, China
| | - Xiujuan Liu
- College of Nursing, Chifeng University, Chifeng, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- Corresponding Author: Department of Pathophysiology, Harbin Medical University, Harbin, China.
| | - Lijie Dong
- Neonatal Intensive Care Unit, Harbin Children’s Hospital, Harbin, China
- Corresponding Author: Neonatal Intensive Care Unit, Harbin Children’s Hospital, Harbin, China.
| |
Collapse
|
21
|
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: 90] [Impact Index Per Article: 30.0] [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
|
22
|
Monosomy X in isogenic human iPSC-derived trophoblast model impacts expression modules preserved in human placenta. Proc Natl Acad Sci U S A 2022; 119:e2211073119. [PMID: 36161909 DOI: 10.1073/pnas.2211073119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mammalian sex chromosomes encode homologous X/Y gene pairs that were retained on the Y chromosome in males and escape X chromosome inactivation (XCI) in females. Inferred to reflect X/Y pair dosage sensitivity, monosomy X is a leading cause of miscarriage in humans with near full penetrance. This phenotype is shared with many other mammals but not the mouse, which offers sophisticated genetic tools to generate sex chromosomal aneuploidy but also tolerates its developmental impact. To address this critical gap, we generated X-monosomic human induced pluripotent stem cells (hiPSCs) alongside otherwise isogenic euploid controls from male and female mosaic samples. Phased genomic variants in these hiPSC panels enable systematic investigation of X/Y dosage-sensitive features using in vitro models of human development. Here, we demonstrate the utility of these validated hiPSC lines to test how X/Y-linked gene dosage impacts a widely used model for human syncytiotrophoblast development. While these isogenic panels trigger a GATA2/3- and TFAP2A/C-driven trophoblast gene circuit irrespective of karyotype, differential expression implicates monosomy X in altered levels of placental genes and in secretion of placental growth factor (PlGF) and human chorionic gonadotropin (hCG). Remarkably, weighted gene coexpression network modules that significantly reflect these changes are also preserved in first-trimester chorionic villi and term placenta. Our results suggest monosomy X may skew trophoblast cell type composition and function, and that the combined haploinsufficiency of the pseudoautosomal region likely plays a key role in these changes.
Collapse
|
23
|
Braun AE, Mitchel OR, Gonzalez TL, Sun T, Flowers AE, Pisarska MD, Winn VD. Sex at the interface: the origin and impact of sex differences in the developing human placenta. Biol Sex Differ 2022; 13:50. [PMID: 36114567 PMCID: PMC9482177 DOI: 10.1186/s13293-022-00459-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/02/2022] [Indexed: 11/20/2022] Open
Abstract
The fetal placenta is a source of hormones and immune factors that play a vital role in maintaining pregnancy and facilitating fetal growth. Cells in this extraembryonic compartment match the chromosomal sex of the embryo itself. Sex differences have been observed in common gestational pathologies, highlighting the importance of maternal immune tolerance to the fetal compartment. Over the past decade, several studies examining placentas from term pregnancies have revealed widespread sex differences in hormone signaling, immune signaling, and metabolic functions. Given the rapid and dynamic development of the human placenta, sex differences that exist at term (37–42 weeks gestation) are unlikely to align precisely with those present at earlier stages when the fetal–maternal interface is being formed and the foundations of a healthy or diseased pregnancy are established. While fetal sex as a variable is often left unreported in studies performing transcriptomic profiling of the first-trimester human placenta, four recent studies have specifically examined fetal sex in early human placental development. In this review, we discuss the findings from these publications and consider the evidence for the genetic, hormonal, and immune mechanisms that are theorized to account for sex differences in early human placenta. We also highlight the cellular and molecular processes that are most likely to be impacted by fetal sex and the evolutionary pressures that may have given rise to these differences. With growing recognition of the fetal origins of health and disease, it is important to shed light on sex differences in early prenatal development, as these observations may unlock insight into the foundations of sex-biased pathologies that emerge later in life. Placental sex differences exist from early prenatal development, and may help explain sex differences in pregnancy outcomes. Transcriptome profiling of early to mid-gestation placenta reveals that immune signaling is a hub of early prenatal sex differences. Differentially expressed genes between male and female placenta fall into the following functional associations: chromatin modification, transcription, splicing, translation, signal transduction, metabolic regulation, cell death and autophagy regulation, ubiquitination, cell adhesion and cell–cell interaction. Placental sex differences likely reflect the interaction of cell-intrinsic chromosome complement with extrinsic endocrine signals from the fetal compartment that accompany gonadal differentiation. Understanding the mechanisms behind sex differences in placental development and function will provide key insight into molecular targets that can be modulated to improve sex-biased obstetrical complications.
Collapse
|
24
|
Mirzakhani H, Weiss ST. Fetal sex and risk of preeclampsia: Dose maternal race matter? J Matern Fetal Neonatal Med 2022; 35:3379-3387. [PMID: 32924669 PMCID: PMC7954987 DOI: 10.1080/14767058.2020.1818221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/21/2020] [Accepted: 08/30/2020] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To examine whether maternal race could affect the relationship between fetal sex and preeclampsia. MATERIAL AND METHODS This study was a cohort analysis using prospectively collected data from pregnant women who participated in the Vitamin Antenatal Asthma Reduction Trial (VDAART). Preeclampsia was the secondary outcome of VDAART. We examined the association of fetal sex with preeclampsia and its potential interaction with maternal race in 813 pregnant women (8% with preeclampsia) in logistic regression models with adjustment for preterm birth (<37 weeks of gestation), maternal age, education, and body mass index at enrollment and clinical center. We further conducted a race stratified analysis and also examined whether any observed association was dependent on the gestational age at delivery and prematurity. RESULTS In an analysis of all races combined, preeclampsia was not more common among pregnant women with a male fetus compared to those with a female fetus (odds ratio [OR] = 1.3, 95% CI = 0.81, 2.24). There was an interaction between African American race and fetal sex in association with preeclampsia after adjustment for preterm delivery and other potential confounders (p = .014). In race stratified analyses, we observed higher odds of preeclampsia among African American pregnant women who carried male fetuses after adjustment for preterm delivery and other potential confounders (adjusted OR = 2.4, 95% CI = 1.12, 5.60). CONCLUSION We observed fetal sexual dimorphic differences in the occurrence of preeclampsia in African American women, but not in Whites. Information on fetal sex may ultimately improve the prediction of pre-eclampsia in African American mothers, who might be at higher risk for this adverse condition in pregnancy.
Collapse
Affiliation(s)
- Hooman Mirzakhani
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Partners Center for Personalized Medicine, Partners Health Care, Boston, MA, USA
| |
Collapse
|
25
|
Mitochondria Targeted Antioxidant Significantly Alleviates Preeclampsia Caused by 11β-HSD2 Dysfunction via OPA1 and MtDNA Maintenance. Antioxidants (Basel) 2022; 11:antiox11081505. [PMID: 36009224 PMCID: PMC9404992 DOI: 10.3390/antiox11081505] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/03/2022] Open
Abstract
We have previously demonstrated that placental 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) dysfunction contributes to PE pathogenesis. We sought to elucidate molecular mechanisms underlying 11β-HSD2 dysfunction-induced PE and to seek potential therapeutic targets using a 11β-HSD2 dysfunction-induced PE-like rat model as well as cultured extravillous trophoblasts (EVTs) since PE begins with impaired function of EVTs. In 11β-HSD2 dysfunction-induced PE-like rat model, we revealed that placental mitochondrial dysfunction occurred, which was associated with mitDNA instability and impaired mitochondrial dynamics, such as decreased optic atrophy 1 (OPA1) expression. MitoTEMPO treatment significantly alleviated the hallmark of PE-like features and improved mitDNA stability and mitochondrial dynamics in the placentas of rat PE-like model. In cultured human EVTs, we found that 11β-HSD2 dysfunction led to mitochondrial dysfunction and disrupted mtDNA stability. MitoTEMPO treatment improved impaired invasion and migration induced by 11β-HSD2 dysfunction in cultured EVTs. Further, we revealed that OPA1 was one of the key factors that mediated 11β-HSD2 dysfunction-induced excess ROS production, mitochondrial dysfunction and mtDNA reduction. Our data indicates that 11β-HSD2 dysfunction causes mitochondrial dysfunctions, which impairs trophoblast function and subsequently results in PE development. Our study immediately highlights that excess ROS is a potential therapeutic target for PE.
Collapse
|
26
|
Sovio U, Clayton GL, Cook E, Gaccioli F, Charnock-Jones DS, Lawlor DA, Smith GCS. Metabolomic Identification of a Novel, Externally Validated Predictive Test for Gestational Diabetes Mellitus. J Clin Endocrinol Metab 2022; 107:e3479-e3486. [PMID: 35436338 PMCID: PMC9282248 DOI: 10.1210/clinem/dgac240] [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: 01/11/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Undiagnosed gestational diabetes mellitus (GDM) is a major preventable cause of stillbirth. In the United Kingdom, women are selected for diagnostic testing for GDM based on risk factors, including body mass index (BMI) > 30 kg/m2. OBJECTIVE To improve the prediction of GDM using metabolomics. METHODS We performed metabolomics on maternal serum from the Pregnancy Outcome Prediction (POP) study at 12 and 20 weeks of gestational age (wkGA; 185 GDM cases and 314 noncases). Predictive metabolites were internally validated using the 28 wkGA POP study serum sample and externally validated using 24- to 28-wkGA fasting plasma from the Born in Bradford (BiB) cohort (349 GDM cases and 2347 noncases). The predictive ability of a model including the metabolites was compared with BMI > 30 kg/m2 in the POP study. RESULTS Forty-seven predictive metabolites were identified using the 12- and 20-wkGA samples. At 28 wkGA, 4 of these [mannose, 4-hydroxyglutamate, 1,5-anhydroglucitol, and lactosyl-N-palmitoyl-sphingosine (d18:1/16:0)] independently increased the bootstrapped area under the receiver operating characteristic curve (AUC) by >0.01. All 4 were externally validated in the BiB samples (P = 2.6 × 10-12, 2.2 × 10-13, 6.9 × 10-28, and 2.6 × 10-17, respectively). In the POP study, BMI > 30 kg/m2 had a sensitivity of 28.7% (95% CI 22.3-36.0%) and a specificity of 85.4% whereas at the same level of specificity, a predictive model using age, BMI, and the 4 metabolites had a sensitivity of 60.2% (95% CI 52.6-67.4%) and an AUC of 0.82 (95% CI 0.78-0.86). CONCLUSIONS We identified 4 strongly and independently predictive metabolites for GDM that could have clinical utility in screening for GDM.
Collapse
Affiliation(s)
- Ulla Sovio
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Gemma L Clayton
- NIHR Bristol Biomedical Research Centre, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Emma Cook
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Francesca Gaccioli
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Deborah A Lawlor
- NIHR Bristol Biomedical Research Centre, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
27
|
Phung TN, Olney KC, Pinto BJ, Silasi M, Perley L, O’Bryan J, Kliman HJ, Wilson MA. X chromosome inactivation in the human placenta is patchy and distinct from adult tissues. HGG ADVANCES 2022; 3:100121. [PMID: 35712697 PMCID: PMC9194956 DOI: 10.1016/j.xhgg.2022.100121] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
In humans, one of the X chromosomes in genetic females is inactivated by a process called X chromosome inactivation (XCI). Variation in XCI across the placenta may contribute to observed sex differences and variability in pregnancy outcomes. However, XCI has predominantly been studied in human adult tissues. Here, we sequenced and analyzed DNA and RNA from two locations from 30 full-term pregnancies. Implementing an allele-specific approach to examine XCI, we report evidence that XCI in the human placenta is patchy, with large patches of either maternal or paternal X chromosomes inactivated. Further, using similar measurements, we show that this is in contrast to adult tissues, which generally exhibit mosaic X inactivation, where bulk samples exhibit both maternal and paternal X chromosome expression. Further, by comparing skewed samples in placenta and adult tissues, we identify genes that are uniquely inactivated or expressed in the placenta compared with adult tissues, highlighting the need for tissue-specific maps of XCI.
Collapse
Affiliation(s)
- Tanya N. Phung
- Center for Evolution and Medicine, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
| | - Kimberly C. Olney
- Center for Evolution and Medicine, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
| | - Brendan J. Pinto
- Center for Evolution and Medicine, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
- Department of Zoology, Milwaukee Public Museum, Milwaukee, WI 53233, USA
| | - Michelle Silasi
- Department of Maternal-Fetal Medicine, Mercy Hospital St. Louis, St. Louis, MO 63141, USA
| | - Lauren Perley
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jane O’Bryan
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Harvey J. Kliman
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Melissa A. Wilson
- Center for Evolution and Medicine, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
- The Biodesign Center for Mechanisms of Evolution, Arizona State University, PO Box 874501, Tempe, AZ 85282, USA
| |
Collapse
|
28
|
Placental sex-dependent spermine synthesis regulates trophoblast gene expression through acetyl-coA metabolism and histone acetylation. Commun Biol 2022; 5:586. [PMID: 35705689 PMCID: PMC9200719 DOI: 10.1038/s42003-022-03530-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 05/24/2022] [Indexed: 11/08/2022] Open
Abstract
Placental function and dysfunction differ by sex but the mechanisms are unknown. Here we show that sex differences in polyamine metabolism are associated with escape from X chromosome inactivation of the gene encoding spermine synthase (SMS). Female placental trophoblasts demonstrate biallelic SMS expression, associated with increased SMS mRNA and enzyme activity. Polyamine depletion in primary trophoblasts reduced glycolysis and oxidative phosphorylation resulting in decreased acetyl-coA availability and global histone hypoacetylation in a sex-dependent manner. Chromatin-immunoprecipitation sequencing and RNA-sequencing identifies progesterone biosynthesis as a target of polyamine regulated gene expression, and polyamine depletion reduced progesterone release in male trophoblasts. The effects of polyamine depletion can be attributed to spermine as SMS-silencing recapitulated the effects on energy metabolism, histone acetylation, and progesterone release. In summary, spermine metabolism alters trophoblast gene expression through acetyl-coA biosynthesis and histone acetylation, and SMS escape from X inactivation explains some features of human placental sex differences.
Collapse
|
29
|
Mashak B, Bagheri RB, Noorani G, Soleimanifakhr S, Ataei M. Comparison of Hemodynamic and Biochemical Factors and Pregnancy Complications in Women with/without Preeclampsia. MAEDICA 2022; 17:363-370. [PMID: 36032590 PMCID: PMC9375891 DOI: 10.26574/maedica.2022.17.2.363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Background:Preeclampsia is the second most common cause of maternal mortality in the world after hemorrhage. The present study was conducted to compare the hemodynamic and biochemical levels and pregnancy complications in women with preeclampsia and normal blood pressure. Methods:This cross-sectional descriptive study was conducted on two groups of healthy mothers and mothers with preeclampsia. The research sample included 147 people selected among all mothers referred to Kamali Educational and Medical Center of Alborz. The relationship of preeclampsia and its severity with indices such as age, maternal and fetal weight, body mass index, Apgar score, liver enzymes, laboratory indices, Doppler ultrasound, economic status and other hemodynamic and biochemical indices was examined. Results:The mean age of patients with normal blood pressure and preeclampsia was 29.2 and 29.9 years, respectively. In the control group, no history of hospitalization in an intensive care unit (ICU) was reported, while in the case group, 28% of mothers were admitted to ICU. In the control group, 93% of the fetal middle cerebral arterial (MCA) index, 95% of UA index, 93% of SD index and 95% of CPR index were normal, while in the case group, 67% of MCA index, 65% of the umbilical arterial (UA) index, 70% of SD index and 36% of CPR index were normal. The mean uric acid was about 32% higher in the case group than the control group. The mean neonatal weight was about 20% higher in mothers with normal blood pressure (2836 g in the control group and 2345 g in the case group). In the multivariate logistic regression, platelet (OR=1, P=.018), pulse rate of mother (OR=1.198, P=.044), uric acid (OR=2.057, P<.001) and LDH (OR=1.006, P=.015) were significant predictors of preeclampsia. Conclusion:By examining the indices at different ages of pregnancy, the occurrence of preeclampsia can be predicted at the appropriate time and its complications for both the mother and fetus can be thus prevented. Platelet, pulse rate of the mother, uric acid and LDH were significant predictors of preeclampsia, of which just uric acid was a strong predictor, with odds ratio (OR)=2.057 - for example, for one unit increase in uric acid, the odds of preeclampsia increase by about two times. Preeclampsia may cause low birth weight as well.
Collapse
Affiliation(s)
- Banafsheh Mashak
- Department of Anesthesiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Ramesh Baradaran Bagheri
- Infertility Fellowship, Department of Obstetrics and Gynecology, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Safoora Soleimanifakhr
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mina Ataei
- Infertility Fellowship, Department of Obstetrics and Gynecology, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
30
|
Sandovici I, Fernandez-Twinn DS, Hufnagel A, Constância M, Ozanne SE. Sex differences in the intergenerational inheritance of metabolic traits. Nat Metab 2022; 4:507-523. [PMID: 35637347 DOI: 10.1038/s42255-022-00570-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/05/2022] [Indexed: 02/02/2023]
Abstract
Strong evidence suggests that early-life exposures to suboptimal environmental factors, including those in utero, influence our long-term metabolic health. This has been termed developmental programming. Mounting evidence suggests that the growth and metabolism of male and female fetuses differ. Therefore, sexual dimorphism in response to pre-conception or early-life exposures could contribute to known sex differences in susceptibility to poor metabolic health in adulthood. However, until recently, many studies, especially those in animal models, focused on a single sex, or, often in the case of studies performed during intrauterine development, did not report the sex of the animal at all. In this review, we (a) summarize the evidence that male and females respond differently to a suboptimal pre-conceptional or in utero environment, (b) explore the potential biological mechanisms that underlie these differences and (c) review the consequences of these differences for long-term metabolic health, including that of subsequent generations.
Collapse
Affiliation(s)
- Ionel Sandovici
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonia Hufnagel
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Miguel Constância
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
31
|
Mayrink J, Leite DF, Nobrega GM, Costa ML, Cecatti JG. Prediction of pregnancy-related hypertensive disorders using metabolomics: a systematic review. BMJ Open 2022; 12:e054697. [PMID: 35470187 PMCID: PMC9039389 DOI: 10.1136/bmjopen-2021-054697] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 04/05/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To determine the accuracy of metabolomics in predicting hypertensive disorders in pregnancy. DESIGN Systematic review of observational studies. DATA SOURCES AND STUDY ELIGIBILITY CRITERIA An electronic literature search was performed in June 2019 and February 2022. Two researchers independently selected studies published between 1998 and 2022 on metabolomic techniques applied to predict the condition; subsequently, they extracted data and performed quality assessment. Discrepancies were dealt with a third reviewer. The primary outcome was pre-eclampsia. Cohort or case-control studies were eligible when maternal samples were taken before diagnosis of the hypertensive disorder. STUDY APPRAISAL AND SYNTHESIS METHODS Data on study design, maternal characteristics, how hypertension was diagnosed, metabolomics details and metabolites, and accuracy were independently extracted by two authors. RESULTS Among 4613 initially identified studies on metabolomics, 68 were read in full text and 32 articles were included. Studies were excluded due to duplicated data, study design or lack of identification of metabolites. Metabolomics was applied mainly in the second trimester; the most common technique was liquid-chromatography coupled to mass spectrometry. Among the 122 different metabolites found, there were 23 amino acids and 21 fatty acids. Most of the metabolites were involved with ammonia recycling; amino acid metabolism; arachidonic acid metabolism; lipid transport, metabolism and peroxidation; fatty acid metabolism; cell signalling; galactose metabolism; nucleotide sugars metabolism; lactose degradation; and glycerolipid metabolism. Only citrate was a common metabolite for prediction of early-onset and late-onset pre-eclampsia. Vitamin D was the only metabolite in common for pre-eclampsia and gestational hypertension prediction. Meta-analysis was not performed due to lack of appropriate standardised data. CONCLUSIONS AND IMPLICATIONS Metabolite signatures may contribute to further insights into the pathogenesis of pre-eclampsia and support screening tests. Nevertheless, it is mandatory to validate such methods in larger studies with a heterogeneous population to ascertain the potential for their use in clinical practice. PROSPERO REGISTRATION NUMBER CRD42018097409.
Collapse
Affiliation(s)
- Jussara Mayrink
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Debora F Leite
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Guilherme M Nobrega
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Maria Laura Costa
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Jose Guilherme Cecatti
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| |
Collapse
|
32
|
Sovio U, Goulding N, McBride N, Cook E, Gaccioli F, Charnock-Jones DS, Lawlor DA, Smith GCS. A Maternal Serum Metabolite Ratio Predicts Large for Gestational Age Infants at Term: A Prospective Cohort Study. J Clin Endocrinol Metab 2022; 107:e1588-e1597. [PMID: 34897472 PMCID: PMC8947792 DOI: 10.1210/clinem/dgab842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Indexed: 12/02/2022]
Abstract
CONTEXT Excessive birth weight is associated with maternal and neonatal complications. However, ultrasonically estimated large for gestational age (LGA; >90th percentile) predicts these complications poorly. OBJECTIVE To determine whether a maternal serum metabolite ratio developed for fetal growth restriction (FGR) is predictive of birth weight across the whole range, including LGA at birth. METHODS Metabolites were measured using ultrahigh performance liquid chromatography-tandem mass spectroscopy. The 4-metabolite ratio was previously derived from an analysis of FGR cases and a random subcohort from the Pregnancy Outcome Prediction study. Here, we evaluated its relationship at 36 weeks of gestational age (wkGA) with birth weight in the subcohort (n = 281). External validation in the Born in Bradford (BiB) study (n = 2366) used the metabolite ratio at 24 to 28 wkGA. RESULTS The inverse of the metabolite ratio at 36 wkGA predicted LGA at term [the area under the receiver operating characteristic curve (AUROCC) = 0.82, 95% CI 0.73 to 0.91, P = 6.7 × 10-5]. The ratio was also inversely associated with birth weight z score (linear regression, beta = -0.29 SD, P = 2.1 × 10-8). Analysis in the BiB cohort confirmed that the ratio at 24 to 28 wkGA predicted LGA (AUROCC = 0.60, 95% CI 0.54 to 0.67, P = 8.6 × 10-5) and was inversely associated with birth weight z score (beta = -0.12 SD, P = 1.3 × 10-9). CONCLUSIONS A metabolite ratio which is strongly predictive of FGR is equally predictive of LGA birth weight and is inversely associated with birth weight across the whole range.
Collapse
Affiliation(s)
- Ulla Sovio
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Neil Goulding
- NIHR Bristol Biomedical Research Centre, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Nancy McBride
- NIHR Bristol Biomedical Research Centre, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Emma Cook
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Francesca Gaccioli
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Deborah A Lawlor
- NIHR Bristol Biomedical Research Centre, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
33
|
Aye IL, Aiken CE, Charnock-Jones DS, Smith GC. Placental energy metabolism in health and disease-significance of development and implications for preeclampsia. Am J Obstet Gynecol 2022; 226:S928-S944. [PMID: 33189710 DOI: 10.1016/j.ajog.2020.11.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023]
Abstract
The placenta is a highly metabolically active organ fulfilling the bioenergetic and biosynthetic needs to support its own rapid growth and that of the fetus. Placental metabolic dysfunction is a common occurrence in preeclampsia although its causal relationship to the pathophysiology is unclear. At the outset, this may simply be seen as an "engine out of fuel." However, placental metabolism plays a vital role beyond energy production and is linked to physiological and developmental processes. In this review, we discuss the metabolic basis for placental dysfunction and propose that the alterations in energy metabolism may explain many of the placental phenotypes of preeclampsia such as reduced placental and fetal growth, redox imbalance, oxidative stress, altered epigenetic and gene expression profiles, and the functional consequences of these aberrations. We propose that placental metabolic reprogramming reflects the dynamic physiological state allowing the tissue to adapt to developmental changes and respond to preeclampsia stress, whereas the inability to reprogram placental metabolism may result in severe preeclampsia phenotypes. Finally, we discuss common tested and novel therapeutic strategies for treating placental dysfunction in preeclampsia and their impact on placental energy metabolism as possible explanations into their potential benefits or harm.
Collapse
|
34
|
Christians JK. The Placenta's Role in Sexually Dimorphic Fetal Growth Strategies. Reprod Sci 2021; 29:1895-1907. [PMID: 34699045 DOI: 10.1007/s43032-021-00780-3] [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: 05/28/2021] [Accepted: 10/19/2021] [Indexed: 12/27/2022]
Abstract
Fetal sex affects the risk of pregnancy complications and the long-term effects of prenatal environment on health. Some have hypothesized that growth strategies differ between the sexes, whereby males prioritize growth whereas females are more responsive to their environment. This review evaluates the role of the placenta in such strategies, focusing on (1) mechanisms underlying sexual dimorphism in gene expression, (2) the nature and extent of sexual dimorphism in placental gene expression, (3) sexually dimorphic responses to nutrient supply, and (4) sexual dimorphism in morphology and histopathology. The sex chromosomes contribute to sex differences in placental gene expression, and fetal hormones may play a role later in development. Sexually dimorphic placental gene expression may contribute to differences in the prevalence of complications such as preeclampsia, although this link is not clear. Placental responses to nutrient supply frequently show sexual dimorphism, but there is no consistent pattern where one sex is more responsive. There are sex differences in the prevalence of placental histopathologies, and placental changes in pregnancy complications, but also many similarities. Overall, no clear patterns support the hypothesis that females are more responsive to the maternal environment, or that males prioritize growth. While male fetuses are at greater risk of a variety of complications, total prenatal mortality is higher in females, such that males exposed to early insults may be more likely to survive and be observed in studies of adverse outcomes. Going forward, robust statistical approaches to test for sex-dependent effects must be more widely adopted to reduce the incidence of spurious results.
Collapse
Affiliation(s)
- Julian K Christians
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada. .,Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada. .,British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada. .,Women's Health Research Institute, BC Women's Hospital and Health Centre, Vancouver, BC, Canada.
| |
Collapse
|
35
|
Bucher M, Kadam L, Ahuna K, Myatt L. Differences in Glycolysis and Mitochondrial Respiration between Cytotrophoblast and Syncytiotrophoblast In-Vitro: Evidence for Sexual Dimorphism. Int J Mol Sci 2021; 22:ijms221910875. [PMID: 34639216 PMCID: PMC8509198 DOI: 10.3390/ijms221910875] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 01/18/2023] Open
Abstract
In the placenta the proliferative cytotrophoblast cells fuse into the terminally differentiated syncytiotrophoblast layer which undertakes several energy-intensive functions including nutrient uptake and transfer and hormone synthesis. We used Seahorse glycolytic and mitochondrial stress tests on trophoblast cells isolated at term from women of healthy weight to evaluate if cytotrophoblast (CT) and syncytiotrophoblast (ST) have different bioenergetic strategies, given their different functions. Whereas there are no differences in basal glycolysis, CT have significantly greater glycolytic capacity and reserve than ST. In contrast, ST have significantly higher basal, ATP-coupled and maximal mitochondrial respiration and spare capacity than CT. Consequently, under stress conditions CT can increase energy generation via its higher glycolytic capacity whereas ST can use its higher and more efficient mitochondrial respiration capacity. We have previously shown that with adverse in utero conditions of diabetes and obesity trophoblast respiration is sexually dimorphic. We found no differences in glycolytic parameters between sexes and no difference in mitochondrial respiration parameters other than increases seen upon syncytialization appear to be greater in females. There were differences in metabolic flexibility, i.e., the ability to use glucose, glutamine, or fatty acids, seen upon syncytialization between the sexes with increased flexibility in female trophoblast suggesting a better ability to adapt to changes in nutrient supply.
Collapse
|
36
|
Longitudinal Plasma Metabolomics Profile in Pregnancy-A Study in an Ethnically Diverse U.S. Pregnancy Cohort. Nutrients 2021; 13:nu13093080. [PMID: 34578958 PMCID: PMC8471130 DOI: 10.3390/nu13093080] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/30/2022] Open
Abstract
Amino acids, fatty acids, and acylcarnitine metabolites play a pivotal role in maternal and fetal health, but profiles of these metabolites over pregnancy are not completely established. We described longitudinal trajectories of targeted amino acids, fatty acids, and acylcarnitines in pregnancy. We quantified 102 metabolites and combinations (37 fatty acids, 37 amino acids, and 28 acylcarnitines) in plasma samples from pregnant women in the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies—Singletons cohort (n = 214 women at 10–14 and 15–26 weeks, 107 at 26–31 weeks, and 103 at 33–39 weeks). We used linear mixed models to estimate metabolite trajectories and examined variation by body mass index (BMI), race/ethnicity, and fetal sex. After excluding largely undetected metabolites, we analyzed 77 metabolites and combinations. Levels of 13 of 15 acylcarnitines, 7 of 25 amino acids, and 18 of 37 fatty acids significantly declined over gestation, while 8 of 25 amino acids and 10 of 37 fatty acids significantly increased. Several trajectories appeared to differ by BMI, race/ethnicity, and fetal sex although no tests for interactions remained significant after multiple testing correction. Future studies merit longitudinal measurements to capture metabolite changes in pregnancy, and larger samples to examine modifying effects of maternal and fetal characteristics.
Collapse
|
37
|
McBride N, Yousefi P, Sovio U, Taylor K, Vafai Y, Yang T, Hou B, Suderman M, Relton C, Smith GCS, Lawlor DA. Do Mass Spectrometry-Derived Metabolomics Improve the Prediction of Pregnancy-Related Disorders? Findings from a UK Birth Cohort with Independent Validation. Metabolites 2021; 11:530. [PMID: 34436471 PMCID: PMC8399752 DOI: 10.3390/metabo11080530] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 12/01/2022] Open
Abstract
Many women who experience gestational diabetes (GDM), gestational hypertension (GHT), pre-eclampsia (PE), have a spontaneous preterm birth (sPTB) or have an offspring born small/large for gestational age (SGA/LGA) do not meet the criteria for high-risk pregnancies based upon certain maternal risk factors. Tools that better predict these outcomes are needed to tailor antenatal care to risk. Recent studies have suggested that metabolomics may improve the prediction of these pregnancy-related disorders. These have largely been based on targeted platforms or focused on a single pregnancy outcome. The aim of this study was to assess the predictive ability of an untargeted platform of over 700 metabolites to predict the above pregnancy-related disorders in two cohorts. We used data collected from women in the Born in Bradford study (BiB; two sub-samples, n = 2000 and n = 1000) and the Pregnancy Outcome Prediction study (POPs; n = 827) to train, test and validate prediction models for GDM, PE, GHT, SGA, LGA and sPTB. We compared the predictive performance of three models: (1) risk factors (maternal age, pregnancy smoking, BMI, ethnicity and parity) (2) mass spectrometry (MS)-derived metabolites (n = 718 quantified metabolites, collected at 26-28 weeks' gestation) and (3) combined risk factors and metabolites. We used BiB for the training and testing of the models and POPs for independent validation. In both cohorts, discrimination for GDM, PE, LGA and SGA improved with the addition of metabolites to the risk factor model. The models' area under the curve (AUC) were similar for both cohorts, with good discrimination for GDM (AUC (95% CI) BiB 0.76 (0.71, 0.81) and POPs 0.76 (0.72, 0.81)) and LGA (BiB 0.86 (0.80, 0.91) and POPs 0.76 (0.60, 0.92)). Discrimination was improved for the combined models (compared to the risk factors models) for PE and SGA, with modest discrimination in both studies (PE-BiB 0.68 (0.58, 0.78) and POPs 0.66 (0.60, 0.71); SGA-BiB 0.68 (0.63, 0.74) and POPs 0.64 (0.59, 0.69)). Prediction for sPTB was poor in BiB and POPs for all models. In BiB, calibration for the combined models was good for GDM, LGA and SGA. Retained predictors include 4-hydroxyglutamate for GDM, LGA and PE and glycerol for GDM and PE. MS-derived metabolomics combined with maternal risk factors improves the prediction of GDM, PE, LGA and SGA, with good discrimination for GDM and LGA. Validation across two very different cohorts supports further investigation on whether the metabolites reflect novel causal paths to GDM and LGA.
Collapse
Affiliation(s)
- Nancy McBride
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK; (P.Y.); (K.T.); (M.S.); (C.R.); (D.A.L.)
- NIHR Bristol Biomedical Research Centre, University of Bristol, Bristol BS8 2BN, UK
- Department of Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Paul Yousefi
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK; (P.Y.); (K.T.); (M.S.); (C.R.); (D.A.L.)
- Department of Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Ulla Sovio
- NIHR Cambridge Biomedical Research Centre, Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge CB2 0QQ, UK; (U.S.); (G.C.S.S.)
| | - Kurt Taylor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK; (P.Y.); (K.T.); (M.S.); (C.R.); (D.A.L.)
| | - Yassaman Vafai
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford BD9 6DA, UK; (Y.V.); (T.Y.); (B.H.)
| | - Tiffany Yang
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford BD9 6DA, UK; (Y.V.); (T.Y.); (B.H.)
| | - Bo Hou
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford BD9 6DA, UK; (Y.V.); (T.Y.); (B.H.)
| | - Matthew Suderman
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK; (P.Y.); (K.T.); (M.S.); (C.R.); (D.A.L.)
- Department of Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Caroline Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK; (P.Y.); (K.T.); (M.S.); (C.R.); (D.A.L.)
- Department of Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Gordon C. S. Smith
- NIHR Cambridge Biomedical Research Centre, Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge CB2 0QQ, UK; (U.S.); (G.C.S.S.)
| | - Deborah A. Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK; (P.Y.); (K.T.); (M.S.); (C.R.); (D.A.L.)
- NIHR Bristol Biomedical Research Centre, University of Bristol, Bristol BS8 2BN, UK
- Department of Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| |
Collapse
|
38
|
Tsompanidis A, Aydin E, Padaigaitė E, Richards G, Allison C, Hackett G, Austin T, Holt R, Baron-Cohen S. Maternal steroid levels and the autistic traits of the mother and infant. Mol Autism 2021; 12:51. [PMID: 34238355 PMCID: PMC8268382 DOI: 10.1186/s13229-021-00453-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prenatal sex steroids have been associated with autism in several clinical and epidemiological studies. It is unclear how this relates to the autistic traits of the mother and how early this can be detected during pregnancy and postnatal development. METHODS Maternal serum was collected from pregnant women (n = 122) before or during their first ultrasound appointment [mean = 12.7 (SD = 0.7) weeks]. Concentrations of the following were measured via immunoassays: testosterone, estradiol, dehydroepiandrosterone sulphate, progesterone; and sex hormone-binding globulin which was used to compute the free fractions of estradiol (FEI) and testosterone (FTI). Standardised human choriogonadotropin (hCG) and pregnancy-associated plasma protein A (PAPP-A) values were obtained from clinical records corresponding to the same serum samples. Mothers completed the Autism Spectrum Quotient (AQ) and for their infants, the Quantitative Checklist for Autism in Toddlers (Q-CHAT) when the infants were between 18 and 20 months old. RESULTS FEI was positively associated with maternal autistic traits in univariate (n = 108, Pearson's r = 0.22, p = 0.019) and multiple regression models (semipartial r = 0.19, p = 0.048) controlling for maternal age and a diagnosis of PCOS. Maternal estradiol levels significantly interacted with fetal sex in predicting infant Q-CHAT scores, with a positive relationship in males but not females (n = 100, interaction term: semipartial r = 0.23, p = 0.036) after controlling for maternal AQ and other covariates. The opposite was found for standardised hCG values and Q-CHAT scores, with a positive association in females but not in males (n = 151, interaction term: r = -0.25, p = 0.005). LIMITATIONS Sample size of this cohort was small, with potential ascertainment bias given elective recruitment. Clinical covariates were controlled in multiple regression models, but additional research is needed to confirm the statistically significant findings in larger cohorts. CONCLUSION Maternal steroid factors during pregnancy are associated with autistic traits in mothers and their infants.
Collapse
Affiliation(s)
- A Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.
| | - E Aydin
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - E Padaigaitė
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.,Department of Clinical Psychological Science, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - G Richards
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.,School of Psychology, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - C Allison
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - G Hackett
- The Rosie Hospital, Cambridge University Hospitals Foundation Trust, Cambridge, UK
| | - T Austin
- The Rosie Hospital, Cambridge University Hospitals Foundation Trust, Cambridge, UK
| | - R Holt
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - S Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
39
|
Prater M, Hamilton RS, Wa Yung H, Sharkey AM, Robson P, Abd Hamid NE, Jauniaux E, Charnock-Jones DS, Burton GJ, Cindrova-Davies T. RNA-Seq reveals changes in human placental metabolism, transport and endocrinology across the first-second trimester transition. Biol Open 2021; 10:268993. [PMID: 34100896 PMCID: PMC8214423 DOI: 10.1242/bio.058222] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
The human placenta is exposed to major environmental changes towards the end of the first trimester associated with full onset of the maternal arterial placental circulation. Changes include a switch from histotrophic to hemotrophic nutrition, and a threefold rise in the intraplacental oxygen concentration. We evaluated their impact on trophoblast development and function using RNA-sequencing (RNA-Seq) and DNA-methylation analyses performed on the same chorionic villous samples at 7-8 (n=8) and 13-14 (n=6) weeks of gestation. Reads were adjusted for fetal sex. Most DEGs were associated with protein processing in the endoplasmic reticulum (ER), hormone secretion, transport, extracellular matrix, vasculogenesis, and reactive oxygen species metabolism. Transcripts higher in the first trimester were associated with synthesis and ER processing of peptide hormones, and glycolytic pathways. Transcripts encoding proteins mediating transport of oxygen, lipids, protein, glucose, and ions were significantly increased in the second trimester. The motifs of CBX3 and BCL6 were significantly overrepresented, indicating the involvement of these transcription factor networks in the regulation of trophoblast migration, proliferation and fusion. These findings are consistent with a high level of cell proliferation and hormone secretion by the early placenta to secure implantation in a physiological low-oxygen environment.
Collapse
Affiliation(s)
- Malwina Prater
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Russell S Hamilton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.,Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Hong Wa Yung
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Andrew M Sharkey
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.,Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Paul Robson
- The Jackson Laboratory, The JAX Center for Genetics of Fertility and Reproduction, 10 Discovery Drive, Farmington, CT 06032, USA.,Genome Institute of Singapore, Singapore 138672, Singapore
| | | | - Eric Jauniaux
- Department of Obstetrics and Gynaecology, EGA Institute for Women's Health, Faculty of Population Health Sciences, University College London, London, WC1E 6BT, UK
| | - D Stephen Charnock-Jones
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.,Department of Obstetrics and Gynaecology, University of Cambridge, The Rosie Hospital, Cambridge, CB2 0SW, UK.,National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Tereza Cindrova-Davies
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| |
Collapse
|
40
|
Gong S, Gaccioli F, Dopierala J, Sovio U, Cook E, Volders PJ, Martens L, Kirk PDW, Richardson S, Smith GCS, Charnock-Jones DS. The RNA landscape of the human placenta in health and disease. Nat Commun 2021; 12:2639. [PMID: 33976128 PMCID: PMC8113443 DOI: 10.1038/s41467-021-22695-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
The placenta is the interface between mother and fetus and inadequate function contributes to short and long-term ill-health. The placenta is absent from most large-scale RNA-Seq datasets. We therefore analyze long and small RNAs (~101 and 20 million reads per sample respectively) from 302 human placentas, including 94 cases of preeclampsia (PE) and 56 cases of fetal growth restriction (FGR). The placental transcriptome has the seventh lowest complexity of 50 human tissues: 271 genes account for 50% of all reads. We identify multiple circular RNAs and validate 6 of these by Sanger sequencing across the back-splice junction. Using large-scale mass spectrometry datasets, we find strong evidence of peptides produced by translation of two circular RNAs. We also identify novel piRNAs which are clustered on Chr1 and Chr14. PE and FGR are associated with multiple and overlapping differences in mRNA, lincRNA and circRNA but fewer consistent differences in small RNAs. Of the three protein coding genes differentially expressed in both PE and FGR, one encodes a secreted protein FSTL3 (follistatin-like 3). Elevated serum levels of FSTL3 in pregnant women are predictive of subsequent PE and FGR. To aid visualization of our placenta transcriptome data, we develop a web application ( https://www.obgyn.cam.ac.uk/placentome/ ).
Collapse
Affiliation(s)
- Sungsam Gong
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Francesca Gaccioli
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Justyna Dopierala
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Functional Genomics, GlaxoSmithKline Limited, Stevenage, Hertfordshire, UK
| | - Ulla Sovio
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emma Cook
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Pieter-Jan Volders
- Computational Omics and Systems Biology Group, Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Lennart Martens
- Computational Omics and Systems Biology Group, Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Paul D W Kirk
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Sylvia Richardson
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
41
|
Hebert JF, Myatt L. Placental mitochondrial dysfunction with metabolic diseases: Therapeutic approaches. Biochim Biophys Acta Mol Basis Dis 2021; 1867:165967. [PMID: 32920120 PMCID: PMC8043619 DOI: 10.1016/j.bbadis.2020.165967] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/25/2022]
Abstract
Both obesity and gestational diabetes mellitus (GDM) lead to poor maternal and fetal outcomes, including pregnancy complications, fetal growth issues, stillbirth, and developmental programming of adult-onset disease in the offspring. Increased placental oxidative/nitrative stress and reduced placental (trophoblast) mitochondrial respiration occur in association with the altered maternal metabolic milieu of obesity and GDM. The effect is particularly evident when the fetus is male, suggesting a sexually dimorphic influence on the placenta. In addition, obesity and GDM are associated with inflexibility in trophoblast, limiting the ability to switch between usage of glucose, fatty acids, and glutamine as substrates for oxidative phosphorylation, again in a sexually dimorphic manner. Here we review mechanisms underlying placental mitochondrial dysfunction: its relationship to maternal and fetal outcomes and the influence of fetal sex. Prevention of placental oxidative stress and mitochondrial dysfunction may improve pregnancy outcomes. We outline pathways to ameliorate deficient mitochondrial respiration, particularly the benefits and pitfalls of mitochondria-targeted antioxidants.
Collapse
Affiliation(s)
- Jessica F Hebert
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, United States of America
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, United States of America.
| |
Collapse
|
42
|
Examining Sex Differences in the Human Placental Transcriptome During the First Fetal Androgen Peak. Reprod Sci 2020; 28:801-818. [PMID: 33150487 DOI: 10.1007/s43032-020-00355-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/11/2020] [Indexed: 01/10/2023]
Abstract
Sex differences in human placenta exist from early pregnancy to term, however, it is unclear whether these differences are driven solely by sex chromosome complement or are subject to differential sex hormonal regulation. Here, we survey the human chorionic villus (CV) transcriptome for sex-linked signatures from 11 to 16 gestational weeks, corresponding to the first window of increasing testis-derived androgen production in male fetuses. Illumina HiSeq RNA sequencing was performed on Lexogen Quantseq 3' libraries derived from CV biopsies (n = 11 females, n = 12 males). Differential expression (DE) was performed to identify sex-linked transcriptional signatures, followed by chromosome mapping, pathway analysis, predicted protein interaction, and post-hoc linear regressions to identify transcripts that trend over time. We observe 322 transcripts DE between male and female CV from 11 to 16 weeks, with 22 transcripts logFC > 1. Contrary to our predictions, the difference between male and female expression of DE autosomal genes was more pronounced at the earlier gestational ages. In females, we found selective upregulation of extracellular matrix components, along with a number of X-linked genes. In males, DE transcripts centered on chromosome 19, with mitochondrial, immune, and pregnancy maintenance-related transcripts upregulated. Among the highest differentially expressed autosomal genes were CCRL2, LGALS13, and LGALS14, which are known to regulate immune cell interactions. Our results provide insight into sex-linked gene expression in late first and early second trimester developing human placenta and lay the groundwork to understand the mechanistic origins of sex differences in prenatal development.
Collapse
|
43
|
Modugno F, Fu Z, Jordan SJ, Group A, Chang-Claude J, Fortner RT, Goodman MT, Moysich KB, Schildkraut JM, Berchuck A, Bandera EV, Qin B, Sutphen R, McLaughlin JR, Menon U, Ramus SJ, Gayther SA, Gentry-Maharaj A, Karpinskyj C, Pearce CL, Wu AH, Risch HA, Webb PM. Offspring sex and risk of epithelial ovarian cancer: a multinational pooled analysis of 12 case-control studies. Eur J Epidemiol 2020; 35:1025-1042. [PMID: 32959149 PMCID: PMC7981786 DOI: 10.1007/s10654-020-00682-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/26/2020] [Indexed: 12/18/2022]
Abstract
While childbearing protects against risk of epithelial ovarian cancer (EOC), few studies have explored the impact on maternal EOC risk of sex of offspring, which may affect the maternal environment during pregnancy. We performed a pooled analysis among parous participants from 12 case-controls studies comprising 6872 EOC patients and 9101 controls. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using multivariable logistic regression for case-control associations and polytomous logistic regression for histotype-specific associations, all adjusted for potential confounders. In general, no associations were found between offspring sex and EOC risk. However, compared to bearing only female offspring, bearing one or more male offspring was associated with increased risk of mucinous EOC (OR = 1.45; 95% CI = 1.01-2.07), which appeared to be limited to women reporting menarche before age 13 compared to later menarche (OR = 1.71 vs 0.99; P-interaction = 0.02). Bearing increasing numbers of male offspring was associated with greater risks of mucinous tumors (OR = 1.31, 1.84, 2.31, for 1, 2 and 3 or more male offspring, respectively; trend-p = 0.005). Stratifying by hormonally-associated conditions suggested that compared to bearing all female offspring, bearing a male offspring was associated with lower risk of endometrioid cancer among women with a history of adult acne, hirsutism, or polycystic ovary syndrome (OR = 0.49, 95% CI = 0.28-0.83) but with higher risk among women without any of those conditions (OR = 1.64 95% CI = 1.14-2.34; P-interaction = 0.003). Offspring sex influences the childbearing-EOC risk relationship for specific histotypes and conditions. These findings support the differing etiologic origins of EOC histotypes and highlight the importance of EOC histotype-specific epidemiologic studies. These findings also suggest the need to better understand how pregnancy affects EOC risk.
Collapse
Grants
- R03 CA092776 NCI NIH HHS
- 191. 211 and 182 Cancer Council NSW
- PPD/RPCI.07 Ovarian Cancer Research Fund
- K22 CA138563 NCI NIH HHS
- R01 CA080742 NCI NIH HHS
- NIH-K07 CA095666, R01-CA83918, NIH-K22-CA138563, P30CA072720 NCI NIH HHS
- K07 CA080668 NCI NIH HHS
- R01-CA58598, N01-CN-55424 and N01-PC-67001 NIH HHS
- P01CA17054, P30CA14089, R01CA61132, N01PC67010, R03CA113148, R03CA115195, N01CN025403, NIH HHS
- 6613-1415-53 National Health Research and Development Program, Health Canada
- R01 CA076016 NCI NIH HHS
- R03 CA110797 NCI NIH HHS
- R01 CA063682 NCI NIH HHS
- K07 CA095666 NCI NIH HHS
- AOCS Peter MacCallum Cancer Centre
- R01 CA126841 NCI NIH HHS
- R01-CA074850; R01-CA080742 NIH HHS
- K07-CA080668,R01-CA95023, MO1-RR000056 R01-CA126841 NCI NIH HHS
- N01 CN025403 NCI NIH HHS
- N01 PC067010 NCI NIH HHS
- R01 CA106414 NCI NIH HHS
- 00-01389V-20170, 2II0200 California Cancer Research Program
- M01 RR000056 NCRR NIH HHS
- P30 CA072720 NCI NIH HHS
- R01 CA095023 NCI NIH HHS
- 199600, 400413 and 105 400281 National Health & Medical Research Council of Australia
- R21 CA095113 NCI NIH HHS
- R03 CA113148 NCI NIH HHS
- R01 CA058598 NCI NIH HHS
- MC_UU_12023/20 Medical Research Council
- R01 CA074850 NCI NIH HHS
- R01 CA063678 NCI NIH HHS
- MR_UU_12023 MRF
- R01 CA063678 and R01 CA063682 NIH HHS
- 01 GB 9401 German Federal Ministry of Education and Research
- P30 CA014089 NCI NIH HHS
- R01 CA083918 NCI NIH HHS
- R03 CA115195 NCI NIH HHS
- R03 CA115214 NCI NIH HHS
- DAMD17-02-1-0669 DOD Peer Reviewed Cancer Research Program
- R01-CA074850; R01-CA080742); Division of Cancer Epidemiology and Genetics, National Cancer Institute
- DAMD17-01-1- 104 0729 Medical Research and Materiel Command
- P01 CA017054 NCI NIH HHS
- R13 CA110770 NCI NIH HHS
- R01-CA76016 NIH HHS
- R01-CA106414-A2 NIH HHS
- CRTG-00-196-01-CCE American Cancer Society
- DAMD17-98-1-8659 DOD Peer Reviewed Cancer Research Program
Collapse
Affiliation(s)
- Francesmary Modugno
- Womens Cancer Research Center, Magee-Womens Research Institute and Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Hospital of UPMC, Suite 2130, 300 Halket Street, Pittsburgh, PA, 15213, USA.
| | - Zhuxuan Fu
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, 15213, USA
| | - Susan J Jordan
- School of Public Health, University of Queensland, Herston, QLD, Australia
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Aocs Group
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Peter MacCallum Cancer Center, Melbourne, VIC, 3000, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, 2145, Australia
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renée T Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Marc T Goodman
- Cancer Prevention and Genetics Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Kirsten B Moysich
- Division of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Joellen M Schildkraut
- Department of Public Health Sciences, University of Virginia, Box 800717, Charlottesville, VA, 22908, USA
| | - Andrew Berchuck
- Department of Gynecologic Oncology, Duke University Medical Center, DUMC 3079, Durham, NC, 27710, USA
| | - Elisa V Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Bo Qin
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Rebecca Sutphen
- Epidemiology Center, College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - John R McLaughlin
- Public Health Ontario, Samuel Lunenfeld Research Institute, Toronto, ON, M5T3L9, Canada
| | - Usha Menon
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, WC1V 6LJ, UK
| | - Susan J Ramus
- Faculty of Medicine, School of Women's and Children's Health, University of NSW Sydney, Sydney, NSW, 2052, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, NSW, 2052, Australia
| | - Simon A Gayther
- Center for Bioinformatics and Functional Genomics and the Cedars Sinai Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Aleksandra Gentry-Maharaj
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, WC1V 6LJ, UK
| | - Chloe Karpinskyj
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, WC1V 6LJ, UK
| | - Celeste L Pearce
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, 90033, USA
| | - Anna H Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, 06510, USA
| | - Penelope M Webb
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| |
Collapse
|
44
|
Association of an Increased Risk of Pre-eclampsia and Fetal Growth Restriction in Singleton and Twin Pregnancies with Female Fetuses. MATERNAL-FETAL MEDICINE 2020. [DOI: 10.1097/fm9.0000000000000069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
45
|
Cissé YM, Chan JC, Nugent BM, Banducci C, Bale TL. Brain and placental transcriptional responses as a readout of maternal and paternal preconception stress are fetal sex specific. Placenta 2020; 100:164-170. [PMID: 32980048 DOI: 10.1016/j.placenta.2020.06.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Despite a wealth of epidemiological evidence that cumulative parental lifetime stress experiences prior to conception are determinant of offspring developmental trajectories, there is a lack of insight on how these previous stress experiences are stored and communicated intergenerationally. Preconception experiences may impact offspring development through alterations in transcriptional regulation of the placenta, a major determinant of offspring growth and sex-specific developmental outcomes. We evaluated the lasting influence of maternal and paternal preconception stress (PCS) on the mid-gestation placenta and fetal brain, utilizing their transcriptomes as proximate readouts of intergenerational impact. METHODS To assess the combined vs. dominant influence of maternal and paternal preconception environment on sex-specific fetal development, we compared transcriptional outcomes using a breeding scheme of one stressed parent, both stressed parents, or no stressed parents as controls. RESULTS Interestingly, offspring sex affected the directionality of transcriptional changes in response to PCS, where male tissues showed a predominant downregulation, and female tissues showed an upregulation. There was also an intriguing effect of parental sex on placental programming where paternal PCS drove more effects in female placentas, while maternal PCS produced more transcriptional changes in male placentas. However, in the fetal brain, maternal PCS produced overall more changes in gene expression than paternal PCS, supporting the idea that the intrauterine environment may have a larger overall influence on the developing brain than it does on shaping the placenta. DISCUSSION Preconception experiences drive changes in the placental and the fetal brain transcriptome at a critical developmental timepoint. While not determinant, these altered transcriptional states may underlie sex-biased risk or resilience to stressful experiences later in life.
Collapse
Affiliation(s)
- Yasmine M Cissé
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Jennifer C Chan
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Bridget M Nugent
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Caitlin Banducci
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States
| | - Tracy L Bale
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, United States.
| |
Collapse
|
46
|
Independent influences of maternal obesity and fetal sex on maternal cardiovascular adaptation to pregnancy: a prospective cohort study. Int J Obes (Lond) 2020; 44:2246-2255. [PMID: 32541920 PMCID: PMC7577853 DOI: 10.1038/s41366-020-0627-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/24/2020] [Accepted: 06/01/2020] [Indexed: 11/21/2022]
Abstract
Background/Objectives Successful pregnancy requires the de novo creation of low-resistance utero-placental and feto-placental circulations and incomplete remodeling of this vasculature can lead to maternal or fetal compromise. Maternal BMI and fetal sex are known to influence vascular compliance and placental development, but it is unknown if these are independent or synergistic effects. Here we aim to investigate the impact of maternal obesity, fetal sex, and any interaction thereof on maternal cardiovascular adaptation to pregnancy, by assessing the physiological drop of uterine artery doppler pulsatility (UtA-PI) and umbilical artery doppler pulsatility index (UA-PI) over gestation. Subjects/Methods Nulliparous women with a singleton pregnancy participating in a prospective cohort study (n = 4212) underwent serial UtA-PI and UA-PI measurements at 20-, 28- and 36-weeks gestation. Linear mixed regression models were employed to investigate the influence of maternal BMI, fetal sex and interactions thereof on the magnitude of change in UtA-PI and UA-PI. Results Throughout gestation, UtA-PI was higher for male fetuses and UA-PI was higher for female fetuses. The physiological drop of UtA-PI was significantly smaller in overweight (change −24.3% [95%CI −22.3, −26.2]) and obese women (change −21.3% [−18.3, −24.3]), compared to normal-weight women (change −25.7% [−24.3, −27.0]) but did not differ by fetal sex. The physiological drop in UA-PI was greater for female than male fetuses (–32.5% [−31.5, −33.5] vs. −30.7% [−29.8, −31.7]) but did not differ by maternal BMI. No interactions between maternal BMI and fetal sex were found. Conclusions Maternal cardiovascular adaptation to pregnancy is independently associated with maternal BMI and fetal sex. Our results imply sexual dimorphism in both maternal cardiovascular adaptation and feto-placental resistance.
Collapse
|
47
|
A maternal serum metabolite ratio predicts fetal growth restriction at term. Nat Med 2020; 26:348-353. [PMID: 32161413 DOI: 10.1038/s41591-020-0804-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 01/30/2020] [Indexed: 11/08/2022]
Abstract
Fetal growth restriction (FGR) is the major single cause of stillbirth1 and is also associated with neonatal morbidity and mortality2,3, impaired health and educational achievement in childhood4,5 and with a range of diseases in later life6. Effective screening and intervention for FGR is an unmet clinical need. Here, we performed ultrahigh performance liquid chromatography-tandem mass spectroscopy (UPLC-MS/MS) metabolomics on maternal serum at 12, 20 and 28 weeks of gestational age (wkGA) using 175 cases of term FGR and 299 controls from the Pregnancy Outcome Prediction (POP) study, conducted in Cambridge, UK, to identify predictive metabolites. Internal validation using 36 wkGA samples demonstrated that a ratio of the products of the relative concentrations of two positively associated metabolites (1-(1-enyl-stearoyl)-2-oleoyl-GPC (P-18:0/18:1) and 1,5-anhydroglucitol) to the product of the relative concentrations of two negatively associated metabolites (5α-androstan-3α,17α-diol disulfate and N1,N12-diacetylspermine) predicted FGR at term. The ratio had approximately double the discrimination as compared to a previously developed angiogenic biomarker7, the soluble fms-like tyrosine kinase 1:placental growth factor (sFLT1:PlGF) ratio (AUC 0.78 versus 0.64, P = 0.0001). We validated the predictive performance of the metabolite ratio in two sub-samples of a demographically dissimilar cohort, Born in Bradford (BiB), conducted in Bradford, UK (P = 0.0002). Screening and intervention using this metabolite ratio in conjunction with ultrasonic imaging at around 36 wkGA could plausibly prevent adverse events through enhanced fetal monitoring and targeted induction of labor.
Collapse
|
48
|
Chassen S, Jansson T. Complex, coordinated and highly regulated changes in placental signaling and nutrient transport capacity in IUGR. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165373. [PMID: 30684642 PMCID: PMC6650384 DOI: 10.1016/j.bbadis.2018.12.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023]
Abstract
The most common cause of intrauterine growth restriction (IUGR) in the developed world is placental insufficiency, a concept often used synonymously with reduced utero-placental and umbilical blood flows. However, placental insufficiency and IUGR are associated with complex, coordinated and highly regulated changes in placental signaling and nutrient transport including inhibition of insulin and mTOR signaling and down-regulation of specific amino acid transporters, Na+/K+-ATPase, the Na+/H+-exchanger, folate and lactate transporters. In contrast, placental glucose transport capacity is unaltered and Ca2+-ATPase activity and the expression of proteins involved in placental lipid transport are increased in IUGR. These findings are not entirely consistent with the traditional view that the placenta is dysfunctional in IUGR, but rather suggest that the placenta adapts to reduce fetal growth in response to an inability of the mother to allocate resources to the fetus. This new model has implications for the understanding of the mechanisms underpinning IUGR and for the development of intervention strategies.
Collapse
Affiliation(s)
- Stephanie Chassen
- Department of Pediatrics, Division of Neonatology, University of Colorado, Anschutz Medical Campus, Aurora, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, USA.
| |
Collapse
|
49
|
Furse S, Fernandez-Twinn DS, Jenkins B, Meek CL, Williams HEL, Smith GCS, Charnock-Jones DS, Ozanne SE, Koulman A. A high-throughput platform for detailed lipidomic analysis of a range of mouse and human tissues. Anal Bioanal Chem 2020; 412:2851-2862. [PMID: 32144454 PMCID: PMC7196091 DOI: 10.1007/s00216-020-02511-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/31/2020] [Accepted: 02/12/2020] [Indexed: 02/02/2023]
Abstract
Lipidomics is of increasing interest in studies of biological systems. However, high-throughput data collection and processing remains non-trivial, making assessment of phenotypes difficult. We describe a platform for surveying the lipid fraction for a range of tissues. These techniques are demonstrated on a set of seven different tissues (serum, brain, heart, kidney, adipose, liver, and vastus lateralis muscle) from post-weaning mouse dams that were either obese (> 12 g fat mass) or lean (<5 g fat mass). This showed that the lipid metabolism in some tissues is affected more by obesity than others. Analysis of human serum (healthy non-pregnant women and pregnant women at 28 weeks' gestation) showed that the abundance of several phospholipids differed between groups. Human placenta from mothers with high and low BMI showed that lean placentae contain less polyunsaturated lipid. This platform offers a way to map lipid metabolism with immediate application in metabolic research and elsewhere. Graphical abstract.
Collapse
Affiliation(s)
- Samuel Furse
- grid.5335.00000000121885934Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ UK ,grid.5335.00000000121885934Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge,, Box 289, Cambridge Biomedical Campus, Cambridge, CB2 0QQ UK
| | - Denise S. Fernandez-Twinn
- grid.5335.00000000121885934Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ UK
| | - Benjamin Jenkins
- grid.5335.00000000121885934Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ UK ,grid.5335.00000000121885934Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge,, Box 289, Cambridge Biomedical Campus, Cambridge, CB2 0QQ UK
| | - Claire L. Meek
- grid.5335.00000000121885934Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ UK ,grid.24029.3d0000 0004 0383 8386Department of Clinical Biochemistry/Wolfson Diabetes & Endocrine Clinic, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ UK
| | - Huw E. L. Williams
- grid.4563.40000 0004 1936 8868Centre for Biomolecular Sciences, School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD UK
| | - Gordon C. S. Smith
- grid.5335.00000000121885934Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, CB2 0SW UK ,grid.5335.00000000121885934Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG UK
| | - D. Stephen Charnock-Jones
- grid.5335.00000000121885934Department of Obstetrics and Gynaecology, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, CB2 0SW UK ,grid.5335.00000000121885934Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG UK
| | - Susan E. Ozanne
- grid.5335.00000000121885934Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ UK
| | - Albert Koulman
- grid.5335.00000000121885934Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ UK ,grid.5335.00000000121885934Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge,, Box 289, Cambridge Biomedical Campus, Cambridge, CB2 0QQ UK
| |
Collapse
|
50
|
Abstract
The placenta is essential for normal in utero development in mammals. In humans, defective placental formation underpins common pregnancy disorders such as pre-eclampsia and fetal growth restriction. The great variation in placental types across mammals means that animal models have been of limited use in understanding human placental development. However, new tools for studying human placental development, including 3D organoids, stem cell culture systems and single cell RNA sequencing, have brought new insights into this field. Here, we review the morphological, molecular and functional aspects of human placental formation, with a focus on the defining cell of the placenta - the trophoblast.
Collapse
Affiliation(s)
- Margherita Y Turco
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ashley Moffett
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| |
Collapse
|